1
|
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
|
2
|
Tang Y, Yang C, Wang C, Wu Y, Xu Z, Ni X. Impaired declarative memory consolidation in children with REM sleep-related obstructive sleep apnea. J Clin Sleep Med 2024; 20:417-425. [PMID: 37889162 PMCID: PMC11019210 DOI: 10.5664/jcsm.10892] [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: 11/05/2022] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 10/28/2023]
Abstract
STUDY OBJECTIVES We explored whether declarative memory consolidation is impaired in children with rapid eye movement sleep-related obstructive sleep apnea (REM-OSA) and investigated the correlation between memory consolidation and sleep-related respiratory parameters. METHODS Participants were children with habitual snoring aged 6-14 years and control children. Participants underwent polysomnography and declarative memory testing. Participants with snoring were categorized as primary snoring (PS), non-rapid eye movement sleep-related obstructive sleep apnea (NREM-OSA), stage-independent (SI)-OSA, and REM-OSA according to obstructive apnea-hypopnea index (OAHI), OAHI in REM sleep (OAHIREM), and OAHI in NREM sleep (OAHINREM). Declarative memory consolidation level was assessed by recall and recognition rates. RESULTS There were 34 controls and 228 children with sleep-disordered breathing: 73 PS, 48 NREM-OSA, 59 SI-OSA, and 48 REM-OSA. Total arousal index was lower in the REM-OSA group than in the NREM-OSA group. In all groups, retest scores were higher than immediate test scores. Recall consolidation in PS, SI-OSA, and REM-OSA groups was lower than for controls and lower in REM-OSA than in NREM-OSA. There were no correlations between recall consolidation or recognition consolidation and OAHI, OAHINREM, oxygen desaturation index in REM sleep, total arousal index, or REM sleep percent. Recognition consolidation was negatively correlated with OAHIREM. CONCLUSIONS Memory consolidation is impaired in children with REM-OSA compared with NREM-OSA and controls. There was no significant correlation between memory consolidation and OAHI, and recognition consolidation was negatively correlated with OAHIREM. It is important to pay attention to the OSA subtype in children. CITATION Tang Y, Yang C, Wang C, Wu Y, Xu Z, Ni X. Impaired declarative memory consolidation in children with REM sleep-related obstructive sleep apnea. J Clin Sleep Med. 2024;20(3):417-425.
Collapse
Affiliation(s)
- Yufen Tang
- Department of Respiratory Medicine, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Chao Yang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern, Beijing Normal University, Beijing, China
| | - Changming Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yunxiao Wu
- Beijing Key Laboratory of Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Zhifei Xu
- Department of Respiratory Medicine, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xin Ni
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| |
Collapse
|
3
|
Titone MK, Hunt C, Bismark A, Nokes B, Lee E, Ramanathan D, Park J, Colvonen P. The effect of obstructive sleep apnea severity on PTSD symptoms during the course of esketamine treatment: a retrospective clinical study. J Clin Sleep Med 2023; 19:2043-2051. [PMID: 37539643 PMCID: PMC10692930 DOI: 10.5664/jcsm.10746] [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: 03/25/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023]
Abstract
STUDY OBJECTIVES Intranasal administration of esketamine is Food and Drug Administration-approved for treatment-resistant depression. In a recent retrospective case series, we show that it has promise in reducing symptoms of posttraumatic stress disorder (PTSD) as well. Untreated obstructive sleep apnea (OSA) is prevalent among veterans with PTSD and has been shown to interfere with other PTSD treatments. In the current study, we examined whether OSA impacts esketamine's effectiveness in reducing symptoms of PTSD or depression. METHODS Participants were 60 veterans with a diagnosis of major depressive disorder and PTSD who received intranasal esketamine treatment at the San Diego Veterans Affairs (VA) Medical Center. We used growth-curve modeling to examine changes in depression and PTSD symptoms following esketamine treatments and, in the subset of individuals screened for OSA (n = 24, all prescribed positive airway pressure therapy), examined the impacts of OSA severity on these trajectories. RESULTS We first showed that both PTSD and depressive symptoms significantly decreased over the course of esketamine treatment. In the subset of veterans screened for OSA, individuals with lower OSA severity reported the greatest reduction in PTSD symptoms, while veterans with the most severe OSA reported the least reduction in PTSD symptoms. Depression response was not affected by severity of OSA in this analysis. CONCLUSIONS Veterans with PTSD and depression tend to benefit from esketamine treatment, but OSA may interfere with esketamine effectiveness. Comorbid OSA should be assessed for and treated to maximize esketamine's benefits in PTSD. CITATION Titone MK, Hunt C, Bismark A, et al. The effect of obstructive sleep apnea severity on PTSD symptoms during the course of esketamine treatment: a retrospective clinical study. J Clin Sleep Med. 2023;19(12):2043-2051.
Collapse
Affiliation(s)
- Madison K. Titone
- VA San Diego Healthcare System, San Diego, California
- Department of Psychiatry, University of California, San Diego, San Diego, California
| | | | | | - Brandon Nokes
- VA San Diego Healthcare System, San Diego, California
| | - Ellen Lee
- VA San Diego Healthcare System, San Diego, California
- Department of Psychiatry, University of California, San Diego, San Diego, California
| | - Dhakshin Ramanathan
- VA San Diego Healthcare System, San Diego, California
- Department of Psychiatry, University of California, San Diego, San Diego, California
| | - Jane Park
- VA San Diego Healthcare System, San Diego, California
| | - Peter Colvonen
- VA San Diego Healthcare System, San Diego, California
- Department of Psychiatry, University of California, San Diego, San Diego, California
| |
Collapse
|
4
|
Badran M, Puech C, Barrow MB, Runion AR, Gozal D. Solriamfetol enhances wakefulness and improves cognition and anxiety in a murine model of OSA. Sleep Med 2023; 107:89-99. [PMID: 37137196 PMCID: PMC11556240 DOI: 10.1016/j.sleep.2023.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/31/2023] [Accepted: 04/09/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a chronic condition characterized by intermittent hypoxia (IH). Excessive daytime sleepiness (EDS) is a common consequence of OSA and is associated with cognitive deficits and anxiety. Modafinil (MOD) and Solriamfetol (SOL) are potent wake-promoting agents clinically used to improve wakefulness in OSA patients with EDS. METHODS Male C57Bl/6J mice were exposed to either IH or room air (RA) controls during the light phase for 16 weeks. Both groups were then randomly assigned to receive once-daily intraperitoneal injections of SOL (200 mg/kg), MOD (200 mg/kg) or vehicle (VEH) for 9 days while continuing IH exposures. Sleep/wake activity was assessed during the dark (active) phase. Novel object recognition (NOR), elevated-plus maze test (EPMT), and forced swim test (FST) were performed before and after drug treatment. RESULTS IH exposure increased dark phase sleep percentage and reduced wake bouts lengths and induced cognitive deficits and anxiogenic effects. Both SOL and MOD treatments decreased sleep propensity under IH conditions, but only SOL promoted improvements in NOR performance (explicit memory) and reduced anxiety-like behaviors. CONCLUSION Chronic IH, a hallmark feature of OSA, induces EDS in young adult mice that is ameliorated by both SOL and MOD. SOL, but not MOD, significantly improves IH-induced cognitive deficits and promotes anxiolytic effects. Thus, SOL could potentially benefit OSA patients beyond EDS management.
Collapse
Affiliation(s)
- Mohammad Badran
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Clementine Puech
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Max B Barrow
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - Alexandra R Runion
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - David Gozal
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA; Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
5
|
Puech C, Badran M, Runion AR, Barrow MB, Cataldo K, Gozal D. Cognitive Impairments, Neuroinflammation and Blood-Brain Barrier Permeability in Mice Exposed to Chronic Sleep Fragmentation during the Daylight Period. Int J Mol Sci 2023; 24:9880. [PMID: 37373028 DOI: 10.3390/ijms24129880] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic condition characterized by intermittent hypoxia (IH) and sleep fragmentation (SF). In murine models, chronic SF can impair endothelial function and induce cognitive declines. These deficits are likely mediated, at least in part, by alterations in Blood-brain barrier (BBB) integrity. Male C57Bl/6J mice were randomly assigned to SF or sleep control (SC) conditions for 4 or 9 weeks and in a subset 2 or 6 weeks of normal sleep recovery. The presence of inflammation and microglia activation were evaluated. Explicit memory function was assessed with the novel object recognition (NOR) test, while BBB permeability was determined by systemic dextran-4kDA-FITC injection and Claudin 5 expression. SF exposures resulted in decreased NOR performance and in increased inflammatory markers and microglial activation, as well as enhanced BBB permeability. Explicit memory and BBB permeability were significantly associated. BBB permeability remained elevated after 2 weeks of sleep recovery (p < 0.01) and returned to baseline values only after 6 weeks. Chronic SF exposures mimicking the fragmentation of sleep that characterizes patients with OSA elicits evidence of inflammation in brain regions and explicit memory impairments in mice. Similarly, SF is also associated with increased BBB permeability, the magnitude of which is closely associated with cognitive functional losses. Despite the normalization of sleep patterns, BBB functional recovery is a protracted process that merits further investigation.
Collapse
Affiliation(s)
- Clementine Puech
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
| | - Mohammad Badran
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
| | - Alexandra R Runion
- Undergraduate Student Research Program, University of Missouri, Columbia, MO 65201, USA
| | - Max B Barrow
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
| | - Kylie Cataldo
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
| | - David Gozal
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, 400 N Keene St., Suite 010, Columbia, MO 65201, USA
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65201, USA
| |
Collapse
|
6
|
Puech C, Badran M, Barrow MB, Runion AR, Gozal D. Solriamfetol improves chronic sleep fragmentation-induced increases in sleep propensity and ameliorates explicit memory in male mice. Sleep 2023; 46:zsad057. [PMID: 36866452 PMCID: PMC10413435 DOI: 10.1093/sleep/zsad057] [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: 10/24/2022] [Revised: 02/15/2023] [Indexed: 03/04/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a highly prevalent condition characterized by episodes of partial or complete breath cessation during sleep that induces sleep fragmentation (SF). One of the frequent manifestations of OSA is the presence of excessive daytime sleepiness (EDS) associated with cognitive deficits. Solriamfetol (SOL) and modafinil (MOD) are wake-promoting agents commonly prescribed to improve wakefulness in OSA patients with EDS. This study aimed to assess the effects of SOL and MOD in a murine model of OSA characterized by periodic SF. Male C57Bl/6J mice were exposed to either control sleep (SC) or SF (mimicking OSA) during the light period (06:00 h to 18:00 h) for 4 weeks, which consistently induces sustained excessive sleepiness during the dark phase. Both groups were then randomly assigned to receive once-daily intraperitoneal injections of SOL (200 mg/kg), MOD (200 mg/kg), or vehicle for 1 week while continuing exposures to SF or SC. Sleep/wake activity and sleep propensity were assessed during the dark phase. Novel Object Recognition test, Elevated-Plus Maze Test, and Forced Swim Test were performed before and after treatment. SOL or MOD decreased sleep propensity in SF, but only SOL induced improvements in explicit memory, while MOD exhibited increased anxiety behaviors. Chronic SF, a major hallmark of OSA, induces EDS in young adult mice that is mitigated by both SOL and MOD. SOL, but not MOD, significantly improves SF-induced cognitive deficits. Increased anxiety behaviors are apparent in MOD-treated mice. Further studies aiming to elucidate the beneficial cognitive effects of SOL are warranted.
Collapse
Affiliation(s)
- Clementine Puech
- Child Health Research Institute, Department of Child Health, University of Missouri School of Medicine, Columbia, MO, USA
| | - Mohammad Badran
- Child Health Research Institute, Department of Child Health, University of Missouri School of Medicine, Columbia, MO, USA
| | - Max B Barrow
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - Alexandra R Runion
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - David Gozal
- Child Health Research Institute, Department of Child Health, University of Missouri School of Medicine, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, USA
| |
Collapse
|
7
|
Ma Y, Niu Z, Ruan L, Xue S, Li N, Yao X, Li Q. Alterations in Amygdala/Hippocampal Volume Ratios in Children with Obstructive Sleep Apnea Syndrome Caused by Adenotonsillar Hypertrophy. Med Sci Monit 2023; 29:e937420. [PMID: 36918755 PMCID: PMC10026529 DOI: 10.12659/msm.937420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Intermittent hypoxemia can cause changes in certain brain structures. However, in pediatric patients with obstructive sleep apnea (OSA) caused by adenotonsillar hypertrophy (ATH), there is only limited information on the effect of ATH-induced OSA on brain structures. This study sought to investigate alterations in amygdala and hippocampal volumes in children with OSA by ATH. MATERIAL AND METHODS Magnetic resonance imaging scans were applied in children who had ATH-induced OSA (ATH/OSA) and in healthy children. Amygdala and hippocampus volumes and adenoid sizes were measured on MRI volumetric images. The ratio of adenoid size/nasopharyngeal depth was used to describe the severity of adenoid hypertrophy. The clinical variables of the involved subjects were investigated. RESULTS One hundred ATH/OSA children and 100 healthy children without ATH/OSA participated in the study. The ATH/OSA children had higher amygdala volumes and amygdala/hippocampus volume ratios but lower hippocampus volumes than healthy controls, and the amygdala/hippocampus volume ratios were correlated with disease duration and hypoxemia conditions. However, our data showed that amygdala/hippocampus volume ratios were not correlated with the ratios of adenoid size/nasopharyngeal depth in the ATH/OSA children. In addition, the ratio of adenoid size/nasopharyngeal depths in ATH/OSA children was higher than that in healthy children in each subgroup based on the age of participants. CONCLUSIONS Compared to healthy controls, amygdala/hippocampus volume ratios are increased in children with ATH/OSA.
Collapse
Affiliation(s)
- Yanshan Ma
- Department of Radiology, Shijiazhuang Hospital of Traditional Chinese Medicine, Shijiazhuang, Hebei, China (mainland)
| | - Zheli Niu
- Department of Nephrology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Lin Ruan
- Department of Nephrology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Sisi Xue
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China (mainland)
| | - Nan Li
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China (mainland)
| | - Xiaoguang Yao
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China (mainland)
| | - Qiang Li
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China (mainland)
| |
Collapse
|
8
|
Cunningham TJ, Kishore D, Guo M, Igue M, Malhotra A, Stickgold R, Djonlagic I. The Effect of Obstructive Sleep Apnea on Sleep-dependent Emotional Memory Consolidation. Ann Am Thorac Soc 2023; 20:296-306. [PMID: 36250951 PMCID: PMC9989861 DOI: 10.1513/annalsats.202204-315oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 10/17/2022] [Indexed: 02/04/2023] Open
Abstract
Rationale: A growing body of evidence suggests that sleep is critical for the adaptive processing and consolidation of emotional information into long-term memory. Previous research has indicated that emotional components of scenes particularly benefit from sleep in healthy groups, yet sleep-dependent emotional memory processes remain unexplored in clinical cohorts, including those with obstructive sleep apnea (OSA). This line of research is important as it will add to the understanding of how disrupted sleep in OSA contributes to both impaired cognition and emotion dysregulation. Objectives: To test the hypothesis that individuals with OSA will have impaired sleep-dependent memory consolidation, with the greatest impact being on memory for emotional content. Methods: In this study, a group of newly diagnosed patients with OSA (n = 26; 10 female; average age, 42.5 years) and a matched group of healthy control subjects (n = 24; 13 female; average age, 37 years) were enrolled in the study at Beth Israel Deaconess Medical Center. Participants encoded scenes with negative or neutral foreground objects placed on neutral backgrounds before a night of polysomnographically recorded sleep. In the morning, they completed a recognition test in which old and new scene objects and backgrounds, presented separately and one at a time, were judged as old, new, or similar compared with what had been previously viewed. Results: Patients with OSA had a deficit in recognition memory for the scenes. Overall recognition (the ability to recognize old items as either old or similar) was impaired across all scene elements, both negative and neutral objects and backgrounds, whereas specific recognition (correctly identifying old items as old) was impaired only for negative objects. Across all participants, successful overall recognition correlated positively with sleep efficiency and rapid eye movement (REM) sleep, whereas successful specific memory recognition correlated only with REM sleep. Conclusions: Our findings indicate that fragmented sleep and reduced REM sleep, both hallmarks of OSA, are associated with disruptions in general memory impairment and veridical memory for emotional content, which could alter emotional regulation and contribute to comorbid emotional distress in OSA.
Collapse
Affiliation(s)
- Tony J. Cunningham
- Center for Sleep and Cognition, Department of Psychiatry and
- Department of Psychology and Neuroscience, Boston College, Chestnut Hill, Massachusetts; and
| | - Divya Kishore
- Department of Neurology, Beth Israel Hospital and Harvard Medical School, Boston, Massachusetts
| | - Meng Guo
- Department of Neurology, Beth Israel Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary, Critical Care, Sleep, and Physiology, University of California San Diego, La Jolla, California
| | - Moroké Igue
- Department of Neurology, Beth Israel Hospital and Harvard Medical School, Boston, Massachusetts
| | - Atul Malhotra
- Division of Pulmonary, Critical Care, Sleep, and Physiology, University of California San Diego, La Jolla, California
| | | | - Ina Djonlagic
- Department of Neurology, Beth Israel Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary, Critical Care, Sleep, and Physiology, University of California San Diego, La Jolla, California
| |
Collapse
|
9
|
Puech C, Badran M, Runion AR, Barrow MB, Qiao Z, Khalyfa A, Gozal D. Explicit memory, anxiety and depressive like behavior in mice exposed to chronic intermittent hypoxia, sleep fragmentation, or both during the daylight period. Neurobiol Sleep Circadian Rhythms 2022; 13:100084. [PMID: 36254342 PMCID: PMC9568859 DOI: 10.1016/j.nbscr.2022.100084] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/01/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic and highly prevalent condition characterized by chronic intermittent hypoxia (IH) and sleep fragmentation (SF), and can lead to a vast array of end-organ morbidities, particularly affecting cardiovascular, metabolic and neurobehavioral functioning. OSA can induce cognitive and behavioral and mood deficits. Male C57Bl/6J 8-week-old mice were housed in custom-designed cages with a silent motorized mechanical sweeper traversing the cage floor at 2-min intervals (SF) during daylight for four weeks. Sleep control (SC) consisted of keeping sweeper immobile. IH consisted of cycling FiO2 21% 90 seconds-6.3% 90s or room air (RA; FiO2 21%) for sixteen weeks and combined SF-IH was conducted for nine weeks. Open field novel object recognition (NOR) testing, elevated-plus maze test (EPMT), and forced swimming test (FST) were performed. SF induced cognitive NOR performance impairments in mice along with reduced anxiety behaviors while IH induced deficits in NOR performance, but increased anxiety behaviors. SF-IH induced impaired performance in NOR test of similar magnitude to IH or SF alone. Combined SF-IH exposures did not affect anxiety behaviors. Thus, both SF an IH altered cognitive function while imposing opposite effects on anxiety behaviors. SF-IH did not magnify the detrimental effects of isolated SF or IH and canceled out the effects on anxiety. Based on these findings, the underlying pathophysiologic processes underlying IH and SF adverse effects on cognitive function appear to differ, while those affecting anxiety counteract each other.
Collapse
Affiliation(s)
- Clementine Puech
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mohammad Badran
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Alexandra R Runion
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - Max B Barrow
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - Zhuanhong Qiao
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Abdelnaby Khalyfa
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - David Gozal
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
10
|
Xu L, Li Q, Ke Y, Yung WH. Chronic Intermittent Hypoxia-Induced Aberrant Neural Activities in the Hippocampus of Male Rats Revealed by Long-Term in vivo Recording. Front Cell Neurosci 2022; 15:784045. [PMID: 35126057 PMCID: PMC8813782 DOI: 10.3389/fncel.2021.784045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic intermittent hypoxia (CIH) occurs in obstructive sleep apnea (OSA), a common sleep-disordered breathing associated with malfunctions in multiple organs including the brain. How OSA-associated CIH impacts on brain activities and functions leading to neurocognitive impairment is virtually unknown. Here, by means of in vivo electrophysiological recordings via chronically implanted multi-electrode arrays in male rat model of OSA, we found that both putative pyramidal neurons and putative interneurons in the hippocampal CA1 subfield were hyper-excitable during the first week of CIH treatment and followed by progressive suppression of neural firing in the longer term. Partial recovery of the neuronal activities was found after normoxia treatment but only in putative pyramidal neurons. These findings correlated well to abnormalities in dendritic spine morphogenesis of these neurons. The results reveal that hippocampal neurons respond to CIH in a complex biphasic and bidirectional manner eventually leading to suppression of firing activities. Importantly, these changes are attributed to a larger extent to impaired functions of putative interneurons than putative pyramidal neurons. Our findings therefore revealed functional and structural damages in central neurons in OSA subjects.
Collapse
Affiliation(s)
- Linhao Xu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Wing-Ho Yung
| | - Wing-Ho Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Ya Ke
| |
Collapse
|
11
|
Duncan M, Guerriero L, Kohler K, Beechem L, Gillis B, Salisbury F, Wessel C, Wang J, Sunderam S, Bachstetter A, O’Hara B, Murphy M. Chronic Fragmentation of the Daily Sleep-Wake Rhythm Increases Amyloid-beta Levels and Neuroinflammation in the 3xTg-AD Mouse Model of Alzheimer's Disease. Neuroscience 2022; 481:111-122. [PMID: 34856352 PMCID: PMC8941625 DOI: 10.1016/j.neuroscience.2021.11.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 01/17/2023]
Abstract
Fragmentation of the daily sleep-wake rhythm with increased nighttime awakenings and more daytime naps is correlated with the risk of development of Alzheimer's disease (AD). To explore whether a causal relationship underlies this correlation, the present study tested the hypothesis that chronic fragmentation of the daily sleep-wake rhythm stimulates brain amyloid-beta (Aβ) levels and neuroinflammation in the 3xTg-AD mouse model of AD. Female 3xTg-AD mice were allowed to sleep undisturbed or were subjected to chronic sleep fragmentation consisting of four daily sessions of enforced wakefulness (one hour each) evenly distributed during the light phase, five days a week for four weeks. Piezoelectric sleep recording revealed that sleep fragmentation altered the daily sleep-wake rhythm to resemble the pattern observed in AD. Levels of amyloid-beta (Aβ40 and Aβ42) determined by ELISA were higher in hippocampal tissue collected from sleep-fragmented mice than from undisturbed controls. In contrast, hippocampal levels of tau and phospho-tau differed minimally between sleep fragmented and undisturbed control mice. Sleep fragmentation also stimulated neuroinflammation as shown by increased expression of markers of microglial activation and proinflammatory cytokines measured by q-RT-PCR analysis of hippocampal samples. No significant effects of sleep fragmentation on Aβ, tau, or neuroinflammation were observed in the cerebral cortex. These studies support the concept that improving sleep consolidation in individuals at risk for AD may be beneficial for slowing the onset or progression of this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- M.J. Duncan
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536,Co-senior authors, address correspondence to M.J. Duncan at
| | - L.E. Guerriero
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - K. Kohler
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536
| | - L.E. Beechem
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536
| | - B.D. Gillis
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536
| | - F. Salisbury
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - C. Wessel
- Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536
| | - J. Wang
- Dept. of Biomedical Engineering, University of Kentucky, Lexington, KY 40506
| | - S. Sunderam
- Dept. of Biomedical Engineering, University of Kentucky, Lexington, KY 40506
| | - A.D. Bachstetter
- Dept. of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536
| | - B.F. O’Hara
- Dept. of Biology, University of Kentucky, Lexington, KY 40506
| | - M.P. Murphy
- Dept. of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536,Sanders-Brown Center on Aging and Alzheimer’s Disease Center, University of Kentucky College of Medicine, Lexington, KY 40536,Co-senior authors, address correspondence to M.J. Duncan at
| |
Collapse
|
12
|
Ulland TK, Ewald AC, Knutson AO, Marino KM, Smith SMC, Watters JJ. Alzheimer's Disease, Sleep Disordered Breathing, and Microglia: Puzzling out a Common Link. Cells 2021; 10:2907. [PMID: 34831129 PMCID: PMC8616348 DOI: 10.3390/cells10112907] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/14/2022] Open
Abstract
Sleep Disordered Breathing (SDB) and Alzheimer's Disease (AD) are strongly associated clinically, but it is unknown if they are mechanistically associated. Here, we review data covering both the cellular and molecular responses in SDB and AD with an emphasis on the overlapping neuroimmune responses in both diseases. We extensively discuss the use of animal models of both diseases and their relative utilities in modeling human disease. Data presented here from mice exposed to intermittent hypoxia indicate that microglia become more activated following exposure to hypoxia. This also supports the idea that intermittent hypoxia can activate the neuroimmune system in a manner like that seen in AD. Finally, we highlight similarities in the cellular and neuroimmune responses between SDB and AD and propose that these similarities may lead to a pathological synergy between SDB and AD.
Collapse
Affiliation(s)
- Tyler K. Ulland
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI 53705, USA; (T.K.U.); (K.M.M.)
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI 53705, USA
| | - Andrea C. Ewald
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| | - Andrew O. Knutson
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| | - Kaitlyn M. Marino
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI 53705, USA; (T.K.U.); (K.M.M.)
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI 53705, USA
| | - Stephanie M. C. Smith
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| | - Jyoti J. Watters
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| |
Collapse
|
13
|
Zhao Y, Yang S, Guo Q, Guo Y, Zheng Y, Ji E. Shashen-Maidong Decoction improved chronic intermittent hypoxia-induced cognitive impairment through regulating glutamatergic signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114040. [PMID: 33794336 DOI: 10.1016/j.jep.2021.114040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/03/2021] [Accepted: 03/13/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Obstructive sleep apnea (OSA) is characterized by chronic intermittent hypoxia (CIH), which is associated with cognitive impairment. Previous study suggested CIH exposure could induce similar symptoms and signs to the clinical features of Deficiency of both Qi and Yin Syndrome (DQYS) in Traditional Chinese Medicine (TCM). Shashen-Maidong Decoction (SMD) has been applied clinically for DQYS for hundred years. However, SMD treatment could be beneficial to CIH induced cognitive impairment is still unclear. AIM OF THE STUDY Therefore, the aim of this study was to investigate the effect of SMD treatment on CIH induced cognitive impairment, and to explore the related neuroprotective mechanism. MATERIALS AND METHODS Mice were exposed to CIH for 5 weeks (8 h/day) and were orally treated with either vehicle or SMD (5.265 g/kg/day) 30 min before CIH exposure. Spatial memory was evaluated by Morris Water Maze and Y-Maze test. Synaptic morphology in hippocampus was observed by Golgi-Cox staining and Electron microscope, and NR2B-ERK signaling pathway were detected by western blotting. RESULTS Our results showed that SMD treatment improved performance in either Morris Water Maze or Y-Maze test in mice exposed to CIH, increased spine density and postsynaptic density (PSD) thickness in hippocampus. SMD treatment suppressed the over-activation of NR2B/CaMKII/SynGAP induced by CIH exposure, enhanced ERK/CREB phosphorylation and increased PSD-95 and BDNF expression. CONCLUSION SMD attenuates the CIH-induced cognitive impairment through regulating NR2B-ERK signaling pathway. Additionally, our findings provided that DQYS may be the potential therapeutic target for neurocognitive diseases in patients with OSA.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Shengchang Yang
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China; Hebei Technology Innovation Center of TCM Formula Preparations, Shijiazhuang, Hebei, People's Republic of China
| | - Qiuhong Guo
- Hebei Technology Innovation Center of TCM Formula Preparations, Shijiazhuang, Hebei, People's Republic of China
| | - Yajing Guo
- Scientific Research Center, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Yuying Zheng
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Ensheng Ji
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China; Hebei Technology Innovation Center of TCM Formula Preparations, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
14
|
Yuan RK, Lopez MR, Ramos-Alvarez MM, Normandin ME, Thomas AS, Uygun DS, Cerda VR, Grenier AE, Wood MT, Gagliardi CM, Guajardo H, Muzzio IA. Differential effect of sleep deprivation on place cell representations, sleep architecture, and memory in young and old mice. Cell Rep 2021; 35:109234. [PMID: 34133936 PMCID: PMC8545463 DOI: 10.1016/j.celrep.2021.109234] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 01/05/2023] Open
Abstract
Poor sleep quality is associated with age-related cognitive decline, and whether reversal of these alterations is possible is unknown. In this study, we report how sleep deprivation (SD) affects hippocampal representations, sleep patterns, and memory in young and old mice. After training in a hippocampus-dependent object-place recognition (OPR) task, control animals sleep ad libitum, although experimental animals undergo 5 h of SD, followed by recovery sleep. Young controls and old SD mice exhibit successful OPR memory, whereas young SD and old control mice are impaired. Successful performance is associated with two cellular phenotypes: (1) "context" cells, which remain stable throughout training and testing, and (2) "object configuration" cells, which remap when objects are introduced to the context and during testing. Additionally, effective memory correlates with spindle counts during non-rapid eye movement (NREM)/rapid eye movement (REM) sigma transitions. These results suggest SD may serve to ameliorate age-related memory deficits and allow hippocampal representations to adapt to changing environments.
Collapse
Affiliation(s)
- Robin K Yuan
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Avenue, Boston, MA, USA; Division of Sleep Medicine, Harvard Medical School, 221 Longwood Avenue, Boston, MA, USA
| | - Matthew R Lopez
- University of Texas at San Antonio, Department of Biology, One UTSA Circle, San Antonio, TX 78249, USA
| | | | - Marc E Normandin
- University of Texas at San Antonio, Department of Biology, One UTSA Circle, San Antonio, TX 78249, USA
| | - Arthur S Thomas
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - David S Uygun
- VA Boston Healthcare System and Department of Psychiatry, Harvard Medical School, West Roxbury, MA 02132, USA
| | - Vanessa R Cerda
- University of Texas at San Antonio, Department of Biology, One UTSA Circle, San Antonio, TX 78249, USA
| | - Amandine E Grenier
- University of Texas at San Antonio, Department of Biology, One UTSA Circle, San Antonio, TX 78249, USA
| | - Matthew T Wood
- University of Texas at San Antonio, Department of Biology, One UTSA Circle, San Antonio, TX 78249, USA
| | - Celia M Gagliardi
- University of Texas at San Antonio, Department of Biology, One UTSA Circle, San Antonio, TX 78249, USA
| | - Herminio Guajardo
- University of Texas at San Antonio, Department of Biology, One UTSA Circle, San Antonio, TX 78249, USA
| | - Isabel A Muzzio
- University of Texas at San Antonio, Department of Biology, One UTSA Circle, San Antonio, TX 78249, USA.
| |
Collapse
|
15
|
Glutamatergic Neurons in the Preoptic Hypothalamus Promote Wakefulness, Destabilize NREM Sleep, Suppress REM Sleep, and Regulate Cortical Dynamics. J Neurosci 2021; 41:3462-3478. [PMID: 33664133 PMCID: PMC8051693 DOI: 10.1523/jneurosci.2718-20.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/24/2021] [Accepted: 02/22/2021] [Indexed: 11/21/2022] Open
Abstract
Clinical and experimental data from the last nine decades indicate that the preoptic area of the hypothalamus is a critical node in a brain network that controls sleep onset and homeostasis. By contrast, we recently reported that a group of glutamatergic neurons in the lateral and medial preoptic area increases wakefulness, challenging the long-standing notion in sleep neurobiology that the preoptic area is exclusively somnogenic. Clinical and experimental data from the last nine decades indicate that the preoptic area of the hypothalamus is a critical node in a brain network that controls sleep onset and homeostasis. By contrast, we recently reported that a group of glutamatergic neurons in the lateral and medial preoptic area increases wakefulness, challenging the long-standing notion in sleep neurobiology that the preoptic area is exclusively somnogenic. However, the precise role of these subcortical neurons in the control of behavioral state transitions and cortical dynamics remains unknown. Therefore, in this study, we used conditional expression of excitatory hM3Dq receptors in these preoptic glutamatergic (Vglut2+) neurons and show that their activation initiates wakefulness, decreases non-rapid eye movement (NREM) sleep, and causes a persistent suppression of rapid eye movement (REM) sleep. We also demonstrate, for the first time, that activation of these preoptic glutamatergic neurons causes a high degree of NREM sleep fragmentation, promotes state instability with frequent arousals from sleep, decreases body temperature, and shifts cortical dynamics (including oscillations, connectivity, and complexity) to a more wake-like state. We conclude that a subset of preoptic glutamatergic neurons can initiate, but not maintain, arousals from sleep, and their inactivation may be required for NREM stability and REM sleep generation. Further, these data provide novel empirical evidence supporting the hypothesis that the preoptic area causally contributes to the regulation of both sleep and wakefulness. SIGNIFICANCE STATEMENT Historically, the preoptic area of the hypothalamus has been considered a key site for sleep generation. However, emerging modeling and empirical data suggest that this region might play a dual role in sleep-wake control. We demonstrate that chemogenetic stimulation of preoptic glutamatergic neurons produces brief arousals that fragment sleep, persistently suppresses REM sleep, causes hypothermia, and shifts EEG patterns toward a “lighter” NREM sleep state. We propose that preoptic glutamatergic neurons can initiate, but not maintain, arousal from sleep and gate REM sleep generation, possibly to block REM-like intrusions during NREM-to-wake transitions. In contrast to the long-standing notion in sleep neurobiology that the preoptic area is exclusively somnogenic, we provide further evidence that preoptic neurons also generate wakefulness.
Collapse
|
16
|
Watts M, Williams G, Lu J, Nithianantharajah J, Claudianos C. MicroRNA-210 Regulates Dendritic Morphology and Behavioural Flexibility in Mice. Mol Neurobiol 2021; 58:1330-1344. [PMID: 33165828 DOI: 10.1007/s12035-020-02197-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/30/2020] [Indexed: 01/05/2023]
Abstract
MicroRNAs are known to be critical regulators of neuronal plasticity. The highly conserved, hypoxia-regulated microRNA-210 (miR-210) has been shown to be associated with long-term memory in invertebrates and dysregulated in neurodevelopmental and neurodegenerative disease models. However, the role of miR-210 in mammalian neuronal function and cognitive behaviour remains unexplored. Here we generated Nestin-cre-driven miR-210 neuronal knockout mice to characterise miR-210 regulation and function using in vitro and in vivo methods. We identified miR-210 localisation throughout neuronal somas and dendritic processes and increased levels of mature miR-210 in response to neural activity in vitro. Loss of miR-210 in neurons resulted in higher oxidative phosphorylation and ROS production following hypoxia and increased dendritic arbour density in hippocampal cultures. Additionally, miR-210 knockout mice displayed altered behavioural flexibility in rodent touchscreen tests, particularly during early reversal learning suggesting processes underlying updating of information and feedback were impacted. Our findings support a conserved, activity-dependent role for miR-210 in neuroplasticity and cognitive function.
Collapse
Affiliation(s)
- Michelle Watts
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Gabrielle Williams
- School of Psychological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - Jing Lu
- School of Psychological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - Jess Nithianantharajah
- The Florey Institute of Neuroscience & Mental Health, Melbourne, VIC, 3052, Australia.
- Florey Department of Neuroscience, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Charles Claudianos
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
- Centre for Mental Health Research, The Australian National University, Canberra, ACT, 0200, Australia.
| |
Collapse
|
17
|
The Dual Orexin Receptor Antagonist DORA-22 Improves Mild Stress-induced Sleep Disruption During the Natural Sleep Phase of Nocturnal Rats. Neuroscience 2021; 463:30-44. [PMID: 33737028 DOI: 10.1016/j.neuroscience.2021.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/17/2022]
Abstract
Dual orexinergic antagonists (DORAs) have been recently developed as a pharmacotherapy alternative to established hypnotics. Hypnotics are largely evaluated in preclinical rodent models in the dark/active period yet should be ideally evaluated in the light/inactive period, analogous to when sleep disruption occurs in humans. We describe here the hypnotic efficacy of DORA-22 in rodent models of sleep disturbance produced by cage changes in the light/inactive period. Rats were administered DORA-22 or the GABA receptor-targeting hypnotic eszopiclone early in the light period, then exposed to six hourly clean cage changes with measurements of NREM sleep onset latency. Both compounds initially promoted sleep (hours 1 and 2), with DORA-22 exhibiting a more rapid hypnotic onset; and exhibited extended efficacy, evident six hours after administration in a sleep latencies test. A common complaint concerning hypnotic use is lingering hypersomnolence, and this is a concern in pharmacotherapy of the elderly. A second study was designed to determine a minimal dose of DORA-22 which would initially promote sleep but exhibit minimal extended hypnotic effect.Animals were administered DORA-22, then exposed for six hours to a single cage previously dirtied by a conspecific, followed by return to home cage. EEG measures indicated that all DORA-22 doses largely promoted sleep in the first hour. The lowest dose (1 mg/kg) did not decrease sleep onset latency at the six-hour timepoint, suggesting no residual hypersomnolence. We described here DORA-22 hypnotic efficacy during the normal sleep period of nocturnal rats, and demonstrate that well-chosen (low) hypnotic doses of DORA-22 may be hypnotically effective yet have minimal lingering effects.
Collapse
|
18
|
Phillips BG, Wang Y, Ambati S, Ma P, Meagher RB. Airways therapy of obstructive sleep apnea dramatically improves aberrant levels of soluble cytokines involved in autoimmune disease. Clin Immunol 2020; 221:108601. [PMID: 33017651 DOI: 10.1016/j.clim.2020.108601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/21/2020] [Accepted: 09/25/2020] [Indexed: 12/17/2022]
Abstract
Obstructive Sleep Apnea (OSA) damages the health of 35% of adult Americans. Disordered sleep results in increased risk of several autoimmune disorders, but the molecular links to autoimmunity are poorly understood. Herein, we identified four cytokines associated with autoimmune disease, whose median serum levels were significantly different for OSA patients receiving airways therapy, from the levels in untreated OSA patients, APRIL (5.2-fold lower, p = 3.5 × 10-11), CD30 (1.6-fold higher, p = 7.7 × 10-5), IFN-Alpha-2 (2.9-fold higher, p = 9.6 × 10-14) and IL-2 (1.9-fold higher, p = 0.0003). Cytokine levels in airways treated patients were similar to the levels in control subjects. t-SNE and UMAP analysis of these high dimensional patient cytokine data identified only two groups, suggesting a similar global response for all four cytokines to airways therapy. Our findings suggest the levels of these four cytokines may be altered by disordered sleep and perhaps by chronic hypoxia. Therapeutic options are discussed.
Collapse
Affiliation(s)
- Bradley G Phillips
- Clinical and Administrative Pharmacy, University of Georgia, Athens, GA 30602, USA; Clinical and Translational Research Unit, University of Georgia, Athens, GA 30602, USA
| | - Ye Wang
- Department of Statistics, University of Georgia, Athens, GA 30602, USA
| | - Suresh Ambati
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Ping Ma
- Department of Statistics, University of Georgia, Athens, GA 30602, USA
| | - Richard B Meagher
- Department of Genetics, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
19
|
|
20
|
Bubu OM, Andrade AG, Umasabor-Bubu OQ, Hogan MM, Turner AD, de Leon MJ, Ogedegbe G, Ayappa I, Jean-Louis G G, Jackson ML, Varga AW, Osorio RS. Obstructive sleep apnea, cognition and Alzheimer's disease: A systematic review integrating three decades of multidisciplinary research. Sleep Med Rev 2019; 50:101250. [PMID: 31881487 DOI: 10.1016/j.smrv.2019.101250] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/29/2022]
Abstract
Increasing evidence links cognitive-decline and Alzheimer's disease (AD) to various sleep disorders, including obstructive sleep apnea (OSA). With increasing age, there are substantial differences in OSA's prevalence, associated comorbidities and phenotypic presentation. An important question for sleep and AD researchers is whether OSA's heterogeneity results in varying cognitive-outcomes in older-adults compared to middle-aged adults. In this review, we systematically integrated research examining OSA and cognition, mild cognitive-impairment (MCI) and AD/AD biomarkers; including the effects of continuous positive airway pressure (CPAP) treatment, particularly focusing on characterizing the heterogeneity of OSA and its cognitive-outcomes. Broadly, in middle-aged adults, OSA is often associated with mild impairment in attention, memory and executive function. In older-adults, OSA is not associated with any particular pattern of cognitive-impairment at cross-section; however, OSA is associated with the development of MCI or AD with symptomatic patients who have a higher likelihood of associated disturbed sleep/cognitive-impairment driving these findings. CPAP treatment may be effective in improving cognition in OSA patients with AD. Recent trends demonstrate links between OSA and AD-biomarkers of neurodegeneration across all age-groups. These distinct patterns provide the foundation for envisioning better characterization of OSA and the need for more sensitive/novel sleep-dependent cognitive assessments to assess OSA-related cognitive-impairment.
Collapse
Affiliation(s)
- Omonigho M Bubu
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA; Department of Population Health, New York University School of Medicine, Center for Healthful Behavior Change, New York, USA; Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, Tampa, FL, USA; Department of Applied Health Sciences, Wheaton College, Wheaton, IL, USA.
| | - Andreia G Andrade
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | | | - Megan M Hogan
- Department of Applied Health Sciences, Wheaton College, Wheaton, IL, USA
| | - Arlener D Turner
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA; Department of Human Services and Psychology, National Louis University, Chicago, IL, USA
| | - Mony J de Leon
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA; Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Gbenga Ogedegbe
- Department of Population Health, New York University School of Medicine, Center for Healthful Behavior Change, New York, USA
| | - Indu Ayappa
- Division of Pulmonary, Critical Care and Sleep Medicine at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girardin Jean-Louis G
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA; Department of Population Health, New York University School of Medicine, Center for Healthful Behavior Change, New York, USA
| | - Melinda L Jackson
- Institute for Breathing and Sleep, Austin Health, Heidelberg, Victoria, Australia; School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Andrew W Varga
- Division of Pulmonary, Critical Care and Sleep Medicine at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ricardo S Osorio
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA.
| |
Collapse
|
21
|
Gamble MC, Katsuki F, McCoy JG, Strecker RE, McKenna JT. The dual orexinergic receptor antagonist DORA-22 improves the sleep disruption and memory impairment produced by a rodent insomnia model. Sleep 2019; 43:5583907. [PMID: 31595304 DOI: 10.1093/sleep/zsz241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/06/2019] [Indexed: 12/16/2022] Open
Abstract
AbstractInsomnia-related sleep disruption can contribute to impaired learning and memory. Treatment of insomnia should ideally improve the sleep profile while minimally affecting mnemonic function, yet many hypnotic drugs (e.g. benzodiazepines) are known to impair memory. Here, we used a rat model of insomnia to determine whether the novel hypnotic drug DORA-22, a dual orexin receptor antagonist, improves mild stress-induced insomnia with minimal effect on memory. Animals were first trained to remember the location of a hidden platform (acquisition) in the Morris Water Maze and then administered DORA-22 (10, 30, or 100 mg/kg doses) or vehicle control. Animals were then subjected to a rodent insomnia model involving two exposures to dirty cages over a 6-hr time period (at time points 0 and 3 hr), followed immediately by a probe trial in which memory of the water maze platform location was evaluated. DORA-22 treatment improved the insomnia-related sleep disruption—wake was attenuated and NREM sleep was normalized. REM sleep amounts were enhanced compared with vehicle treatment for one dose (30 mg/kg). In the first hour of insomnia model exposure, DORA-22 promoted the number and average duration of NREM sleep spindles, which have been previously proposed to play a role in memory consolidation (all doses). Water maze measures revealed probe trial performance improvement for select doses of DORA-22, including increased time spent in the platform quadrant (10 and 30 mg/kg) and time spent in platform location and number of platform crossings (10 mg/kg only). In conclusion, DORA-22 treatment improved insomnia-related sleep disruption and memory consolidation deficits.
Collapse
Affiliation(s)
- Mackenzie C Gamble
- Boston VA Research Institute, Inc., Jamaica Plain, MA
- VA Boston Healthcare System, West Roxbury, MA
| | - Fumi Katsuki
- Boston VA Research Institute, Inc., Jamaica Plain, MA
- VA Boston Healthcare System, West Roxbury, MA
- Department of Psychiatry, Harvard Medical School, West Roxbury, MA
| | - John G McCoy
- Boston VA Research Institute, Inc., Jamaica Plain, MA
- VA Boston Healthcare System, West Roxbury, MA
- Neuroscience Program, Stonehill College, Easton, MA
| | - Robert E Strecker
- Boston VA Research Institute, Inc., Jamaica Plain, MA
- VA Boston Healthcare System, West Roxbury, MA
- Department of Psychiatry, Harvard Medical School, West Roxbury, MA
| | - James Timothy McKenna
- Boston VA Research Institute, Inc., Jamaica Plain, MA
- VA Boston Healthcare System, West Roxbury, MA
- Department of Psychiatry, Harvard Medical School, West Roxbury, MA
| |
Collapse
|
22
|
Carroll CM, Macauley SL. The Interaction Between Sleep and Metabolism in Alzheimer's Disease: Cause or Consequence of Disease? Front Aging Neurosci 2019; 11:258. [PMID: 31616284 PMCID: PMC6764218 DOI: 10.3389/fnagi.2019.00258] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/30/2019] [Indexed: 01/21/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and affects over 45 million people worldwide. Both type-2-diabetes (T2D), a metabolic condition associated with aging, and disrupted sleep are implicated in the pathogenesis of AD, but how sleep and metabolism interact to affect AD progression remains unclear. In the healthy brain, sleep/wake cycles are a well-coordinated interaction between metabolic and neuronal activity, but when disrupted, are associated with a myriad of health-related issues, including metabolic syndrome, cardiovascular disease, T2D, and AD. Therefore, this review will explore our current understanding of the relationship between metabolism, sleep, and AD-related pathology to identify the causes and consequences of disease progression in AD. Moreover, sleep disturbances and metabolic dysfunction could serve as potential therapeutic targets to mitigate the increased risk of AD in individuals with T2D or offer a novel approach for treating AD.
Collapse
Affiliation(s)
| | - Shannon L. Macauley
- Section of Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
23
|
Grubac Z, Sutulovic N, Ademovic A, Velimirovic M, Rasic-Markovic A, Macut D, Petronijevic N, Stanojlovic O, Hrncic D. Short-term sleep fragmentation enhances anxiety-related behavior: The role of hormonal alterations. PLoS One 2019; 14:e0218920. [PMID: 31269081 PMCID: PMC6609147 DOI: 10.1371/journal.pone.0218920] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 06/13/2019] [Indexed: 01/25/2023] Open
Abstract
Introduction The neuroendocrine background of acute sleep fragmentation in obstructive sleep apnea and sleep fragmentation involvement in psychiatric comorbidities, common in these patients, are still largely unknown. The aim of this study was to determine the effects of short-term experimental sleep fragmentation on anxiety -like behavior and hormonal status in rats. Methods Male rats were adapted to treadmill (ON and OFF mode with belt speed set on 0.02m/s and 0.00m/s) and randomized to: 1) treadmill control (TC, only OFF mode); 2) motion, activity control (AC, 10min ON and 30min OFF mode) and 3) sleep fragmentation (SF, 30s ON and 90s OFF mode) group. Six hours later, the animals were tested in the open field, elevated plus maze and light/dark test (n = 8/group). Testosterone, estradiol, progesterone and corticosterone were determined in separate animal cohort immediately upon sleep fragmentation (n = 6/group). Results SF rats showed decreased rearings number, decreased time spent in the central area and increased thigmotaxic index compared to TC and AC rats in the open field test. Similarly, increased anxiety upon sleep fragmentation was observed in the elevated plus maze and the light/dark test. Significantly lower testosterone, estradiol and progesterone levels were determined in SF in comparison to AC and TC groups, while there was no significant difference in the levels of corticosterone. Conclusion Short term sleep fragmentation enhances anxiety-related behavior in rats, which could be partly mediated by the observed hormonal changes presented in the current study in form of testosterone, estradiol and progesterone depletion.
Collapse
Affiliation(s)
- Zeljko Grubac
- Laboratory of Neurophysiology, Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nikola Sutulovic
- Laboratory of Neurophysiology, Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Anida Ademovic
- Laboratory of Neurophysiology, Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milica Velimirovic
- Institute of Clinical and Medical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Rasic-Markovic
- Laboratory of Neurophysiology, Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Djuro Macut
- Clinic of Endocrinology, Diabetes and Metabolic Disease, CCS, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Natasa Petronijevic
- Institute of Clinical and Medical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Olivera Stanojlovic
- Laboratory of Neurophysiology, Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragan Hrncic
- Laboratory of Neurophysiology, Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- * E-mail: ,
| |
Collapse
|
24
|
Duncan MJ, Farlow H, Tirumalaraju C, Yun DH, Wang C, Howard JA, Sanden MN, O'Hara BF, McQuerry KJ, Bachstetter AD. Effects of the dual orexin receptor antagonist DORA-22 on sleep in 5XFAD mice. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:70-80. [PMID: 30859123 PMCID: PMC6396100 DOI: 10.1016/j.trci.2019.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction Sleep disruption is a characteristic of Alzheimer's disease (AD) that may exacerbate disease progression. This study tested whether a dual orexin receptor antagonist (DORA) would enhance sleep and attenuate neuropathology, neuroinflammation, and cognitive deficits in an AD-relevant mouse model, 5XFAD. Methods Wild-type (C57Bl6/SJL) and 5XFAD mice received chronic treatment with vehicle or DORA-22. Piezoelectric recordings monitored sleep and spatial memory was assessed via spontaneous Y-maze alternations. Aβ plaques, Aβ levels, and neuroinflammatory markers were measured by immunohistochemistry, enzyme-linked immunosorbent assay, and real-time polymerase chain reaction, respectively. Results In 5XFAD mice, DORA-22 significantly increased light-phase sleep without reducing Aβ levels, plaque density, or neuroinflammation. Effects of DORA-22 on cognitive deficits could not be determined because the 5XFAD mice did not exhibit deficits. Discussion These findings suggest that DORAs may improve sleep in AD patients. Further investigations should optimize the dose and duration of DORA-22 treatment and explore additional AD-relevant animal models and cognitive tests.
Collapse
Affiliation(s)
- Marilyn J Duncan
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Hannah Farlow
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Chairtra Tirumalaraju
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Do-Hyun Yun
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Chanung Wang
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | - James A Howard
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Madison N Sanden
- Department of Statistics, University of Kentucky, Lexington, KY, USA
| | - Bruce F O'Hara
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | | | - Adam D Bachstetter
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
25
|
McKenna JT, Gamble MC, Anderson‐Chernishof MB, Shah SR, McCoy JG, Strecker RE. A rodent cage change insomnia model disrupts memory consolidation. J Sleep Res 2018; 28:e12792. [PMID: 30461100 DOI: 10.1111/jsr.12792] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/21/2018] [Accepted: 10/18/2018] [Indexed: 11/29/2022]
Affiliation(s)
- James T. McKenna
- Boston VA Research Institute, Inc. Jamaica Plain Massachusetts
- VA Boston Healthcare System West Roxbury Massachusetts
- Department of Psychiatry Harvard Medical School West Roxbury Massachusetts
| | - Mackenzie C. Gamble
- Boston VA Research Institute, Inc. Jamaica Plain Massachusetts
- VA Boston Healthcare System West Roxbury Massachusetts
| | - Marissa B. Anderson‐Chernishof
- Boston VA Research Institute, Inc. Jamaica Plain Massachusetts
- VA Boston Healthcare System West Roxbury Massachusetts
- Department of Psychiatry Harvard Medical School West Roxbury Massachusetts
| | - Sunny R. Shah
- Boston VA Research Institute, Inc. Jamaica Plain Massachusetts
- VA Boston Healthcare System West Roxbury Massachusetts
| | - John G. McCoy
- Boston VA Research Institute, Inc. Jamaica Plain Massachusetts
- VA Boston Healthcare System West Roxbury Massachusetts
- Stonehill College Easton Massachusetts
| | - Robert E. Strecker
- Boston VA Research Institute, Inc. Jamaica Plain Massachusetts
- VA Boston Healthcare System West Roxbury Massachusetts
- Department of Psychiatry Harvard Medical School West Roxbury Massachusetts
| |
Collapse
|
26
|
Watts ME, Pocock R, Claudianos C. Brain Energy and Oxygen Metabolism: Emerging Role in Normal Function and Disease. Front Mol Neurosci 2018; 11:216. [PMID: 29988368 PMCID: PMC6023993 DOI: 10.3389/fnmol.2018.00216] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/01/2018] [Indexed: 01/09/2023] Open
Abstract
Dynamic metabolic changes occurring in neurons are critically important in directing brain plasticity and cognitive function. In other tissue types, disruptions to metabolism and the resultant changes in cellular oxidative state, such as increased reactive oxygen species (ROS) or induction of hypoxia, are associated with cellular stress. In the brain however, where drastic metabolic shifts occur to support physiological processes, subsequent changes to cellular oxidative state and induction of transcriptional sensors of oxidative stress likely play a significant role in regulating physiological neuronal function. Understanding the role of metabolism and metabolically-regulated genes in neuronal function will be critical in elucidating how cognitive functions are disrupted in pathological conditions where neuronal metabolism is affected. Here, we discuss known mechanisms regulating neuronal metabolism as well as the role of hypoxia and oxidative stress during normal and disrupted neuronal function. We also summarize recent studies implicating a role for metabolism in regulating neuronal plasticity as an emerging neuroscience paradigm.
Collapse
Affiliation(s)
- Michelle E Watts
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Charles Claudianos
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia.,Centre for Mental Health Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
27
|
Alex RM, Mousavi ND, Zhang R, Gatchel RJ, Behbehani K. Obstructive sleep apnea: Brain hemodynamics, structure, and function. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/jabr.12101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Raichel M. Alex
- Department of Bioengineering; University of Texas at Arlington; Arlington TX USA
| | | | - Rong Zhang
- Department of Neurology and Neurotherapeutics; University of Texas Southwestern Medical Center; Dallas TX USA
- Institute for Exercise and Environmental Medicine; Texas Health Hospital Dallas; Dallas TX USA
| | - Robert J. Gatchel
- Department of Psychology; University of Texas at Arlington; Arlington TX USA
| | - Khosrow Behbehani
- Department of Bioengineering; University of Texas at Arlington; Arlington TX USA
| |
Collapse
|
28
|
Lee ML, Katsuyama ÂM, Duge LS, Sriram C, Krushelnytskyy M, Kim JJ, de la Iglesia HO. Fragmentation of Rapid Eye Movement and Nonrapid Eye Movement Sleep without Total Sleep Loss Impairs Hippocampus-Dependent Fear Memory Consolidation. Sleep 2016; 39:2021-2031. [PMID: 27568801 DOI: 10.5665/sleep.6236] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/29/2016] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES Sleep is important for consolidation of hippocampus-dependent memories. It is hypothesized that the temporal sequence of nonrapid eye movement (NREM) sleep and rapid eye movement (REM) sleep is critical for the weakening of nonadaptive memories and the subsequent transfer of memories temporarily stored in the hippocampus to more permanent memories in the neocortex. A great body of evidence supporting this hypothesis relies on behavioral, pharmacological, neural, and/or genetic manipulations that induce sleep deprivation or stage-specific sleep deprivation. METHODS We exploit an experimental model of circadian desynchrony in which intact animals are not deprived of any sleep stage but show fragmentation of REM and NREM sleep within nonfragmented sleep bouts. We test the hypothesis that the shortening of NREM and REM sleep durations post-training will impair memory consolidation irrespective of total sleep duration. RESULTS When circadian-desynchronized animals are trained in a hippocampus-dependent contextual fear-conditioning task they show normal short-term memory but impaired long-term memory consolidation. This impairment in memory consolidation is positively associated with the post-training fragmentation of REM and NREM sleep but is not significantly associated with the fragmentation of total sleep or the total amount of delta activity. We also show that the sleep stage fragmentation resulting from circadian desynchrony has no effect on hippocampus-dependent spatial memory and no effect on hippocampus-independent cued fear-conditioning memory. CONCLUSIONS Our findings in an intact animal model, in which sleep deprivation is not a confounding factor, support the hypothesis that the stereotypic sequence and duration of sleep stages play a specific role in long-term hippocampus-dependent fear memory consolidation.
Collapse
Affiliation(s)
- Michael L Lee
- Department of Biology, University of Washington, Seattle, WA.,Graduate Program in Neuroscience, University of Washington, Seattle, WA
| | | | - Leanne S Duge
- Department of Biology, University of Washington, Seattle, WA
| | - Chaitra Sriram
- Department of Biology, University of Washington, Seattle, WA
| | | | - Jeansok J Kim
- Graduate Program in Neuroscience, University of Washington, Seattle, WA.,Department of Psychology, University of Washington, Seattle WA
| | - Horacio O de la Iglesia
- Department of Biology, University of Washington, Seattle, WA.,Graduate Program in Neuroscience, University of Washington, Seattle, WA
| |
Collapse
|
29
|
Repeated pre-training sleep restriction in adolescent rats impaired spatial performance. Sleep Biol Rhythms 2016. [DOI: 10.1007/s41105-016-0080-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
30
|
Daurat A, Sarhane M, Tiberge M. Syndrome d’apnées obstructives du sommeil et cognition : une revue. Neurophysiol Clin 2016; 46:201-15. [DOI: 10.1016/j.neucli.2016.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 04/13/2016] [Accepted: 04/29/2016] [Indexed: 11/29/2022] Open
|
31
|
Sforza E, Roche F. Chronic intermittent hypoxia and obstructive sleep apnea: an experimental and clinical approach. HYPOXIA (AUCKLAND, N.Z.) 2016; 4:99-108. [PMID: 27800512 PMCID: PMC5085272 DOI: 10.2147/hp.s103091] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obstructive sleep apnea (OSA) is a prevalent sleep disorder considered as an independent risk factor for cardiovascular consequences, such as systemic arterial hypertension, ischemic heart disease, cardiac arrhythmias, metabolic disorders, and cognitive dysfunction. The pathogenesis of OSA-related consequence is assumed to be chronic intermittent hypoxia (IH) inducing alterations at the molecular level, oxidative stress, persistent systemic inflammation, oxygen sensor activation, and increase of sympathetic activity. Overall, these mechanisms have an effect on vessel permeability and are considered to be important factors for explaining vascular, metabolic, and cognitive OSA-related consequences. The present review attempts to examine together the research paradigms and clinical studies on the effect of acute and chronic IH and the potential link with OSA. We firstly describe the literature data on the mechanisms activated by acute and chronic IH at the experimental level, which are very helpful and beneficial to explaining OSA consequences. Then, we describe in detail the effect of IH in patients with OSA that we can consider "the human model" of chronic IH. In this way, we can better understand the specific pathophysiological mechanisms proposed to explain the consequences of IH in OSA.
Collapse
Affiliation(s)
- Emilia Sforza
- Service de Physiologie Clinique et de l’Exercice, Pole NOL, CHU, EA SNA-EPIS 4607, Faculté de Médecine J. Lisfranc, UJM Saint-Etienne, Université de Lyon, Saint-Etienne, France
| | - Fréderic Roche
- Service de Physiologie Clinique et de l’Exercice, Pole NOL, CHU, EA SNA-EPIS 4607, Faculté de Médecine J. Lisfranc, UJM Saint-Etienne, Université de Lyon, Saint-Etienne, France
| |
Collapse
|
32
|
Sleep disruption increases seizure susceptibility: Behavioral and EEG evaluation of an experimental model of sleep apnea. Physiol Behav 2015; 155:188-94. [PMID: 26705666 DOI: 10.1016/j.physbeh.2015.12.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 12/24/2022]
Abstract
Sleep disruption accompanies sleep apnea as one of its major symptoms. Obstructive sleep apnea is particularly common in patients with refractory epilepsy, but causing factors underlying this are far from being resolved. Therefore, translational studies regarding this issue are important. Our aim was to investigate the effects of sleep disruption on seizure susceptibility of rats using experimental model of lindane-induced refractory seizures. Sleep disruption in male Wistar rats with implanted EEG electrodes was achieved by treadmill method (belt speed set on 0.02 m/s for working and 0.00 m/s for stop mode, respectively). Animals were assigned to experimental conditions lasting 6h: 1) sleep disruption (sleep interrupted, SI; 30s working and 90 s stop mode every 2 min; 180 cycles in total); 2) activity control (AC, 10 min working and 30 min stop mode, 9 cycles in total); 3) treadmill chamber control (TC, only stop mode). Afterwards, the animals were intraperitoneally treated with lindane (L, 4 mg/kg, SI+L, AC+L and TC+L groups) or dimethylsulfoxide (DMSO, SIc, ACc and TCc groups). Convulsive behavior was assessed by seizure incidence, latency time to first seizure, and its severity during 30 min after drug administration. Number and duration of ictal periods were determined in recorded EEGs. Incidence and severity of lindane-induced seizures were significantly increased, latency time significantly decreased in animals undergoing sleep disruption (SI+L group) compared with the animals from TC+L. Seizure latency was also significantly decreased in SI+L compared to AC+L groups. Number of ictal periods were increased and duration of it presented tendency to increase in SI+L comparing to AC+L. No convulsive signs were observed in TCc, ACc and SIc groups, as well as no ictal periods in EEG. These results indicate sleep disruption facilitates induction of epileptic activity in rodent model of lindane-epilepsy enabling translational research of this phenomenon.
Collapse
|
33
|
Mutluer T, Karakoc Demirkaya S, Abali O. Assessment of sleep problems and related risk factors observed in Turkish children with Autism spectrum disorders. Autism Res 2015; 9:536-42. [PMID: 26459217 DOI: 10.1002/aur.1542] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/28/2015] [Indexed: 01/02/2023]
Abstract
Sleep problems are common and difficult to manage in children with autism spectrum disorders (ASD). Another major adverse impact of sleep problems is that they exacerbate behavioral problems. To assess sleep problems and possible behavioral risk factors in detail, we aimed to compare sleep habits of children with ASD, with healthy children. The relationship between sleep difficulties and concomitant behavioral problems such as repetitive behaviors, hyperactivity, and social withdrawal were also examined. Hundred and seventeen children and adolescents including 64 with the diagnosis of ASD and 53 healthy subjects were enrolled in the study. Diagnostic Interview for ASD was performed according to DSM-IV-TR. Socio-demographical data form and childhood autism rating scale were filled by researchers. Aberrant behavior checklist (ABC), child behavior checklist and pediatric sleep questionnaire (PSQ) were completed by the parents of the children. Children with ASD had higher frequency of sleep problems, snoring, breathing problems, behavioral problems compared with healthy children (for all parameters; P < 0.001). A positive correlation was identified between the total score of PSQ and the total score of ABC (P < 0.05, Spearman correlation coefficient: 0.347). Sleep latency was prolonged in children with ASD compared with healthy subjects (P < 0.001). In accordance with the current literature, children with ASD were subject to sleep problems significantly more than the control group. Identified risk factors for sleep problems in ASD children were behavioral factors such as stereotypies, self-mutilation, hyperactivity, and social withdrawal. Autism Res 2016, 9: 536-542. © 2015 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tuba Mutluer
- Van Regional Education and Research Hospital, Child and Adolescent Psychiatry Clinic
| | | | - Osman Abali
- Department of Child and Adolescent Psychiatry, Istanbul University Istanbul Medical Faculty
| |
Collapse
|
34
|
Wallace E, Kim DY, Kim KM, Chen S, Blair Braden B, Williams J, Jasso K, Garcia A, Rho JM, Bimonte-Nelson H, Maganti R. Differential effects of duration of sleep fragmentation on spatial learning and synaptic plasticity in pubertal mice. Brain Res 2015; 1615:116-128. [PMID: 25957790 DOI: 10.1016/j.brainres.2015.04.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 03/17/2015] [Accepted: 04/12/2015] [Indexed: 12/15/2022]
Abstract
STUDY OBJECTIVE To examine the differential effects of acute and chronic sleep fragmentation (SF) on spatial learning and memory, and hippocampal long-term potentiation (LTP) in pubertal mice. METHODS Two studies were performed during which adolescent C57/Bl6 mice were subjected to acute-SF 24h a day × 3 days or chronic-SF for 12h a day × 2 weeks using a programmable rotating lever that provides tactile stimulus with controls housed in similar cages. Spatial learning and memory was examined using the Morris water maze, and long-term potentiation (LTP) was evaluated after stimulation of Schaffer collaterals in CA1 hippocampus post SF. Actigraphy was used during the period of SF to monitor rest-activity patterns. Electroencephalographic (EEG) recordings were acquired for analysis of vigilance state patterns and delta-power. Serum corticosterone was measured to assess stress levels. RESULTS Acute-SF via tactile stimulation negatively impacted spatial learning, as well as LTP maintenance, compared to controls with no tactile stimulation. While actigraphy showed significantly increased motor activity during SF in both groups, EEG data indicated that overall sleep efficiency did not differ between baseline and SF days, but significant increases in number of wakeful bouts and decreases in average NREM and REM bout lengths were seen during lights-on. Acute sleep fragmentation did not impact corticosterone levels. CONCLUSIONS The current results indicate that, during development in pubertal mice, acute-SF for 24h a day × 3 days negatively impacted spatial learning and synaptic plasticity. Further studies are needed to determine if any inherent long-term homeostatic mechanisms in the adolescent brain afford greater resistance to the deleterious effects of chronic-SF.
Collapse
Affiliation(s)
- Eli Wallace
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Do Young Kim
- Barrow Neurological Institute/St Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Kye-Min Kim
- Barrow Neurological Institute/St Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Stephanie Chen
- Barrow Neurological Institute/St Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - B Blair Braden
- Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Jeremy Williams
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kalene Jasso
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Jong M Rho
- Departments of Pediatrics and Clinical Neurosciences, University of Calgary Faculty of Medicine, Calgary, Canada
| | - Heather Bimonte-Nelson
- Arizona State University, Tempe, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Rama Maganti
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
35
|
Baud MO, Magistretti PJ, Petit JM. Sustained sleep fragmentation induces sleep homeostasis in mice. Sleep 2015; 38:567-79. [PMID: 25325477 DOI: 10.5665/sleep.4572] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 09/14/2014] [Indexed: 01/12/2023] Open
Abstract
STUDY OBJECTIVES Sleep fragmentation (SF) is an integral feature of sleep apnea and other prevalent sleep disorders. Although the effect of repetitive arousals on cognitive performance is well documented, the effects of long-term SF on electroencephalography (EEG) and molecular markers of sleep homeostasis remain poorly investigated. To address this question, we developed a mouse model of chronic SF and characterized its effect on EEG spectral frequencies and the expression of genes previously linked to sleep homeostasis including clock genes, heat shock proteins, and plasticity-related genes. DESIGN N/A. SETTING Animal sleep research laboratory. PARTICIPANTS Sixty-six C57BL6/J adult mice. INTERVENTIONS Instrumental sleep disruption at a rate of 60/h during 14 days. MEASUREMENTS AND RESULTS Locomotor activity and EEG were recorded during 14 days of SF followed by recovery for 2 days. Despite a dramatic number of arousals and decreased sleep bout duration, SF minimally reduced total quantity of sleep and did not significantly alter its circadian distribution. Spectral analysis during SF revealed a homeostatic drive for slow wave activity (SWA; 1-4 Hz) and other frequencies as well (4-40 Hz). Recordings during recovery revealed slow wave sleep consolidation and a transient rebound in SWA, and paradoxical sleep duration. The expression of selected genes was not induced following chronic SF. CONCLUSIONS Chronic SF increased sleep pressure confirming that altered quality with preserved quantity triggers core sleep homeostasis mechanisms. However, it did not induce the expression of genes induced by sleep loss, suggesting that these molecular pathways are not sustainably activated in chronic diseases involving SF.
Collapse
Affiliation(s)
- Maxime O Baud
- Laboratory of Neuroenergetic and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Department of Neurology, University of California at San Francisco (UCSF), San Francisco, CA
| | - Pierre J Magistretti
- Laboratory of Neuroenergetic and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, KSA.,Centre de Neurosciences Psychiatriques, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, Prilly, Switzerland
| | - Jean-Marie Petit
- Laboratory of Neuroenergetic and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Centre de Neurosciences Psychiatriques, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, Prilly, Switzerland
| |
Collapse
|
36
|
Chen Y, Zhao C, Zhang C, Luo L, Yu G. Influence of chronic intermittent hypoxia on growth associated protein 43 expression in the hippocampus of young rats. Neural Regen Res 2015; 7:1241-6. [PMID: 25709622 PMCID: PMC4336958 DOI: 10.3969/j.issn.1673-5374.2012.16.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 04/23/2012] [Indexed: 11/29/2022] Open
Abstract
This study aimed to explore the pathological change to hippocampal neurons and the expression of growth associated protein 43 in 21-day-old young rats following chronic intermittent hypoxia. Hematoxylin-eosin staining results showed varying degrees of degeneration and necrosis in hippocampal neurons depending on the modeling time. Immunohistochemistry revealed that growth associated protein 43 expression in young rats following chronic intermittent hypoxia decreased, but that levels were still higher than those of normal rats at each time point, especially 4 weeks after modeling. During 1–5 weeks after modeling, a slow growth in rat weight was observed. Experimental findings indicate that chronic intermittent hypoxia may induce growth dysfunction and necrosis of hippocampal neurons, as well as increase the expression of growth associated protein 43 in young rats.
Collapse
Affiliation(s)
- Yan Chen
- Department of Physiology, Luzhou Medical College, Luzhou 646000, Sichuan Province, China
| | - Chunling Zhao
- Department of Physiology, Luzhou Medical College, Luzhou 646000, Sichuan Province, China
| | - Chunlai Zhang
- Department of Physiology, Luzhou Medical College, Luzhou 646000, Sichuan Province, China
| | - Lirong Luo
- Functional Experiment Center of Luzhou Medical College, Luzhou 646000, Sichuan Province, China
| | - Guang Yu
- Department of Physiology, Luzhou Medical College, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
37
|
Abstract
Hippocampal electrophysiology and behavioral evidence support a role for sleep in spatial navigational memory, but the role of particular sleep stages is less clear. Although rodent models suggest the importance of rapid eye movement (REM) sleep in spatial navigational memory, a similar role for REM sleep has never been examined in humans. We recruited subjects with severe obstructive sleep apnea (OSA) who were well treated and adherent with continuous positive airway pressure (CPAP). Restricting CPAP withdrawal to REM through real-time monitoring of the polysomnogram provides a novel way of addressing the role of REM sleep in spatial navigational memory with a physiologically relevant stimulus. Individuals spent two different nights in the laboratory, during which subjects performed timed trials before and after sleep on one of two unique 3D spatial mazes. One night of sleep was normally consolidated with use of therapeutic CPAP throughout, whereas on the other night, CPAP was reduced only in REM sleep, allowing REM OSA to recur. REM disruption via this method caused REM sleep reduction and significantly fragmented any remaining REM sleep without affecting total sleep time, sleep efficiency, or slow-wave sleep. We observed improvements in maze performance after a night of normal sleep that were significantly attenuated after a night of REM disruption without changes in psychomotor vigilance. Furthermore, the improvement in maze completion time significantly positively correlated with the mean REM run duration across both sleep conditions. In conclusion, we demonstrate a novel role for REM sleep in human memory formation and highlight a significant cognitive consequence of OSA.
Collapse
|
38
|
Aubrecht TG, Jenkins R, Magalang UJ, Nelson RJ. Influence of gonadal hormones on the behavioral effects of intermittent hypoxia in mice. Am J Physiol Regul Integr Comp Physiol 2014; 308:R489-99. [PMID: 25552660 DOI: 10.1152/ajpregu.00379.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Obstructive sleep apnea (OSA) is characterized by repetitive upper airway obstruction resulting in cyclic intermittent hypoxia (IH) during sleep in affected individuals. OSA occurs more frequently in postmenopausal than premenopausal women and the severity of OSA increases after menopause. Gonadal hormones can influence brain and behavior; testosterone and estrogens in particular can enhance spatial learning and memory. We hypothesized that estrogens may protect mice from IH-induced hippocampal morphological and behavioral changes. To test this hypothesis we exposed intact or gonadectomized male and female mice to room air or IH [15 cycles/h, 8 h/day, fraction of inspired oxygen (FiO 2) nadir of 5%] for a total of 30 days. During the final 4 days of IH, mice were tested for anxiety- and depressive-like behaviors. After cessation of IH exposure mice were tested on the Barnes maze and passive avoidance tests to assess learning and memory. Ovariectomy paired with IH treatment, impaired spatial learning and memory compared to all other female groups. Intact male mice receiving IH treatment also had impaired learning and memory compared with intact or castrated male mice exposed to room air. Learning and memory changes were mirrored by changes in basilar dendritic length of the CA1 region of the hippocampus. These data suggest that estrogens provide protection against IH-induced deficits, whereas androgens partially exacerbate IH-induced deficits on learning and memory.
Collapse
Affiliation(s)
- Taryn G Aubrecht
- Department of Neuroscience and Neuroscience Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio; and
| | - Richelle Jenkins
- Department of Neuroscience and Neuroscience Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio; and
| | - Ulysses J Magalang
- Department of Neuroscience and Neuroscience Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio; and Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Randy J Nelson
- Department of Neuroscience and Neuroscience Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio; and
| |
Collapse
|
39
|
Hypomyelination, memory impairment, and blood-brain barrier permeability in a model of sleep apnea. Brain Res 2014; 1597:28-36. [PMID: 25482664 DOI: 10.1016/j.brainres.2014.11.052] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/16/2014] [Accepted: 11/24/2014] [Indexed: 01/28/2023]
Abstract
We investigated the effect of intermittent hypoxia, mimicking sleep apnea, on axonal integrity, blood-brain barrier permeability, and cognitive function of mice. Forty-seven C57BL mice were exposed to intermittent or sham hypoxia, alternating 30s of progressive hypoxia and 30s of reoxigenation, during 8h/day. The axonal integrity in cerebellum was evaluated by transmission electron microscopy. Short- and long-term memories were assessed by novel object recognition test. The levels of endothelin-1 were measured by ELISA. Blood-brain barrier permeability was quantified by Evans Blue dye. After 14 days, animals exposed to intermittent hypoxia showed hypomyelination in cerebellum white matter and higher serum levels of endothelin-1. The short and long-term memories in novel object recognition test was impaired in the group exposed to intermittent hypoxia as compared to controls. Blood-brain barrier permeability was similar between the groups. These results indicated that hypomyelination and impairment of short- and long-term working memories occurred in C57BL mice after 14 days of intermittent hypoxia mimicking sleep apnea.
Collapse
|
40
|
Topchiy I, Amodeo DA, Ragozzino ME, Waxman J, Radulovacki M, Carley DW. Acute exacerbation of sleep apnea by hyperoxia impairs cognitive flexibility in Brown-Norway rats. Sleep 2014; 37:1851-61. [PMID: 25364080 DOI: 10.5665/sleep.4184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES To determine whether learning deficits occur during acute exacerbation of spontaneous sleep related breathing disorder (SRBD) in rats with high (Brown Norway; BN) and low (Zucker Lean; ZL) apnea propensity. DESIGN Spatial acquisition (3 days) and reversal learning (3 days) in the Morris water maze (MWM) with polysomnography (12:00-08:00): (1) with acute SRBD exacerbation (by 20-h hyperoxia immediately preceding reversal learning) or (2) without SRBD exacerbation (room air throughout). SETTING Randomized, placebo-controlled, repeated-measures design. PARTICIPANTS 14 BN rats; 16 ZL rats. INTERVENTIONS 20-h hyperoxia. MEASUREMENTS AND RESULTS Apneas were detected as cessation of respiration ≥ 2 sec. Swim latency in MWM, apnea indices (AI; apneas/hour of sleep) and percentages of recording time for nonrapid eye movement (NREM), rapid eye movement (REM), and total sleep were assessed. Baseline AI in BN rats was more than double that of ZL rats (22.46 ± 2.27 versus 10.7 ± 0.9, P = 0.005). Hyperoxia increased AI in both BN (34.3 ± 7.4 versus 22.46 ± 2.27) and ZL rats (15.4 ± 2.7 versus 10.7 ± 0.9) without changes in sleep stage percentages. Control (room air) BN and ZL rats exhibited equivalent acquisition and reversal learning. Acute exacerbation of AI by hyperoxia produced a reversal learning performance deficit in BN but not ZL rats. In addition, the percentage of REM sleep and REM apnea index in BN rats during hyperoxia negatively correlated with reversal learning performance. CONCLUSIONS Acute exacerbation of sleep related breathing disorder by hyperoxia impairs reversal learning in a rat strain with high apnea propensity, but not a strain with a low apnea propensity. This suggests a non-linear threshold effect may contribute to the relationships between sleep apnea and cognitive dysfunctions, but strain-specific differences also may be important.
Collapse
Affiliation(s)
- Irina Topchiy
- Center for Narcolepsy, Sleep and Health Research, University of Illinois at Chicago, Chicago, IL ; Department of Biobehavioral and Health Sciences, University of Illinois at Chicago, Chicago, IL
| | - Dionisio A Amodeo
- Department of Psychology, University of Illinois at Chicago, Chicago, IL
| | | | - Jonathan Waxman
- Center for Narcolepsy, Sleep and Health Research, University of Illinois at Chicago, Chicago, IL
| | - Miodrag Radulovacki
- Center for Narcolepsy, Sleep and Health Research, University of Illinois at Chicago, Chicago, IL ; Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
| | - David W Carley
- Center for Narcolepsy, Sleep and Health Research, University of Illinois at Chicago, Chicago, IL ; Department of Medicine, University of Illinois at Chicago, Chicago, IL ; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL ; Department of Biobehavioral and Health Sciences, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
41
|
Diogo LN, Monteiro EC. The efficacy of antihypertensive drugs in chronic intermittent hypoxia conditions. Front Physiol 2014; 5:361. [PMID: 25295010 PMCID: PMC4170135 DOI: 10.3389/fphys.2014.00361] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 09/03/2014] [Indexed: 12/22/2022] Open
Abstract
Sleep apnea/hypopnea disorders include centrally originated diseases and obstructive sleep apnea (OSA). This last condition is renowned as a frequent secondary cause of hypertension (HT). The mechanisms involved in the pathogenesis of HT can be summarized in relation to two main pathways: sympathetic nervous system stimulation mediated mainly by activation of carotid body (CB) chemoreflexes and/or asphyxia, and, by no means the least important, the systemic effects of chronic intermittent hypoxia (CIH). The use of animal models has revealed that CIH is the critical stimulus underlying sympathetic activity and hypertension, and that this effect requires the presence of functional arterial chemoreceptors, which are hyperactive in CIH. These models of CIH mimic the HT observed in humans and allow the study of CIH independently without the mechanical obstruction component. The effect of continuous positive airway pressure (CPAP), the gold standard treatment for OSA patients, to reduce blood pressure seems to be modest and concomitant antihypertensive therapy is still required. We focus this review on the efficacy of pharmacological interventions to revert HT associated with CIH conditions in both animal models and humans. First, we explore the experimental animal models, developed to mimic HT related to CIH, which have been used to investigate the effect of antihypertensive drugs (AHDs). Second, we review what is known about drug efficacy to reverse HT induced by CIH in animals. Moreover, findings in humans with OSA are cited to demonstrate the lack of strong evidence for the establishment of a first-line antihypertensive regimen for these patients. Indeed, specific therapeutic guidelines for the pharmacological treatment of HT in these patients are still lacking. Finally, we discuss the future perspectives concerning the non-pharmacological and pharmacological management of this particular type of HT.
Collapse
Affiliation(s)
- Lucilia N Diogo
- Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa Lisboa, Portugal
| | - Emília C Monteiro
- Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa Lisboa, Portugal
| |
Collapse
|
42
|
Endoplasmic reticulum stress plays critical role in brain damage after chronic intermittent hypoxia in growing rats. Exp Neurol 2014; 257:148-56. [PMID: 24810321 DOI: 10.1016/j.expneurol.2014.04.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 04/17/2014] [Accepted: 04/28/2014] [Indexed: 01/08/2023]
Abstract
Obstructive sleep apnea hypopnea syndrome (OSAHS) in children is associated with multiple system morbidities. Cognitive dysfunction as a result of central nervous system complication has been reported in children with OSAHS. However, the underlying mechanisms are poorly understood. Endoplasmic reticulum stress (ERS)-related apoptosis plays an important role in various diseases of the central nervous system, but very little is known about the role of ERS in mediating pathophysiological reactions to cognitive dysfunction in OSAHS. Chronic intermittent hypoxia (CIH) exposures, modeling OSAHS, across 2 and 4weeks in growing rats made more reference memory errors, working memory errors and total memory errors in the 8-Arm radial maze task, increased significantly TUNEL positive cells, upregulated the unfolded protein response in the hippocampus and prefrontal cortex as evidenced by increased phosphorylation of PKR-like endoplasmic reticulum kinase, inositol-requiring enzyme l and some downstream products. A selective inhibitor of eukaryotic initiation factor-2a dephosphorylation, salubrinal, prevented C/EBP-homologous protein activation in the hippocampus and prefrontal cortex throughout hypoxia/reoxygenation exposure. Our findings suggest that ERS mediated cell apoptosis may be one of the underlying mechanisms of cognitive dysfunction in OSAHS children. Further, a specific ERS inhibitor Salubrinal should be tested for neuroprotection against CIH-induced injury.
Collapse
|
43
|
Li Y, Panossian LA, Zhang J, Zhu Y, Zhan G, Chou YT, Fenik P, Bhatnagar S, Piel DA, Beck SG, Veasey S. Effects of chronic sleep fragmentation on wake-active neurons and the hypercapnic arousal response. Sleep 2014; 37:51-64. [PMID: 24470695 DOI: 10.5665/sleep.3306] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
STUDY OBJECTIVES Delayed hypercapnic arousals may occur in obstructive sleep apnea. The impaired arousal response is expected to promote more pronounced oxyhemoglobin desaturations. We hypothesized that long-term sleep fragmentation (SF) results in injury to or dysfunction of wake-active neurons that manifests, in part, as a delayed hypercapnic arousal response. DESIGN Adult male mice were implanted for behavioral state recordings and randomly assigned to 4 weeks of either orbital platform SF (SF4wk, 30 events/h) or control conditions (Ct4wk) prior to behavioral, histological, and locus coeruleus (LC) whole cell electrophysiological evaluations. MEASUREMENTS AND RESULTS SF was successfully achieved across the 4 week study, as evidenced by a persistently increased arousal index, P < 0.01 and shortened sleep bouts, P < 0.05, while total sleep/wake times and plasma corticosterone levels were unaffected. A multiple sleep latency test performed at the onset of the dark period showed a reduced latency to sleep in SF4wk mice (P < 0.05). The hypercapnic arousal latency was increased, Ct4wk 64 ± 5 sec vs. SF4wk 154 ± 6 sec, P < 0.001, and remained elevated after a 2 week recovery (101 ± 4 sec, P < 0.001). C-fos activation in noradrenergic, orexinergic, histaminergic, and cholinergic wake-active neurons was reduced in response to hypercapnia (P < 0.05-0.001). Catecholaminergic and orexinergic projections into the cingulate cortex were also reduced in SF4wk (P < 0.01). In addition, SF4wk resulted in impaired LC neuron excitability (P < 0.01). CONCLUSIONS Four weeks of sleep fragmentation (SF4wk) impairs arousal responses to hypercapnia, reduces wake neuron projections and locus coeruleus neuronal excitability, supporting the concepts that some effects of sleep fragmentation may contribute to impaired arousal responses in sleep apnea, which may not reverse immediately with therapy.
Collapse
Affiliation(s)
- Yanpeng Li
- Department of Neurology, Neuroscience Research Center, Shanghai Changzheng Hospital, the Affiliated Hospital to the Second Military Medical University, Shanghai City, China ; Center for Sleep and Circadian Neurobiology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Lori A Panossian
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Jing Zhang
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Yan Zhu
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Guanxia Zhan
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Yu-Ting Chou
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Polina Fenik
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Seema Bhatnagar
- Department of Anesthesiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - David A Piel
- Department of Anesthesiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Sheryl G Beck
- Department of Anesthesiology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Sigrid Veasey
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
44
|
Rahman A, Languille S, Lamberty Y, Babiloni C, Perret M, Bordet R, Blin OJ, Jacob T, Auffret A, Schenker E, Richardson J, Pifferi F, Aujard F. Sleep deprivation impairs spatial retrieval but not spatial learning in the non-human primate grey mouse lemur. PLoS One 2013; 8:e64493. [PMID: 23717620 PMCID: PMC3661499 DOI: 10.1371/journal.pone.0064493] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 04/15/2013] [Indexed: 12/04/2022] Open
Abstract
A bulk of studies in rodents and humans suggest that sleep facilitates different phases of learning and memory process, while sleep deprivation (SD) impairs these processes. Here we tested the hypothesis that SD could alter spatial learning and memory processing in a non-human primate, the grey mouse lemur (Microcebus murinus), which is an interesting model of aging and Alzheimer's disease (AD). Two sets of experiments were performed. In a first set of experiments, we investigated the effects of SD on spatial learning and memory retrieval after one day of training in a circular platform task. Eleven male mouse lemurs aged between 2 to 3 years were tested in three different conditions: without SD as a baseline reference, 8 h of SD before the training and 8 h of SD before the testing. The SD was confirmed by electroencephalographic recordings. Results showed no effect of SD on learning when SD was applied before the training. When the SD was applied before the testing, it induced an increase of the amount of errors and of the latency prior to reach the target. In a second set of experiments, we tested the effect of 8 h of SD on spatial memory retrieval after 3 days of training. Twenty male mouse lemurs aged between 2 to 3 years were tested in this set of experiments. In this condition, the SD did not affect memory retrieval. This is the first study that documents the disruptive effects of the SD on spatial memory retrieval in this primate which may serve as a new validated challenge to investigate the effects of new compounds along physiological and pathological aging.
Collapse
Affiliation(s)
- Anisur Rahman
- Mécanismes Adaptatifs et Evolution, UMR 7179 Centre National de la Recherche Scientifique, Muséum National d'Histoire Naturelle, Brunoy, France
| | - Solène Languille
- Mécanismes Adaptatifs et Evolution, UMR 7179 Centre National de la Recherche Scientifique, Muséum National d'Histoire Naturelle, Brunoy, France
| | - Yves Lamberty
- UCB Pharma s.a., Neuroscience Therapeutic Area, Braine l'Alleud, Belgium
| | - Claudio Babiloni
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy, and IRCCS San Raffalele Pisana, Rome, Italy
| | - Martine Perret
- Mécanismes Adaptatifs et Evolution, UMR 7179 Centre National de la Recherche Scientifique, Muséum National d'Histoire Naturelle, Brunoy, France
| | - Regis Bordet
- Département de Pharmacologie Médicale, EA 1046, Université Lille Nord de France, UDSL, Faculté de Médecine, CHU, Lille, France
| | - Olivier J. Blin
- CPCET-CIC, AP-HM Timone, INT, UMR 7289, CNRS - Aix Marseille Université, Marseille, France
| | - Tom Jacob
- Johnson and Johnson Pharmaceutical Research and Development, A Division of Janssen Pharmaceutica, Beerse, Belgium
| | - Alexandra Auffret
- CPCET-CIC, AP-HM Timone, INT, UMR 7289, CNRS - Aix Marseille Université, Marseille, France
| | | | - Jill Richardson
- GlaxoSmithKline, R&D China U.K. Group, Stevenage, Hertfordshire, United Kingdom
| | - Fabien Pifferi
- Mécanismes Adaptatifs et Evolution, UMR 7179 Centre National de la Recherche Scientifique, Muséum National d'Histoire Naturelle, Brunoy, France
| | - Fabienne Aujard
- Mécanismes Adaptatifs et Evolution, UMR 7179 Centre National de la Recherche Scientifique, Muséum National d'Histoire Naturelle, Brunoy, France
| |
Collapse
|
45
|
Aubrecht TG, Weil ZM, Magalang UJ, Nelson RJ. Dim light at night interacts with intermittent hypoxia to alter cognitive and affective responses. Am J Physiol Regul Integr Comp Physiol 2013; 305:R78-86. [PMID: 23657638 DOI: 10.1152/ajpregu.00100.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Obstructive sleep apnea (OSA) and dim light at night (dLAN) have both been independently associated with alterations in mood and cognition. We aimed to determine whether dLAN would interact with intermittent hypoxia (IH), a condition characteristic of OSA, to alter the behavioral, cognitive, and affective responses. Adult male mice were housed in either standard lighting conditions (14:10-h light-dark cycle; 150 lux:0 lux) or dLAN (150 lux:5 lux). Mice were then exposed to IH (15 cycles/h, 8 h/day, FiO2 nadir of 5%) for 3 wk, then tested in assays of affective and cognitive responses; brains were collected for dendritic morphology and PCR analysis. Exposure to dLAN and IH increased anxiety-like behaviors, as assessed in the open field, elevated plus maze, and the light/dark box. dLAN and IH increased depressive-like behaviors in the forced swim test. IH impaired learning and memory performance in the passive avoidance task; however, no differences were observed in spatial working memory, as assessed by y-maze or object recognition. IH combined with dLAN decreased cell body area in the CA1 and CA3 regions of the hippocampus. Overall, IH decreased apical spine density in the CA3, whereas dLAN decreased spine density in the CA1 of the hippocampus. TNF-α gene expression was not altered by IH or lighting condition, whereas VEGF expression was increased by dLAN. The combination of IH and dLAN provokes negative effects on hippocampal dendritic morphology, affect, and cognition, suggesting that limiting nighttime exposure to light in combination with other established treatments may be of benefit to patients with OSA.
Collapse
Affiliation(s)
- Taryn G Aubrecht
- Department of Neuroscience and Institute of Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|
46
|
Toth LA, Bhargava P. Animal models of sleep disorders. Comp Med 2013; 63:91-104. [PMID: 23582416 PMCID: PMC3625050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 10/21/2012] [Accepted: 11/25/2012] [Indexed: 06/02/2023]
Abstract
Problems with sleep affect a large part of the general population, with more than half of all people in the United States reporting difficulties with sleep or insufficient sleep at various times and about 40 million affected chronically. Sleep is a complex physiologic process that is influenced by many internal and environmental factors, and problems with sleep are often related to specific personal circumstances or are based on subjective reports from the affected person. Although human subjects are used widely in the study of sleep and sleep disorders, the study of animals has been invaluable in developing our understanding about the physiology of sleep and the underlying mechanisms of sleep disorders. Historically, the use of animals for the study of sleep disorders has arguably been most fruitful for the condition of narcolepsy, in which studies of dogs and mice revealed previously unsuspected mechanisms for this condition. The current overview considers animal models that have been used to study 4 of the most common human sleep disorders-insomnia, narcolepsy, restless legs syndrome, and sleep apnea-and summarizes considerations relevant to the use of animals for the study of sleep and sleep disorders. Animal-based research has been vital to the elucidation of mechanisms that underlie sleep, its regulation, and its disorders and undoubtedly will remain crucial for discovering and validating sleep mechanisms and testing interventions for sleep disorders.
Collapse
Affiliation(s)
- Linda A Toth
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA.
| | | |
Collapse
|
47
|
Quintessential Risk Factors: Their Role in Promoting Cognitive Dysfunction and Alzheimer’s Disease. Neurochem Res 2012; 37:2627-58. [DOI: 10.1007/s11064-012-0854-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 07/21/2012] [Indexed: 12/13/2022]
|
48
|
Yang SR, Sun H, Huang ZL, Yao MH, Qu WM. Repeated sleep restriction in adolescent rats altered sleep patterns and impaired spatial learning/memory ability. Sleep 2012; 35:849-59. [PMID: 22654204 DOI: 10.5665/sleep.1888] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES To investigate possible differences in the effect of repeated sleep restriction (RSR) during adolescence and adulthood on sleep homeostasis and spatial learning and memory ability. DESIGN The authors examined electroencephalograms of rats as they were subjected to 4-h daily sleep deprivation that continued for 7 consecutive days and assessed the spatial learning and memory by Morris water maze test (WMT). PARTICIPANTS Adolescent and adult rats. MEASUREMENTS AND RESULTS Adolescent rats exhibited a similar amount of rapid eye movement (REM) and nonrapid eye movement (NREM) sleep with higher slow wave activity (SWA, 0.5-4 Hz) and fewer episodes and conversions with prolonged durations, indicating they have better sleep quality than adult rats. After RSR, adult rats showed strong rebound of REM sleep by 31% on sleep deprivation day 1; this value was 37% on sleep deprivation day 7 in adolescents compared with 20-h baseline level. On sleep deprivation day 7, SWA in adult and adolescent rats increased by 47% and 33%, and such elevation lasted for 5 h and 7 h, respectively. Furthermore, the authors investigated the effects of 4-h daily sleep deprivation immediately after the water maze training sessions on spatial cognitive performance. Adolescent rats sleep-restricted for 7 days traveled a longer distance to find the hidden platform during the acquisition training and had fewer numbers of platform crossings in the probe trial than those in the control group, something that did not occur in the sleep-deprived adult rats. CONCLUSIONS Repeated sleep restriction (RSR) altered sleep profiles and mildly impaired spatial learning and memory capability in adolescent rats.
Collapse
Affiliation(s)
- Su-Rong Yang
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | | | | | | | | |
Collapse
|
49
|
Disrupted sleep without sleep curtailment induces sleepiness and cognitive dysfunction via the tumor necrosis factor-α pathway. J Neuroinflammation 2012; 9:91. [PMID: 22578011 PMCID: PMC3411474 DOI: 10.1186/1742-2094-9-91] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 05/11/2012] [Indexed: 12/14/2022] Open
Abstract
Background Sleepiness and cognitive dysfunction are recognized as prominent consequences of sleep deprivation. Experimentally induced short-term sleep fragmentation, even in the absence of any reductions in total sleep duration, will lead to the emergence of excessive daytime sleepiness and cognitive impairments in humans. Tumor necrosis factor (TNF)-α has important regulatory effects on sleep, and seems to play a role in the occurrence of excessive daytime sleepiness in children who have disrupted sleep as a result of obstructive sleep apnea, a condition associated with prominent sleep fragmentation. The aim of this study was to examine role of the TNF-α pathway after long-term sleep fragmentation in mice. Methods The effect of chronic sleep fragmentation during the sleep-predominant period on sleep architecture, sleep latency, cognitive function, behavior, and inflammatory markers was assessed in C57BL/6 J and in mice lacking the TNF-α receptor (double knockout mice). In addition, we also assessed the above parameters in C57BL/6 J mice after injection of a TNF-α neutralizing antibody. Results Mice subjected to chronic sleep fragmentation had preserved sleep duration, sleep state distribution, and cumulative delta frequency power, but also exhibited excessive sleepiness, altered cognitive abilities and mood correlates, reduced cyclic AMP response element-binding protein phosphorylation and transcriptional activity, and increased phosphodiesterase-4 expression, in the absence of AMP kinase-α phosphorylation and ATP changes. Selective increases in cortical expression of TNF-α primarily circumscribed to neurons emerged. Consequently, sleepiness and cognitive dysfunction were absent in TNF-α double receptor knockout mice subjected to sleep fragmentation, and similarly, treatment with a TNF-α neutralizing antibody abrogated sleep fragmentation-induced learning deficits and increases in sleep propensity. Conclusions Taken together, our findings show that recurrent arousals during sleep, as happens during sleep apnea, induce excessive sleepiness via activation of inflammatory mechanisms, and more specifically TNF-α-dependent pathways, despite preserved sleep duration.
Collapse
|
50
|
Pathogenesis of cognitive dysfunction in patients with obstructive sleep apnea: a hypothesis with emphasis on the nucleus tractus solitarius. SLEEP DISORDERS 2012; 2012:251096. [PMID: 23470865 PMCID: PMC3581091 DOI: 10.1155/2012/251096] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 12/13/2011] [Accepted: 12/22/2011] [Indexed: 02/06/2023]
Abstract
OSA is characterized by the quintessential triad of intermittent apnea, hypoxia, and hypoxemia due to pharyngeal collapse. This paper highlights the upstream mechanisms that may trigger cognitive decline in OSA. Three interrelated steps underpin cognitive dysfunction in OSA patients. First, several risk factors upregulate peripheral inflammation; these crucial factors promote neuroinflammation, cerebrovascular endothelial dysfunction, and oxidative stress in OSA. Secondly, the neuroinflammation exerts negative impact globally on the CNS, and thirdly, important foci in the neocortex and brainstem are rendered inflamed and dysfunctional. A strong link is known to exist between neuroinflammation and neurodegeneration. A unique perspective delineated here underscores the importance of dysfunctional brainstem nuclei in etiopathogenesis of cognitive decline in OSA patients. Nucleus tractus solitarius (NTS) is the central integration hub for afferents from upper airway (somatosensory/gustatory), respiratory, gastrointestinal, cardiovascular (baroreceptor and chemoreceptor) and other systems. The NTS has an essential role in sympathetic and parasympathetic systems also; it projects to most key brain regions and modulates numerous physiological functions. Inflamed and dysfunctional NTS and other key brainstem nuclei may play a pivotal role in triggering memory and cognitive dysfunction in OSA. Attenuation of upstream factors and amelioration of the NTS dysfunction remain important challenges.
Collapse
|