1
|
Zhang X, Xu H, Yin S, Gozal D, Khalyfa A. Obstructive sleep apnea and memory impairments: Clinical characterization, treatment strategies, and mechanisms. Sleep Med Rev 2025; 81:102092. [PMID: 40286536 DOI: 10.1016/j.smrv.2025.102092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Obstructive sleep apnea (OSA), is associated with dysfunction in the cardiovascular, metabolic and neurological systems. However, the relationship between OSA and memory impairment, intervention effects, and underlying pathways are not well understood. This review summarizes recent advances in the clinical characterization, treatment strategies, and mechanisms of OSA-induced memory impairments. OSA patients may exhibit significant memory declines, including impairments in working memory from visual and verbal sources. The underlying mechanisms behind OSA-related memory impairment are complex and multifactorial with poorly understood aspects that require further investigation. Neuroinflammation, oxidative stress, neuronal damage, synaptic plasticity, and blood-brain barrier dysfunction, as observed under exposures to intermittent hypoxia and sleep fragmentation are likely contributors to learning and memory dysfunction. Continuous positive airway pressure treatment can provide remarkable relief from memory impairment in OSA patients. Other treatments are emerging but need to be rigorously evaluated for cognitive improvement. Clinically, reliable and objective diagnostic tools are necessary for accurate diagnosis and clinical characterization of cognitive impairments in OSA patients. The complex links between gut-brain axis, epigenetic landscape, genetic susceptibility, and OSA-induced memory impairments suggest new directions for research. Characterization of clinical phenotypic clusters can facilitate advances in precision medicine to predict and treat OSA-related memory deficits.
Collapse
Affiliation(s)
- Xiaoman Zhang
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Huajun Xu
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shankai Yin
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - David Gozal
- Department of Pediatrics and Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Abdelnaby Khalyfa
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
2
|
Liu S, Liu X, Ke M, Wang J. Sleep fragmentation impairs cognitive function and exacerbates Alzheimer's disease-related pathology in a mouse model by disrupting mitochondrial biogenesis. Exp Neurol 2025; 386:115153. [PMID: 39832661 DOI: 10.1016/j.expneurol.2025.115153] [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: 09/25/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
A large proportion of Alzheimer's disease (AD) patients suffer from various types of chronic sleep disturbances, including sleep fragmentation (SF). In addition, impaired mitochondrial biogenesis is an important feature of AD, but whether it is altered in sleep disorders has not been fully elucidated. Hence, we aimed to investigate the relationship between SF and mitochondrial biogenesis and the possible impact of SF on AD-related pathology. In this study, thirty-six 9-month-old 3xTgAD model mice and thirty-six 9-month-old wild-type (WT) C57BL/6 J mice were divided into a control group (6 weeks of normal sleep), a SF group (6 weeks of SF) and a SF + recovery sleep group (6 weeks of SF followed by 2 weeks of recovery sleep). Cognitive functions were assessed by behavioural experiments. Mitochondrial structure and function and the activity of a classic mitochondrial biogenesis signalling pathway were investigated using transmission electron microscopy (TEM), reverse transcription quantitative polymerase chain reaction (RT-qPCR), immunofluorescence and Western blotting. Markers of AD-related pathology, including the levels of amyloid β (Aβ) and tau proteins, were assessed by immunofluorescence and Western blotting. The expression of insulin-degrading enzyme (IDE) was assessed by Western blotting. We found that long-term SF impaired the cognitive functions of the mice. In addition, chronic SF reduced the expression of mitochondrial respiratory chain components, the number of mitochondria, the fluorescence intensity of COX-IV, the level of mitochondrial DNA (mtDNA) and the expression of crucial regulators of the AMPK/SIRT-1/PGC-1α signalling pathway in the mouse prefrontal cortex and hippocampus, while recovery sleep could partly abrogate these effects. Moreover, SF reduced the protein level of IDE and increased the Aβ burden and tau hyperphosphorylation. This study demonstrates that chronic SF can negatively regulate the AMPK/SIRT-1/PGC-1α signalling pathway to disrupt mitochondrial biogenesis in the brains of mice, which may subsequently exacerbate AD-related pathology by decreasing the expression of IDE.
Collapse
Affiliation(s)
- Shunjie Liu
- Department of Neurology, The Second People's Hospital of Foshan, Foshan 528000, Guangdong Province, China
| | - Xingyi Liu
- Department of Neurology, The Second People's Hospital of Foshan, Foshan 528000, Guangdong Province, China
| | - Man Ke
- Department of Neurology, The Second People's Hospital of Foshan, Foshan 528000, Guangdong Province, China
| | - Jinliang Wang
- Department of Neurology, The Second People's Hospital of Foshan, Foshan 528000, Guangdong Province, China.
| |
Collapse
|
3
|
Puech C, Badran M, Barrow MB, Gozal D. Cognitive Function, Sleep, and Neuroinflammatory Markers in Mice Exposed to Very Long-Term Intermittent Hypoxia. Int J Mol Sci 2025; 26:1815. [PMID: 40076441 PMCID: PMC11899729 DOI: 10.3390/ijms26051815] [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: 12/13/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 03/14/2025] Open
Abstract
Chronic intermittent hypoxia (IH) is one of the hallmark features of obstructive sleep apnea (OSA) and adversely affects neurocognitive and behavioral functioning. However, how the duration of IH correlates with its deleterious effects remains unexplored. We aimed to assess the effects of IH over a prolonged period of time mimicking untreated OSA. Male C57Bl/6J mice were exposed to IH for 96 weeks. Sleep activity was acquired using a piezoelectric system. Novel object recognition (NOR) and the elevated plus maze test (EPMT) were conducted as measures of cognitive function and anxiety, respectively. Brain inflammation was evaluated by a panel of inflammation marker assays. All tests were performed after 16 and 96 weeks of IH exposure. After 96 weeks, sleep percentages during the dark phase decreased in both IH and room air (RA) compared to 16-week exposure (RA: p = 0.0214; IH: p = 0.0188). In addition to age-dependent declines in NOR performance, the mice after 96 weeks of IH exposure had lower NOR preference scores than RA controls (p = 0.0070). The time spent in open arms of the EPMT was reduced in mice exposed to IH compared to RA. Inflammatory marker expression increased in IH-exposed mice. Thus, aging and IH induce similar alterations in sleep, cognition, and neuroinflammation. However, the effects of aging are exacerbated by concurrent IH, suggesting that OSA is a disease associated with an acceleration in biological aging.
Collapse
Affiliation(s)
- Clementine Puech
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65201, USA; (C.P.); (M.B.); (M.B.B.)
| | - Mohammad Badran
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65201, USA; (C.P.); (M.B.); (M.B.B.)
| | - Max B. Barrow
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65201, USA; (C.P.); (M.B.); (M.B.B.)
| | - David Gozal
- Department of Pediatrics and Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| |
Collapse
|
4
|
Balsamo F, Meneo D, Berretta E, Baglioni C, Gelfo F. Could sleep be a brain/cognitive/neural reserve-builder factor? A systematic review on the cognitive effects of sleep modulation in animal models. Neurosci Biobehav Rev 2025; 169:106015. [PMID: 39828234 DOI: 10.1016/j.neubiorev.2025.106015] [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: 08/02/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
The brain/cognitive/neural reserve concept suggests that lifelong experiences, from early life through adulthood, make the brain more resilient to neuronal damage. Modifiable lifestyle factors, such as sleep, can support the development and enhance such a reserve, helping to counteract age- or disease-related brain changes and their impact on cognition. Sleep plays a crucial role in cognitive functioning, and disruptions or disorders may increase neurodegenerative risks. This systematic review aims to explore how functional and disturbed sleep impacts cognitive functions and neuromorphological mechanisms in rodents, aiming to better understand its role in brain/cognitive/neural reserve development. This systematic review, registered on PROSPERO (ID: CRD42023423901) and conducted according to PRISMA-P guidelines, searched PubMed, Scopus, Web of Science, and Embase databases for studies up to June 2022, with terms related to sleep, rodents, and cognitive functions. Of the 28,666 articles identified, 142 met the inclusion criteria. Main results showed significant cognitive decline after sleep deprivation, especially in memory performance. These findings supports the importance of sleep as a critical factor in modulating brain/cognitive/neural reserve.
Collapse
Affiliation(s)
- Francesca Balsamo
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy; IRCCS Fondazione Santa Lucia, Rome 00179, Italy.
| | - Debora Meneo
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy
| | | | - Chiara Baglioni
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy; Department of Clinical Psychology and Psychophysiology/Sleep, Medicine, Centre for Mental Disorders, University Medical Centre, Freiburg, Germany
| | - Francesca Gelfo
- Department of Human Sciences, Guglielmo Marconi University, Rome 00193, Italy; IRCCS Fondazione Santa Lucia, Rome 00179, Italy.
| |
Collapse
|
5
|
Chang HHV, Fesshaye AS, Tidmore A, Sanford LD, Britten RA. Sleep Fragmentation Results in Novel Set-shifting Decrements in GCR-exposed Male and Female Rats. Radiat Res 2025; 203:18-25. [PMID: 39576067 DOI: 10.1667/rade-24-00146.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025]
Abstract
The prolonged exposure to multiple spaceflight stressors during long-duration missions to the Moon and Mars will be challenging to the physical and mental health of the astronauts. Ground-based studies have reported that attentional set-shifting task (ATSET) performance is impaired after space radiation (SR) exposure. At certain times during deep-space missions, astronauts will likely have to contend with the combined impacts of SR and sleep perturbation. In rats, poor quality, fragmented sleep adversely impacts performance in multiple cognitive tasks, including the ATSET task. While both SR and sleep perturbations independently cause cognitive performance deficits, the incidence, severity and exact nature of those decrements following combined exposure to these flight stressors is largely unknown. This study established the impact that a single night of fragmented sleep has on ATSET performance in both male and female rats exposed to 10 cGy of galactic cosmic ray simulation (GCRsim). The GCRsim beam is a complex beam that mimics the mass and energy spectra of the SR particles that an astronaut will be exposed to within the spacecraft. Rats that had no obvious ATSET performance decrements when normally rested were subjected to fragmented sleep and their ATSET performance reassessed. Sleep fragmentation resulted in significant ATSET performance decrements in GCRsim-exposed rats, with specific performance decrements being observed in stages where attention or cue shifting is extensively used. Performance decrements in these stages are rarely observed after SR exposure. While both male and female rats exhibited latent sleep-related performance decrements, these were sex dependent, with male and female rats exhibiting different types of performance decrements (either reduced processing speed or task completion efficiency) in different stages of the ATSET task. This study suggests that SR-induced cognitive impairment may not be fully evident in normally rested rats, with an underestimation of both the incidence and nature of performance decrements that could occur when multiple space flight stressors are present. These data suggest that that there may be synergistic interactions between multiple space flight stressors that may not be easily predicted from their independent actions.
Collapse
Affiliation(s)
| | | | | | - Larry D Sanford
- Biomedical and Translational Sciences
- Center for Integrative Neuroscience and Inflammatory Diseases, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, Virginia
| | - Richard A Britten
- Radiation Oncology
- Center for Integrative Neuroscience and Inflammatory Diseases, Macon and Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, Virginia
| |
Collapse
|
6
|
Tang M, Wu Y, Liang J, Yang S, Huang Z, Hu J, Yang Q, Liu F, Li S. Gut microbiota has important roles in the obstructive sleep apnea-induced inflammation and consequent neurocognitive impairment. Front Microbiol 2024; 15:1457348. [PMID: 39712898 PMCID: PMC11659646 DOI: 10.3389/fmicb.2024.1457348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
Obstructive sleep apnea (OSA) is a state of sleep disorder, characterized by repetitive episodes of apnea and chronic intermittent hypoxia. OSA has an extremely high prevalence worldwide and represents a serious challenge to public health, yet its severity is frequently underestimated. It is now well established that neurocognitive dysfunction, manifested as deficits in attention, memory, and executive functions, is a common complication observed in patients with OSA, whereas the specific pathogenesis remains poorly understood, despite the likelihood of involvement of inflammation. Here, we provide an overview of the current state of the art, demonstrating the intimacy of OSA with inflammation and cognitive impairment. Subsequently, we present the recent findings on the investigation of gut microbiota alteration in the OSA conditions, based on both patients-based clinical studies and animal models of OSA. We present an insightful discussion on the role of changes in the abundance of specific gut microbial members, including short-chain fatty acid (SCFA)-producers and/or microbes with pathogenic potential, in the pathogenesis of inflammation and further cognitive dysfunction. The transplantation of fecal microbiota from the mouse model of OSA can elicit inflammation and neurobehavioral disorders in naïve mice, thereby validating the causal relationship to inflammation and cognitive abnormality. This work calls for greater attention on OSA and the associated inflammation, which require timely and effective therapy to protect the brain from irreversible damage. This work also suggests that modification of the gut microbiota using prebiotics, probiotics or fecal microbiota transplantation may represent a potential adjuvant therapy for OSA.
Collapse
Affiliation(s)
- Mingxing Tang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Yongliang Wu
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Junyi Liang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Shuai Yang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Zuofeng Huang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Jing Hu
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Qiong Yang
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Fei Liu
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| | - Shuo Li
- Department of Otolaryngology, Shenzhen Nanshan People’s Hospital, Shenzhen, China
- Department of Otolaryngology, The 6th Affiliated Hospital, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
7
|
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
|
8
|
Sriram S, Carstens K, Dewing W, Fiacco TA. Astrocyte regulation of extracellular space parameters across the sleep-wake cycle. Front Cell Neurosci 2024; 18:1401698. [PMID: 38988660 PMCID: PMC11233815 DOI: 10.3389/fncel.2024.1401698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/07/2024] [Indexed: 07/12/2024] Open
Abstract
Multiple subfields of neuroscience research are beginning to incorporate astrocytes into current frameworks of understanding overall brain physiology, neuronal circuitry, and disease etiology that underlie sleep and sleep-related disorders. Astrocytes have emerged as a dynamic regulator of neuronal activity through control of extracellular space (ECS) volume and composition, both of which can vary dramatically during different levels of sleep and arousal. Astrocytes are also an attractive target of sleep research due to their prominent role in the glymphatic system, a method by which toxic metabolites generated during wakefulness are cleared away. In this review we assess the literature surrounding glial influences on fluctuations in ECS volume and composition across the sleep-wake cycle. We also examine mechanisms of astrocyte volume regulation in glymphatic solute clearance and their role in sleep and wake states. Overall, findings highlight the importance of astrocytes in sleep and sleep research.
Collapse
Affiliation(s)
- Sandhya Sriram
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| | - Kaira Carstens
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| | - Wayne Dewing
- Undergraduate Major in Neuroscience, University of California, Riverside, Riverside, CA, United States
| | - Todd A Fiacco
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
9
|
Fu X, Wan XJ, Liu JY, Sun Q, Shen Y, Li J, Mao CJ, Ma QH, Wang F, Liu CF. Effects of sleep fragmentation on white matter pathology in a rat model of cerebral small vessel disease. Sleep 2024; 47:zsad225. [PMID: 37638817 DOI: 10.1093/sleep/zsad225] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/10/2023] [Indexed: 08/29/2023] Open
Abstract
STUDY OBJECTIVES Mounting evidence indicated the correlation between sleep and cerebral small vessel disease (CSVD). However, little is known about the exact causality between poor sleep and white matter injury, a typical signature of CSVD, as well as the underlying mechanisms. METHODS Spontaneously hypertensive rats (SHR) and control Wistar Kyoto rats were subjected to sleep fragmentation (SF) for 16 weeks. The effects of chronic sleep disruption on the deep white matter and cognitive performance were observed. RESULTS SHR were validated as a rat model for CSVD. Fragmented sleep induced strain-dependent white matter abnormalities, characterized by reduced myelin integrity, impaired oligodendrocytes precursor cells (OPC) maturation and pro-inflammatory microglial polarization. Partially reversible phenotypes of OPC and microglia were observed in parallel following sleep recovery. CONCLUSIONS Long-term SF-induced pathological effects on the deep white matter in a rat model of CSVD. The pro-inflammatory microglial activation and the block of OPC maturation may be involved in the mechanisms linking sleep to white matter injury.
Collapse
Affiliation(s)
- Xiang Fu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xiao-Jie Wan
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jun-Yi Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Qian Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yun Shen
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Li
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Quan-Hong Ma
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
10
|
Thomas RJ. A matter of fragmentation. Sleep 2024; 47:zsae030. [PMID: 38285604 DOI: 10.1093/sleep/zsae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Indexed: 01/31/2024] Open
Affiliation(s)
- Robert Joseph Thomas
- Professor of Medicine, Harvard Medical School, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
11
|
Hong SH, Lee DB, Yoon DW, Kim J. Melatonin Improves Glucose Homeostasis and Insulin Sensitivity by Mitigating Inflammation and Activating AMPK Signaling in a Mouse Model of Sleep Fragmentation. Cells 2024; 13:470. [PMID: 38534314 PMCID: PMC10969771 DOI: 10.3390/cells13060470] [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: 02/20/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Sleep fragmentation (SF) can increase inflammation and production of reactive oxygen species (ROS), leading to metabolic dysfunction. SF is associated with inflammation of adipose tissue and insulin resistance. Several studies have suggested that melatonin may have beneficial metabolic effects due to activating AMP-activated protein kinase (AMPK). However, it is unclear whether melatonin affects the AMPK signaling pathway in SF-induced metabolic dysfunction. Therefore, we hypothesize that SF induces metabolic impairment and inflammation in white adipose tissue (WAT), as well as altered intracellular homeostasis. We further hypothesize that these conditions could be improved by melatonin treatment. We conducted an experiment using adult male C57BL/6 mice, which were divided into three groups: control, SF, and SF with melatonin treatment (SF+Mel). The SF mice were housed in SF chambers, while the SF+Mel mice received daily oral melatonin. After 12 weeks, glucose tolerance tests, insulin tolerance tests, adipose tissue inflammation tests, and AMPK assessments were performed. The SF mice showed increased weight gain, impaired glucose regulation, inflammation, and decreased AMPK in WAT compared to the controls. Melatonin significantly improved these outcomes by mitigating SF-induced metabolic dysfunction, inflammation, and AMPK downregulation in adipose tissue. The therapeutic efficacy of melatonin against cardiometabolic impairments in SF may be due to its ability to restore adipose tissue homeostatic pathways.
Collapse
Affiliation(s)
- Seok Hyun Hong
- Sleep Medicine Institute, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea; (S.H.H.); (D.-B.L.)
- Department of Biomedical Laboratory Science, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea
| | - Da-Been Lee
- Sleep Medicine Institute, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea; (S.H.H.); (D.-B.L.)
- Department of Health and Safety Convergence Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Dae-Wui Yoon
- Sleep Medicine Institute, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea; (S.H.H.); (D.-B.L.)
- Department of Biomedical Laboratory Science, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea
| | - Jinkwan Kim
- Sleep Medicine Institute, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea; (S.H.H.); (D.-B.L.)
- Department of Biomedical Laboratory Science, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
12
|
Jackson CD, Badran M, Gozal D, Brown CR, Khalyfa A. Sleep fragmentation disrupts Lyme arthritis resolution in mice. Sleep Med 2024; 114:196-202. [PMID: 38219655 DOI: 10.1016/j.sleep.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 01/16/2024]
Abstract
STUDY OBJECTIVES Lyme arthritis is a common late-stage complication of infection by Borrelia burgdorferi, the agent of Lyme disease. Patients with Lyme arthritis report increased levels of sleep disturbance associated with pain. Using a mouse model of experimental Lyme arthritis, we investigated the effect of disrupted sleep on the development and resolution of joint inflammation. METHODS Lyme arthritis-susceptible C3H/HeJ mice (n = 10/group) were infected with B. burgdorferi and were left either alone (control) or subjected to sleep fragmentation (SF). Arthritis development or resolution were monitored. The impact of SF on immune and inflammatory parameters such as arthritis severity scores, anti-borrelia antibody production, and bacterial clearance was measured. We also determined the effect of SF on arthritis resolution in C3H mice deficient in leukotriene (LT) B4 signaling (BLT1/2-/-) who display delayed Lyme arthritis resolution. RESULTS SF had no significant impact on Lyme arthritis development or inflammatory parameters regardless of whether SF treatment began 1 week prior to or congruent with infection. However, initiation of SF at the peak of arthritis resulted in a significant delay in arthritis resolution as measured by joint edema, arthritis severity scores, and decreased bacterial clearance from the joint. This was accompanied by significant changes in joint cytokine transcription levels (e.g., increased TNFα and decreased IL-4). SF has no significant impact on Lyme arthritis resolution in the BLT1/2-/- mice. CONCLUSIONS Poor sleep, especially near the peak of arthritis inflammation, may delay initiation of resolution programs possibly through altering cytokine production and host immune responses, leading to defects in spirochete clearance and prolonged disease.
Collapse
Affiliation(s)
- Christa D Jackson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| | - Mohammad Badran
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO, USA.
| | - David Gozal
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO, USA; Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA; Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA.
| | - Charles R Brown
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| | - Abdelnaby Khalyfa
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
13
|
Moriarty AK, Waseem TC, Keeter WC, Ma SD, Bai R, Ivanov AV, Kirk CL, Mussbacher M, Sanford LD, Galkina EV. Disturbed Sleep Supports Neutrophil Activation and Promotes Atherosclerosis and Plaque Necrosis. Circ Res 2023; 133:1056-1059. [PMID: 37947098 PMCID: PMC10842439 DOI: 10.1161/circresaha.123.322858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Affiliation(s)
- Alina K. Moriarty
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Tayab C. Waseem
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - W. Coles Keeter
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Shelby D. Ma
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Robin Bai
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Aleksandr V. Ivanov
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Cassandra L. Kirk
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Marion Mussbacher
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Larry D. Sanford
- Department of Anatomy and Pathology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Elena V. Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
- Center for Integrative Neuroscience and Inflammatory Diseases Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| |
Collapse
|
14
|
Candow DG, Forbes SC, Ostojic SM, Prokopidis K, Stock MS, Harmon KK, Faulkner P. "Heads Up" for Creatine Supplementation and its Potential Applications for Brain Health and Function. Sports Med 2023; 53:49-65. [PMID: 37368234 PMCID: PMC10721691 DOI: 10.1007/s40279-023-01870-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2023] [Indexed: 06/28/2023]
Abstract
There is emerging interest regarding the potential beneficial effects of creatine supplementation on indices of brain health and function. Creatine supplementation can increase brain creatine stores, which may help explain some of the positive effects on measures of cognition and memory, especially in aging adults or during times of metabolic stress (i.e., sleep deprivation). Furthermore, creatine has shown promise for improving health outcome measures associated with muscular dystrophy, traumatic brain injury (including concussions in children), depression, and anxiety. However, whether any sex- or age-related differences exist in regard to creatine and indices of brain health and function is relatively unknown. The purpose of this narrative review is to: (1) provide an up-to-date summary and discussion of the current body of research focusing on creatine and indices of brain health and function and (2) discuss possible sex- and age-related differences in response to creatine supplementation on brain bioenergetics, measures of brain health and function, and neurological diseases.
Collapse
Affiliation(s)
- Darren G Candow
- Aging Muscle & Bone Health Laboratory, Faculty of Kinesiology & Health Studies, University of Regina, 3737 Wascana Parkway, Regina, SK, S4S 0A2, Canada.
| | - Scott C Forbes
- Department of Physical Education Studies, Brandon University, Brandon, MB, Canada
| | - Sergej M Ostojic
- Department of Nutrition and Public Health, University of Agder, Kristiansand, Norway
| | | | - Matt S Stock
- School of Kinesiology and Rehabilitation Sciences, University of Central Florida, Orlando, FL, USA
| | - Kylie K Harmon
- Department of Exercise Science, Syracuse University, New York, NY, USA
| | - Paul Faulkner
- Department of Psychology, University of Roehampton, London, UK
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
Cognitive and Brain Gray Matter Changes in Children with Obstructive Sleep Apnea: A Voxel-Based Morphological Study. Neuropediatrics 2023; 54:139-146. [PMID: 36473490 DOI: 10.1055/a-1993-3985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND To explore the neural difference between children with obstructive sleep apnea (OSA) and healthy controls, together with the relation between this difference and clinical severity indicator of children with OSA. METHODS Twenty-seven children with OSA (7.6 ± 2.5 years, apnea hypopnea index [AHI]: 9.7 ± 5.3 events/h) and 30 healthy controls (7.8 ± 2.6 years, AHI: 1.7 ± 1.2 events/h) were recruited and matched with age, gender, and handedness. All children underwent 3.0 T magnetic resonance imaging of the brain and cognitive testing evaluating. Volumetric segmentation of cortical and subcortical structures and voxel-based morphometry were performed. Pearson's correlation analysis was performed between these features of gray matter volume (GMV) and obstructive apnea index (OAI) among children with OSA. RESULTS In the comparison of children's Wechsler test scores of full-scale intelligence quotient and verbal intelligence quotient, the OSA group was significantly lower than the control group (p < 0.05). Compared with the control group, the GMV of many brain regions in the OSA group was significantly decreased (p < 0.05). In the correlation analysis of GMV and OAI in OSA group, right inferior frontal gyrus volume was significantly negatively correlated with OAI (r = - 0.49, p = 0.02). CONCLUSION Children with OSA presented abnormal neural activities in some brain regions and impaired cognitive functions. This finding suggests an association between the OSA and decreased GMV in children.
Collapse
|
19
|
Yang DF, Huang WC, Wu CW, Huang CY, Yang YCSH, Tung YT. Acute sleep deprivation exacerbates systemic inflammation and psychiatry disorders through gut microbiota dysbiosis and disruption of circadian rhythms. Microbiol Res 2023; 268:127292. [PMID: 36608535 DOI: 10.1016/j.micres.2022.127292] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/23/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Acute sleep deprivation (ASD) is often observed in shift workers and characterized by drowsiness and unrelenting exhaustion. The physiological and psychological effects of ASD include anxiety, depression, cognitive impairment, systemic inflammation, stress responses, and disruptions of gut microbiota. However, the mechanisms involved in the ASD-associated circadian dysregulations with regard to gut dysbiosis, systemic inflammation, physiological modulation, and psychiatry disorders remain unclear. The aim of this study was to investigate whether central nervous system disorders induced by ASD are related to inflammation, barrier dysfunction, and circadian dysregulation. We also assessed impacts on microbiota succession. Male C57BL/6 mice were randomly allocated to the control and sleep deprivation (SD) groups. Mice in the SD group were subjected to 72 h of paradoxical SD using the modified multiple-platform method for ASD induction (72 h rapid eye movement-SD). The effects of ASD on dietary consumption, behaviors, cytokines, microbiota, and functional genes were determined. The appetite of the SD group was significantly higher than that of the control group, but the body weight was significantly lower than that of the control group. The anxiety-like behaviors were found in the SD group. Alpha and beta diversity of microbiota showed significant decrease after ASD induction; the relative abundance of Candidatus_Arthromitus and Enterobacter was increased, whereas that abundance of Lactobacillus, Muribaculum, Monoglobus, Parasutterella, and others was decreased in the SD group. These effects were accompanied by reduction in fecal propionic acid. In the proximal colon, the SD group exhibited significantly higher inflammation (tumor necrosis factor-α [TNF-α]) and dysregulation of the circadian rhythms (brain and muscle ARNT-like 1 [BMAL1] and cryptochrome circadian regulator 1 [CRY1]) and tight junction genes (occludin [OCLN]) than the control group. Gut barrier dysfunction slightly increased the plasma concentration of lipopolysaccharide and significantly elevated TNF-α. Inflammatory signals might be transduced through the brain via TNF receptor superfamily member 1 A (TNFRSF1A), which significantly increased the levels of microglia activation marker (ionized calcium-binding adapter molecule 1 [IBA1]) and chemokine (intercellular adhesion molecule 1 [ICAM1]) in the cerebral cortex. The serotonin receptor (5-hydroxytryptamine 1A receptor [5-HT1AR]) was significantly downregulated in the hippocampus. In summary, 72 h of rapid eye movement-SD induced physiological and psychological stress, which led to disruption of the circadian rhythms and gut microbiota dysbiosis; these effects were related to decrement of short chain fatty acids, gut inflammation, and hyperpermeability. The microbiota may be utilized as preventive and therapeutic strategies for ASD from the perspectives of medicine and nutrition.
Collapse
Affiliation(s)
- Deng-Fa Yang
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110, Taiwan.
| | - Wen-Ching Huang
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 112, Taiwan.
| | - Changwei W Wu
- Graduate Institute of Mind, Brain and Consciousness, Taipei Medical University, Taipei 110, Taiwan; Brain and Consciousness Research Center, Shuang Ho Hospital-Taipei Medical University, New Taipei 235, Taiwan.
| | - Ching-Ying Huang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan.
| | - Yu-Chen S H Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 110, Taiwan.
| | - Yu-Tang Tung
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan.
| |
Collapse
|
20
|
Sleep Fragmentation Accelerates Carcinogenesis in a Chemical-Induced Colon Cancer Model. Int J Mol Sci 2023; 24:ijms24054547. [PMID: 36901981 PMCID: PMC10003038 DOI: 10.3390/ijms24054547] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Aims of this study were to test whether sleep fragmentation (SF) increased carcinogenesis and to investigate the possible mechanisms of carcinogenesis in a chemical-induced colon cancer model. In this study, eight-week-old C57BL/6 mice were divided into Home cage (HC) and SF groups. After the azoxymethane (AOM) injection, the mice in the SF group were subjected to SF for 77 days. SF was accomplished in a sleep fragmentation chamber. In the second protocol, mice were divided into 2% dextran sodium sulfate (DSS)-treated, HC, and SF groups and were exposed to the HC or SF procedures. Immunohistochemical and immunofluorescent stainings were conducted to determine the level of 8-OHdG and reactive oxygen species (ROS), respectively. Quantitative real-time polymerase chain reaction was used to assess the relative expression of inflammatory and ROS-generating genes. The number of tumors and average tumor size were significantly higher in the SF group than in the HC group. The intensity (%) of the 8-OHdG stained area was significantly higher in the SF group than in the HC group. The fluorescence intensity of ROS was significantly higher in the SF group than the HC group. SF accelerated cancer development in a murine AOM/DSS-induced model of colon cancer, and the increased carcinogenesis was associated with ROS- and oxidative stress-induced DNA damage.
Collapse
|
21
|
Blackwell AA, Tracz JA, Fesshaye AS, Tidmore A, Osterlund Oltmanns JR, Schaeffer EA, Lake RI, Wallace DG, Britten RA. Fine motor deficits exhibited in rat string-pulling behavior following exposure to sleep fragmentation and deep space radiation. Exp Brain Res 2023; 241:427-440. [PMID: 36574036 DOI: 10.1007/s00221-022-06527-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/06/2022] [Indexed: 12/28/2022]
Abstract
Deep space flight missions will expose astronauts to multiple stressors, including sleep fragmentation and space radiation. There is debate over whether sleep disruptions are an issue in deep space. While these stressors independently impair sensorimotor function, the combined effects on performance are currently unknown. String-pulling behavior involves highly organized bimanual reach-to-grasp and withdraw movements. This behavior was examined under rested wakeful conditions and immediately following one session of sleep fragmentation in Sham and irradiated rats 3 months after exposure (10 cGy 4Helium or 5-ion simulated Galactic Cosmic Radiation). Sleep fragmentation disrupted several aspects of string-pulling behavior, such that rats' ability to grasp the string was reduced, reach endpoint concentration was more variable, and distance traveled by the nose increased in the Y-range compared to rested wakeful performance. Overall, irradiated rats missed the string more than Sham rats 3 months post-exposure. Irradiated rats also exhibited differential impairments at 3 months, with additional deficits unveiled after sleep fragmentation. 4Helium-exposed rats took longer to approach the string after sleep fragmentation. Further, rats exposed to 4Helium traveled shorter withdraw distances 3 months after irradiation, while this only emerged in the other irradiated group after sleep fragmentation. These findings identify sleep fragmentation as a risk for fine motor dysfunction in Sham and irradiated conditions, in addition to radiation exposure. There may be complex temporal alterations in performance that are stressor- and ion-dependent. Thus, it is critical to implement appropriate models of multi-flight stressors and performance assessments in preparation for future deep space flight missions.
Collapse
Affiliation(s)
- Ashley A Blackwell
- Department of Radiation Oncology, Eastern Virginia Medical School, 700 W. Olney Rd., Lewis Hall, Norfolk, VA, 23507, USA. .,Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA.
| | - Jovanna A Tracz
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Arriyam S Fesshaye
- Department of Radiation Oncology, Eastern Virginia Medical School, 700 W. Olney Rd., Lewis Hall, Norfolk, VA, 23507, USA
| | - Alyssa Tidmore
- Department of Radiation Oncology, Eastern Virginia Medical School, 700 W. Olney Rd., Lewis Hall, Norfolk, VA, 23507, USA
| | | | - Ericka A Schaeffer
- Department of Psychology, Northern Illinois University, DeKalb, IL, 60115, USA
| | - Rami I Lake
- Department of Psychology, Northern Illinois University, DeKalb, IL, 60115, USA
| | - Douglas G Wallace
- Department of Psychology, Northern Illinois University, DeKalb, IL, 60115, USA
| | - Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, 700 W. Olney Rd., Lewis Hall, Norfolk, VA, 23507, USA.,Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA, 23507, USA.,Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| |
Collapse
|
22
|
Gabryelska A, Turkiewicz S, Ditmer M, Sochal M. Neurotrophins in the Neuropathophysiology, Course, and Complications of Obstructive Sleep Apnea-A Narrative Review. Int J Mol Sci 2023; 24:1808. [PMID: 36768132 PMCID: PMC9916304 DOI: 10.3390/ijms24031808] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/04/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a disorder characterized by chronic intermittent hypoxia and sleep fragmentation due to recurring airway collapse during sleep. It is highly prevalent in modern societies, and due to its pleiotropic influence on the organism and numerous sequelae, it burdens patients and physicians. Neurotrophins (NTs), proteins that modulate the functioning and development of the central nervous system, such as brain-derived neurotrophic factor (BDNF), have been associated with OSA, primarily due to their probable involvement in offsetting the decline in cognitive functions which accompanies OSA. However, NTs influence multiple aspects of biological functioning, such as immunity. Thus, extensive evaluation of their role in OSA might enlighten the mechanism behind some of its elusive features, such as the increased risk of developing an immune-mediated disease or the association of OSA with cardiovascular diseases. In this review, we examine the interactions between NTs and OSA and discuss their contribution to OSA pathophysiology, complications, as well as comorbidities.
Collapse
Affiliation(s)
- Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland
| | | | | | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland
| |
Collapse
|
23
|
Tan S, Gao H, Sun J, Li N, Zhang Y, Yang L, Wang M, Wang Q, Zhai Q. CD33/TREM2 Signaling Mediates Sleep Deprivation-Induced Memory Impairment by Regulating Microglial Phagocytosis. Neuromolecular Med 2023:10.1007/s12017-023-08733-6. [PMID: 36639554 DOI: 10.1007/s12017-023-08733-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023]
Abstract
Sleep deprivation causes significant memory impairment in healthy adults. Extensive research has focused on identifying the biological mechanisms underlying memory impairment. Microglia-mediated synaptic elimination plays an indispensable role in sleep deprivation. Here, the potential role of the CD33/TREM2 signaling pathway in modulating memory decline during chronic sleep restriction (CSR) was evaluated. In this study, adult male C57BL/6 mice were sleep-restricted using an automated sleep deprivation apparatus for 20 h per day for 7 days. The Y-maze test revealed that spontaneous alternation was significantly reduced in CSR mice compared with control mice. The percentage of exploratory preference for the novel object in CSR mice was significantly decreased compared with that in control mice. These memory deficits correlated with aberrant microglial activation and increased phagocytic ability. Moreover, in CSR mice, the CD33 protein level in hippocampal tissue was significantly downregulated, but the TREM2 protein level was increased. In BV2 microglial cells, downregulation of CD33 increased TREM2 expression and improved microglial phagocytosis. Then, the sialic ligand monosialo-ganglioside 1 (GM1, 20 mg/kg, i.p.) was administered to mice once a day during CSR. Our results further showed that GM1 activated CD33 and consequently disturbed TREM2-mediated microglial phagocytosis. Finally, GM1 reversed CSR-induced synaptic loss and memory impairment via the CD33/TREM2 signaling pathway in the CA1 region of the hippocampus. This study provides novel evidence that activating CD33 and/or inhibiting TREM2 activity represent potential therapies for sleep loss-induced memory deficits through the modulation of microglial phagocytosis.
Collapse
Affiliation(s)
- Shuwen Tan
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hui Gao
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jianyu Sun
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Na Li
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yuxin Zhang
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Liu Yang
- Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Min Wang
- Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Qian Zhai
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
24
|
Yao CA, Chen IL, Chen CY, Torng PL, Su TC. Association between Wakeup Frequency at Night and Atherogenic Dyslipidemia: Evidence for Sex Differences. J Atheroscler Thromb 2023; 30:87-99. [PMID: 35444101 PMCID: PMC9899702 DOI: 10.5551/jat.63254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM This study aimed to determine whether sleep disturbance, defined as the wakeup frequency at night, is associated with atherogenic dyslipidemia and to explore possible sex differences. METHODS A total of 1,368 adults aged 19-70 years were included in the study of lifestyles and atherogenic dyslipidemia at the National Taiwan University Hospital in the period of 2008-2012. They completed a questionnaire regarding lifestyle information and sleep quality, including sleep hour duration, use of sleeping pills, and wakeup frequency during nighttime sleep. The measured lipid profiles included total cholesterol, triglycerides, low- and high-density lipoprotein cholesterol (LDL-C and HDL-C, respectively), non-HDL-C, and small dense LDL-C (sdLDL-C). Multivariate logistic regression was performed to determine habitual interrupted sleep and the odds ratio of atherogenic dyslipidemia following adjustment for conventional risk factors and for sex-based subgroup analysis. RESULTS A wakeup frequency ≥ 3 times per night was independently associated with an increased risk [odds ratio (95% confidence interval)] of dyslipidemia was 1.96 (1.17-3.28), and non-HDL-C ≥ 160 mg/dL was 1.78 (1.09-2.89). A higher wakeup frequency was associated with increased atherogenic dyslipidemia in women than in men. The multivariate adjusted relative risks for non-HDL ≥ 160 mg/dL and cholesterol ≥ 200 mg/dL were 3.05 (1.27-7.34) and 4.01(1.29-12.45) for female individuals with insomnia and those with a wakeup frequency ≥ 2 times per night, respectively. CONCLUSION A higher wakeup frequency was associated with atherogenic dyslipidemia in Taiwanese adults, particularly in women. This study also provided another evidence of increasing cardiovascular diseases in subjects with habitual interrupted sleep.
Collapse
Affiliation(s)
- Chien-An Yao
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - I-Ling Chen
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Yen Chen
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei, Taiwan,Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Pao-Ling Torng
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ta-Chen Su
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei, Taiwan,Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan,Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
25
|
Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Bryukhovetskiy I, Manzhulo I, Wiklund L, Sharma HS. Nanowired Delivery of Cerebrolysin Together with Antibodies to Amyloid Beta Peptide, Phosphorylated Tau, and Tumor Necrosis Factor Alpha Induces Superior Neuroprotection in Alzheimer's Disease Brain Pathology Exacerbated by Sleep Deprivation. ADVANCES IN NEUROBIOLOGY 2023; 32:3-53. [PMID: 37480458 DOI: 10.1007/978-3-031-32997-5_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Sleep deprivation induces amyloid beta peptide and phosphorylated tau deposits in the brain and cerebrospinal fluid together with altered serotonin metabolism. Thus, it is likely that sleep deprivation is one of the predisposing factors in precipitating Alzheimer's disease (AD) brain pathology. Our previous studies indicate significant brain pathology following sleep deprivation or AD. Keeping these views in consideration in this review, nanodelivery of monoclonal antibodies to amyloid beta peptide (AβP), phosphorylated tau (p-tau), and tumor necrosis factor alpha (TNF-α) in sleep deprivation-induced AD is discussed based on our own investigations. Our results suggest that nanowired delivery of monoclonal antibodies to AβP with p-tau and TNF-α induces superior neuroprotection in AD caused by sleep deprivation, not reported earlier.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei Province, China
| | - Dafin F Muresanu
- Department Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Department Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - José Vicente Lafuente
- LaNCE, Department Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
26
|
Sleep-Disturbance-Induced Microglial Activation Involves CRH-Mediated Galectin 3 and Autophagy Dysregulation. Cells 2022; 12:cells12010160. [PMID: 36611953 PMCID: PMC9818437 DOI: 10.3390/cells12010160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Chronic sleep disturbances (CSDs) including insomnia, insufficient sleep time, and poor sleep quality are major public health concerns around the world, especially in developed countries. CSDs are major health risk factors linked to multiple neurodegenerative and neuropsychological diseases. It has been suggested that CSDs could activate microglia (Mg) leading to increased neuroinflammation levels, which ultimately lead to neuronal dysfunction. However, the detailed mechanisms underlying CSD-mediated microglial activation remain mostly unexplored. In this study, we used mice with three-weeks of sleep fragmentation (SF) to explore the underlying pathways responsible for Mg activation. Our results revealed that SF activates Mg in the hippocampus (HP) but not in the striatum and prefrontal cortex (PFc). SF increased the levels of corticotropin-releasing hormone (CRH) in the HP. In vitro mechanism studies revealed that CRH activation of Mg involves galectin 3 (Gal3) upregulation and autophagy dysregulation. CRH could disrupt lysosome membrane integrity resulting in lysosomal cathepsins leakage. CRHR2 blockage mitigated CRH-mediated effects on microglia in vitro. SF mice also show increased Gal3 levels and autophagy dysregulation in the HP compared to controls. Taken together, our results show that SF-mediated hippocampal Mg activation involves CRH mediated galectin 3 and autophagy dysregulation. These findings suggest that targeting the hippocampal CRH system might be a novel therapeutic approach to ameliorate CSD-mediated neuroinflammation and neurodegenerative diseases.
Collapse
|
27
|
Pain and Analgesic related insomnia. Pain Manag Nurs 2022; 24:254-264. [DOI: 10.1016/j.pmn.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/11/2022] [Accepted: 11/15/2022] [Indexed: 12/23/2022]
|
28
|
Mishra I, Pullum KB, Eads KN, Strunjas AR, Ashley NT. Peripheral Sympathectomy Alters Neuroinflammatory and Microglial Responses to Sleep Fragmentation in Female Mice. Neuroscience 2022; 505:111-124. [PMID: 36240943 PMCID: PMC9671838 DOI: 10.1016/j.neuroscience.2022.09.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/05/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
Sleep loss, either induced by obstructive sleep apnea or other forms of sleep dysfunction, induces an inflammatory response, as commonly measured by increased circulating levels of pro-inflammatory cytokines. Increased catecholamines from sympathetic nervous system (SNS) activation regulates this peripheral inflammation. However, the role that catecholamines play in mediating neuroinflammation from sleep perturbations is undescribed. The aims of this study were to determine (i) the effect of peripheral SNS inhibition upon neuroinflammatory responses to sleep fragmentation (SF) and (ii) whether homeostasis can be restored after 1 week of recovery sleep. We measured gene expression levels of pro- and anti-inflammatory cytokines and microglial activity in brain (prefrontal cortex, hippocampus and hypothalamus) of female mice that were subjected to acute SF for 24 hours, chronic SF for 8 weeks, or 7 days of recovery after chronic SF. In each experiment, SF and control mice were peripherally sympathectomized with 6-hydroxydopamine (6-OHDA) or injected with vehicle. SF elevated cytokine mRNA expression in brain and increased microglial density and cell area in some regions. In addition, chronic SF promoted hyper-ramification in resting microglia upon exposure to chronic, but not acute, SF. Effects of chronic SF were more pronounced than acute SF, and 1 week of recovery was not sufficient to alleviate neuroinflammation. Importantly, 6-OHDA treatment significantly alleviated SF-induced inflammation and microglial responses. This study provides evidence of SNS regulation of neural inflammation from SF, suggesting a potential role for therapeutics that could mitigate neuroinflammatory responses to sleep dysfunction.
Collapse
Affiliation(s)
- Ila Mishra
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA; Harrington Discovery Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Keelee B Pullum
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kristen N Eads
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA
| | - Anna R Strunjas
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA
| | - Noah T Ashley
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101, USA.
| |
Collapse
|
29
|
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: 13] [Impact Index Per Article: 4.3] [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
|
30
|
Sur B, Lee B. Myricetin prevents sleep deprivation-induced cognitive impairment and neuroinflammation in rat brain via regulation of brain-derived neurotropic factor. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:415-425. [PMID: 36302617 PMCID: PMC9614391 DOI: 10.4196/kjpp.2022.26.6.415] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 11/07/2022]
Abstract
Memory formation in the hippocampus is formed and maintained by circadian clock genes during sleep. Sleep deprivation (SD) can lead to memory impairment and neuroinflammation, and there remains no effective pharmacological treatment for these effects. Myricetin (MYR) is a common natural flavonoid that has various pharmacological activities. In this study, we investigated the effects of MYR on memory impairment, neuroinflammation, and neurotrophic factors in sleep-deprived rats. We analyzed SD-induced cognitive and spatial memory, as well as pro-inflammatory cytokine levels during SD. SD model rats were intraperitoneally injected with 10 and 20 mg/kg/day MYR for 14 days. MYR administration significantly ameliorated SD-induced cognitive and spatial memory deficits; it also attenuated the SD-induced inflammatory response associated with nuclear factor kappa B activation in the hippocampus. In addition, MYR enhanced the mRNA expression of brain-derived neurotropic factor (BDNF) in the hippocampus. Our results showed that MYR improved memory impairment by means of anti-inflammatory activity and appropriate regulation of BDNF expression. Our findings suggest that MYR is a potential functional ingredient that protects cognitive function from SD.
Collapse
Affiliation(s)
- Bongjun Sur
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Korea
| | - Bombi Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 02447, Korea,Center for Converging Humanities, Kyung Hee University, Seoul 02447, Korea,Correspondence Bombi Lee, E-mail:
| |
Collapse
|
31
|
Ensminger DC, Wheeler ND, Al Makki R, Eads KN, Ashley NT. Contrasting effects of sleep fragmentation and angiotensin-II treatment upon pro-inflammatory responses of mice. Sci Rep 2022; 12:14763. [PMID: 36042284 PMCID: PMC9427781 DOI: 10.1038/s41598-022-19166-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/25/2022] [Indexed: 11/30/2022] Open
Abstract
Disordered sleep promotes inflammation in brain and peripheral tissues, but the mechanisms that regulate these responses are poorly understood. One hypothesis is that activation of the sympathetic nervous system (SNS) from sleep loss elevates blood pressure to promote vascular sheer stress leading to inflammation. As catecholamines produced from SNS activation can directly regulate inflammation, we pharmacologically altered blood pressure using an alternative approach-manipulation of the renin-angiotensin system (RAS). Male C57BL6/J mice were treated with angiotensin or captopril to elevate and reduce blood pressure, respectively and then exposed to 24-h of sleep fragmentation (SF) or allowed to sleep (control). Pro- and anti-inflammatory cytokine gene expression and as endothelial adhesion gene expression as well as serum glucocorticoids (corticosterone) were measured. RAS manipulation elevated cytokines and endothelial adhesion expression in heart and aorta while SF increased cytokine expression in peripheral tissues, but not brain. However, there were interactive effects of angiotensin-II and SF upon cytokine gene expression in hippocampus and hypothalamus, but not prefrontal cortex. SF, but not RAS manipulation, elevated serum corticosterone concentration. These findings highlight the contrasting effects of RAS manipulation and SF, implying that inflammation from SF is acting on different pathways that are largely independent of RAS manipulation.
Collapse
Affiliation(s)
- David C Ensminger
- Department of Biology, Western Kentucky University, Bowling Green, KY, USA.
- Department of Biological Sciences, San José State University, San Jose, CA, USA.
| | - Nicholas D Wheeler
- Department of Biology, Western Kentucky University, Bowling Green, KY, USA
- College of Veterinary Medicine, Mississippi State University, Starkville, MS, USA
| | - Reem Al Makki
- Department of Biological Sciences, San José State University, San Jose, CA, USA
| | - Kristen N Eads
- School of Physician Assistant Studies, Lipscomb University, Nashville, TN, USA
| | - Noah T Ashley
- Department of Biology, Western Kentucky University, Bowling Green, KY, USA
| |
Collapse
|
32
|
Neurocognitive Consequences in Children with Sleep Disordered Breathing: Who Is at Risk? CHILDREN 2022; 9:children9091278. [PMID: 36138586 PMCID: PMC9497121 DOI: 10.3390/children9091278] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022]
Abstract
Sleep-disordered breathing (SDB) is a prevalent disease in children characterized by snoring and narrowing of the upper airway leading to gas exchange abnormalities during sleep as well as sleep fragmentation. SDB has been consistently associated with problematic behaviors and adverse neurocognitive consequences in children but causality and determinants of susceptibility remain incompletely defined. Since the 1990s several studies have enlightened these associations and consistently reported poorer academic performance, lower scores on neurocognitive tests, and behavioral abnormalities in children suffering from SDB. However, not all children with SDB develop such consequences, and severity of SDB based on standard diagnostic indices has often failed to discriminate among those children with or without neurocognitive risk. Accordingly, a search for discovery of markers and clinically useful tools that can detect those children at risk for developing cognitive and behavioral deficits has been ongoing. Here, we review the advances in this field and the search for possible detection approaches and unique phenotypes of children with SDB who are at greater risk of developing neurocognitive consequences.
Collapse
|
33
|
Popadic V, Brajkovic M, Klasnja S, Milic N, Rajovic N, Lisulov DP, Divac A, Ivankovic T, Manojlovic A, Nikolic N, Memon L, Brankovic M, Popovic M, Sekulic A, Macut JB, Markovic O, Djurasevic S, Stojkovic M, Todorovic Z, Zdravkovic M. Correlation of Dyslipidemia and Inflammation With Obstructive Sleep Apnea Severity. Front Pharmacol 2022; 13:897279. [PMID: 35694268 PMCID: PMC9179947 DOI: 10.3389/fphar.2022.897279] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/28/2022] [Indexed: 12/14/2022] Open
Abstract
Introduction: Obstructive sleep apnea (OSA) is a serious condition linked with various metabolic disorders and associated with increased all-cause and cardiovascular mortality. Although the potential mechanisms of pathophysiological processes related to OSA are relatively well known, the data regarding the correlation between obstructive sleep apnea, dyslipidemia, and systemic inflammation are still inconclusive. Methods: The study was conducted as a retrospective cohort study including 328 patients with newly diagnosed obstructive sleep apnea during the period between April 2018, and May 2020, in University Clinical Hospital Center “Bezanijska kosa”, Belgrade, Serbia. Polysomnography was performed in all patients according to the protocol. Numerous demographic, antropometric, laboratory, and clinical data were correlated to Apnea-Hypopnea Index (AHI) as a dependent variable, with a particular review on the relation between lipid abnormalities, inflammatory parameters, and obstructive sleep apnea severity. Multivariate logistic regression model was used to assess predictors of severe OSA (AHI ≥30 per hour). Results: A total of 328 patients were included in the study. The mean age of the patients was 54.0 ± 12.5 years and more than two-thirds were male (68.8%). The majority of the patients had an AHI of at least 30 events per hour. Patients with severe OSA were more frequently male, obese, hypertensive and hyperlipidemic, and had increased neck circumference (both male and female patients). One hundred and thirty-two patients had metabolic syndrome. Patients with severe OSA more frequently had metabolic syndrome and significantly higher levels of glucose, creatinine, uric acid, AST, ALT, CK, microalbumine/creatinine ratio, triglyceride, total cholesterol, HDL, total cholеsterol to HDL‐C ratio, CRP, and ESR. In the multivariate linear regression model with AHI (≥30 per hour) as a dependent variable, of demographic and clinical data, triglycerides ≥1.7 mmol/L and CRP >5 mg/L were significantly associated with AHI≥30 per hour. Conclusion: The present study on 328 patients with newly diagnosed obstructive sleep apnea revealed significant relation of lipid abnormalities, inflammatory markers, and other clinically important data with obstructive sleep apnea severity. These results can lead to a better understanding of the underlying pathophysiological processes and open the door to a new world of potentially useful therapeutic modalities.
Collapse
Affiliation(s)
- Viseslav Popadic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
- *Correspondence: Viseslav Popadic,
| | - Milica Brajkovic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Slobodan Klasnja
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Natasa Milic
- Institute for Medical Statistics and Informatics, Faculty of Medicine University of Belgrade, Belgrade, Serbia
- Department of Internal Medicine,Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MI, United States
| | - Nina Rajovic
- Institute for Medical Statistics and Informatics, Faculty of Medicine University of Belgrade, Belgrade, Serbia
| | | | - Anica Divac
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Tatjana Ivankovic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Andrea Manojlovic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Novica Nikolic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Lidija Memon
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Marija Brankovic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Maja Popovic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Ana Sekulic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelica Bjekic Macut
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Olivera Markovic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Maja Stojkovic
- School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Zoran Todorovic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marija Zdravkovic
- University Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
34
|
Roodsari SK, Cheng Y, Reed KM, Wellman LL, Sanford LD, Kim WK, Guo ML. Sleep Disturbance Alters Cocaine-Induced Locomotor Activity: Involvement of Striatal Neuroimmune and Dopamine Signaling. Biomedicines 2022; 10:biomedicines10051161. [PMID: 35625897 PMCID: PMC9138453 DOI: 10.3390/biomedicines10051161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
Sleep disorders have high comorbidity with drug addiction and function as major risk factors for developing drug addiction. Recent studies have indicated that both sleep disturbance (SD) and abused drugs could activate microglia, and that increased neuroinflammation plays a critical role in the pathogenesis of both diseases. Whether microglia are involved in the contribution of chronic SDs to drug addiction has never been explored. In this study, we employed a mouse model of sleep fragmentation (SF) with cocaine treatment and examined their locomotor activities, as well as neuroinflammation levels and dopamine signaling in the striatum, to assess their interaction. We also included mice with, or without, SF that underwent cocaine withdrawal and challenge. Our results showed that SF significantly blunted cocaine-induced locomotor stimulation while having marginal effects on locomotor activity of mice with saline injections. Meanwhile, SF modulated the effects of cocaine on neuroimmune signaling in the striatum and in ex vivo isolated microglia. We did not observe differences in dopamine signaling in the striatum among treatment groups. In mice exposed to cocaine and later withdrawal, SF reduced locomotor sensitivity and also modulated neuroimmune and dopamine signaling in the striatum. Taken together, our results suggested that SF was capable of blunting cocaine-induced psychoactive effects through modulating neuroimmune and dopamine signaling. We hypothesize that SF could affect neuroimmune and dopamine signaling in the brain reward circuitry, which might mediate the linkage between sleep disorders and drug addiction.
Collapse
Affiliation(s)
- Soheil Kazemi Roodsari
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (S.K.R.); (Y.C.); (K.M.R.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
| | - Yan Cheng
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (S.K.R.); (Y.C.); (K.M.R.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
| | - Kirstin M. Reed
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (S.K.R.); (Y.C.); (K.M.R.)
| | - Laurie L. Wellman
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Larry D. Sanford
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Woong-Ki Kim
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ming-Lei Guo
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (S.K.R.); (Y.C.); (K.M.R.)
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (L.L.W.); (L.D.S.); (W.-K.K.)
- Correspondence: ; Tel.: +1-757-446-5891
| |
Collapse
|
35
|
Gozal D. Brain structure-function relationships in sleep apnea among obese children: no time to waste! Sleep 2022; 45:zsac055. [PMID: 35554580 DOI: 10.1093/sleep/zsac055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Affiliation(s)
- David Gozal
- Department of Child Health and the Child Health Research Institute, and the Comprehensive Sleep Medicine Program, University of Missouri School of Medicine, Columbia, MO 65201, USA
| |
Collapse
|
36
|
Sleep-Disordered Breathing Risk with Comorbid Insomnia Is Associated with Mild Cognitive Impairment. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12052414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Introduction: Few studies have evaluated the combined association between SDB with comorbid insomnia and mild cognitive impairment (MCI). To test the hypothesis that SDB with comorbid insomnia is associated with greater odds of MCI than either sleep disorder independently, we used ADNI data to evaluate cross-sectional associations between SDB risk with comorbid insomnia status and MCI. Methods: Participants with normal cognition or MCI were included. Insomnia was defined by self-report. SDB risk was assessed by modified STOP-BANG. Logistic regression models evaluated associations between four sleep disorder subgroups (low risk for SDB alone, low risk for SDB with insomnia, high risk for SDB alone, and high risk for SDB with insomnia) and MCI. Models adjusted for age, sex, BMI, APOE4 genotype, race, ethnicity, education, marital status, hypertension, cardiovascular disease, stroke, alcohol abuse, and smoking. Results: The sample (n = 1391) had a mean age of 73.5 ± 7.0 years, 44.9% were female, 72.0% were at low risk for SDB alone, 13.8% at low risk for SDB with insomnia, 10.1% at high risk for SDB alone, and 4.1% at high risk for SDB with insomnia. Only high risk for SDB with comorbid insomnia was associated with higher odds of MCI (OR 3.22, 95% CI 1.57–6.60). Conclusion: Studies are needed to evaluate SDB with comorbid insomnia as a modifiable risk factor for MCI.
Collapse
|
37
|
TNF signaling pathway-mediated microglial activation in the PFC underlies acute paradoxical sleep deprivation-induced anxiety-like behaviors in mice. Brain Behav Immun 2022; 100:254-266. [PMID: 34915154 DOI: 10.1016/j.bbi.2021.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/08/2021] [Accepted: 12/09/2021] [Indexed: 12/24/2022] Open
Abstract
Acute sleep deprivation is a common condition in modern life and increases anxiety symptoms in healthy individuals. The neuroinflammatory response induced by microglial activation could be an important contributing factor, but its underlying molecular mechanisms are still unclear. In the present study, we first found that acute paradoxical sleep deprivation (PSD) induced by the modified multiple platform method (MMPM) for 6 h led to anxiety-like behavior in mice, as verified by the open field test, elevated plus maze test, light-dark box test, and marble burying test. In addition, bioinformatic analysis suggested an important relationship between acute sleep deprivation and brain inflammatory signaling pathways. Key genes enriched in the TNF signaling pathway were confirmed to be altered during acute PSD by qPCR and Western blot analyses, including the upregulation of the prostaglandin-endoperoxide synthase 2 (Ptgs2) and suppressor of cytokine signaling 3 protein (Socs3) genes and the downregulation of the cysteine-aspartic acid protease 3 (Casp3) gene. Furthermore, we found that microglial cells in the prefrontal cortex (PFC) were activated with significant branch structure changes and that the cell body area was increased in the PSD model. Finally, we found that minocycline, a tetracycline with anti-inflammatory properties, may ameliorate the anxiogenic effect and microglial activation. Our study reveals significant correlations of anxiety-like behavior, microglial activation, and inflammation during acute PSD.
Collapse
|
38
|
Koinis-Mitchell D, Marshall GD, Kopel SJ, Belanger NMS, Ayala-Figueroa J, Echevarria S, Millman R, Zheng T, Weathers J, Gredvig CA, Carskadon MA. Experimental methods to study sleep disruption and immune balance in urban children with asthma. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2022; 3:zpac003. [PMID: 35355783 PMCID: PMC8947185 DOI: 10.1093/sleepadvances/zpac003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/08/2021] [Indexed: 11/13/2022]
Abstract
Study Objectives We describe research methods developed to examine effects of sleep disruption on changes in immune balance, lung function, and cognitive performance in a sample of urban, ethnically diverse children with persistent asthma. Two case examples (8- and 10-year-old males) are presented to highlight methods of the current study and illustrate effects of experimentally disrupted sleep on the immune balance profile (Th1/Th2 cytokines), key sleep variables from polysomnography data, and lung function in our sample. Methods Children follow an individualized structured sleep schedule consistent with their habitual sleep need (≥9.5 hours' time in bed) for six days before a laboratory-based experimental sleep protocol. Children then spend two successive nights in the sleep lab monitored by polysomnography: a baseline night consisting of uninterrupted sleep, and a disruption night, during which they are awoken for 2 minutes between 20-minute intervals of uninterrupted sleep. Evening and morning blood draws bracket baseline and disruption nights for immune biomarker assessment. Results A shift towards immune imbalance following the sleep disruption protocol was observed in these illustrative cases. Conclusions Data from these case examples provide evidence that the experimental protocol caused disruptions in sleep as observed on polysomnography and had the hypothesized downstream effects on immune balance associated with clinical asthma control. Documenting the effects of sleep disruption on immune function in children with persistent asthma is a crucial step towards understanding associations between sleep, immune balance, and asthma outcomes and provides important information for developing novel interventions for youth with asthma and suboptimal sleep. Clinical Trials Not applicable.
Collapse
Affiliation(s)
- Daphne Koinis-Mitchell
- Department of Pediatrics, Hasbro Children’s Hospital, Providence, RI, USA
- Bradley-Hasbro Children’s Research Center, Rhode Island Hospital, Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Gailen D Marshall
- Division of Allergy, Asthma and Clinical Immunology, Department of Medicine, The University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Sheryl J Kopel
- Department of Pediatrics, Hasbro Children’s Hospital, Providence, RI, USA
- Bradley-Hasbro Children’s Research Center, Rhode Island Hospital, Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Nicole M S Belanger
- Department of Pediatrics, Hasbro Children’s Hospital, Providence, RI, USA
- Bradley-Hasbro Children’s Research Center, Rhode Island Hospital, Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Jesús Ayala-Figueroa
- Department of Pediatrics, Hasbro Children’s Hospital, Providence, RI, USA
- Bradley-Hasbro Children’s Research Center, Rhode Island Hospital, Providence, RI, USA
| | - Sofia Echevarria
- Department of Pediatrics, Hasbro Children’s Hospital, Providence, RI, USA
- Bradley-Hasbro Children’s Research Center, Rhode Island Hospital, Providence, RI, USA
| | - Richard Millman
- Department of Pediatrics, Hasbro Children’s Hospital, Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Tao Zheng
- Department of Pediatrics, Hasbro Children’s Hospital, Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Jessica Weathers
- EP Bradley Hospital Sleep and Chronobiology Research Laboratory, Providence, RI, USA
| | - Caroline A Gredvig
- EP Bradley Hospital Sleep and Chronobiology Research Laboratory, Providence, RI, USA
| | - Mary A Carskadon
- Warren Alpert Medical School of Brown University, Providence, RI, USA
- EP Bradley Hospital Sleep and Chronobiology Research Laboratory, Providence, RI, USA
| |
Collapse
|
39
|
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
|
40
|
Miola A, Dal Porto V, Meda N, Perini G, Solmi M, Sambataro F. Secondary Mania induced by TNF-α inhibitors: A systematic review. Psychiatry Clin Neurosci 2022; 76:15-21. [PMID: 34590391 PMCID: PMC9298409 DOI: 10.1111/pcn.13302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/09/2021] [Accepted: 09/13/2021] [Indexed: 12/01/2022]
Abstract
A growing number of studies support a bidirectional relationship between inflammation and bipolar disorders. Tumor necrosis factor-α (TNF-α) inhibitors have recently attracted interest as potential therapeutic compounds for treating depressive symptoms, but the risk for triggering mood switches in patients with or without bipolar disorders remains controversial. Thus, we conducted a systematic review to study the anti-TNF-α medication-induced manic or hypomanic episodes. PubMed, Scopus, Medline, and Embase databases were screened for a comprehensive literature search from inception until November 2020, using The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Out of the initial 75 references, the screening resulted in the inclusion of four case reports (each describing one patient) and a cohort study (in which 40 patients out of 7600-0.53% - experienced elated mood episodes after infliximab administration). Of these 44 patients, 97.7% experienced a manic episode and 2.3% hypomania. 93.2% of patients had no history of psychiatric disorder or psychotropic treatment. Only 6.8% had a history of psychiatric disorders with the affective spectrum (4.6% dysthymia and 2.3% bipolar disorder). The time of onset of manic or hypomanic symptoms varied across TNF-α inhibitors with an early onset for Infliximab and a later onset for Adalimumab and Etanercept. These findings suggest that medications targeting the TNF-α pathway may trigger a manic episode in patients with or without affective disorders. However, prospective studies are needed to evaluate the relative risk of such side effects and identify the population susceptible to secondary mania.
Collapse
Affiliation(s)
- Alessandro Miola
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy.,Casa di Cura Parco dei Tigli, Padova, Italy
| | | | - Nicola Meda
- Department of Medicine, University of Padova, Padova, Italy
| | - Giulia Perini
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy.,Casa di Cura Parco dei Tigli, Padova, Italy
| | - Marco Solmi
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada.,Department of Mental Health, The Ottawa Hospital, Ottawa, ON, Canada.,Clinical Epidemiology Program, Ottawa Hospital Research Institute (OHRI), University of Ottawa, Ottawa, ON, Canada
| | - Fabio Sambataro
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy
| |
Collapse
|
41
|
Damian A, Gozal D. Pediatric Obstructive Sleep Apnea: What’s in a Name? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1384:63-78. [PMID: 36217079 DOI: 10.1007/978-3-031-06413-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Obstructive sleep apnea is a highly prevalent disease across the lifespan and imposes substantial morbidities, some of which may become irreversible if the condition is not diagnosed and treated in a timely fashion. Here, we focus on the clinical and epidemiological characteristics of pediatric obstructive sleep apnea, describe some of the elements that by virtue of their presence facilitate the emergence of disrupted sleep and breathing and its downstream consequences, and also discuss the potential approaches to diagnosis in at-risk children.
Collapse
Affiliation(s)
- Allan Damian
- Departments of Neurology, University of Missouri School of Medicine, Columbia, MO, USA
- Comprehensive Sleep Medicine Program, University of Missouri School of Medicine, Columbia, MO, USA
| | - David Gozal
- Comprehensive Sleep Medicine Program, University of Missouri School of Medicine, Columbia, MO, USA.
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA.
| |
Collapse
|
42
|
Abstract
Multiple Sclerosis (MS) is a common neuroinflammatory disorder which is associated with disabling clinical consequences. The MS disease process may involve neural centers implicated in the control of breathing, leading to ventilatory disturbances during both wakefulness and sleep. In this chapter, a brief overview of MS disease mechanisms and clinical sequelae including sleep disorders is provided. The chapter then focuses on obstructive sleep apnea-hypopnea (OSAH) which is the most prevalent respiratory control abnormality encountered in ambulatory MS patients. The diagnosis, prevalence, and clinical consequences as well as data on effects of OSAH treatment in MS patients are discussed, including the impact on the disabling symptom of fatigue and other clinical sequelae. We also review pathophysiologic mechanisms contributing to OSAH in MS, and in turn mechanisms by which OSAH may impact on the MS disease process, resulting in a bidirectional relationship between these two conditions. We then discuss central sleep apnea, other respiratory control disturbances, and the pathogenesis and management of respiratory muscle weakness and chronic hypoventilation in MS. We also provide a brief overview of Neuromyelitis Optica Spectrum Disorders and review current data on respiratory control disturbances and sleep-disordered breathing in that condition.
Collapse
Affiliation(s)
- R John Kimoff
- Respiratory Division and Sleep Laboratory, McGill University Health Centre, McGill University, Montreal, QC, Canada; Respiratory Epidemiology and Clinical Research Unit, Research Institute of McGill University Health Centre, Montreal, QC, Canada.
| | - Marta Kaminska
- Respiratory Division and Sleep Laboratory, McGill University Health Centre, McGill University, Montreal, QC, Canada; Respiratory Epidemiology and Clinical Research Unit, Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Daria Trojan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University Health Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
43
|
Differential Effect of Light and Dark Period Sleep Fragmentation on Composition of Gut Microbiome and Inflammation in Mice. Life (Basel) 2021; 11:life11121283. [PMID: 34947814 PMCID: PMC8709399 DOI: 10.3390/life11121283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/17/2022] Open
Abstract
Bi-directional interactions amongst the gut microbiota, immune system, and brain function are thought to be critical mediators of health and disease. The role sleep plays in mediating these interactions is not known. We assessed the effects of sleep fragmentation (SF) on the microbiota–gut–brain axis. Male C57BL/6NCrl mice (4 to 5 per cage, fed standard lab chow) experienced SF via mechanical stimulation at 2 min intervals during the light (SF) and dark (DD, dark disturbances) periods. Home cage (HC) controls were undisturbed. After 10 days, fecal samples were collected at light onset, midday, light offset, and midnight. Samples were also collected after 10 days without SF. Subsequently, the mice were randomized across groups and allowed 20 additional days of recovery followed by 10 days of SF or DD. To assess effects on the microbiota, 16S rRNA sequencing was used, and mesenteric lymph nodes (MLNs) and cortex and medial prefrontal cortex were analyzed using cytokine arrays. SF and DD produced significant alterations in the microbiota compared to HC, and DD had greater impact than SF on some organisms. SF produced marked suppression in MLNs of chemokines that regulate inflammation (CCL3, CCL4 and their receptor CCR5) and maintain the immune mucosal barrier (Cxcl13) at the same time that cortical cytokines (IL-33) indicated neuroinflammation. DD effects on immune responses were similar to HC. These data suggest that SF alters the microbiome and suppresses mucosal immunity at the same time that mediators of brain inflammation are upregulated. The translational implications for potential application to clinical care are compelling.
Collapse
|
44
|
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
|
45
|
Cheng Y, Kim WK, Wellman LL, Sanford LD, Guo ML. Short-Term Sleep Fragmentation Dysregulates Autophagy in a Brain Region-Specific Manner. Life (Basel) 2021; 11:life11101098. [PMID: 34685469 PMCID: PMC8538758 DOI: 10.3390/life11101098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 01/17/2023] Open
Abstract
In this study, we investigated autophagy, glial activation status, and corticotropin releasing factor (CRF) signaling in the brains of mice after 5 days of sleep fragmentation (SF). Three different brain regions including the striatum, hippocampus, and frontal cortex were selected for examination based on roles in sleep regulation and sensitivity to sleep disruption. For autophagy, we monitored the levels of various autophagic induction markers including beclin1, LC3II, and p62 as well as the levels of lysosomal associated membrane protein 1 and 2 (LAMP1/2) and the transcription factor EB (TFEB) which are critical for lysosome function and autophagy maturation stage. For the status of microglia and astrocytes, we determined the levels of Iba1 and GFAP in these brain regions. We also measured the levels of CRF and its cognate receptors 1 and 2 (CRFR1/2). Our results showed that 5 days of SF dysregulated autophagy in the striatum and hippocampus but not in the frontal cortex. Additionally, 5 days of SF activated microglia in the striatum but not in the hippocampus or frontal cortex. In the striatum, CRFR2 but not CRFR1 was significantly increased in SF-experienced mice. CRF did not alter its mRNA levels in any of the three brain regions assessed. Our findings revealed that autophagy processes are sensitive to short-term SF in a region-specific manner and suggest that autophagy dysregulation may be a primary initiator for brain changes and functional impairments in the context of sleep disturbances and disorders.
Collapse
Affiliation(s)
- Yan Cheng
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Drug Addiction Laboratory, Department Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Woong-Ki Kim
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Laurie L. Wellman
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Larry D. Sanford
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Sleep Research Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ming-Lei Guo
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, VA 23507, USA; (Y.C.); (W.-K.K.); (L.L.W.); (L.D.S.)
- Drug Addiction Laboratory, Department Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Correspondence: ; Tel.: +1-757-446-5891
| |
Collapse
|
46
|
Anxiogenic Potential of Experimental Sleep Fragmentation Is Duration-Dependent and Mediated via Oxidative Stress State. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2262913. [PMID: 34471462 PMCID: PMC8405322 DOI: 10.1155/2021/2262913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/04/2021] [Indexed: 11/28/2022]
Abstract
Sleep architecture alterations, among which sleep fragmentation is highly prevalent, represent risk factors for a variety of diseases, ranging from cardiovascular to brain disorders, including anxiety. What mediates anxiety occurrence upon sleep fragmentation is still a matter of debate. We hypothesized that the sleep fragmentation effects on anxiety are dependent on its duration and mediated by increased oxidative stress and alterations in the number of parvalbumin (PV+) interneurons in the hippocampus. Sleep was fragmented in rats by the treadmill method during a period of 14 days (SF group). Rats with undisturbed sleep in the treadmill (TC group) and those receiving equal amounts of treadmill belt motion (EC group) served as controls. To assess anxiety, we subjected rats to the open field, elevated plus maze, and light-dark tests on the 0, 7th, and 14th day. Upon the last test, brain structures were sampled for oxidative stress assessment and PV+ interneuron immunohistochemistry. The results of ethological tests of anxiety-linked behavior suggested duration-dependent anxiogenic potential of sleep fragmentation. Rats' anxiety-linked behavior upon sleep fragmentation significantly correlated with oxidative stress. The rats with fragmented sleep (SF) showed significantly higher oxidative stress in the hippocampus, thalamus, and cortex, compared to controls (TC and EC), while the antioxidant enzymes' activity was significantly decreased. No significant differences were observed in hippocampal PV+ interneurons among these groups. Our results showed that duration of sleep fragmentation is a significant determinant of anxiety-linked behavior, and these effects are mediated through oxidative distress in the brain. Herein, it is revealed that the sleep fragmentation-oxidative stress-anxiety axis contributes to our better understanding of pathophysiological processes, occurring due to disrupted sleep patterns.
Collapse
|
47
|
Liu T, Ouyang R. Effect of continuous positive air pressure on cognitive impairment associated with obstructive sleep apnea. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:865-871. [PMID: 34565731 PMCID: PMC10929983 DOI: 10.11817/j.issn.1672-7347.2021.190600] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Indexed: 11/03/2022]
Abstract
Obstructive sleep apnea (OSA) is a kind of sleep-related breathing disorder, involving multiple organs and systems, which can lead to cognitive impairment. At present, the pathophysiological mechanism of cognitive impairment related to OSA is not clear. It is still unknown whether continuous positive airway pressure (CPAP) has therapeutic effect on cognitive impairment in patients with OSA. These patients repeatedly experience intermittent hypoxia and have sleep fragmentation, which results in abnormal brain structure and function, characterizing by extensive cognitive impairment. Appropriate CPAP can correct the abnormal pathophysiological process of OSA patients, restore brain structure and function to a certain extent, and improve cognitive function. Domestic OSA patients have poor acceptance and compliance to CPAP, while the therapeutic effect of CPAP depends on the timing of treatment and compliance, so many patients do not get effective treatment. Systematically expounding the influence of CPAP on the cognitive function of patients with OSA can help clinicians and patients improve their understanding of CPAP treatment and establish a correct concept of early and standardized treatment.
Collapse
Affiliation(s)
- Ting Liu
- Department of Respiratory and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Ruoyun Ouyang
- Department of Respiratory and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
48
|
Ji T, Li X, Chen J, Ren X, Mei L, Qiu Y, Zhang J, Wang S, Xu Z, Li H, Zheng L, Peng Y, Liu Y, Ni X, Tai J, Liu J. Brain function in children with obstructive sleep apnea: a resting-state fMRI study. Sleep 2021; 44:6155746. [PMID: 33675225 DOI: 10.1093/sleep/zsab047] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/16/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE To explore the neural difference between children with obstructive sleep apnea (OSA) and healthy controls, together with the relation between this difference and cognitive dysfunction of children with OSA. METHODS Twenty children with OSA (7.2 ± 3.1 years, apnea hypopnea index (AHI): 16.5 ± 16.6 events/h) and 29 healthy controls (7.7 ± 2.8 years, AHI: 1.7 ± 1.2 events/h) were recruited and matched with age, gender, and handedness. All children underwent resting-state fMRI (rs-fMRI) and T1-wighted imaging. Some children were sedated for MRI scanning. We compared amplitude of low frequency fluctuation (ALFF) and regional homogeneity (ReHo) of children with OSA with those of healthy controls. During resting-state, the former reflects the intensity of the spontaneous neural activities, whereas the latter reflects temporal similarity of the spontaneous neural activities within a local brain region. Pearson correlation analysis was performed between these features of rs-fMRI and cognitive scores among children with OSA. RESULTS Compared with controls, children with OSA showed decreased ALFF in the left angular gyrus but increased ALFF in the right insula, and decreased ReHo in the left medial superior frontal gyrus, right lingual gyrus, and left precuneus. Additionally, among children with OSA, the ReHo value in the right lingual gyrus was negatively correlated with FIQ and VIQ, whereas that in the left medial superior frontal gyrus was positively correlated with VIQ. CONCLUSIONS Children with OSA presented abnormal neural activities in some brain regions and impaired cognitive functions with the former possibly being the neural mechanism of the latter.
Collapse
Affiliation(s)
- Tingting Ji
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaodan Li
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jun Chen
- Beijing Engineering Research Center of Pediatric Surgery, Engineering and Translational Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xuemin Ren
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medical Science and Engineering, Beihang University, Beijing, China.,Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the, People's Republic of China, Beijing, China
| | - Lin Mei
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yue Qiu
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Shengcai Wang
- Department of Otolaryngology, Head and Neck Surgery, 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.,Department of Sleep Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hongbin Li
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Li Zheng
- Department of Sleep Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yun Peng
- Department of Radiology, Imaging Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yue Liu
- Department of Radiology, Imaging Center, 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.,Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jun Tai
- Department of Otorhinolaryngology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Jiangang Liu
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medical Science and Engineering, Beihang University, Beijing, China.,Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the, People's Republic of China, Beijing, China
| |
Collapse
|
49
|
Silva Junior JF, Eckeli AL, Ribeiro CCC, Batista RFL, da Silva AAM, Alves CMC. Influence of excessive daily sleeping and sleep quality on BDNF and NGF serum levels in adolescents. Sleep Med 2021; 84:415-423. [PMID: 34329829 DOI: 10.1016/j.sleep.2021.06.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 01/09/2023]
Abstract
INTRODUCTION The brain-derived neurotrophic factor (BDNF) and neural growth factor (NGF) are widely expressed in the brain and play an important role in neuroplasticity, neurogenesis, and increased neuronal connections. Previous studies have shown that reduced serum levels of these proteins are associated with disorders in human sleep. OBJECTIVE Current study evaluates the prevalence in adolescents of excessive daytime sleepiness (EDS) and sleep quality, and analyzes the influence of these factors on BDNF and NGF serum levels. METHODS A cross-section population-based study was conducted with data from a Brazilian birth cohort, with a sample of five hundred and thirteen 18-19-year-old adolescents. Sleep quality was assessed by the Pittsburgh Sleep Quality Index and EDS by Epworth Sleepiness Scale. Neurotrophins serum levels were measured by Luminex™ technology kits. Analysis consisted of marginal structural models which compared people who were exposed and not exposed to sleep quality and EDS. RESULTS Poor sleep quality and EDS were detected in 62.57% and 36.35% of the sample. Adolescents with poor sleep quality and EDS had -0.39 (p-value = 0.049) and -0.51 pg/ml in NGF (p-value = 0.009). Individuals with self-reported sleep disorder had lower serum levels of NGF (Coef. -0.41, p-value = 0.045). CONCLUSION High prevalence of EDS and low sleep quality in a population of adolescents were evidenced. Poor sleep quality and EDS were associated with lower NGF levels, whilst adolescents with self-reported sleep disorder had lower serum levels of NGF.
Collapse
Affiliation(s)
| | - Alan Luiz Eckeli
- Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | | | | |
Collapse
|
50
|
Jung YJ, Oh E. Is REM sleep behavior disorder a friend or foe of obstructive sleep apnea? Clinical and etiological implications for neurodegeneration. J Clin Sleep Med 2021; 17:1305-1312. [PMID: 33660615 DOI: 10.5664/jcsm.9144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
NONE Rapid eye movement sleep behavior disorder (RBD) is a parasomnia characterized by loss of muscle atonia during rapid eye movement sleep, associated with complex motor enactment of dreams. Obstructive sleep apnea (OSA) is a relatively common sleep disorder characterized by repetitive episodes of upper airway obstruction while sleeping, which can result in hypoxemia and sleep fragmentation. Even though the nature of RBD and OSA is different, OSA may sometimes be accompanied by RBD symptoms. Accordingly, it is reasonable to distinguish these 2 sleep disorders in people with dream enactment behaviors. Although RBD and OSA share similar sleep phenomena, their association has yet to be elucidated. Herein we draw attention to various RBD-mimicking conditions, RBD combined with OSA, and the relationship between RBD and OSA. Furthermore, the clinical implications of OSA in neurodegeneration and the optimized management of RBD combined with OSA are also discussed in this review.
Collapse
Affiliation(s)
- Yu Jin Jung
- Department of Neurology, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|