1
|
Gaucher J, Vial G, Bouyon S, Briançon-Marjollet A, Faury G, Arnaud C, Bailly S, Tamisier R, Kinouchi K, Baldi P, Gozal D, Pépin JL. On the imperative of including 24-h and longitudinal multidimensional physiological phenotyping in rodent models of sleep apnea. Sleep Med 2025; 127:133-137. [PMID: 39847827 DOI: 10.1016/j.sleep.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Affiliation(s)
- Jonathan Gaucher
- Grenoble Alpes University, HP2 Laboratory, INSERM U1300, Grenoble Alpes University Hospital, Grenoble, France.
| | - Guillaume Vial
- Grenoble Alpes University, HP2 Laboratory, INSERM U1300, Grenoble Alpes University Hospital, Grenoble, France
| | - Sophie Bouyon
- Grenoble Alpes University, HP2 Laboratory, INSERM U1300, Grenoble Alpes University Hospital, Grenoble, France
| | - Anne Briançon-Marjollet
- Grenoble Alpes University, HP2 Laboratory, INSERM U1300, Grenoble Alpes University Hospital, Grenoble, France
| | - Gilles Faury
- Grenoble Alpes University, HP2 Laboratory, INSERM U1300, Grenoble Alpes University Hospital, Grenoble, France
| | - Claire Arnaud
- Grenoble Alpes University, HP2 Laboratory, INSERM U1300, Grenoble Alpes University Hospital, Grenoble, France
| | - Sébastien Bailly
- Grenoble Alpes University, HP2 Laboratory, INSERM U1300, Grenoble Alpes University Hospital, Grenoble, France
| | - Renaud Tamisier
- Grenoble Alpes University, HP2 Laboratory, INSERM U1300, Grenoble Alpes University Hospital, Grenoble, France
| | - Kenichiro Kinouchi
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, Department of Computer Science, University of California, Irvine, CA, 92697, USA
| | - David Gozal
- Department of Pediatrics and Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25701, USA
| | - Jean-Louis Pépin
- Grenoble Alpes University, HP2 Laboratory, INSERM U1300, Grenoble Alpes University Hospital, Grenoble, France.
| |
Collapse
|
2
|
Yu X, Zhou X, He Z, He B, Wan K, Wei M, Guo T, Han Y. Sleep and APOE-ε4 have a synergistic effect on plasma biomarkers and longitudinal cognitive decline in older adults. CNS Neurosci Ther 2024; 30:e14558. [PMID: 38421124 PMCID: PMC10850800 DOI: 10.1111/cns.14558] [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: 08/24/2023] [Revised: 10/31/2023] [Accepted: 11/25/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Sleep disorders are prevalent among patients with Alzheimer's disease (AD), and the APOE ε4 genotype is a key genetic risk factor for sporadic AD. However, the combined effect of the genotype and sleep disorders on cognitive decline remains uncertain. METHODS A total of 972 participants were drawn from the SILCODE cohort, comprising 655 without the ε4 allele (APOE-) and 317 with ε4 allele (APOE+). Data were collected, including neuropsychological assessments, sleep measurements, plasma biomarkers, and PET imaging. A Sleep Composite Index (SCI) was created, categorizing participants into high risk (Sleep+) and low risk (Sleep-). RESULTS Significant predictions of dementia risk associated with plasma p-tau181, neurofilament light chain (NfL), and SCI. Individuals with both Sleep+ and APOE+ had a higher risk of dementia compared to those with Sleep-. The Sleep+/APOE+ group had higher plasma NfL levels than the Sleep-/APOE- group. Similar trends emerged in plasma NfL levels among the Aβ PET-positive subgroup. Plasma NfL levels explained 23% of the relationship between SCI and cognitive impairment. CONCLUSION Our study highlights sleep disorder was associated with cognitive decline, with plasma NfL playing a partial mediating role. These findings explain how sleep disorders affect cognitive function and emphasize the importance of healthy sleep for older adults.
Collapse
Affiliation(s)
- Xianfeng Yu
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Xia Zhou
- Department of NeurologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Zhengbo He
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Beiqi He
- School of Information and Communication EngineeringHainan UniversityHaikouChina
| | - Ke Wan
- Department of NeurologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Min Wei
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Tengfei Guo
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Ying Han
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
- School of Information and Communication EngineeringHainan UniversityHaikouChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- National Clinical Research Center for Geriatric DisordersBeijingChina
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Schneider G. Obstructive Sleep Apnea - Influence on the Cardiovascular System and Cognition. Laryngorhinootologie 2023; 102:S101-S114. [PMID: 37130534 PMCID: PMC10184569 DOI: 10.1055/a-1963-9957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Kardiovaskuläre und kognitive Erkrankungen sind ebenso wie die obstruktive Schlafapnoe sehr häufige Krankheiten mit einer erheblichen Beeinträchtigung der Lebensqualität und einer deutlichen sozioökonomischen Bedeutung. Die Auswirkungen einer unbehandelten obstruktiven Schlafapnoe (OSA) auf das kardiovaskuläre und kognitive Erkrankungsrisiko und die Therapieeffekte einer OSA sind für die meisten kardiovaskulären und kognitiven Folgeerkrankungen wissenschaftlich nachgewiesen. Für die klinische Praxis besteht ein deutlicher Bedarf nach mehr Interdisziplinarität. Aus schlafmedizinischer Sicht müssen bei der Therapieindikation das individuelle kardiovaskuläre und kognitive Risiko berücksichtigt und kognitive Erkrankungen bei der Beurteilung der Therapieintoleranz und residuellen Symptomatik beachtet werden. Aus internistischer Sicht sollte bei Patienten mit schlecht einstellbarem Hypertonus, Vorhofflimmern, koronarer Herzkrankheit und Schlaganfall die Abklärung einer OSA in die Diagnostik integriert werden. Bei Patienten mit milder kognitiver Beeinträchtigung, Alzheimer-Krankheit und Depression können sich die typischen Symptome wie Fatigue, Tagesmüdigkeit und Reduktion der kognitiven Leistungen mit OSA-Symptomen überschneiden. Die Diagnostik einer OSA sollte in die Abklärung dieser Krankheitsbilder integriert werden, da eine Therapie der OSA die kognitiven Beeinträchtigungen reduzieren und die Lebensqualität verbessern kann.
Collapse
|
6
|
Wang Y, Guo H, He F. Circadian disruption: from mouse models to molecular mechanisms and cancer therapeutic targets. Cancer Metastasis Rev 2023; 42:297-322. [PMID: 36513953 DOI: 10.1007/s10555-022-10072-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022]
Abstract
The circadian clock is a timekeeping system for numerous biological rhythms that contribute to the regulation of numerous homeostatic processes in humans. Disruption of circadian rhythms influences physiology and behavior and is associated with adverse health outcomes, especially cancer. However, the underlying molecular mechanisms of circadian disruption-associated cancer initiation and development remain unclear. It is essential to construct good circadian disruption models to uncover and validate the detailed molecular clock framework of circadian disruption in cancer development and progression. Mouse models are the most widely used in circadian studies due to their relatively small size, fast reproduction cycle, easy genome manipulation, and economic practicality. Here, we reviewed the current mouse models of circadian disruption, including suprachiasmatic nuclei destruction, genetic engineering, light disruption, sleep deprivation, and other lifestyle factors in our understanding of the crosstalk between circadian rhythms and oncogenic signaling, as well as the molecular mechanisms of circadian disruption that promotes cancer growth. We focused on the discoveries made with the nocturnal mouse, diurnal human being, and cell culture and provided several circadian rhythm-based cancer therapeutic strategies.
Collapse
Affiliation(s)
- Yu Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haidong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Feng He
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
7
|
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: 16] [Impact Index Per Article: 5.3] [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
|
8
|
Factors Influencing Alzheimer's Disease Risk: Whether and How They are Related to the APOE Genotype. Neurosci Bull 2022; 38:809-819. [PMID: 35149974 PMCID: PMC9276873 DOI: 10.1007/s12264-021-00814-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/25/2021] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease featuring progressive cognitive impairment. Although the etiology of late-onset AD remains unclear, the close association of AD with apolipoprotein E (APOE), a gene that mainly regulates lipid metabolism, has been firmly established and may shed light on the exploration of AD pathogenesis and therapy. However, various confounding factors interfere with the APOE-related AD risk, raising questions about our comprehension of the clinical findings concerning APOE. In this review, we summarize the most debated factors interacting with the APOE genotype and AD pathogenesis, depict the extent to which these factors relate to APOE-dependent AD risk, and discuss the possible underlying mechanisms.
Collapse
|
9
|
Atayde AL, Fischer CE, Schweizer TA, Munoz DG. Neuropsychiatric Inventory-Questionnaire Assessed Nighttime Behaviors in Cognitively Asymptomatic Patients with Pathologically Confirmed Alzheimer's Disease Predict More Rapid Cognitive Deterioration. J Alzheimers Dis 2022; 86:1137-1147. [PMID: 35180114 PMCID: PMC9664561 DOI: 10.3233/jad-215276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND The temporal relationship between sleep, Alzheimer's disease (AD), and cognitive impairment remains to be further elucidated. OBJECTIVE First, we aim to determine whether the Neuropsychiatric Inventory-Questionnaire (NPI-Q) assessed nighttime behaviors prior to cognitive decline influence the rate of cognitive deterioration in pathologically confirmed AD, and second, to assess the possible interactions with APOE allele and cerebral amyloid angiopathy (CAA). METHODS The rate of cognitive decline between cognitively asymptomatic participants from the National Alzheimer Coordinating Center who eventually received a neuropathologic diagnosis of AD with (+NTB) or without (-NTB) nighttime behaviors were compared using independent samples t-test. Participants were stratified by APOE carrier and CAA status. Demographic and patient characteristics were assessed using descriptive statistics, and the independent samples t-test was used for continuous variables and chi-square test for categorical variables. The significance level was set at p≤0.05. RESULTS The rate of cognitive decline was greater in +NTB (n = 74; 3.30 points/year) than -NTB (n = 330; 2.45 points/year) (p = 0.016), even if there was no difference in cognitive status at onset. This difference was restricted to APOE ɛ4 carriers (p = 0.049) and positive CAA participants (p = 0.020). Significance was not reached in non-carriers (p = 0.186) and negative CAA (p = 0.364). APOE and CAA were not differentially distributed between the NTB groups. CONCLUSION NPI-Q assessed nighttime behaviors, a surrogate for sleep disturbances, are associated with more rapidly deteriorating cognition in patients with AD neuropathology who are also carriers of APOE ɛ4 or show CAA.
Collapse
Affiliation(s)
- Adrienne L. Atayde
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, Canada
| | - Corinne E. Fischer
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Faculty of Medicine, Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Tom A. Schweizer
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Canada
- Division of Neurosurgery, St. Michael’s Hospital, Toronto, Canada
| | - David G. Munoz
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, 209 Victoria Street, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Division of Pathology, St. Michael’s Hospital, Toronto, Canada
| |
Collapse
|
10
|
Framnes-DeBoer SN, Jones AA, Kang MY, Propsom K, Nelson LR, Arble DM. The timing of intermittent hypoxia differentially affects macronutrient intake and energy substrate utilization in mice. Am J Physiol Endocrinol Metab 2021; 321:E543-E550. [PMID: 34459217 DOI: 10.1152/ajpendo.00183.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sleep apnea is a common sleep disorder characterized by periodic breathing cessation and intermittent hypoxia (IH). Although previous studies have demonstrated that IH alone can influence metabolic outcomes such as body weight, it remains unclear how the timing of IH can specifically affect these outcomes. Here, we examine how pairing 10-h periods of IH to either the animals' resting phase (e.g., IH during the day) or active phase (e.g., IH during the night) differentially affects body weight, macronutrient selection, energy expenditure, respiratory exchange rate, and glucose tolerance. We find that in contrast to mice exposed to IH during the night, mice exposed to IH during the day preferentially decrease their carbohydrate intake and switch to fat metabolism. Moreover, when the IH stimulus was removed, mice that had been exposed to day IH continued to eat a minimal amount of carbohydrates and consumed a higher percentage of kilocalorie from fat for at least 5 days. These data demonstrate that food choice and substrate utilization are secondary to the timing of IH but not IH itself. Taken together, these data have key clinical implications for individuals with sleep apnea and particularly those who are also experiencing circadian disruption such as night-shift workers.NEW & NOTEWORTHY Pairing repeated hypoxic episodes to a mouse's resting phase during the day preferentially decreases carbohydrate intake and results in a switch to metabolic fat oxidation. These data indicate that the timing of intermittent hypoxia should be considered when calculating sleep apnea's effects on metabolic outcomes.
Collapse
Affiliation(s)
| | - Aaron A Jones
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Michelle Y Kang
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Kat Propsom
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Lauren R Nelson
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| | - Deanna M Arble
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin
| |
Collapse
|
11
|
Burtscher J, Mallet RT, Burtscher M, Millet GP. Hypoxia and brain aging: Neurodegeneration or neuroprotection? Ageing Res Rev 2021; 68:101343. [PMID: 33862277 DOI: 10.1016/j.arr.2021.101343] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022]
Abstract
The absolute reliance of the mammalian brain on oxygen to generate ATP renders it acutely vulnerable to hypoxia, whether at high altitude or in clinical settings of anemia or pulmonary disease. Hypoxia is pivotal to the pathogeneses of myriad neurological disorders, including Alzheimer's, Parkinson's and other age-related neurodegenerative diseases. Conversely, reduced environmental oxygen, e.g. sojourns or residing at high altitudes, may impart favorable effects on aging and mortality. Moreover, controlled hypoxia exposure may represent a treatment strategy for age-related neurological disorders. This review discusses evidence of hypoxia's beneficial vs. detrimental impacts on the aging brain and the molecular mechanisms that mediate these divergent effects. It draws upon an extensive literature search on the effects of hypoxia/altitude on brain aging, and detailed analysis of all identified studies directly comparing brain responses to hypoxia in young vs. aged humans or rodents. Special attention is directed toward the risks vs. benefits of hypoxia exposure to the elderly, and potential therapeutic applications of hypoxia for neurodegenerative diseases. Finally, important questions for future research are discussed.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland; Institute of Sport Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland.
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, CH-1015, Lausanne, Switzerland
| |
Collapse
|
12
|
Cortese R, Khalyfa A, Bao R, Gozal D. Gestational sleep apnea perturbations induce metabolic disorders by divergent epigenomic regulation. Epigenomics 2021; 13:751-765. [PMID: 33929266 DOI: 10.2217/epi-2020-0435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Late-gestational sleep fragmentation (LG-SF) and intermittent hypoxia (LG-IH), two hallmarks of obstructive sleep apnea, lead to metabolic dysfunction in the offspring. We investigated specific biological processes that are epigenetically regulated by LG-SF and LG-IH. Materials & methods: We analyzed DNA methylation profiles in offspring visceral white adipose tissues by MeDIP-chip followed by pathway analysis. Results: We detected 1187 differentially methylated loci (p < 0.01) between LG-SF and LG-IH. Epigenetically regulated genes in LG-SF offspring were associated with lipid and glucose metabolism, whereas those in LG-IH were related to inflammatory signaling and cell proliferation. Conclusion: While LG-SF and LG-IH will result in equivalent phenotypic alterations in offspring, each paradigm appears to operate through epigenetic regulation of different biological processes.
Collapse
Affiliation(s)
- Rene Cortese
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Abdelnaby Khalyfa
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Riyue Bao
- Hillman Cancer Center, UPMC, Pittsburgh, PA 15232, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David Gozal
- Department of Child Health, Child Health Research Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
13
|
Cubillos-Zapata C, Almendros I, Díaz-García E, Toledano V, Casitas R, Galera R, López-Collazo E, Farre R, Gozal D, García-Rio F. Differential effect of intermittent hypoxia and sleep fragmentation on PD-1/PD-L1 upregulation. Sleep 2021; 43:5647611. [PMID: 31782790 DOI: 10.1093/sleep/zsz285] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/27/2019] [Indexed: 12/16/2022] Open
Abstract
Immunosurveillance is compromised in patients with obstructive sleep apnea (OSA) as reflected by overexpression of the programmed death cell receptor and its ligand (PD-1/PD-L1) coinhibitory axis. However, the contributions of intermittent hypoxia (IH) and sleep fragmentation (SF) are unclear. We therefore evaluated the expression of PD-1 and PD-L1 on immune cells from mice subjected to IH or SF, and in human cells exposed to IH, oxidative stress, or both conditions. Six-week-old male C57BL/6J mice were exposed to either IH or SF using previously established in vivo models. Moreover, human peripheral blood mononuclear cells (PBMC) were cultured overnight under normoxia, IH, hydrogen peroxide (H2O2), or both. Murine splenocytes and human PBMC were isolated, and labeled using surface-specific antibodies for flow cytometry analysis. Compared to control mice, IH induced higher expression of PD-L1 on F4/80 cells and of PD-1 on CD4+ and CD8+ T-cells, whereas no significant changes emerged after SF. In vitro models of IH and oxidative stress showed similar changes for expression of PD-L1 on human monocytes and PD-1 on CD4+ T-cells. Furthermore, H2O2 increased PD-1 expression on CD8+ T-cells, compromising their cytotoxic capacity assessed by perforin expression, similar to IH. No evidence of synergistic effects was apparent. Therefore, PD-1/PD-L1 upregulation reported in patients with OSA appears to be preferentially mediated by IH rather than SF.
Collapse
Affiliation(s)
- Carolina Cubillos-Zapata
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Respiratory Diseases Group, Respiratory Service, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Isaac Almendros
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unitat de Biofísica i Bioenginyeria, Facultad de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain
| | - Elena Díaz-García
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Respiratory Diseases Group, Respiratory Service, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Victor Toledano
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,The Innate Immune Response Group, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Raquel Casitas
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Respiratory Diseases Group, Respiratory Service, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Raúl Galera
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Respiratory Diseases Group, Respiratory Service, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Eduardo López-Collazo
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,The Innate Immune Response Group, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Ramón Farre
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unitat de Biofísica i Bioenginyeria, Facultad de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO
| | - Francisco García-Rio
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Respiratory Diseases Group, Respiratory Service, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain.,Departamento de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
14
|
Romanella SM, Roe D, Tatti E, Cappon D, Paciorek R, Testani E, Rossi A, Rossi S, Santarnecchi E. The Sleep Side of Aging and Alzheimer's Disease. Sleep Med 2021; 77:209-225. [PMID: 32912799 PMCID: PMC8364256 DOI: 10.1016/j.sleep.2020.05.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 01/23/2023]
Abstract
As we age, sleep patterns undergo significant modifications in micro and macrostructure, worsening cognition and quality of life. These are associated with remarkable brain changes, like deterioration in synaptic plasticity, gray and white matter, and significant modifications in hormone levels. Sleep alterations are also a core component of mild cognitive impairment (MCI) and Alzheimer's Disease (AD). AD night time is characterized by a gradual decrease in slow-wave activity and a substantial reduction of REM sleep. Sleep abnormalities can accelerate AD pathophysiology, promoting the accumulation of amyloid-β (Aβ) and phosphorylated tau. Thus, interventions that target sleep disturbances in elderly people and MCI patients have been suggested as a possible strategy to prevent or decelerate conversion to dementia. Although cognitive-behavioral therapy and pharmacological medications are still first-line treatments, despite being scarcely effective, new interventions have been proposed, such as sensory stimulation and Noninvasive Brain Stimulation (NiBS). The present review outlines the current state of the art of the relationship between sleep modifications in healthy aging and the neurobiological mechanisms underlying age-related changes. Furthermore, we provide a critical analysis showing how sleep abnormalities influence the prognosis of AD pathology by intensifying Aβ and tau protein accumulation. We discuss potential therapeutic strategies to target sleep disruptions and conclude that there is an urgent need for testing new therapeutic sleep interventions.
Collapse
Affiliation(s)
- S M Romanella
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy
| | - D Roe
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - E Tatti
- Department of Molecular, Cellular & Biomedical Sciences, CUNY, School of Medicine, New York, NY, USA
| | - D Cappon
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - R Paciorek
- Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - E Testani
- Sleep Medicine Center, Department of Neurology, Policlinico Santa Maria Le Scotte, Siena, Italy
| | - A Rossi
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy; Human Physiology Section, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - S Rossi
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy; Human Physiology Section, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - E Santarnecchi
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Italy; Berenson-Allen Center for Noninvasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Pires GN, Benedetto L, Cortese R, Gozal D, Gulia KK, Kumar VM, Tufik S, Andersen ML. Effects of sleep modulation during pregnancy in the mother and offspring: Evidences from preclinical research. J Sleep Res 2020; 30:e13135. [PMID: 32618040 DOI: 10.1111/jsr.13135] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022]
Abstract
Disturbed sleep during gestation may lead to adverse outcomes for both mother and child. Animal research plays an important role in providing insights into this research field by enabling ethical and methodological requirements that are not possible in humans. Here, we present an overview and discuss the main research findings related to the effects of prenatal sleep deprivation in animal models. Using systematic review approaches, we retrieved 42 articles dealing with some type of sleep alteration. The most frequent research topics in this context were maternal sleep deprivation, maternal behaviour, offspring behaviour, development of sleep-wake cycles in the offspring, hippocampal neurodevelopment, pregnancy viability, renal physiology, hypertension and metabolism. This overview indicates that the number of basic studies in this field is growing, and provides biological plausibility to suggest that sleep disturbances might be detrimental to both mother and offspring by promoting increased risk at the behavioural, hormonal, electrophysiological, metabolic and epigenetic levels. More studies on the effects of maternal sleep deprivation are needed, in light of their major translational perspective.
Collapse
Affiliation(s)
- Gabriel Natan Pires
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Luciana Benedetto
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rene Cortese
- Department of Child Health and Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - David Gozal
- Department of Child Health and Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - Kamalesh K Gulia
- Division of Sleep Research, Biomedical Technology Wing - Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | | | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Monica Levy Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Khalyfa A, Castro-Grattoni AL, Gozal D. Cardiovascular morbidities of obstructive sleep apnea and the role of circulating extracellular vesicles. Ther Adv Respir Dis 2020; 13:1753466619895229. [PMID: 31852426 PMCID: PMC6923690 DOI: 10.1177/1753466619895229] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Obstructive sleep apnea (OSA) is characterized by recurrent upper airway collapse
during sleep resulting in impaired blood gas exchange, namely intermittent
hypoxia (IH) and hypercapnia, fragmented sleep (SF), increased oxidative stress
and systemic inflammation. Among a myriad of potential associated morbidities,
OSA has been particularly implicated as mechanistically contributing to the
prevalence and severity of cardiovascular diseases (CVD). However, the benefits
of continuous positive airway pressure (CPAP), which is generally employed in
OSA treatment, to either prevent or improve CVD outcomes remain unconvincing,
suggesting that the pathophysiological mechanisms underlying the incremental CVD
risk associated with OSA are not clearly understood. One of the challenges in
development of non-invasive diagnostic assays is the ability to identify
clinically and mechanistically relevant biomarkers. Circulating extracellular
vesicles (EVs) and their cargos reflect underlying changes in cellular
homeostasis and can provide insights into how cells and systems cope with
physiological perturbations by virtue of the identity and abundance of miRNAs,
mRNAs, proteins, and lipids that are packaged in the EVs under normal as well as
diseased states, such as OSA. EVs can not only provide unique insights into
coordinated cellular responses at the organ or systemic level, but can also
serve as reporters of the effects of OSA in CVD, either by their properties
enabling regeneration and repair of injured vascular cells or by damaging them.
Here, we highlight recent progress in the pathological CVD consequences of OSA,
and explore the putative roles of EVs in OSA-associated CVD, along with emerging
diagnostic and therapeutic opportunities. The reviews of this paper are available via the supplemental material
section.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - Anabel L Castro-Grattoni
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - David Gozal
- Department of Child Health and MU Women's and Children's Hospital, University of Missouri School of Medicine, 400 N. Keene Street, Suite 010, Columbia, MO 65201, USA
| |
Collapse
|
17
|
Shi Y, Luo H, Liu H, Hou J, Feng Y, Chen J, Xing L, Ren X. Related biomarkers of neurocognitive impairment in children with obstructive sleep apnea. Int J Pediatr Otorhinolaryngol 2019; 116:38-42. [PMID: 30554705 DOI: 10.1016/j.ijporl.2018.10.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Different experiment approaches have demonstrated that children with obstructive sleep apnea (OSA) exhibit neurocognitive and behavioral deficits. This review summarized the potential biomarkers of OSA-associated neurocognitive impairment in children. METHODS A scoping review of studies on children with OSA that evaluated the potential value of different markers in identifying neurocognitive impairment was undertaken. Additionally, the biomarkers were categorized according to the different research methods, including brain imaging studies, serological indicators and urine markers. RESULTS Majority of the studies that evaluated blood biomarkers, plasma insulin growth factor-1 (IGF-1) and Alzheimer's disease (AD)-related biomarkers appeared to exhibit a favorable profile, and could discriminate between OSA children with or without neurocognitive impairments. Brain imaging studies and urinary neurotransmitters could also be helpful for screening OSA cognitive morbidity in children. CONCLUSION Due to limited research methods available in children, the cognitive susceptibility of children with OSA has been rarely studied. The main reason for this may be the limited research methods in children. Numerous study populations of children and complex psychological tests are required, which involve major labor and costs.Multi-center prospective studies are needed to identify suitable biomarkers for the timely prediction and effective intervention to prevent neurocognitive impairment in children with OSA and to explore further opportunities in this arena.
Collapse
Affiliation(s)
- Yewen Shi
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Huanan Luo
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Haiqin Liu
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Jin Hou
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Yani Feng
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Jinwei Chen
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Liang Xing
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Xiaoyong Ren
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
18
|
Nair D, Ramesh V, Gozal D. Cognitive Deficits Are Attenuated in Neuroglobin Overexpressing Mice Exposed to a Model of Obstructive Sleep Apnea. Front Neurol 2018; 9:426. [PMID: 29922222 PMCID: PMC5996123 DOI: 10.3389/fneur.2018.00426] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Obstructive sleep apnea (OSA) is a highly prevalent disease manifesting as intermittent hypoxia during sleep (IH) and is increasingly recognized as being independently associated with neurobehavioral deficits. These deficits may be due to increased apoptosis in the hippocampus and cerebral cortex, as well as increased oxidative stress and inflammation. It has been reported that neuroglobin (Ngb) is upregulated in response to hypoxia-ischemia insults and exhibits a protective role in ischemia-reperfusion brain injury. We hypothesized that transgenic overexpression of Ngb would attenuate spatial learning deficits in a murine model of OSA. Methods:Wild-type mice and Ngb overexpressing male mice (Ngb-TG) were randomly assigned to either IH or room air (RA) exposures. The effects of IH during the light period on performance in a water maze spatial task were assessed, as well as anxiety and depressive-like behaviors using elevated plus maze (EPM) and forced swim tests. Cortical tissues from all the mice were extracted for biochemical studies for lipid peroxidation. Results:Ngb TG mice exhibited increased Ngb immunoreactivity in brain tissues and IH did not elicit significant changes in Ngb expression in either Ngb-TG mice or WT mice. On a standard place training task in the water maze, Ngb-TG mice displayed preserved spatial learning, and were protected from the reduced spatial learning performances observed in WT mice exposed to IH. Furthermore, anxiety and depression levels were enhanced in WT mice exposed to IH as compared to RA controls, while alterations emerged in Ngb-TG mice exposed to IH. Furthermore, WT mice, but not Ngb-TG mice had significantly elevated levels of malondialdehyde in cortical lysates following IH exposures. Conclusions:In a murine model of OSA, oxidative stress responses and neurocognitive and behavioral impairments induced by IH during sleep are attenuated by the neuroprotective effects of Ngb.
Collapse
Affiliation(s)
- Deepti Nair
- Section of Sleep Medicine, Biological Sciences Division, Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, United States.,Atlantic Health System, Morristown, NJ, United States.,Biomedical Research Institute of New Jersey, Cedar Knolls, NJ, United States
| | - Vijay Ramesh
- Section of Sleep Medicine, Biological Sciences Division, Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, United States
| | - David Gozal
- Section of Sleep Medicine, Biological Sciences Division, Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
19
|
Chong SYC, Xin L, Ptáček LJ, Fu YH. Disorders of sleep and circadian rhythms. HANDBOOK OF CLINICAL NEUROLOGY 2018; 148:531-538. [PMID: 29478598 DOI: 10.1016/b978-0-444-64076-5.00034-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sleep is fundamental to the survival of humans. However, knowledge regarding the role of sleep and its regulation is poorly understood. Genetics in flies, mice, and humans has led to a detailed understanding of some aspects of circadian regulation. Sleep homeostasis (the effect of increasing periods of wakefulness on our sleep propensity) is largely not understood. Sleep homeostasis is distinct from, but also linked to, the circadian clock. It is only in the last two decades that our understanding of some sleep disorders has been revealed. These breakthroughs were mostly fueled by intensive investigation using genetic tools. Although modern human genetics has revolutionized scientific research of neurologic disorders beginning ~35 years ago, studies of sleep and sleep disorders have lagged behind those of many neurologic diseases. This is due to the complexity in phenotyping behaviors like sleep and the fact that sleep is strongly influenced by environmental and other factors. We have long been aware that the amount of sleep required by individuals is normally distributed in the general population with small proportions of people being natural short or natural long sleepers. However, it has been less than a decade since Mendelian families of natural short sleepers have been recognized. Recent work has made significant advances and mechanistic insights of several sleep disorders as well as familial natural short sleepers by using ever-improving human genetic and cellular molecular tools. Given recent advances into genetic and biologic understanding of sleep, the hope of understanding this indispensable process is closer. Ultimately, our growing understanding will lead to more effective treatments of human sleep disorders.
Collapse
Affiliation(s)
- S Y Christin Chong
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Lijuan Xin
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Louis J Ptáček
- Department of Neurology, University of California, San Francisco, CA, United States; Howard Hughes Medical Institute, San Francisco, CA, United States
| | - Ying-Hui Fu
- Department of Neurology, University of California, San Francisco, CA, United States.
| |
Collapse
|
20
|
Kincheski GC, Valentim IS, Clarke JR, Cozachenco D, Castelo-Branco MTL, Ramos-Lobo AM, Rumjanek VMBD, Donato J, De Felice FG, Ferreira ST. Chronic sleep restriction promotes brain inflammation and synapse loss, and potentiates memory impairment induced by amyloid-β oligomers in mice. Brain Behav Immun 2017; 64:140-151. [PMID: 28412140 DOI: 10.1016/j.bbi.2017.04.007] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/02/2017] [Accepted: 04/10/2017] [Indexed: 12/24/2022] Open
Abstract
It is increasingly recognized that sleep disturbances and Alzheimer's disease (AD) share a bidirectional relationship. AD patients exhibit sleep problems and alterations in the regulation of circadian rhythms; conversely, poor quality of sleep increases the risk of development of AD. The aim of the current study was to determine whether chronic sleep restriction potentiates the brain impact of amyloid-β oligomers (AβOs), toxins that build up in AD brains and are thought to underlie synapse damage and memory impairment. We further investigated whether alterations in levels of pro-inflammatory mediators could play a role in memory impairment in sleep-restricted mice. We found that a single intracerebroventricular (i.c.v.) infusion of AβOs disturbed sleep pattern in mice. Conversely, chronically sleep-restricted mice exhibited higher brain expression of pro-inflammatory mediators, reductions in levels of pre- and post-synaptic marker proteins, and exhibited increased susceptibility to the impact of i.c.v. infusion of a sub-toxic dose of AβOs (1pmol) on performance in the novel object recognition memory task. Sleep-restricted mice further exhibited an increase in brain TNF-α levels in response to AβOs. Interestingly, memory impairment in sleep-restricted AβO-infused mice was prevented by treatment with the TNF-α neutralizing monoclonal antibody, infliximab. Results substantiate the notion of a dual relationship between sleep and AD, whereby AβOs disrupt sleep/wake patterns and chronic sleep restriction increases brain vulnerability to AβOs, and point to a key role of brain inflammation in increased susceptibility to AβOs in sleep-restricted mice.
Collapse
Affiliation(s)
- Grasielle C Kincheski
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Isabela S Valentim
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vivian M B D Rumjanek
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
21
|
Jorba I, Menal MJ, Torres M, Gozal D, Piñol-Ripoll G, Colell A, Montserrat JM, Navajas D, Farré R, Almendros I. Ageing and chronic intermittent hypoxia mimicking sleep apnea do not modify local brain tissue stiffness in healthy mice. J Mech Behav Biomed Mater 2017; 71:106-113. [DOI: 10.1016/j.jmbbm.2017.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 01/08/2023]
|
22
|
Degeneration and energy shortage in the suprachiasmatic nucleus underlies the circadian rhythm disturbance in ApoE -/- mice: implications for Alzheimer's disease. Sci Rep 2016; 6:36335. [PMID: 27824104 PMCID: PMC5099891 DOI: 10.1038/srep36335] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 10/05/2016] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) patients suffer sleep disorders and circadian rhythm disturbances (CRDs). The underlying mechanisms are incompletely understood, and treatments are lacking. In this study, we characterized the locomotor activity, clock gene expression, morphological degeneration and energy metabolism of suprachiasmatic nucleus (SCN), together with retinal light sensing, in ApoE-/- mice, a model for AD. Compared with the control C57BL/6J mice, ApoE-/- mice exhibited disordered circadian locomotor activity under dim light and constant darkness, with impaired re-entrainment to phase change schedules. Decreased retinal melanopsin expression, together with amyloidosis and tau deposition, was evident in ApoE-/- mice. Mitochondrial and synaptic deterioration, altered SIRT1-mediated energy metabolism and clock gene expression were also observed in ApoE-/- SCN. Supplementation with fat or ketone bodies but not glucose, or intraperitoneal administration of nicotinamide, restored the locomotor rhythmicity and circadian expression of SIRT1 and clock genes, as well as reducing neurodegeneration. Taken together, ApoE deficiency induced degeneration and a significant disturbance in the SCN rhythmicity. Decline of retinal light sensing and SCN structural and metabolic deteriorations represented the major pathologies accounting for the CRDs in ApoE-/- mice. Our curative experiments may help develop future therapies to treat the CRDs and sleep disorders in AD patients.
Collapse
|
23
|
D'Rozario AL, Cross NE, Vakulin A, Bartlett DJ, Wong KKH, Wang D, Grunstein RR. Quantitative electroencephalogram measures in adult obstructive sleep apnea - Potential biomarkers of neurobehavioural functioning. Sleep Med Rev 2016; 36:29-42. [PMID: 28385478 DOI: 10.1016/j.smrv.2016.10.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/15/2016] [Accepted: 10/08/2016] [Indexed: 10/20/2022]
Abstract
Obstructive sleep apnea (OSA) results in significantly impaired cognitive functioning and increased daytime sleepiness in some patients leading to increased risk of motor vehicle and workplace accidents and reduced productivity. Clinicians often face difficulty in identifying which patients are at risk of neurobehavioural dysfunction due to wide inter-individual variability, and disparity between symptoms and conventional metrics of disease severity such as the apnea hypopnea index. Quantitative electroencephalogram (EEG) measures are determinants of awake neurobehavioural function in healthy subjects. However, the potential value of quantitative EEG (qEEG) measurements as biomarkers of neurobehavioural function in patients with OSA has not been examined. This review summarises the existing literature examining qEEG in OSA patients including changes in brain activity during wake and sleep states, in relation to daytime sleepiness, cognitive impairment and OSA treatment. It will speculate on the mechanisms which may underlie changes in EEG activity and discuss the potential utility of qEEG as a clinically useful predictor of neurobehavioural function in OSA.
Collapse
Affiliation(s)
- Angela L D'Rozario
- CIRUS Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; School of Psychology, Faculty of Science, Brain and Mind Centre and Charles Perkins Centre, The University of Sydney, Australia; Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital & Sydney Local Health District, Sydney, NSW, Australia.
| | - Nathan E Cross
- CIRUS Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Andrew Vakulin
- CIRUS Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; Adelaide Institute for Sleep Health: A Flinders Centre of Research Excellence, School of Medicine, Faculty of Medicine, Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, Australia
| | - Delwyn J Bartlett
- CIRUS Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, NSW, Australia
| | - Keith K H Wong
- CIRUS Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital & Sydney Local Health District, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, NSW, Australia
| | - David Wang
- CIRUS Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital & Sydney Local Health District, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, NSW, Australia
| | - Ronald R Grunstein
- CIRUS Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital & Sydney Local Health District, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, NSW, Australia
| |
Collapse
|
24
|
Kheirandish-Gozal L, Philby MF, Alonso-Álvarez ML, Terán-Santos J, Gozal D. Biomarkers of Alzheimer Disease in Children with Obstructive Sleep Apnea: Effect of Adenotonsillectomy. Sleep 2016; 39:1225-32. [PMID: 27070140 DOI: 10.5665/sleep.5838] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/02/2016] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVE Obese children are at increased risk for developing obstructive sleep apnea (OSA), and both of these conditions are associated with an increased risk for end-organ morbidities. Both OSA and obesity (OB) have been associated with increased risk for Alzheimer disease (AD). This study aimed to assess whether OSA and OB lead to increased plasma levels of 2 AD markers amyloid β protein 42 (Aβ42) and pre-senilin 1 (PS1). METHODS Fasting morning plasma samples from otherwise healthy children with a diagnosis of OB, OSA, or both (OSA+OB), and controls, and in a subset of children with OSA after adenotonsillectomy (T&A) were assayed for Aβ42 and PS1 levels using commercial enzyme-linked immunosorbent assay kits. RESULTS 286 children (mean age of 7.2 ± 2.7 y) were evaluated. Compared to control subjects, OB children had similar Aβ42 (108.3 ± 31.7 pg/mL versus 83.6 ± 14.6 pg/mL) and PS1 levels (0.89 ± 0.44 ng/mL versus 0.80 ± 0.29 pg/mL). However, OSA children (Aβ42: 186.2 ± 66.7 pg/mL; P < 0.001; PS1: 3.42 ± 1.46 ng/mL; P < 0.001), and particularly OSA+OB children had significant elevations in both Aβ42 (349.4 ± 112.9 pg/mL; P < 0.001) and PS1 (PS1: 4.54 ± 1.16 ng/mL; P < 0.001) circulating concentrations. In a subset of 24 children, T&A resulted in significant reductions of Aβ42 (352.0 ± 145.2 versus 151.9 ± 81.4 pg/mL; P < 0.0001) and PS1 (4.82 ± 1.09 versus 2.02 ± 1.18 ng/mL; P < 0.0001). CONCLUSIONS Thus, OSA, and particularly OSA+OB, are associated with increased plasma levels of AD biomarkers, which decline upon treatment of OSA in a representative, yet not all- encompassing subset of patients, suggesting that OSA may accelerate AD-related processes even in early childhood. However, the cognitive and overall health-related implications of these findings remain to be defined.
Collapse
Affiliation(s)
- Leila Kheirandish-Gozal
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Mona F Philby
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - María Luz Alonso-Álvarez
- Sleep Unit, CIBER of Respiratory Diseases, Instituto Carlos III, CIBERES, Hospital Universitario de Burgos (HUBU), Burgos, Spain
| | - Joaquin Terán-Santos
- Sleep Unit, CIBER of Respiratory Diseases, Instituto Carlos III, CIBERES, Hospital Universitario de Burgos (HUBU), Burgos, Spain
| | - David Gozal
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| |
Collapse
|
25
|
Gileles-Hillel A, Kheirandish-Gozal L, Gozal D. Biological plausibility linking sleep apnoea and metabolic dysfunction. Nat Rev Endocrinol 2016; 12:290-8. [PMID: 26939978 DOI: 10.1038/nrendo.2016.22] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obstructive sleep apnoea (OSA) is a very common disorder that affects 10-25% of the general population. In the past two decades, OSA has emerged as a cardiometabolic risk factor in both paediatric and adult populations. OSA-induced metabolic perturbations include dyslipidaemia, atherogenesis, liver dysfunction and abnormal glucose metabolism. The mainstay of treatment for OSA is adenotonsillectomy in children and continuous positive airway pressure therapy in adults. Although these therapies are effective at resolving the sleep-disordered breathing component of OSA, they do not always produce beneficial effects on metabolic function. Thus, a deeper understanding of the underlying mechanisms by which OSA influences metabolic dysfunction might yield improved therapeutic approaches and outcomes. In this Review, we summarize the evidence obtained from animal models and studies of patients with OSA of potential mechanistic pathways linking the hallmarks of OSA (intermittent hypoxia and sleep fragmentation) with metabolic dysfunction. Special emphasis is given to adipose tissue dysfunction induced by sleep apnoea, which bears a striking resemblance to adipose dysfunction resulting from obesity. In addition, important gaps in current knowledge and promising lines of future investigation are identified.
Collapse
Affiliation(s)
- Alex Gileles-Hillel
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Knapp Center for Biomedical Discovery, Room 4100, 900 East 57th Street, Mailbox 4, Chicago, Illinois 60637-1470, USA
| | - Leila Kheirandish-Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Knapp Center for Biomedical Discovery, Room 4100, 900 East 57th Street, Mailbox 4, Chicago, Illinois 60637-1470, USA
| | - David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Knapp Center for Biomedical Discovery, Room 4100, 900 East 57th Street, Mailbox 4, Chicago, Illinois 60637-1470, USA
| |
Collapse
|
26
|
Cortese R, Almendros I, Wang Y, Gozal D. Tumor circulating DNA profiling in xenografted mice exposed to intermittent hypoxia. Oncotarget 2016; 6:556-69. [PMID: 25415227 PMCID: PMC4381615 DOI: 10.18632/oncotarget.2785] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/15/2014] [Indexed: 01/06/2023] Open
Abstract
Intermittent hypoxia (IH) a hallmark characteristic of obstructive sleep apnea (OSA), is proposed as a major determinant of processes involving tumor growth, invasion and metastasis. To examine whether circulating DNA (cirDNA) in blood plasma reflects changes in tumor cells under IH conditions, we used a xenografted murine model. Mice engrafted with TC1 epithelial lung cancer cells and controls were exposed to IH or room air (RA) conditions. Plasma cirDNA amounts were significantly increased in mice exposed to IH (p<0.05). Significant associations between plasma cirDNA concentrations and tumor size, weight and invasiveness also emerged (p<0.05). Using a methylation microarray-based approach, we identified 2,094 regions showing significant differential cirDNA modifications. Systems biology analyses revealed an association with molecular pathways deregulated in cancer progression and with distal and TSS-associated transcription factor binding sites. We detected clusters of highly variable regions in chromosomes 7, 13, 14 and X, which may highlight hotspots for DNA deletions. Single locus displayed high intragroup variation, suggesting cellular heterogeneity within the tissue may be associated to cirDNA release. Thus, exposures to IH increase the shedding of cirDNA into circulation, which carries epigenetic modifications that may characterize cell populations within the tumor that preferentially release their DNA upon IH exposure.
Collapse
Affiliation(s)
- Rene Cortese
- Department of Pediatrics, Section of Pediatric Sleep Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Isaac Almendros
- Department of Pediatrics, Section of Pediatric Sleep Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Yang Wang
- Department of Pediatrics, Section of Pediatric Sleep Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - David Gozal
- Department of Pediatrics, Section of Pediatric Sleep Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| |
Collapse
|
27
|
Waller KL, Mortensen EL, Avlund K, Osler M, Fagerlund B, Lauritzen M, Jennum P. Subjective sleep quality and daytime sleepiness in late midlife and their association with age-related changes in cognition. Sleep Med 2016; 17:165-73. [DOI: 10.1016/j.sleep.2015.01.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 12/04/2014] [Accepted: 01/05/2015] [Indexed: 12/13/2022]
|
28
|
Miller MA. The Role of Sleep and Sleep Disorders in the Development, Diagnosis, and Management of Neurocognitive Disorders. Front Neurol 2015; 6:224. [PMID: 26557104 PMCID: PMC4615953 DOI: 10.3389/fneur.2015.00224] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/12/2015] [Indexed: 12/31/2022] Open
Abstract
It is becoming increasingly apparent that sleep plays an important role in the maintenance, disease prevention, repair, and restoration of both mind and body. The sleep and wake cycles are controlled by the pacemaker activity of the superchiasmic nucleus in the hypothalamus but can be disrupted by diseases of the nervous system causing disordered sleep. A lack of sleep has been associated with an increase in all-cause mortality. Likewise, sleep disturbances and sleep disorders may disrupt neuronal pathways and have an impact on neurological diseases. Sleep deprivation studies in normal subjects demonstrate that a lack of sleep can cause attention and working memory impairment. Moreover, untreated sleep disturbances and sleep disorders such as obstructive sleep apnoe (OSA) can also lead to cognitive impairment. Poor sleep and sleep disorders may present a significant risk factor for the development of dementia. In this review, the underlying mechanisms and the role of sleep and sleep disorders in the development of neurocognitive disorders [dementia and mild cognitive impairment (MCI)] and how the presence of sleep disorders could direct the process of diagnosis and management of neurocognitive disorders will be discussed.
Collapse
|
29
|
Das D, Medina B, Baktir MA, Mojabi FS, Fahimi A, Ponnusamy R, Salehi A. Increased incidence of intermittent hypoxemia in the Ts65Dn mouse model of Down syndrome. Neurosci Lett 2015; 604:91-6. [PMID: 26240993 DOI: 10.1016/j.neulet.2015.07.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 01/13/2023]
Abstract
In addition to nervous system, cardiovascular and respiratory systems are primarily affected in Down syndrome (DS). The Ts65Dn mouse model is widely used to recapitulate cognitive dysfunction in DS. While these mice consistently show failure in learning and memory along with functional and structural abnormalities in the hippocampal region, the underlying mechanisms behind cognitive dysfunction remain to be fully elucidated. Convergent evidence implicates chronic episodes of hypoxemia in cognitive dysfunction in people with DS. Using an infra-red detection system to assess oxygen saturation in free-moving mice, we assessed arterial blood oxygenation in both adolescent and adult Ts65Dn mice and found a significant increase in the incidence of hypoxemia in both groups. Notably, the severity of hypoxemia increased during the dark cycle, suggesting a link between hypoxemia and increased motor activity. Postmortem analysis showed significant increase in the expression of mitochondrial Cox4i2, the terminal enzyme of the mitochondrial respiratory chain and oxygen response element. Altogether these data suggest early and chronic occurrence of hypoxemia in the Ts65Dn mouse model of DS, which can contribute to cognitive dysfunction in these mice.
Collapse
Affiliation(s)
- Devsmita Das
- VA Palo Alto Health Care System, Palo Alto, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, CA 94305, USA
| | - Brian Medina
- VA Palo Alto Health Care System, Palo Alto, CA 94305, USA
| | | | - Fatemeh S Mojabi
- VA Palo Alto Health Care System, Palo Alto, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, CA 94305, USA
| | - Atoossa Fahimi
- VA Palo Alto Health Care System, Palo Alto, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, CA 94305, USA
| | - Ravikumar Ponnusamy
- VA Palo Alto Health Care System, Palo Alto, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, CA 94305, USA
| | - Ahmad Salehi
- VA Palo Alto Health Care System, Palo Alto, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, CA 94305, USA.
| |
Collapse
|
30
|
Kang D, Lim HK, Jung WS, Jeong JH, Kim TW, Han JH, Lee CU, Hong SC. Sleep and Alzheimer’s Disease. SLEEP MEDICINE RESEARCH 2015. [DOI: 10.17241/smr.2015.6.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
31
|
Gozal D, Farré R, Nieto FJ. Obstructive sleep apnea and cancer: Epidemiologic links and theoretical biological constructs. Sleep Med Rev 2015; 27:43-55. [PMID: 26447849 DOI: 10.1016/j.smrv.2015.05.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/21/2015] [Accepted: 05/25/2015] [Indexed: 12/19/2022]
Abstract
Sleep disorders have emerged as highly prevalent conditions in the last 50-75 y. Along with improved understanding of such disorders, the realization that perturbations in sleep architecture and continuity may initiate, exacerbate or modulate the phenotypic expression of multiple diseases including cancer has gained increased attention. Furthermore, the intermittent hypoxia that is attendant to sleep disordered breathing, has recently been implicated in increased incidence and more adverse prognosis of cancer. The unifying conceptual framework linking these associations proposes that increased sympathetic activity and/or alterations in immune function, particularly affecting innate immune cellular populations, underlie the deleterious effects of sleep disorders on tumor biology. In this review, the epidemiological evidence linking disrupted sleep and intermittent hypoxia to oncological outcomes, and the potential biological underpinnings of such associations as illustrated by experimental murine models will be critically appraised. The overarching conclusion appears supportive in the formulation of an hypothetical framework, in which fragmented sleep and intermittent hypoxia may promote changes in multiple signalosomes and transcription factors that can not only initiate malignant transformation, but will also alter the tumor microenvironment, disrupt immunosurveillance, and thus hasten tumor proliferation and increase local and metastatic invasion. Future bench-based experimental studies as well as carefully conducted and controlled clinical epidemiological studies appear justified for further exploration of these hypotheses.
Collapse
Affiliation(s)
- David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA.
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona-IDIBAPS, Barcelona, Spain; CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - F Javier Nieto
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
32
|
Sleep and Alzheimer's disease. Sleep Med Rev 2015; 19:29-38. [DOI: 10.1016/j.smrv.2014.03.007] [Citation(s) in RCA: 301] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 03/03/2014] [Accepted: 03/25/2014] [Indexed: 11/18/2022]
|
33
|
Lim DC, Brady DC, Po P, Chuang LP, Marcondes L, Kim EY, Keenan BT, Guo X, Maislin G, Galante RJ, Pack AI. Simulating obstructive sleep apnea patients' oxygenation characteristics into a mouse model of cyclical intermittent hypoxia. J Appl Physiol (1985) 2014; 118:544-57. [PMID: 25429097 DOI: 10.1152/japplphysiol.00629.2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mouse models of cyclical intermittent hypoxia (CIH) are used to study the consequences of both hypoxia and oxidative stress in obstructive sleep apnea (OSA). Whether or not a mouse model of CIH that simulates OSA patients' oxygenation characteristics would translate into improved patient care remains unanswered. First we identified oxygenation characteristics using the desaturation and resaturation time in 47 OSA subjects from the Molecular Signatures of Obstructive Sleep Apnea Cohort (MSOSA). We observe that a cycle of intermittent hypoxia is not sinusoidal; specifically, desaturation time increases in an almost linear relationship to the degree of hypoxia (nadir), whereas resaturation time is somewhat constant (∼15 s), irrespective of the nadir. Second, we modified the Hycon mouse model of CIH to accommodate a 15-s resaturation time. Using this modified CIH model, we explored whether a short resaturation schedule (15 s), which includes the characteristics of OSA patients, had a different effect on levels of oxidative stress (i.e., urinary 8,12-iso-iPF2α-VI levels) compared with sham and a long resaturation schedule (90 s), a schedule that is not uncommon in rodent models of CIH. Results suggest that shorter resaturation time may result in a higher level of 8,12-iso-iPF2α-VI compared with long resaturation or sham conditions. Therefore, simulating the rodent model of CIH to reflect this and other OSA patients' oxygenation characteristics may be worthy of consideration to better understand the effects of hypoxia, oxidative stress, and their interactions.
Collapse
Affiliation(s)
- Diane C Lim
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania;
| | - Daniel C Brady
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Pengse Po
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Li Pang Chuang
- Department of Thoracic Medicine and Department of Sleep Center, Chang Gung Memorial Hospital, Taipei, Taiwan and Graduate Institute of Clinical Medical Sciences, Chang Gung University, Tauyan, Taiwan; and
| | | | - Emily Y Kim
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brendan T Keenan
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaofeng Guo
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Greg Maislin
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Raymond J Galante
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Allan I Pack
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Carreras A, Zhang SX, Peris E, Qiao Z, Gileles-Hillel A, Li RC, Wang Y, Gozal D. Chronic sleep fragmentation induces endothelial dysfunction and structural vascular changes in mice. Sleep 2014; 37:1817-24. [PMID: 25364077 DOI: 10.5665/sleep.4178] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
STUDY OBJECTIVES Sleep fragmentation (SF) is a common occurrence and constitutes a major characteristic of obstructive sleep apnea (OSA). SF has been implicated in multiple OSA-related morbidities, but it is unclear whether SF underlies any of the cardiovascular morbidities of OSA. We hypothesized that long-term SF exposures may lead to endothelial dysfunction and altered vessel wall structure. METHODS AND RESULTS Adult male C57BL/6J mice were fed normal chow and exposed to daylight SF or control sleep (CTL) for 20 weeks. Telemetric blood pressure and endothelial function were assessed weekly using a modified laser-Doppler hyperemic test. Atherosclerotic plaques, elastic fiber disruption, lumen area, wall thickness, foam cells, and macrophage recruitment, as well as expression of senescence-associated markers were examined in excised aortas. Increased latencies to reach baseline perfusion levels during the post-occlusive period emerged in SF mice with increased systemic BP values starting at 8 weeks of SF and persisting thereafter. No obvious atherosclerotic plaques emerged, but marked elastic fiber disruption and fiber disorganization were apparent in SF-exposed mice, along with increases in the number of foam cells and macrophages in the aorta wall. Senescence markers showed reduced TERT and cyclin A and increased p16INK4a expression, with higher IL-6 plasma levels in SF-exposed mice. CONCLUSIONS Long-term sleep fragmentation induces vascular endothelial dysfunction and mild blood pressure increases. Sleep fragmentation also leads to morphologic vessel changes characterized by elastic fiber disruption and disorganization, increased recruitment of inflammatory cells, and altered expression of senescence markers, thereby supporting a role for sleep fragmentation in the cardiovascular morbidity of OSA.
Collapse
Affiliation(s)
- Alba Carreras
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Shelley X Zhang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Eduard Peris
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Zhuanhong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Alex Gileles-Hillel
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Richard C Li
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Yang Wang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| |
Collapse
|
35
|
Pan W, Kastin AJ. Can sleep apnea cause Alzheimer's disease? Neurosci Biobehav Rev 2014; 47:656-69. [DOI: 10.1016/j.neubiorev.2014.10.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/17/2014] [Accepted: 10/22/2014] [Indexed: 11/29/2022]
|
36
|
Khalyfa A, Wang Y, Zhang SX, Qiao Z, Abdelkarim A, Gozal D. Sleep fragmentation in mice induces nicotinamide adenine dinucleotide phosphate oxidase 2-dependent mobilization, proliferation, and differentiation of adipocyte progenitors in visceral white adipose tissue. Sleep 2014; 37:999-1009. [PMID: 24790279 DOI: 10.5665/sleep.3678] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Chronic sleep fragmentation (SF) without sleep curtailment induces increased adiposity. However, it remains unclear whether mobilization, proliferation, and differentiation of adipocyte progenitors (APs) occurs in visceral white adipose tissue (VWAT), and whether nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2) activity plays a role. METHODS Changes in VWAT depot cell size and AP proliferation were assessed in wild-type and Nox2 null male mice exposed to SF and control sleep (SC). To assess mobilization, proliferation, and differentiation of bone marrow mesenchymal stem cells (BM-MSC), Sca-1+ bone marrow progenitors were isolated from GFP+ or RFP+ mice, and injected intravenously to adult male mice (C57BL/6) previously exposed to SF or SC. RESULTS In comparison with SC, SF was associated with increased weight accrual at 3 w and thereafter, larger subcutaneous and visceral fat depots, and overall adipocyte size at 8 w. Increased global AP numbers in VWAT along with enhanced AP BrDU labeling in vitro and in vivo emerged in SF. Systemic injections of GFP+ BM-MSC resulted in increased AP in VWAT, as well as in enhanced differentiation into adipocytes in SF-exposed mice. No differences occurred between SF and SC in Nox2 null mice for any of these measurements. CONCLUSIONS Chronic sleep fragmentation (SF) induces obesity in mice and increased proliferation and differentiation of adipocyte progenitors (AP) in visceral white adipose tissue (VWAT) that are mediated by increased Nox2 activity. In addition, enhanced migration of bone marrow mesenchymal stem cells from the systemic circulation into VWAT, along with AP differentiation, proliferation, and adipocyte formation occur in SF-exposed wild-type but not in oxidase 2 (Nox2) null mice. Thus, Nox2 may provide a therapeutic target to prevent obesity in the context of sleep disorders.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Yang Wang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Shelley X Zhang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Zhuanhong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Amal Abdelkarim
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| |
Collapse
|
37
|
Kinugawa K, Nguyen-Michel VH, Mariani J. [Obstructive sleep apnea syndrome: a cause of cognitive disorders in the elderly?]. Rev Med Interne 2014; 35:664-9. [PMID: 24630586 DOI: 10.1016/j.revmed.2014.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 12/19/2013] [Accepted: 02/03/2014] [Indexed: 11/19/2022]
Abstract
Obstructive sleep apnea syndrome is a chronic disease characterized by repeated upper airway obstructions during sleep, resulting in fragmented sleep with arousals, nocturnal intermittent hypoxemia and diurnal dysfunctions. Despite its high prevalence in elderly, sleep apnea syndrome seems to be underestimated and difficult to be recognized because of the lack of clinical symptoms specificity in this population. Among the numerous consequences of the obstructive sleep apnea syndrome, cognitive impairment prevails on the attention, executive functions and memory. Neuroimaging studies in human and experimental models allowed to highlight neural correlates of these cognitive dysfunctions in obstructive sleep apnea syndrome. The obstructive sleep apnea syndrome with cognitive impairment shares some features with Alzheimer's disease, involving genetic predisposition ApoE4, hippocampus and synaptic plasticity abnormalities. In this context, the question arises whether obstructive sleep apnea syndrome is a possible etiological or aggravating factor of cognitive decline in elderly with mild cognitive impairment or Alzheimer's disease. Although there are conflicting results in studies evaluating therapeutic efficiency of continuous positive air pressure, obstructive sleep apnea syndrome seems nevertheless as a correctable factor, at least for its impact on some cognitive consequences. Looking for sleep apnea syndrome in elderly with cognitive decline should be considered in a global, diagnosis and therapeutic management.
Collapse
Affiliation(s)
- K Kinugawa
- Unité d'explorations fonctionnelles du sujet âgé, hôpital Charles-Foix, Assistance publique-Hôpitaux de Paris, 7, avenue de la République, 94005 Ivry-sur-Seine, France; UMR 8256 « Adaptation Biologique et Vieillissement », UPMC/CNRS - Sorbonne universités, 4, place Jussieu, 75005 Paris, France; Institut de la longévité Charles-Foix, 7, avenue de la République, 92005 Ivry-sur-Seine, France.
| | - V H Nguyen-Michel
- Unité d'explorations fonctionnelles du sujet âgé, hôpital Charles-Foix, Assistance publique-Hôpitaux de Paris, 7, avenue de la République, 94005 Ivry-sur-Seine, France
| | - J Mariani
- Unité d'explorations fonctionnelles du sujet âgé, hôpital Charles-Foix, Assistance publique-Hôpitaux de Paris, 7, avenue de la République, 94005 Ivry-sur-Seine, France; UMR 8256 « Adaptation Biologique et Vieillissement », UPMC/CNRS - Sorbonne universités, 4, place Jussieu, 75005 Paris, France; Institut de la longévité Charles-Foix, 7, avenue de la République, 92005 Ivry-sur-Seine, France
| |
Collapse
|
38
|
Lim ASP, Yu L, Kowgier M, Schneider JA, Buchman AS, Bennett DA. Modification of the relationship of the apolipoprotein E ε4 allele to the risk of Alzheimer disease and neurofibrillary tangle density by sleep. JAMA Neurol 2014; 70:1544-51. [PMID: 24145819 DOI: 10.1001/jamaneurol.2013.4215] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
IMPORTANCE The apolipoprotein E (APOE [GenBank, 348; OMIM, 107741]) ε4 allele is a common and well-established genetic risk factor for Alzheimer disease (AD). Sleep consolidation is also associated with AD risk, and previous work suggests that APOE genotype and sleep may interact to influence cognitive function. OBJECTIVE To determine whether better sleep consolidation attenuates the relationship of the APOE genotype to the risk of incident AD and the burden of AD pathology. DESIGN, SETTING, AND PARTICIPANTS A prospective longitudinal cohort study with up to 6 years of follow-up was conducted. Participants included 698 community-dwelling older adults without dementia (mean age, 81.7 years; 77% women) in the Rush Memory and Aging Project. EXPOSURES We used up to 10 days of actigraphic recording to quantify the degree of sleep consolidation and ascertained APOE genotype. MAIN OUTCOMES AND MEASURES Participants underwent annual evaluation for AD during a follow-up period of up to 6 years. Autopsies were performed on 201 participants who died, and β-amyloid (Aβ) and neurofibrillary tangles were identified by immunohistochemistry and quantified. RESULTS During the follow-up period, 98 individuals developed AD. In a series of Cox proportional hazards regression models, better sleep consolidation attenuated the effect of the ε4 allele on the risk of incident AD (hazard ratio, 0.67; 95% CI, 0.46-0.97; P = .04 per allele per 1-SD increase in sleep consolidation). In a series of linear mixed-effect models, better sleep consolidation also attenuated the effect of the ε4 allele on the annual rate of cognitive decline. In individuals who died, better sleep consolidation attenuated the effect of the ε4 allele on neurofibrillary tangle density (interaction estimate, -0.42; SE = 0.17; P = .02), which accounted for the effect of sleep consolidation on the association between APOE genotype and cognition proximate to death. CONCLUSIONS AND RELEVANCE Better sleep consolidation attenuates the effect of APOE genotype on incident AD and development of neurofibrillary tangle pathology. Assessment of sleep consolidation may identify APOE+ individuals at high risk for incident AD, and interventions to enhance sleep consolidation should be studied as potentially useful means to reduce the risk of AD and development of neurofibrillary tangles in APOE ε4+ individuals.
Collapse
Affiliation(s)
- Andrew S P Lim
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
39
|
Tan HL, Kheirandish-Gozal L, Gozal D. The promise of translational and personalised approaches for paediatric obstructive sleep apnoea: an 'Omics' perspective. Thorax 2014; 69:474-80. [PMID: 24550060 DOI: 10.1136/thoraxjnl-2013-204640] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obstructive sleep apnoea (OSA) can result in significant morbidities including the cardiovascular, metabolic and neurocognitive systems. These effects are purportedly mediated via activation of inflammatory cascades and the induction of oxidative stress, ultimately resulting in cellular injury and dysfunction. While great advances have been made in sleep medicine research in the past decades, there are still wide gaps in our knowledge concerning the exact underlying pathophysiological mechanisms of OSA and consequences. Without resolving these issues, the reasons why patients with a similar severity of OSA can have markedly different clinical presentation and end-organ morbidity, that is, phenotype, will continue to remain elusive. This review aims to highlight the recent exciting discoveries in genotype-phenotype interactions, epigenetics, genomics and proteomics related to OSA. Just as PCR revolutionised the field of genetics, the potential power of 'Omics' promises to transform the field of sleep medicine, and provide critical insights into the downstream pathological cascades inherent to OSA, thereby enabling personalised diagnosis and management for this highly prevalent sleep disorder.
Collapse
Affiliation(s)
- Hui-Leng Tan
- Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, , London, UK
| | | | | |
Collapse
|
40
|
Hakim F, Wang Y, Zhang SXL, Zheng J, Yolcu ES, Carreras A, Khalyfa A, Shirwan H, Almendros I, Gozal D. Fragmented sleep accelerates tumor growth and progression through recruitment of tumor-associated macrophages and TLR4 signaling. Cancer Res 2014; 74:1329-37. [PMID: 24448240 DOI: 10.1158/0008-5472.can-13-3014] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Sleep fragmentation (SF) is a highly prevalent condition and a hallmark of sleep apnea, a condition that has been associated with increased cancer incidence and mortality. In this study, we examined the hypothesis that sleep fragmentation promotes tumor growth and progression through proinflammatory TLR4 signaling. In the design, we compared mice that were exposed to sleep fragmentation one week before engraftment of syngeneic TC1 or LL3 tumor cells and tumor analysis four weeks later. We also compared host contributions through the use of mice genetically deficient in TLR4 or its effector molecules MYD88 or TRIF. We found that sleep fragmentation enhanced tumor size and weight compared with control mice. Increased invasiveness was apparent in sleep fragmentation tumors, which penetrated the tumor capsule into surrounding tissues, including adjacent muscle. Tumor-associated macrophages (TAM) were more numerous in sleep fragmentation tumors, where they were distributed in a relatively closer proximity to the tumor capsule compared with control mice. Although tumors were generally smaller in both MYD88(-/-) and TRIF(-/-) hosts, the more aggressive features produced by sleep fragmentation persisted. In contrast, these more aggressive features produced by sleep fragmentation were abolished completely in TLR4(-/-) mice. Our findings offer mechanistic insights into how sleep perturbations can accelerate tumor growth and invasiveness through TAM recruitment and TLR4 signaling pathways.
Collapse
Affiliation(s)
- Fahed Hakim
- Authors' Affiliations: Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, The University of Chicago, Chicago, Illinois; and Department of Microbiology and Immunology, The University of Louisville, Louisville, Kentucky
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sherwani SI, Aldana C, Usmani S, Adin C, Kotha S, Khan M, Eubank T, Scherer PE, Parinandi N, Magalang UJ. Intermittent hypoxia exacerbates pancreatic β-cell dysfunction in a mouse model of diabetes mellitus. Sleep 2013; 36:1849-58. [PMID: 24293759 DOI: 10.5665/sleep.3214] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
STUDY OBJECTIVES The effects of intermittent hypoxia (IH) on pancreatic function in the presence of diabetes and the underlying mechanisms are unclear. We hypothesized that IH would exacerbate pancreatic β-cell dysfunction and alter the fatty acids in the male Tallyho/JngJ (TH) mouse, a rodent model of type 2 diabetes. DESIGN TH mice were exposed for 14 d to either 8 h of IH or intermittent air (IA), followed by an intraperitoneal glucose tolerance test (IPGTT) and tissue harvest. The effect of IH on insulin release was determined by using a β3-adrenergic receptor (AR) agonist. MEASUREMENTS AND RESULTS During IH, pancreatic tissue pO2 decreased from 20.4 ± 0.9 to 5.7 ± 2.6 mm Hg, as determined by electron paramagnetic resonance oximetry. TH mice exposed to IH exhibited higher plasma glucose levels during the IPGTT (P < 0.001) while the insulin levels tended to be lower (P = 0.06). Pancreatic islets of the IH group showed an enhancement of the caspase-3 staining (P = 0.002). IH impaired the β-AR agonist-mediated insulin release (P < 0.001). IH increased the levels of the total free fatty acids and saturated fatty acids (palmitic and stearic acids), and decreased levels of the monounsaturated fatty acids in the pancreas and plasma. Ex vivo exposure of pancreatic islets to palmitic acid suppressed insulin secretion and decreased islet cell viability. CONCLUSIONS Intermittent hypoxia increases pancreatic apoptosis and exacerbates dysfunction in a polygenic rodent model of diabetes. An increase in free fatty acids and a shift in composition towards long chain saturated fatty acid species appear to mediate these effects.
Collapse
Affiliation(s)
- Shariq I Sherwani
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH ; Dorothy M. Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, OH
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hita-Yañez E, Atienza M, Cantero JL. Polysomnographic and subjective sleep markers of mild cognitive impairment. Sleep 2013; 36:1327-34. [PMID: 23997365 PMCID: PMC3738041 DOI: 10.5665/sleep.2956] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES Growing evidence suggests that sleep disturbances precede by years the clinical onset of Alzheimer disease (AD). The goal of the current study is to determine whether changes in polysomnographic (PSG) sleep patterns accompany subjective sleep complaints in patients with mild cognitive impairment (MCI). We further examine whether meaningful changes in objective sleep physiology are predicted by self-reported sleep measures in MCI patients, and whether incipient neurodegeneration contributes to exacerbate sleep misperception. DESIGN SETTING AND PARTICIPANTS Overnight PSG recordings and self-reported sleep measures were obtained from 25 healthy elderly (HE) subjects and 25 patients with MCI at the sleep laboratory. RESULTS Both PSG and self-reported sleep measures confirmed that sleep is altered in patients with MCI. Whereas subjective sleep responses predicted fragmentation of slow wave sleep (SWS) in HE individuals, this relationship was not evident in MCI patients. Furthermore, patients with MCI showed significant discrepancies in the estimation of sleep onset latency when compared with HE subjects. CONCLUSIONS Sleep is significantly impaired in patients with mild cognitive impairment at both the objective and subjective level, which may be used as a surrogate marker of preclinical Alzheimer disease. Taken together, these findings aid in the development of novel therapeutic strategies devoted to improve sleep in the elderly population at risk of developing Alzheimer disease.
Collapse
Affiliation(s)
- Eva Hita-Yañez
- Laboratory of Functional Neuroscience, Spanish Network of Excellence for Research on Neurodegenerative Diseases (CIBERNED), University Pablo de Olavide, Seville, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Spanish Network of Excellence for Research on Neurodegenerative Diseases (CIBERNED), University Pablo de Olavide, Seville, Spain
| | - Jose L. Cantero
- Laboratory of Functional Neuroscience, Spanish Network of Excellence for Research on Neurodegenerative Diseases (CIBERNED), University Pablo de Olavide, Seville, Spain
| |
Collapse
|
43
|
Nair D, Ramesh V, Li RC, Schally AV, Gozal D. Growth hormone releasing hormone (GHRH) signaling modulates intermittent hypoxia-induced oxidative stress and cognitive deficits in mouse. J Neurochem 2013; 127:531-40. [PMID: 23815362 DOI: 10.1111/jnc.12360] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/23/2013] [Accepted: 06/25/2013] [Indexed: 11/27/2022]
Abstract
Intermittent hypoxia (IH) during sleep, such as occurs in obstructive sleep apnea (OSA), leads to degenerative changes in the hippocampus, and is associated with spatial learning deficits in adult mice. In both patients and murine models of OSA, the disease is associated with suppression of growth hormone (GH) secretion, which is actively involved in the growth, development, and function of the central nervous system (CNS). Recent work showed that exogenous GH therapy attenuated neurocognitive deficits elicited by IH during sleep in rats. Here, we show that administration of the Growth Hormone Releasing Hormone (GHRH) agonist JI-34 attenuates IH-induced neurocognitive deficits, anxiety, and depression in mice along with reduction in oxidative stress markers such as MDA and 8-hydroxydeoxyguanosine, and increases in hypoxia inducible factor-1α DNA binding and up-regulation of insulin growth factor-1 and erythropoietin expression. In contrast, treatment with a GHRH antagonist (MIA-602) during intermittent hypoxia did not affect any of the IH-induced deleterious effects in mice. Thus, exogenous GHRH administered as the formulation of a GHRH agonist may provide a viable therapeutic intervention to protect IH-vulnerable brain regions from OSA-associated neurocognitive dysfunction. Sleep apnea, characterized by chronic intermittent hypoxia (IH), is associated with substantial cognitive and behavioral deficits. Here, we show that administration of a GHRH agonist (JI-34) reduces oxidative stress, increases both HIF-1α nuclear binding and downstream expression of IGF1 and erythropoietin (EPO) in hippocampus and cortex, and markedly attenuates water maze performance deficits in mice exposed to intermittent hypoxia during sleep.
Collapse
Affiliation(s)
- Deepti Nair
- Department of Pediatrics, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | |
Collapse
|
44
|
Gozal D. CrossTalk proposal: the intermittent hypoxia attending severe obstructive sleep apnoea does lead to alterations in brain structure and function. J Physiol 2013; 591:379-81. [PMID: 23322286 DOI: 10.1113/jphysiol.2012.241216] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
45
|
Kheirandish-Gozal L, Gozal D. Genotype-phenotype interactions in pediatric obstructive sleep apnea. Respir Physiol Neurobiol 2013; 189:338-43. [PMID: 23563156 DOI: 10.1016/j.resp.2013.03.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 03/27/2013] [Accepted: 03/27/2013] [Indexed: 02/05/2023]
Abstract
Pediatric sleep disordered breathing (PSDB) is not only a very frequent condition affecting 2-4% of all children, but is also associated with an increased risk for a variety of manifestations underlying end-organ injury and dysfunction that impose both immediate and potentially long-term morbidities and corresponding inherent elevations in healthcare costs. One of the major problems with the creation of valid algorithms aiming to stratify diagnostic and treatment prioritization lies in our current inability to predict and identify those children who are most at-risk for PSDB-induced adverse consequences. Thus, improved our understanding of the mechanisms governing phenotype variance in PSDB is essential. Here, we examine some of the potential underpinnings of phenotypic variability in PSDB, and further propose a conceptual framework aimed at facilitating the process of advancing knowledge in this frequent disorder.
Collapse
Affiliation(s)
- Leila Kheirandish-Gozal
- Sections of Pediatric Sleep Medicine and Pediatric Pulmonology, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL, United States.
| | | |
Collapse
|
46
|
|