1
|
Khandayataray P, Murthy MK. Exploring the nexus: Sleep disorders, circadian dysregulation, and Alzheimer's disease. Neuroscience 2025; 574:21-41. [PMID: 40189132 DOI: 10.1016/j.neuroscience.2025.03.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/10/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
We reviewed the connections among Alzheimer's disease (AD), sleep deprivation, and circadian rhythm disorders. Evidence is mounting that disrupted sleep and abnormal circadian rhythms are not merely symptoms of AD, but are also involved in accelerating the disease. Amyloid-beta (Aβ) accumulates, a feature of AD, and worsens with sleep deprivation because glymphatic withdrawal is required to clear toxic proteins from the brain. In addition, disturbances in circadian rhythm can contribute to the induction of neuroinflammation and oxidative stress, thereby accelerating neurodegenerative processes. While these interactions are bidirectional, Alzheimer's pathology further disrupts sleep and circadian function in a vicious cycle that worsens cognitive decline, which is emphasized in the review. The evidence that targeting sleep and circadian mechanisms may serve as therapeutic strategies for AD was strengthened by this study through the analysis of the molecular and physiological pathways. Further work on this nexus could help unravel the neurobiological mechanisms common to the onset of Alzheimer's and disrupted sleep and circadian regulation, which could result in earlier intervention to slow or prevent the onset of the disease.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab 140401, India.
| |
Collapse
|
2
|
Gorgoni M, Fasiello E, Leonori V, Galbiati A, Scarpelli S, Alfonsi V, Annarumma L, Casoni F, Castronovo V, Ferini-Strambi L, De Gennaro L. K-Complex morphological alterations in insomnia disorder and their relationship with sleep state misperception. Sleep 2025; 48:zsaf040. [PMID: 39951438 DOI: 10.1093/sleep/zsaf040] [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: 10/21/2024] [Revised: 02/07/2025] [Indexed: 02/16/2025] Open
Abstract
Insomnia disorder (ID) is characterized by electroencephalographic indexes of hyperarousal, often associated with the underestimation of sleep duration (i.e. sleep state misperception). Albeit non-rapid eye movement sleep K-complexes (KCs) are involved in sleep protection and arousal, only a few studies investigated their alterations in ID with heterogenous findings, and results about their possible relationship with sleep state misperception are missing. The study aims to assess KCs in ID and their relationship with sleep state misperception, also considering their correlation with sleep architecture (i.e. the large-scale organization of sleep). Nineteen ID patients (12 F; age: 42.4 ± 12.1 years) and 18 healthy controls (HC; 10 F; age: 41.6 ± 11.9 years) underwent a night of home polysomnography and completed sleep diaries upon awakening. KC density, amplitude, and area under the curve were assessed in midline frontal, central, and parietal derivations. Sleep state misperception was investigated by considering polysomnographic and subjective total sleep time (TST). We found reduced anterior KC morphology (i.e. amplitude and area under the curve) in ID patients compared to HCs, which was associated with TST underestimation. KC morphology was negatively associated with N3 latency, sleep fragmentation and arousal indexes, and positively related with N3 percentage and sleep efficiency. Our findings suggest an impaired sleep protection mechanism expressed by altered KCs morphology in ID involved in sleep state misperception. The observed correlations support the view of KC as the forerunner of slow-wave sleep and protector of sleep continuity. A better understanding of sleep-protecting mechanisms alteration as a predisposing and/or maintaining factor of ID is needed.
Collapse
Affiliation(s)
- Maurizio Gorgoni
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Elisabetta Fasiello
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
- Department of Clinical Neurosciences, Neurology-Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valerio Leonori
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Andrea Galbiati
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
- Department of Clinical Neurosciences, Neurology-Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Scarpelli
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Valentina Alfonsi
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | | | - Francesca Casoni
- Department of Clinical Neurosciences, Neurology-Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vincenza Castronovo
- Department of Clinical Neurosciences, Neurology-Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Ferini-Strambi
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
- Department of Clinical Neurosciences, Neurology-Sleep Disorders Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi De Gennaro
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
3
|
Lüthi A, Nedergaard M. Anything but small: Microarousals stand at the crossroad between noradrenaline signaling and key sleep functions. Neuron 2025; 113:509-523. [PMID: 39809276 DOI: 10.1016/j.neuron.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 09/16/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Continuous sleep restores the brain and body, whereas fragmented sleep harms cognition and health. Microarousals (MAs), brief (3- to 15-s-long) wake intrusions into sleep, are clinical markers for various sleep disorders. Recent rodent studies show that MAs during healthy non-rapid eye movement (NREM) sleep are driven by infraslow fluctuations of noradrenaline (NA) in coordination with electrophysiological rhythms, vasomotor activity, cerebral blood volume, and glymphatic flow. MAs are hence part of healthy sleep dynamics, raising questions about their biological roles. We propose that MAs bolster NREM sleep's benefits associated with NA fluctuations, according to an inverted U-shaped curve. Weakened noradrenergic fluctuations, as may occur in neurodegenerative diseases or with sleep aids, reduce MAs, whereas exacerbated fluctuations caused by stress fragment NREM sleep and collapse NA signaling. We suggest that MAs are crucial for the restorative and plasticity-promoting functions of sleep and advance our insight into normal and pathological arousal dynamics from sleep.
Collapse
Affiliation(s)
- Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
4
|
Mullins AE, Pehel S, Parekh A, Kam K, Bubu OM, Tolbert TM, Rapoport DM, Ayappa I, Varga AW, Osorio RS. The stability of slow-wave sleep and EEG oscillations across two consecutive nights of laboratory polysomnography in cognitively normal older adults. J Sleep Res 2025; 34:e14281. [PMID: 38937887 PMCID: PMC11671611 DOI: 10.1111/jsr.14281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024]
Abstract
Laboratory polysomnography provides gold-standard measures of sleep physiology, but multi-night investigations are resource intensive. We assessed the night-to-night stability via reproducibility metrics for sleep macrostructure and electroencephalography oscillations in a group of cognitively normal adults attending two consecutive polysomnographies. Electroencephalographies were analysed using an automatic algorithm for detection of slow-wave activity, spindle and K-complex densities. Average differences between nights for sleep macrostructure, electroencephalography oscillations and sleep apnea severity were assessed, and test-retest reliability was determined using two-way intraclass correlations. Agreement was calculated using the smallest real differences between nights for all measures. Night 2 polysomnographies showed significantly greater time in bed, total sleep time (6.3 hr versus 6.8 hr, p < 0.001) and percentage of rapid eye movement sleep (17.5 versus 19.7, p < 0.001). Intraclass correlations were low for total sleep time, percentage of rapid eye movement sleep and sleep efficiency, moderate for percentage of slow-wave sleep and percentage of non-rapid eye movement 2 sleep, good for slow-wave activity and K-complex densities, and excellent for spindles and apnea-hypopnea index with hypopneas defined according to 4% oxygen desaturation criteria only. The smallest real difference values were proportionally high for most sleep macrostructure measures, indicating moderate agreement, and proportionally lower for most electroencephalography microstructure variables. Slow waves, K-complexes, spindles and apnea severity indices are highly reproducible across two consecutive nights of polysomnography. In contrast, sleep macrostructure measures all demonstrated poor reproducibility as indicated by low intraclass correlation values and moderate agreement. Although there were average differences in percentage of rapid eye movement sleep and total sleep time, these were numerically small and perhaps functionally or clinically less significant. One night of in-laboratory polysomnography is enough to provide stable, reproducible estimates of an individual's sleep concerning measures of slow-wave activity, spindles, K-complex densities and apnea severity.
Collapse
Affiliation(s)
- Anna E. Mullins
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Shayna Pehel
- Center for Sleep and Brain Health, Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Ankit Parekh
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Korey Kam
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Omonigho M. Bubu
- Center for Sleep and Brain Health, Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas M. Tolbert
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - David M. Rapoport
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Indu Ayappa
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrew W. Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ricardo S. Osorio
- Center for Sleep and Brain Health, Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
5
|
Embang JEG, Tan YHV, Ng YX, Loyola GJP, Wong LW, Guo Y, Dong Y. Role of sleep and neurochemical biomarkers in synaptic plasticity related to neurological and psychiatric disorders: A scoping review. J Neurochem 2025; 169:e16270. [PMID: 39676063 DOI: 10.1111/jnc.16270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 12/17/2024]
Abstract
Sleep is vital for maintaining physical and mental well-being, impacting cognitive functions like memory and learning through neuroplasticity. Sleep disturbances prevalent in neurological and psychiatric disorders exacerbate cognitive decline, imposing societal burdens. Exploring the relationship between sleep and neuroplasticity elucidates the mechanisms influencing cognition, particularly amidst the prevalent sleep disturbances in these clinical populations. While existing reviews provide valuable insights, gaps remain in understanding the neurophysiological mechanisms underlying sleep and cognitive function. This scoping review aims to investigate the characteristic patterns of sleep parameters and neurochemical biomarkers in reflecting neuroplasticity changes related to neurological and psychiatric disorders and to explore how these markers interact and influence cognition at the molecular level. Studies involving adults and older adults were included, excluding animal models and the paediatric population. Selected studies explored the relationship between sleep parameter or neurochemical biomarker changes and cognitive impairment, reflecting underlying neuroplasticity changes. Peer-reviewed articles, clinical trials, theses, and dissertations in English were included while excluding secondary research and non-peer-reviewed sources. A three-step search strategy was executed following the updated Joanna Briggs Institute methodology for scoping reviews. Published studies were retrieved from nine databases, grey literature, expert recommendations, and hand-searching of the included studies' bibliography. A basic qualitative content synthesis of 34 studies was conducted per JBI's scoping review guidance. Slow-wave and Rapid-Eye Movement sleep, sleep spindles, sleep cycle disruption, K-Complex(KC) density, Hippocampal sEEG, BDNF, IL-6, iNOS mRNA expression, plasma serotonin, CSF Aβ-42, t-tau and p-tau proteins, and serum cortisol revealed associations with cognitive dysfunction. Examining the relationship between sleep parameters, neurochemical biomarkers, and cognitive function reveals neuronal mechanisms that guide potential therapeutic interventions and enhance quality patient care.
Collapse
Affiliation(s)
- Johann Emilio Gonzales Embang
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Division of Nursing, National University Hospital, Singapore City, Singapore
- National University Health System, Singapore City, Singapore
| | - Ying Hui Valerie Tan
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Division of Nursing, National University Hospital, Singapore City, Singapore
- National University Health System, Singapore City, Singapore
| | - Yu Xuan Ng
- National University Health System, Singapore City, Singapore
- Division of Nursing, Alexandra Hospital, Singapore City, Singapore
| | - Gerard Jude Ponce Loyola
- College of Medicine, University of the Philippines, Manila, Philippines
- Philippine General Hospital, Manila, Philippines
| | - Lik-Wei Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Yuqing Guo
- Sue & Bill Gross School of Nursing, University of California, Irvine, California, USA
| | - Yanhong Dong
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| |
Collapse
|
6
|
Han F, Liu X, Yang Y, Liu X. Sex-specific age-related differences in cerebrospinal fluid clearance assessed by resting-state functional magnetic resonance imaging. Neuroimage 2024; 302:120905. [PMID: 39461604 DOI: 10.1016/j.neuroimage.2024.120905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024] Open
Abstract
Cerebrospinal fluid (CSF) flow may assist the clearance of brain wastes, such as amyloid-β (Aβ) and tau, and thus play an important role in aging and dementias. However, a lack of non-invasive tools to assess the CSF dynamics-related clearance in humans hindered the understanding of the relevant changes in healthy aging. The global infra-slow (<0.1 Hz) brain activity measured by the global mean resting-state fMRI signal (gBOLD) was recently found to be coupled by large CSF movements. This coupling has been found to correlate with various pathologies of Alzheimer's disease (AD), particularly Aβ pathology, linking it to waste clearance. Using resting-state fMRI data from a group of 719 healthy aging participants, we examined the sex-specific differences of the gBOLD-CSF coupling over a wide age range between 36-100 years of age. We found that this coupling index remains stable before around age 55 and then starts to decline afterward, particularly in females. Menopause may contribute to the accelerated decline in females.
Collapse
Affiliation(s)
- Feng Han
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
| | - Xufu Liu
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
| | - Yifan Yang
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
| | - Xiao Liu
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA; Institute for Computational and Data Sciences, The Pennsylvania State University, PA, USA.
| |
Collapse
|
7
|
Liu X. Decoupling Between Brain Activity and Cerebrospinal Fluid Movement in Neurological Disorders. J Magn Reson Imaging 2024; 60:1743-1752. [PMID: 37991132 PMCID: PMC11109023 DOI: 10.1002/jmri.29148] [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: 09/01/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023] Open
Abstract
Recent research has identified a link between the global mean signal of resting-state functional MRI (fMRI) and macro-scale cerebrospinal fluid movement, indicating the potential link between this resting-state dynamic and brain waste clearance. Consistent with this notion, the strength of this coupling has been associated with multiple neurodegenerative disease pathologies, especially the build-up of toxic proteins. This article aimed to review the latest advancements in this research area, emphasizing studies on spontaneous global brain activity that is tightly linked to the global mean resting-state fMRI signal, and aimed to discuss potential mechanisms through which this activity and associated physiological modulations might affect brain waste clearance. The available evidence supports the presence of a highly organized global brain activity that is linked to arousal and memory systems. This global brain dynamic, along with its associated physiological modulations, has the potential to influence brain waste clearance through multiple pathways through multiple pathways. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Institute for Computational and Data Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
8
|
Kegyes-Brassai AC, Pierson-Bartel R, Bolla G, Kamondi A, Horvath AA. Disruption of sleep macro- and microstructure in Alzheimer's disease: overlaps between neuropsychology, neurophysiology, and neuroimaging. GeroScience 2024:10.1007/s11357-024-01357-z. [PMID: 39333449 DOI: 10.1007/s11357-024-01357-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/14/2024] [Indexed: 09/29/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia, often associated with impaired sleep quality and disorganized sleep structure. This study aimed to characterize changes in sleep macrostructure and K-complex density in AD, in relation to neuropsychological performance and brain structural changes. We enrolled 30 AD and 30 healthy control participants, conducting neuropsychological exams, brain MRI, and one-night polysomnography. AD patients had significantly reduced total sleep time (TST), sleep efficiency, and relative durations of non-rapid eye movement (NREM) stages 2 (S2), 3 (S3), and rapid eye movement (REM) sleep (p < 0.01). K-complex (KC) density during the entire sleep period and S2 (p < 0.001) was significantly decreased in AD. We found strong correlations between global cognitive performance and relative S3 (p < 0.001; r = 0.86) and REM durations (p < 0.001; r = 0.87). TST and NREM stage 1 (S1) durations showed a moderate negative correlation with amygdaloid and hippocampal volumes (p < 0.02; r = 0.51-0.55), while S3 and REM sleep had a moderate positive correlation with cingulate cortex volume (p < 0.02; r = 0.45-0.61). KC density strongly correlated with global cognitive function (p < 0.001; r = 0.66) and the thickness of the anterior cingulate cortex (p < 0.05; r = 0.45-0.47). Our results indicate significant sleep organization changes in AD, paralleling cognitive decline. Decreased slow wave sleep and KCs are strongly associated with cingulate cortex atrophy. Since sleep changes are prominent in early AD, they may serve as prognostic markers or therapeutic targets.
Collapse
Affiliation(s)
| | | | - Gergo Bolla
- School of PhD Studies, Semmelweis University, Budapest, Hungary
- Neurocognitive Research Centre, Nyírő Gyula National Institute of Psychiatry, and Addictology, Budapest, Hungary
| | - Anita Kamondi
- Neurocognitive Research Centre, Nyírő Gyula National Institute of Psychiatry, and Addictology, Budapest, Hungary
- Department of Neurosurgery and Neurointervention, Semmelweis University, Budapest, Hungary
- Department of Neurology, Semmelweis University, Budapest, Hungary
| | - Andras Attila Horvath
- Neurocognitive Research Centre, Nyírő Gyula National Institute of Psychiatry, and Addictology, Budapest, Hungary
- Department of Anatomy Histology and Embryology, Semmelweis University, Budapest, Hungary
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
9
|
Gonzales PNG, Villaraza SG, Rosa JACD. The association between sleep and Alzheimer's disease: a systematic review. Dement Neuropsychol 2024; 18:e20230049. [PMID: 39193464 PMCID: PMC11348879 DOI: 10.1590/1980-5764-dn-2023-0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/20/2024] [Accepted: 05/11/2024] [Indexed: 08/29/2024] Open
Abstract
It is estimated that 45% of individuals with cognitive impairment experience sleep disturbances prior to the onset of cognitive symptoms. Assessing sleeping problems and enhancing sleep quality are critical first steps to reduce the risk of cognitive impairment. Objective To review existing literature based on predefined eligibility criteria to understand the connection between sleep disturbance and Alzheimer's disease. Methods A thorough and systematic evaluation of numerous studies was carried out to assess one or more of the following epidemiological factors: (1) sleep disorders, (2) cognitive impairment, and (3) risk estimates for cognitive impairment due to sleep. Results Studies suggest that individuals who experience memory loss may encounter sleep disturbances before noticing other symptoms. Numerous sleep disorders, such as excessive and inadequate sleep duration, poor sleep quality, circadian rhythm abnormalities, insomnia, and obstructive sleep apnea were found to increase the risk of cognitive dysfunction and dementia. Additionally, lower sleep quality and shorter sleep duration have been linked to higher cerebral-β-amyloid levels. Objective evidence for the development of cognitive impairment is provided by the architecture of sleep stages. Patients experiencing sleep problems may benefit from specific types of sleep medicine as a preventative measure against cognitive decline. Conclusion Sleep disorders can have adverse effects on cognitive health. The duration and quality of sleep are fundamental factors for maintaining a healthy brain as we age. Proper sleep can aid prevent cognitive impairment, particularly Alzheimer's disease and dementia.
Collapse
|
10
|
Zhuang D, Yu N, Han S, Zhang X, Ju C. The Kv7 channel opener Retigabine reduces neuropathology and alleviates behavioral deficits in APP/PS1 transgenic mice. Behav Brain Res 2024; 471:115137. [PMID: 38971432 DOI: 10.1016/j.bbr.2024.115137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Hyperexcitability of neuronal networks is central to the pathogenesis of Alzheimer's disease (AD). Pharmacological activation of Kv7 channels is an effective way to reduce neuronal firing. Our results showed that that pharmacologically activating the Kv7 channel with Retigabine (RTG) can alleviate cognitive impairment in mice without affecting spontaneous activity. RTG could also ameliorate damage to the Nissl bodies in cortex and hippocampal CA and DG regions in 9-month-old APP/PS1 mice. Additionally, RTG could reduce the Aβ plaque number in the hippocampus and cortex of both 6-month-old and 9-month-old mice. By recordings of electroencephalogram, we showed that a decrease in the number of abnormal discharges in the brains of the AD model mice when the Kv7 channel was opened. Moreover, Western blot analysis revealed a reduction in the expression of the p-Tau protein in both the hippocampus and cortex upon Kv7 channel opening. These findings suggest that Kv7 channel opener RTG may ameliorate cognitive impairment in AD, most likely by reducing brain excitability.
Collapse
Affiliation(s)
- Dongpei Zhuang
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| | - Nan Yu
- Department of Pharmacy, Qingdao Eighth People's Hospital, China.
| | - Shuo Han
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| | - Xinyao Zhang
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| | - Chuanxia Ju
- Department of Pharmacology, School of Pharmacy, Qingdao University Qingdao Medical College, China.
| |
Collapse
|
11
|
Gauld C, Hartley S, Micoulaud-Franchi JA, Royant-Parola S. Sleep Health Analysis Through Sleep Symptoms in 35,808 Individuals Across Age and Sex Differences: Comparative Symptom Network Study. JMIR Public Health Surveill 2024; 10:e51585. [PMID: 38861716 PMCID: PMC11200043 DOI: 10.2196/51585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/28/2023] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Sleep health is a multidimensional construct that includes objective and subjective parameters and is influenced by individual sleep-related behaviors and sleep disorders. Symptom network analysis allows modeling of the interactions between variables, enabling both the visualization of relationships between different factors and the identification of the strength of those relationships. Given the known influence of sex and age on sleep health, network analysis can help explore sets of mutually interacting symptoms relative to these demographic variables. OBJECTIVE This study aimed to study the centrality of symptoms and compare age and sex differences regarding sleep health using a symptom network approach in a large French population that feels concerned about their sleep. METHODS Data were extracted from a questionnaire provided by the Réseau Morphée health network. A network analysis was conducted on 39 clinical variables related to sleep disorders and sleep health. After network estimation, statistical analyses consisted of calculating inferences of centrality, robustness (ie, testifying to a sufficient effect size), predictability, and network comparison. Sleep clinical variable centralities within the networks were analyzed by both sex and age using 4 age groups (18-30, 31-45, 46-55, and >55 years), and local symptom-by-symptom correlations determined. RESULTS Data of 35,808 participants were obtained. The mean age was 42.7 (SD 15.7) years, and 24,964 (69.7%) were women. Overall, there were no significant differences in the structure of the symptom networks between sexes or age groups. The most central symptoms across all groups were nonrestorative sleep and excessive daytime sleepiness. In the youngest group, additional central symptoms were chronic circadian misalignment and chronic sleep deprivation (related to sleep behaviors), particularly among women. In the oldest group, leg sensory discomfort and breath abnormality complaint were among the top 4 central symptoms. Symptoms of sleep disorders thus became more central with age than sleep behaviors. The high predictability of central nodes in one of the networks underlined its importance in influencing other nodes. CONCLUSIONS The absence of structural difference between networks is an important finding, given the known differences in sleep between sexes and across age groups. These similarities suggest comparable interactions between clinical sleep variables across sexes and age groups and highlight the implication of common sleep and wake neural circuits and circadian rhythms in understanding sleep health. More precisely, nonrestorative sleep and excessive daytime sleepiness are central symptoms in all groups. The behavioral component is particularly central in young people and women. Sleep-related respiratory and motor symptoms are prominent in older people. These results underscore the importance of comprehensive sleep promotion and screening strategies tailored to sex and age to impact sleep health.
Collapse
Affiliation(s)
| | - Sarah Hartley
- Sleep Center, APHP Hôpital Raymond Poincaré, Université de Versailles Saint-Quentin en Yvelines, Garches, France
- Réseau Morphée, Garches, France
| | - Jean-Arthur Micoulaud-Franchi
- Services of Functional Exploration of the Nervous System, University Sleep Clinic, University Hospital of Bordeaux, Bordeaux, France
- Unité Sommeil, Addiction, Neuropsychiatrie, Centre national de la recherche scientifique Unité Mixte de Recherche-6033, Bordeaux, France
| | | |
Collapse
|
12
|
Slutsky I. Linking activity dyshomeostasis and sleep disturbances in Alzheimer disease. Nat Rev Neurosci 2024; 25:272-284. [PMID: 38374463 DOI: 10.1038/s41583-024-00797-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2024] [Indexed: 02/21/2024]
Abstract
The presymptomatic phase of Alzheimer disease (AD) starts with the deposition of amyloid-β in the cortex and begins a decade or more before the emergence of cognitive decline. The trajectory towards dementia and neurodegeneration is shaped by the pathological load and the resilience of neural circuits to the effects of this pathology. In this Perspective, I focus on recent advances that have uncovered the vulnerability of neural circuits at early stages of AD to hyperexcitability, particularly when the brain is in a low-arousal states (such as sleep and anaesthesia). Notably, this hyperexcitability manifests before overt symptoms such as sleep and memory deficits. Using the principles of control theory, I analyse the bidirectional relationship between homeostasis of neuronal activity and sleep and propose that impaired activity homeostasis during sleep leads to hyperexcitability and subsequent sleep disturbances, whereas sleep disturbances mitigate hyperexcitability via negative feedback. Understanding the interplay among activity homeostasis, neuronal excitability and sleep is crucial for elucidating the mechanisms of vulnerability to and resilience against AD pathology and for identifying new therapeutic avenues.
Collapse
Affiliation(s)
- Inna Slutsky
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
13
|
Gorgoni M, Cenani J, Scarpelli S, D'Atri A, Alfonsi V, Annarumma L, Pietrogiacomi F, Ferrara M, Marra C, Rossini PM, De Gennaro L. The role of the sleep K-complex on the conversion from mild cognitive impairment to Alzheimer's disease. J Sleep Res 2024; 33:e14046. [PMID: 37718942 DOI: 10.1111/jsr.14046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/24/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023]
Abstract
The present literature points to an alteration of the human K-complex during non-rapid eye movement sleep in Alzheimer's disease. Nevertheless, the few findings on the K-complex changes in mild cognitive impairment and their possible predictive role on the Alzheimer's disease conversion show mixed findings, lack of replication, and a main interest for the frontal region. The aim of the present study was to assess K-complex measures in amnesic mild cognitive impairment subsequently converted in Alzheimer's disease over different cortical regions, comparing them with healthy controls and stable amnesic mild cognitive impairment. We assessed baseline K-complex density, amplitude, area under the curve and overnight changes in frontal, central and parietal midline derivations of 12 amnesic mild cognitive impairment subsequently converted in Alzheimer's disease, 12 stable amnesic mild cognitive impairment and 12 healthy controls. We also assessed delta electroencephalogram power, to determine if K-complex alterations in amnesic mild cognitive impairment occur with modification of the electroencephalogram power in the frequency range of the slow-wave activity. We found a reduced parietal K-complex density in amnesic mild cognitive impairment subsequently converted in Alzheimer's disease compared with stable amnesic mild cognitive impairment and healthy controls, without changes in K-complex morphology and overnight modulation. Both amnesic mild cognitive impairment groups showed decreased slow-wave sleep percentage compared with healthy controls. No differences between groups were observed in slow-wave activity power. Our findings suggest that K-complex alterations in mild cognitive impairment may be observed earlier in parietal regions, likely mirroring the topographical progression of Alzheimer's disease-related brain pathology, and express a frontal predominance only in a full-blown phase of Alzheimer's disease. Consistently with previous results, such K-complex modification occurs in the absence of significant electroencephalogram power changes in the slow oscillations range.
Collapse
Affiliation(s)
- Maurizio Gorgoni
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- Body and Action Lab, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Jessica Cenani
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Serena Scarpelli
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Aurora D'Atri
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | | | | | - Michele Ferrara
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Camillo Marra
- Institute of Neurology, Catholic University, Rome, Italy
| | - Paolo Maria Rossini
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, Rome, Italy
| | - Luigi De Gennaro
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- Body and Action Lab, IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
14
|
Sun H, Adra N, Ayub MA, Ganglberger W, Ye E, Fernandes M, Paixao L, Fan Z, Gupta A, Ghanta M, Moura Junior VF, Rosand J, Westover MB, Thomas RJ. Assessing Risk of Health Outcomes From Brain Activity in Sleep: A Retrospective Cohort Study. Neurol Clin Pract 2024; 14:e200225. [PMID: 38173542 PMCID: PMC10759032 DOI: 10.1212/cpj.0000000000200225] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/04/2023] [Indexed: 01/05/2024]
Abstract
Background and Objectives Patterns of electrical activity in the brain (EEG) during sleep are sensitive to various health conditions even at subclinical stages. The objective of this study was to estimate sleep EEG-predicted incidence of future neurologic, cardiovascular, psychiatric, and mortality outcomes. Methods This is a retrospective cohort study with 2 data sets. The Massachusetts General Hospital (MGH) sleep data set is a clinic-based cohort, used for model development. The Sleep Heart Health Study (SHHS) is a community-based cohort, used as the external validation cohort. Exposure is good, average, or poor sleep defined by quartiles of sleep EEG-predicted risk. The outcomes include ischemic stroke, intracranial hemorrhage, mild cognitive impairment, dementia, atrial fibrillation, myocardial infarction, type 2 diabetes, hypertension, bipolar disorder, depression, and mortality. Diagnoses were based on diagnosis codes, brain imaging reports, medications, cognitive scores, and hospital records. We used the Cox survival model with death as the competing risk. Results There were 8673 participants from MGH and 5650 from SHHS. For all outcomes, the model-predicted 10-year risk was within the 95% confidence interval of the ground truth, indicating good prediction performance. When comparing participants with poor, average, and good sleep, except for atrial fibrillation, all other 10-year risk ratios were significant. The model-predicted 10-year risk ratio closely matched the observed event rate in the external validation cohort. Discussion The incidence of health outcomes can be predicted by brain activity during sleep. The findings strengthen the concept of sleep as an accessible biological window into unfavorable brain and general health outcomes.
Collapse
Affiliation(s)
- Haoqi Sun
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Noor Adra
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Muhammad Abubakar Ayub
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Wolfgang Ganglberger
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Elissa Ye
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Marta Fernandes
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Luis Paixao
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Ziwei Fan
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Aditya Gupta
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Manohar Ghanta
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Valdery F Moura Junior
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Jonathan Rosand
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - M Brandon Westover
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Robert J Thomas
- Department of Neurology (HS, NA, MAA, WG, EY, MF, LP, ZF, AG, MG, VFMJ, JR, MBW), Massachusetts General Hospital; Henry and Allison McCance Center for Brain Health at Mass General (HS, VFMJ, JR, MBW); Department of Neurology (HS, WG, AG, MG, VFMJ, MBW), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (MAA), Louisiana State University Health Sciences Center, Shreveport, LA; Department of Neurology (LP), Washington University School of Medicine in St. Louis, MO; and Division of Pulmonary (RJT), Critical Care and Sleep, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
15
|
De Gennaro L, Cordone S. Mounting Evidence on the Relationship between Sleep and Alzheimer's Disease. J Integr Neurosci 2024; 23:21. [PMID: 38287863 DOI: 10.31083/j.jin2301021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 12/27/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Affiliation(s)
- Luigi De Gennaro
- Department of Psychology, University of Rome "Sapienza", 00185 Rome, Italy
| | - Susanna Cordone
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| |
Collapse
|
16
|
Tapia-Rivas NI, Estévez PA, Cortes-Briones JA. A robust deep learning detector for sleep spindles and K-complexes: towards population norms. Sci Rep 2024; 14:263. [PMID: 38167626 PMCID: PMC10762090 DOI: 10.1038/s41598-023-50736-7] [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: 07/21/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024] Open
Abstract
Sleep spindles (SSs) and K-complexes (KCs) are brain patterns involved in cognitive functions that appear during sleep. Large-scale sleep studies would benefit from precise and robust automatic sleep event detectors, capable of adapting the variability in both electroencephalography (EEG) signals and expert annotation rules. We introduce the Sleep EEG Event Detector (SEED), a deep learning system that outperforms existing approaches in SS and KC detection, reaching an F1-score of 80.5% and 83.7%, respectively, on the MASS2 dataset. SEED transfers well and requires minimal fine-tuning for new datasets and annotation styles. Remarkably, SEED substantially reduces the required amount of annotated data by using a novel pretraining approach that leverages the rule-based detector A7. An analysis of 11,224 subjects revealed that SEED's detections provide better estimates of SS population statistics than existing approaches. SEED is a powerful resource for obtaining sleep-event statistics that could be useful for establishing population norms.
Collapse
Affiliation(s)
| | - Pablo A Estévez
- Department of Electrical Engineering, University of Chile, Santiago, Chile.
- Millennium Institute of Intelligent Healthcare Engineering, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - José A Cortes-Briones
- Schizophrenia and Neuropharmacology Research Group at Yale (SNRGY), Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
17
|
Hanert A, Schönfeld R, Weber FD, Nowak A, Döhring J, Philippen S, Granert O, Burgalossi A, Born J, Berg D, Göder R, Häussermann P, Bartsch T. Reduced overnight memory consolidation and associated alterations in sleep spindles and slow oscillations in early Alzheimer's disease. Neurobiol Dis 2024; 190:106378. [PMID: 38103701 DOI: 10.1016/j.nbd.2023.106378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Spatial navigation critically underlies hippocampal-entorhinal circuit function that is early affected in Alzheimer's disease (AD). There is growing evidence that AD pathophysiology dynamically interacts with the sleep/wake cycle impairing hippocampal memory. To elucidate sleep-dependent consolidation in a cohort of symptomatic AD patients (n = 12, 71.25 ± 2.16 years), we tested hippocampal place learning by means of a virtual reality task and verbal memory by a word-pair association task before and after a night of sleep. Our results show an impaired overnight memory retention in AD compared with controls in the verbal task, together with a significant reduction of sleep spindle activity (i.e., lower amplitude of fast sleep spindles, p = 0.016) and increased duration of the slow oscillation (SO; p = 0.019). Higher spindle density, faster down-to-upstate transitions within SOs, and the time delay between SOs and nested spindles predicted better memory performance in healthy controls but not in AD patients. Our results show that mnemonic processing and memory consolidation in AD is slightly impaired as reflected by dysfunctional oscillatory dynamics and spindle-SO coupling during NonREM sleep. In this translational study based on experimental paradigms in animals and extending previous work in healthy aging and preclinical disease stages, our results in symptomatic AD further deepen the understanding of the memory decline within a bidirectional relationship of sleep and AD pathology.
Collapse
Affiliation(s)
- Annika Hanert
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Robby Schönfeld
- Institute of Psychology, Division of Clinical Psychology, Martin-Luther-University Halle-Wittenberg, 06099 Halle (Saale), Germany
| | - Frederik D Weber
- Institute for Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72074 Tübingen, Germany; Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6525 EN Nijmegen, the Netherlands; Department of Sleep and Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, the Netherlands
| | - Alexander Nowak
- Department of Psychiatry and Psychotherapy, Sleep Laboratory, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Juliane Döhring
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany; Institute for General Medicine, University Hospital of Schleswig-Holstein, 24105 Kiel, Germany
| | - Sarah Philippen
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Oliver Granert
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Andrea Burgalossi
- Institute of Neurobiology, Werner-Reichardt Center for Integrative Neuroscience, University of Tübingen, 72074 Tübingen, Germany
| | - Jan Born
- Institute for Medical Psychology and Behavioral Neurobiology, University of Tübingen, 72074 Tübingen, Germany
| | - Daniela Berg
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Robert Göder
- Department of Psychiatry and Psychotherapy, Sleep Laboratory, University Hospital of Schleswig Holstein, 24105 Kiel, Germany
| | - Peter Häussermann
- Department of Geriatric Psychiatry, LVR Klinik Köln, Academic Teaching Hospital, University of Cologne, Köln, Germany
| | - Thorsten Bartsch
- Department of Neurology, Memory Disorders and Plasticity Group, University Hospital of Schleswig Holstein, 24105 Kiel, Germany.
| |
Collapse
|
18
|
Gu Y, Gagnon JF, Kaminska M. Sleep electroencephalography biomarkers of cognition in obstructive sleep apnea. J Sleep Res 2023; 32:e13831. [PMID: 36941194 DOI: 10.1111/jsr.13831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 03/23/2023]
Abstract
Obstructive sleep apnea has been associated with cognitive impairment and may be linked to disorders of cognitive function. These associations may be a result of intermittent hypoxaemia, sleep fragmentation and changes in sleep microstructure in obstructive sleep apnea. Current clinical metrics of obstructive sleep apnea, such as the apnea-hypopnea index, are poor predictors of cognitive outcomes in obstructive sleep apnea. Sleep microstructure features, which can be identified on sleep electroencephalography of traditional overnight polysomnography, are increasingly being characterized in obstructive sleep apnea and may better predict cognitive outcomes. Here, we summarize the literature on several major sleep electroencephalography features (slow-wave activity, sleep spindles, K-complexes, cyclic alternating patterns, rapid eye movement sleep quantitative electroencephalography, odds ratio product) identified in obstructive sleep apnea. We will review the associations between these sleep electroencephalography features and cognition in obstructive sleep apnea, and examine how treatment of obstructive sleep apnea affects these associations. Lastly, evolving technologies in sleep electroencephalography analyses will also be discussed (e.g. high-density electroencephalography, machine learning) as potential predictors of cognitive function in obstructive sleep apnea.
Collapse
Affiliation(s)
- Yusing Gu
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jean-François Gagnon
- Department of Psychology, Université du Québec à Montréal, Montréal, Québec, Canada
- Center for Advanced Research in Sleep Medicine, CIUSSS-NÎM - Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Marta Kaminska
- Respiratory Epidemiology and Clinical Research Unit, Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
- Respiratory Division & Sleep Laboratory, McGill University Health Centre, Montreal, Québec, Canada
| |
Collapse
|
19
|
Yao AY, Halloran PJ, Ge Y, Singh N, Zhou J, Galske J, He W, Yan R, Hu X. Bace1 Deletion in the Adult Reverses Epileptiform Activity and Sleep-wake Disturbances in AD Mice. J Neurosci 2023; 43:6197-6211. [PMID: 37536983 PMCID: PMC10476643 DOI: 10.1523/jneurosci.2124-22.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023] Open
Abstract
Alzheimer's disease (AD) increases the risk for seizures and sleep disorders. We show here that germline deletion of β-site amyloid precursor protein (APP) cleaving enzyme-1 (BACE1) in neurons, but not in astrocytes, increased epileptiform activity. However, Bace1 deletion at adult ages did not alter the normal EEG waveform, indicating less concern for BACE1 inhibition in patients. Moreover, we showed that deletion of Bace1 in the adult was able to reverse epileptiform activity in 5xFAD mice. Intriguingly, treating 5xFAD and APPNL-G-F/NL-G-F (APP KI) mice of either sex with one BACE1 inhibitor Lanabecestat (AZD3293) dramatically increased epileptiform spiking, likely resulting from an off-target effect. We also monitored sleep-wake pathologies in these mice and showed increased wakefulness, decreased non-rapid eye movement sleep, and rapid eye movement sleep in both 5xFAD and APP KI mice; BACE1 inhibition in the adult 5xFAD mice reversed plaque load and sleep disturbances, but this was not seen in APP KI mice. Further studies with and without BACE1 inhibitor treatment showed different levels of plaque-associated microgliosis and activated microglial proteins in 5xFAD mice compared with APP KI mice. Together, BACE1 inhibition should be developed to avoid off-target effect for achieving benefits in reducing epileptic activity and sleep disturbance in Alzheimer's patients.SIGNIFICANCE STATEMENT BACE1 is widely recognized as a therapeutic target for treating Alzheimer's disease patients. However, BACE1 inhibitors failed in clinical trials because of inability to show cognitive improvement in patients. Here we show that BACE1 inhibition actually reduces sleep disturbances and epileptic seizures; both are seen in AD patients. We further showed that one of clinically tested BACE1 inhibitors does have off-target effects, and development of safer BACE1 inhibitors will be beneficial to AD patients. Results from this study will provide useful guidance for additional drug development.
Collapse
Affiliation(s)
- Annie Y Yao
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Patrick J Halloran
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Yingying Ge
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Neeraj Singh
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - John Zhou
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - James Galske
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Wanxia He
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Xiangyou Hu
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030
| |
Collapse
|
20
|
Han F, Liu X, Yang Y, Liu X. Sex-specific age-related changes in glymphatic function assessed by resting-state functional magnetic resonance imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.02.535258. [PMID: 37034667 PMCID: PMC10081329 DOI: 10.1101/2023.04.02.535258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
The glymphatic system that clears out brain wastes, such as amyloid-β (Aβ) and tau, through cerebrospinal fluid (CSF) flow may play an important role in aging and dementias. However, a lack of non-invasive tools to assess the glymphatic function in humans hindered the understanding of the glymphatic changes in healthy aging. The global infra-slow (<0.1 Hz) brain activity measured by the global mean resting-state fMRI signal (gBOLD) was recently found to be coupled by large CSF movements. This coupling has been used to measure the glymphatic process and found to correlate with various pathologies of Alzheimer's disease (AD), including Aβ pathology. Using resting-state fMRI data from a large group of 719 healthy aging participants, we examined the sex-specific changes of the gBOLD-CSF coupling, as a measure of glymphatic function, over a wide age range between 36-100 years old. We found that this coupling index remains stable before around age 55 and then starts to decline afterward, particularly in females. Menopause may contribute to the accelerated decline in females.
Collapse
Affiliation(s)
- Feng Han
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
| | - Xufu Liu
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
| | - Yifan Yang
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
| | - Xiao Liu
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
- Institute for Computational and Data Sciences, The Pennsylvania State University, PA, USA
| |
Collapse
|
21
|
Tao P, Svetnik V, Bliwise DL, Zammit G, Lines C, Herring WJ. Comparison of polysomnography in people with Alzheimer's disease and insomnia versus non-demented elderly people with insomnia. Sleep Med 2023; 101:515-521. [PMID: 36529106 DOI: 10.1016/j.sleep.2022.11.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/16/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND We used baseline polysomnography (PSG) data obtained during the clinical program development for suvorexant to compare the PSG profiles of people with Alzheimer's disease and insomnia (ADI) versus age-matched elderly individuals with insomnia (EI). METHODS Sleep laboratory baseline PSG data from participants age 55-80 years from 2 trials in people with insomnia and a trial in people with ADI were included. ADI participants had dementia of mild-to-moderate severity. Diagnostic criteria for insomnia, exclusion for other sleep problems, PSG recording procedures, and endpoint derivations were similar across the trials. All participants underwent a night of in-laboratory PSG prior to the baseline night to allow for screening/adaptation. Participants in the EI and ADI groups were compared with regard to sleep architecture, sleep micro-structure, and quantitative EEG power spectral endpoints. The analysis was performed on a post hoc basis using propensity score matching to compare sleep parameters separately in women and men while accounting for age group and total sleep time. RESULTS A total of 837 EI and 239 ADI participants were included, with the majority in each population (∼65%) being women. Compared to EI, those with ADI had a lower percentage of time spent in slow wave sleep (and a corresponding higher percentage of time spent in the lighter N1 sleep), a lower number of spindles per minute of N2 sleep, and lower absolute EEG power during NREM sleep, particularly in the lower-frequency bands. Trends for lower REM sleep percentage in ADI did not reach statistical significance. CONCLUSIONS Our findings in this large data set, in which the influence of sleep problems was effectively subtracted out (since both groups had insomnia), provide strong confirmatory support of results from previous smaller studies in indicating that AD of mild-to-moderate severity is associated with less slow wave sleep, spindles, and lower-frequency EEG power. TRIAL REGISTRATION ClinicalTrials.gov, numbers NCT01097616, NCT01097629, NCT02750306.
Collapse
Affiliation(s)
| | | | - Donald L Bliwise
- Sleep Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Gary Zammit
- Clinilabs Drug Development Corporation, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | |
Collapse
|
22
|
Azami H, Moguilner S, Penagos H, Sarkis RA, Arnold SE, Gomperts SN, Lam AD. EEG Entropy in REM Sleep as a Physiologic Biomarker in Early Clinical Stages of Alzheimer's Disease. J Alzheimers Dis 2023; 91:1557-1572. [PMID: 36641682 PMCID: PMC10039707 DOI: 10.3233/jad-221152] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is associated with EEG changes across the sleep-wake cycle. As the brain is a non-linear system, non-linear EEG features across behavioral states may provide an informative physiologic biomarker of AD. Multiscale fluctuation dispersion entropy (MFDE) provides a sensitive non-linear measure of EEG information content across a range of biologically relevant time-scales. OBJECTIVE To evaluate MFDE in awake and sleep EEGs as a potential biomarker for AD. METHODS We analyzed overnight scalp EEGs from 35 cognitively normal healthy controls, 23 participants with mild cognitive impairment (MCI), and 19 participants with mild dementia due to AD. We examined measures of entropy in wake and sleep states, including a slow-to-fast-activity ratio of entropy (SFAR-entropy). We compared SFAR-entropy to linear EEG measures including a slow-to-fast-activity ratio of power spectral density (SFAR-PSD) and relative alpha power, as well as to cognitive function. RESULTS SFAR-entropy differentiated dementia from MCI and controls. This effect was greatest in REM sleep, a state associated with high cholinergic activity. Differentiation was evident in the whole brain EEG and was most prominent in temporal and occipital regions. Five minutes of REM sleep was sufficient to distinguish dementia from MCI and controls. Higher SFAR-entropy during REM sleep was associated with worse performance on the Montreal Cognitive Assessment. Classifiers based on REM sleep SFAR-entropy distinguished dementia from MCI and controls with high accuracy, and outperformed classifiers based on SFAR-PSD and relative alpha power. CONCLUSION SFAR-entropy measured in REM sleep robustly discriminates dementia in AD from MCI and healthy controls.
Collapse
Affiliation(s)
- Hamed Azami
- Massachusetts Alzheimer's Disease Research Center, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Sebastian Moguilner
- Massachusetts Alzheimer's Disease Research Center, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Hector Penagos
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rani A. Sarkis
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Steven E. Arnold
- Massachusetts Alzheimer's Disease Research Center, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Stephen N. Gomperts
- Massachusetts Alzheimer's Disease Research Center, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Alice D. Lam
- Massachusetts Alzheimer's Disease Research Center, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
23
|
Khasawneh N, Fraiwan M, Fraiwan L. Detection of K-complexes in EEG waveform images using faster R-CNN and deep transfer learning. BMC Med Inform Decis Mak 2022; 22:297. [PMID: 36397034 PMCID: PMC9673349 DOI: 10.1186/s12911-022-02042-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022] Open
Abstract
Background The electroencephalography (EEG) signal carries important information about the electrical activity of the brain, which may reveal many pathologies. This information is carried in certain waveforms and events, one of which is the K-complex. It is used by neurologists to diagnose neurophysiologic and cognitive disorders as well as sleep studies.
Existing detection methods largely depend on tedious, time-consuming, and error-prone manual inspection of the EEG waveform. Methods In this paper, a highly accurate K-complex detection system is developed. Based on multiple convolutional neural network (CNN) feature extraction backbones and EEG waveform images, a regions with faster regions with convolutional neural networks (Faster R-CNN) detector was designed, trained, and tested. Extensive performance evaluation was performed using four deep transfer learning feature extraction models (AlexNet, ResNet-101, VGG19 and Inceptionv3). The dataset was comprised of 10948 images of EEG waveforms, with the location of the K-complexes included as separate text files containing the bounding boxes information. Results The Inceptionv3 and VGG19-based detectors performed consistently high (i.e., up to 99.8% precision and 0.2% miss rate) over different testing scenarios, in which the number of training images was varied from 60% to 80% and the positive overlap threshold was increased from 60% to 90%. Conclusions Our automated method appears to be a highly accurate automatic K-complex detection in real-time that can aid practitioners in speedy EEG inspection. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12911-022-02042-x.
Collapse
|
24
|
Gorgoni M, Galbiati A. Non-REM sleep electrophysiology in REM sleep behaviour disorder: A narrative mini-review. Neurosci Biobehav Rev 2022; 142:104909. [DOI: 10.1016/j.neubiorev.2022.104909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/22/2022] [Accepted: 10/06/2022] [Indexed: 10/31/2022]
|
25
|
Casagrande M, Forte G, Favieri F, Corbo I. Sleep Quality and Aging: A Systematic Review on Healthy Older People, Mild Cognitive Impairment and Alzheimer’s Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19148457. [PMID: 35886309 PMCID: PMC9325170 DOI: 10.3390/ijerph19148457] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/27/2022] [Accepted: 07/07/2022] [Indexed: 02/01/2023]
Abstract
Aging is characterized by changes in the structure and quality of sleep. When the alterations in sleep become substantial, they can generate or accelerate cognitive decline, even in the absence of overt pathology. In fact, impaired sleep represents one of the earliest symptoms of Alzheimer’s disease (AD). This systematic review aimed to analyze the studies on sleep quality in aging, also considering mild cognitive impairment (MCI) and AD. The review process was conducted according to the PRISMA statement. A total of 71 studies were included, and the whole sample had a mean age that ranged from 58.3 to 93.7 years (62.8–93.7 healthy participants and 61.8–86.7 pathological populations). Of these selected studies, 33 adopt subjective measurements, 31 adopt objective measures, and 10 studies used both. Pathological aging showed a worse impoverishment of sleep than older adults, in both subjective and objective measurements. The most common aspect compromised in AD and MCI were REM sleep, sleep efficiency, sleep latency, and sleep duration. These results underline that sleep alterations are associated with cognitive impairment. In conclusion, the frequency and severity of sleep disturbance appear to follow the evolution of cognitive impairment. The overall results of objective measures seem more consistent than those highlighted by subjective measurements.
Collapse
Affiliation(s)
- Maria Casagrande
- Department of Dynamic and Clinical Psychology and Health Studies, Sapienza University of Rome, 00185 Roma, Italy;
- Correspondence: (M.C.); (I.C.)
| | - Giuseppe Forte
- Department of Dynamic and Clinical Psychology and Health Studies, Sapienza University of Rome, 00185 Roma, Italy;
- Body and Action Laboratory, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179 Rome, Italy;
| | - Francesca Favieri
- Body and Action Laboratory, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179 Rome, Italy;
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Roma, Italy
| | - Ilaria Corbo
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Roma, Italy
- Correspondence: (M.C.); (I.C.)
| |
Collapse
|
26
|
Chen Q, Shen W, Sun H, Zhang H, Liu C, Chen Z, Yu L, Cai X, Ke J, Li L, Zhang L, Fang Q. The effect of coupled inhibitory-facilitatory repetitive transcranial magnetic stimulation on shaping early reorganization of the motor network after stroke. Brain Res 2022; 1790:147959. [PMID: 35654120 DOI: 10.1016/j.brainres.2022.147959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 11/29/2022]
Abstract
Neural plasticity is a major factor driving cortical reorganization after stroke. This study aimed to evaluate functional connectivity (FC) changes in the cortical motor network after coupled inhibitory-facilitatory repetitive transcranial magnetic stimulation (rTMS) treatment and to assess the correlation between FC changes and functional recovery, further characterizing the neural mechanisms underlying the beneficial effects of rTMS. We randomly divided 63 patients with acute stroke into four groups: (1) Group A received coupled inhibitory-facilitatory rTMS [1 Hz over the contralesional primary motor cortex (M1) and 10 Hz over ipsilesional M1]; (2) Group B received a contralesional sham stimulation and ipsilesional 10 Hz stimulation; (3) Group C received a contralesional 1 Hz rTMS and ipsilesional sham stimulation; and (4) Group D received bilateral sham stimulation only. Standardized rehabilitation therapy was performed immediately after rTMS, and each group was treated with their respective treatment modalities for 4 weeks. Twenty-four hours before and after the intervention, participants underwent resting-state functional magnetic resonance imaging. Additional functional assessments were conducted at baseline, after treatment, and at the 3 month follow-up. The rTMS treatment significantly changed the FCs of intra- and inter-hemispheric cortical motor networks in the rTMS groups (A and B) compared with the sham group (Group D). This effect was more pronounced in Group A, which displayed a changed FC between the contralesional postcentral gyrus and contralesional superior parietal gyrus, between the contralesional precentral gyrus and contralesional postcentral gyrus, and between the ipsilesional postcentral gyrus and contralesional superior parietal gyrus, when compared with Groups B and C. Importantly, FC changes were significantly correlated with improvement of motor function. In the early stages of ischemic stroke, coupled rTMS was more conducive to motor recovery by modulating the FCs of intra-hemispheric and inter-hemispheric motor networks. Our results suggested that FC changes were related to motor function recovery for early-stage cerebral stroke patients treated with coupled rTMS. These findings could help to understand the mechanism of coupled rTMS and further the use of this therapy as an adjunct rehabilitation technique in motor recovery.
Collapse
Affiliation(s)
- Qingmei Chen
- Department of Physical Medicine &Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China; Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Wenjun Shen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Haiwei Sun
- Department of Emergency Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Hanjun Zhang
- Department of Physical Medicine &Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Chuandao Liu
- Department of Physical Medicine &Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Zhiguo Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Liqiang Yu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Xiuying Cai
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Jun Ke
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Li Li
- Department of Physical Medicine &Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China.
| | - Lichi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China.
| |
Collapse
|
27
|
Roy J, Wong KY, Aquili L, Uddin MS, Heng BC, Tipoe GL, Wong KH, Fung ML, Lim LW. Role of melatonin in Alzheimer's disease: From preclinical studies to novel melatonin-based therapies. Front Neuroendocrinol 2022; 65:100986. [PMID: 35167824 DOI: 10.1016/j.yfrne.2022.100986] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 01/21/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022]
Abstract
Melatonin and novel melatonin-based therapies such as melatonin-containing hybrid molecules, melatonin analogues, and melatonin derivatives have been investigated as potential therapeutics against Alzheimer's disease (AD) pathogenesis. In this review, we examine the developmental trends of melatonin therapies for AD from 1997 to 2021. We then highlight the neuroprotective mechanisms of melatonin therapy derived from preclinical studies. These mechanisms include the alleviation of amyloid-related burden, neurofibrillary tangle accumulation, oxidative stress, neuroinflammation, apoptosis, mitochondrial dysfunction, and impaired neuroplasticity and neurotransmission. We further illustrate the beneficial effects of melatonin on behavior in animal models of AD. Next, we discuss the clinical effects of melatonin on sleep, cognition, behavior, psychiatric symptoms, electroencephalography findings, and molecular biomarkers in patients with mild cognitive impairment and AD. We then explore the effectiveness of novel melatonin-based therapies. Lastly, we discuss the limitations of current melatonin therapies for AD and suggest two emerging research themes for future study.
Collapse
Affiliation(s)
- Jaydeep Roy
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kan Yin Wong
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Luca Aquili
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; College of Science, Health, Engineering and Education, Discipline of Psychology, Murdoch University, Perth, Australia
| | - Md Sahab Uddin
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Boon Chin Heng
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Peking University School of Stomatology, Beijing, China
| | - George Lim Tipoe
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kah Hui Wong
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Man Lung Fung
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
28
|
Toward noninvasive brain stimulation 2.0 in Alzheimer's disease. Ageing Res Rev 2022; 75:101555. [PMID: 34973457 PMCID: PMC8858588 DOI: 10.1016/j.arr.2021.101555] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/01/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022]
Abstract
Noninvasive brain stimulation techniques (NiBS) have gathered substantial interest in the study of dementia, considered their possible role in help defining diagnostic biomarkers of altered neural activity for early disease detection and monitoring of its pathophysiological course, as well as for their therapeutic potential of boosting residual cognitive functions. Nevertheless, current approaches suffer from some limitations. In this study, we review and discuss experimental NiBS applications that might help improve the efficacy of future NiBS uses in Alzheimer's Disease (AD), including perturbation-based biomarkers for early diagnosis and disease tracking, solutions to enhance synchronization of oscillatory electroencephalographic activity across brain networks, enhancement of sleep-related memory consolidation, image-guided stimulation for connectome control, protocols targeting interneuron pathology and protein clearance, and finally hybrid-brain models for in-silico modeling of AD pathology and personalized target selection. The present work aims to stress the importance of multidisciplinary, translational, model-driven interventions for precision medicine approaches in AD.
Collapse
|
29
|
Wu EQ, Peng XY, Chen SD, Zhao XY, Tang ZR. Detecting Alzheimer’s Dementia Degree. IEEE Trans Cogn Dev Syst 2022. [DOI: 10.1109/tcds.2020.3015131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
30
|
Neeti, Ranjan R, Sahana BC. Automatic Identification of K-complex in Sleep EEG Signal for Clinical Practice. 2022 IEEE DELHI SECTION CONFERENCE (DELCON) 2022:1-8. [DOI: 10.1109/delcon54057.2022.9753300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Neeti
- National Institute of Technology,Department of ECE,Patna,Bihar,India,800005
| | - Rakesh Ranjan
- National Institute of Technology,Department of ECE,Patna,Bihar,India,800005
| | | |
Collapse
|
31
|
Mutti C, Misirocchi F, Zilioli A, Rausa F, Pizzarotti S, Spallazzi M, Parrino L. Sleep and brain evolution across the human lifespan: A mutual embrace. FRONTIERS IN NETWORK PHYSIOLOGY 2022; 2:938012. [PMID: 36926070 PMCID: PMC10013002 DOI: 10.3389/fnetp.2022.938012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022]
Abstract
Sleep can be considered a window to ascertain brain wellness: it dynamically changes with brain maturation and can even indicate the occurrence of concealed pathological processes. Starting from prenatal life, brain and sleep undergo an impressive developmental journey that accompanies human life throughout all its steps. A complex mutual influence rules this fascinating course and cannot be ignored while analysing its evolution. Basic knowledge on the significance and evolution of brain and sleep ontogenesis can improve the clinical understanding of patient's wellbeing in a more holistic perspective. In this review we summarized the main notions on the intermingled relationship between sleep and brain evolutionary processes across human lifespan, with a focus on sleep microstructure dynamics.
Collapse
Affiliation(s)
- Carlotta Mutti
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Francesco Misirocchi
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Alessandro Zilioli
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Francesco Rausa
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Silvia Pizzarotti
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Marco Spallazzi
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Liborio Parrino
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| |
Collapse
|
32
|
Chen QM, Yao FR, Sun HW, Chen ZG, Ke J, Liao J, Cai XY, Yu LQ, Wu ZY, Wang Z, Pan X, Liu HY, Li L, Zhang QQ, Ling WH, Fang Q. Combining inhibitory and facilitatory repetitive transcranial magnetic stimulation (rTMS) treatment improves motor function by modulating GABA in acute ischemic stroke patients. Restor Neurol Neurosci 2021; 39:419-434. [PMID: 34924405 DOI: 10.3233/rnn-211195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The combination of inhibitory and facilitatory repetitive transcranial magnetic stimulation (rTMS) can improve motor function of stroke patients with undefined mechanism. It has been demonstrated that rTMS exhibits a neuro-modulatory effect by regulating the major inhibitory neurotransmitter γ-aminobutyric acid (GABA) in other diseases. OBJECTIVES To evaluate the effect of combined inhibitory and facilitatory rTMS on GABA in the primary motor cortex (M1) for treating motor dysfunction after acute ischemic stroke. METHODS 44 ischemic stroke patients with motor dysfunction were randomly divided into two groups. The treatment group was stimulated with 10 Hz rTMS at the ipsilesional M1 and 1 Hz rTMS at the contralesional M1. The sham group received bilateral sham stimulation at the motor cortices. The GABA level in the bilateral M1 was measured by proton magnetic resonance spectroscopy (1H-MRS) at 24 hours before and after rTMS stimulation. Motor function was measured using the Fugl-Meyer Assessment (FMA). The clinical assessments were performed before and after rTMS and after 3 months. RESULTS The treatment group exhibited a greater improvement in motor function 24 hours after rTMS compared to the sham group. The increased improvement in motor function lasted for at least 3 months after treatment. Following 4 weeks of rTMS, the GABA level in the ipsilesional M1 of the treatment group was significantly decreased compared to the sham group. Furthermore, the change of FMA score for motor function was negatively correlated to the change of the GABA:Cr ratio. Finally, the effect of rTMS on motor function outcome was partially mediated by GABA level change in response to the treatment (27.7%). CONCLUSIONS Combining inhibitory and facilitatory rTMS can decrease the GABA level in M1, which is correlated to the improvement of motor function. Thus, the GABA level in M1 may be a potential biomarker for treatment strategy decisions regarding rTMS neuromodulatory interventions.
Collapse
Affiliation(s)
- Qing-Mei Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,Department of Physical Medicine & Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Fei-Rong Yao
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Hai-Wei Sun
- Department of Emergency Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhi-Guo Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jun Ke
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Juan Liao
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiu-Ying Cai
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Li-Qiang Yu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhen-Yan Wu
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhi Wang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xi Pan
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Hao-Yu Liu
- Department of Physical Medicine & Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Li Li
- Department of Physical Medicine & Rehabilitation, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Quan-Quan Zhang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Wei-Hua Ling
- Department of Emergency Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
33
|
Galbiati A, Carli G, Fasiello E, Casoni F, Zucconi M, De Gennaro L, Perani D, Ferini-Strambi L. Exploring the functional role and neural correlates of K-complexes in isolated rapid eye movement sleep behavior disorder. Cortex 2021; 145:105-114. [PMID: 34710676 DOI: 10.1016/j.cortex.2021.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/10/2021] [Accepted: 08/23/2021] [Indexed: 02/05/2023]
Abstract
Underlying neural mechanisms and cognitive implications of non-Rapid Eye Movement (NREM) sleep in isolated Rapid Eye Movement (REM) sleep behavior disorder (iRBD) are not yet fully elucidated. This study aims to evaluate brain metabolic connectivity of the anterior default mode network (ADMN) underlying a waveform that is an hallmark of NREM sleep, namely K-complex (KC) and their implication for neuropsychological functioning in iRBD patients. Combining polysomnographic and multivariate molecular imaging (FDG-PET) approaches may provide crucial insights regarding KCs role in the prodromal stages of synucleinopathies. We applied a seed-based interregional correlation analysis on FDG-PET data. iRBD patients with cognitive decline displayed a reduced KC density (KCd) in comparison to patients without cognitive impairments. KCd showed a significant positive correlation with global cognitive functioning, specifically with visuo-spatial and executive performances, two cognitive domains known to be relevant in predicting conversion into neurodegenerative disorders. Increased KCd was associated with a more preserved ADMN connectivity. Our study underlines the importance of NREM sleep in prodromal stages of synucleinopathies, and future investigations might clarify its role in iRBD.
Collapse
Affiliation(s)
- Andrea Galbiati
- "Vita-Salute" San Raffaele University, Milan, Italy; IRCCS San Raffaele Scientific Institute, Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, Milan, Italy.
| | - Giulia Carli
- "Vita-Salute" San Raffaele University, Milan, Italy; In vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Fasiello
- IRCCS San Raffaele Scientific Institute, Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, Milan, Italy; Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Francesca Casoni
- IRCCS San Raffaele Scientific Institute, Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, Milan, Italy
| | - Marco Zucconi
- IRCCS San Raffaele Scientific Institute, Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, Milan, Italy
| | - Luigi De Gennaro
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Daniela Perani
- "Vita-Salute" San Raffaele University, Milan, Italy; In vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Nuclear Medicine Unit, San Raffaele Hospital, Milan, Italy
| | - Luigi Ferini-Strambi
- "Vita-Salute" San Raffaele University, Milan, Italy; IRCCS San Raffaele Scientific Institute, Department of Clinical Neurosciences, Neurology - Sleep Disorders Center, Milan, Italy
| |
Collapse
|
34
|
Falter A, Van Den Bossche MJA. How non-rapid eye movement sleep and Alzheimer pathology are linked. World J Psychiatry 2021; 11:1027-1038. [PMID: 34888171 PMCID: PMC8613758 DOI: 10.5498/wjp.v11.i11.1027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder characterized by the presence of senile plaques and neurofibrillary tangles. Research attempts to identify characteristic factors that are associated with the presence of the AD pathology on the one hand and that increase the risk of developing AD on the other. Changes in non-rapid eye movement (NREM) sleep may meet both requirements for various reasons. First, NREM-sleep is important for optimal memory function. In addition, studies report that the presence of AD pathology is associated with NREM-sleep changes. Finally, more and more results appear to suggest that sleep problems are not only a symptom of AD but can also increase the risk of AD. Several of these studies suggest that it is primarily a lack of NREM-sleep that is responsible for this increased risk. However, the majority investigated sleep only through subjective reporting, as a result of which NREM-sleep could not be analyzed separately. The aim of this literature study is therefore to present the results of the studies that relate the AD pathology and NREM-sleep (registered by electroencephalography). Furthermore, we try to evaluate whether NREM-sleep analysis could be used to support the diagnosis of AD and whether NREM-sleep deficiency could be a causal factor in the development of AD.
Collapse
Affiliation(s)
- Annelies Falter
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven 3000, Belgium
| | - Maarten J A Van Den Bossche
- Department of Geriatric Psychiatry, University Psychiatric Center KU Leuven, Leuven 3000, Belgium
- Center for Neuropsychiatry, Research Group Psychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
35
|
Jagirdar R, Fu CH, Park J, Corbett BF, Seibt FM, Beierlein M, Chin J. Restoring activity in the thalamic reticular nucleus improves sleep architecture and reduces Aβ accumulation in mice. Sci Transl Med 2021; 13:eabh4284. [PMID: 34731016 PMCID: PMC8985235 DOI: 10.1126/scitranslmed.abh4284] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sleep disruptions promote increases of amyloid β (Aβ) and tau in the brain and increase Alzheimer’s disease (AD) risk, but the precise mechanisms that give rise to sleep disturbances have yet to be defined. The thalamic reticular nucleus (TRN) is essential for sleep maintenance and for the regulation of slow-wave sleep (SWS). We examined the TRN in transgenic mice that express mutant human amyloid precursor protein (APP) and found reduced neuronal activity, increased sleep fragmentation, and decreased SWS time as compared to nontransgenic littermates. Selective activation of the TRN using excitatory DREADDs restored sleep maintenance, increased time in SWS, and reduced amyloid plaque load in both hippocampus and cortex. Our findings suggest that the TRN may play a major role in symptoms associated with AD. Enhancing TRN activity might be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Rohan Jagirdar
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Chia-Hsuan Fu
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Jin Park
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Brian F. Corbett
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Frederik M. Seibt
- Department of Neurobiology and Anatomy, McGovern Medical School at UTHealth, Houston, TX 77030
| | - Michael Beierlein
- Department of Neurobiology and Anatomy, McGovern Medical School at UTHealth, Houston, TX 77030
| | - Jeannie Chin
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
36
|
Sakellariou DF, Vakrinou A, Koutroumanidis M, Richardson MP. Neurocraft: software for microscale brain network dynamics. Sci Rep 2021; 11:20716. [PMID: 34671076 PMCID: PMC8528833 DOI: 10.1038/s41598-021-99195-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/28/2021] [Indexed: 11/08/2022] Open
Abstract
The brain operates at millisecond timescales but despite of that, the study of its functional networks is approached with time invariant methods. Equally, for a variety of brain conditions treatment is delivered with fixed temporal protocols unable to monitor and follow the rapid progression and therefore the cycles of a disease. To facilitate the understanding of brain network dynamics we developed Neurocraft, a user friendly software suite. Neurocraft features a highly novel signal processing engine fit for tracking evolving network states with superior time and frequency resolution. A variety of analytics like dynamic connectivity maps, force-directed representations and propagation models, allow for the highly selective investigation of transient pathophysiological dynamics. In addition, machine-learning tools enable the unsupervised investigation and selection of key network features at individual and group-levels. For proof of concept, we compared six seizure-free and non seizure-free focal epilepsy patients after resective surgery using Neurocraft. The network features were calculated using 50 intracranial electrodes on average during at least 120 epileptiform discharges lasting less than one second, per patient. Powerful network differences were detected in the pre-operative data of the two patient groups (effect size = 1.27), suggesting the predictive value of dynamic network features. More than one million patients are treated with cardiac and neuro modulation devices that are unable to track the hourly or daily changes in a subject's disease. Decoding the dynamics of transition from normal to abnormal states may be crucial in the understanding, tracking and treatment of neurological conditions. Neurocraft provides a user-friendly platform for the research of microscale brain dynamics and a stepping stone for the personalised device-based adaptive neuromodulation in real-time.
Collapse
Affiliation(s)
- Dimitris Fotis Sakellariou
- Real World Solutions, IQVIA, London, N1 9JY, UK.
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- Department of Neurophysiology and Epilepsy, St Thomas' Hospital NHS Trust, London, UK.
| | - Angeliki Vakrinou
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Neurophysiology and Epilepsy, St Thomas' Hospital NHS Trust, London, UK
| | | | - Mark Phillip Richardson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
37
|
Kuang H, Zhu YG, Zhou ZF, Yang MW, Hong FF, Yang SL. Sleep disorders in Alzheimer's disease: the predictive roles and potential mechanisms. Neural Regen Res 2021; 16:1965-1972. [PMID: 33642368 PMCID: PMC8343328 DOI: 10.4103/1673-5374.308071] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/12/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022] Open
Abstract
Sleep disorders are common in patients with Alzheimer's disease, and can even occur in patients with amnestic mild cognitive impairment, which appears before Alzheimer's disease. Sleep disorders further impair cognitive function and accelerate the accumulation of amyloid-β and tau in patients with Alzheimer's disease. At present, sleep disorders are considered as a risk factor for, and may be a predictor of, Alzheimer's disease development. Given that sleep disorders are encountered in other types of dementia and psychiatric conditions, sleep-related biomarkers to predict Alzheimer's disease need to have high specificity and sensitivity. Here, we summarize the major Alzheimer's disease-specific sleep changes, including abnormal non-rapid eye movement sleep, sleep fragmentation, and sleep-disordered breathing, and describe their ability to predict the onset of Alzheimer's disease at its earliest stages. Understanding the mechanisms underlying these sleep changes is also crucial if we are to clarify the role of sleep in Alzheimer's disease. This paper therefore explores some potential mechanisms that may contribute to sleep disorders, including dysregulation of the orexinergic, glutamatergic, and γ-aminobutyric acid systems and the circadian rhythm, together with amyloid-β accumulation. This review could provide a theoretical basis for the development of drugs to treat Alzheimer's disease based on sleep disorders in future work.
Collapse
Affiliation(s)
- Huang Kuang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yu-Ge Zhu
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhi-Feng Zhou
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi Province, China
| | - Mei-Wen Yang
- Department of Nurse, Nanchang University Hospital, Nanchang, Jiangxi Province, China
| | - Fen-Fang Hong
- Department of Experimental Teaching Center, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
38
|
Gorgoni M, Scarpelli S, Annarumma L, D’Atri A, Alfonsi V, Ferrara M, De Gennaro L. The Regional EEG Pattern of the Sleep Onset Process in Older Adults. Brain Sci 2021; 11:1261. [PMID: 34679326 PMCID: PMC8534130 DOI: 10.3390/brainsci11101261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 02/05/2023] Open
Abstract
Healthy aging is characterized by macrostructural sleep changes and alterations of regional electroencephalographic (EEG) sleep features. However, the spatiotemporal EEG pattern of the wake-sleep transition has never been described in the elderly. The present study aimed to assess the topographical and temporal features of the EEG during the sleep onset (SO) in a group of 36 older participants (59-81 years). The topography of the 1 Hz bins' EEG power and the time course of the EEG frequency bands were assessed. Moreover, we compared the delta activity and delta/beta ratio between the older participants and a group of young adults. The results point to several peculiarities in the elderly: (a) the generalized post-SO power increase in the slowest frequencies did not include the 7 Hz bin; (b) the alpha power revealed a frequency-specific pattern of post-SO modifications; (c) the sigma activity exhibited only a slight post-SO increase, and its highest bins showed a frontotemporal power decrease. Older adults showed a generalized reduction of delta power and delta/beta ratio in both pre- and post-SO intervals compared to young adults. From a clinical standpoint, the regional EEG activity may represent a target for brain stimulation techniques to reduce SO latency and sleep fragmentation.
Collapse
Affiliation(s)
- Maurizio Gorgoni
- Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy; (S.S.); (V.A.); (L.D.G.)
| | - Serena Scarpelli
- Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy; (S.S.); (V.A.); (L.D.G.)
| | | | - Aurora D’Atri
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.D.); (M.F.)
| | - Valentina Alfonsi
- Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy; (S.S.); (V.A.); (L.D.G.)
| | - Michele Ferrara
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.D.); (M.F.)
| | - Luigi De Gennaro
- Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy; (S.S.); (V.A.); (L.D.G.)
- Body and Action Lab, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| |
Collapse
|
39
|
Sewell KR, Erickson KI, Rainey-Smith SR, Peiffer JJ, Sohrabi HR, Brown BM. Relationships between physical activity, sleep and cognitive function: A narrative review. Neurosci Biobehav Rev 2021; 130:369-378. [PMID: 34506842 DOI: 10.1016/j.neubiorev.2021.09.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/23/2021] [Accepted: 09/03/2021] [Indexed: 11/19/2022]
Abstract
Physical activity and exercise can improve cognitive function and reduce the risk for dementia. Other lifestyle factors, including sleep, are associated with cognitive function and dementia risk, and exercise is an effective therapeutic strategy for improving sleep. Based on these associations, it has been hypothesised that sleep might be an important mediator for the effects of exercise on cognition. Here, we review the current literature to evaluate whether sleep and physical activity are independently or jointly associated with cognitive function. The extant literature in this area is minimal, and the causal relationships between physical activity, sleep and cognition have not been examined. A small number of cross-sectional studies in this area suggest that physical activity may attenuate some of the negative impact that poor sleep has on cognition, and also that sleep may be a mechanism through which physical activity improves cognitive abilities. Further research may enable the development of individually tailored intervention programs to result in the greatest cognitive benefit, ultimately delaying the onset of Alzheimer's disease.
Collapse
Affiliation(s)
- Kelsey R Sewell
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA, Australia.
| | - Kirk I Erickson
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA, Australia; Department of Psychology, University of Pittsburgh, 4200 Fifth Ave, PA, United States
| | - Stephanie R Rainey-Smith
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, 8 Verdun Street, Nedlands, WA, Australia; Centre For Healthy Ageing, Health Futures Institute, Murdoch University, 90 South Street, Murdoch, WA, Australia; School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, Australia; School of Psychological Science, University of Western Australia, 35 Stirling Highway, Crawley, WA, Australia
| | - Jeremiah J Peiffer
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA, Australia; Centre For Healthy Ageing, Health Futures Institute, Murdoch University, 90 South Street, Murdoch, WA, Australia
| | - Hamid R Sohrabi
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, 8 Verdun Street, Nedlands, WA, Australia; Centre For Healthy Ageing, Health Futures Institute, Murdoch University, 90 South Street, Murdoch, WA, Australia; School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, Australia; Department of Biomedical Sciences, Macquarie University, Balaclava Road, NSW, Australia
| | - Belinda M Brown
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South Street, Murdoch, WA, Australia; Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, 8 Verdun Street, Nedlands, WA, Australia; Centre For Healthy Ageing, Health Futures Institute, Murdoch University, 90 South Street, Murdoch, WA, Australia; School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, Australia
| |
Collapse
|
40
|
Thomas RJ, Kim H, Maillard P, DeCarli CS, Heckman EJ, Karjadi C, Ang TFA, Au R. Digital sleep measures and white matter health in the Framingham Heart Study. EXPLORATION OF MEDICINE 2021; 2:253-267. [PMID: 34927164 PMCID: PMC8682916 DOI: 10.37349/emed.2021.00045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/18/2021] [Indexed: 01/23/2023] Open
Abstract
AIM Impaired sleep quality and sleep oxygenation are common sleep pathologies. This study assessed the impact of these abnormalities on white matter (WM) integrity in an epidemiological cohort. METHODS The target population was the Framingham Heart Study Generation-2/Omni-1 Cohorts. Magnetic resonance imaging (diffusion tensor imaging) was used to assess WM integrity. Wearable digital devices were used to assess sleep quality: the (M1-SleepImage™ system) and the Nonin WristOx for nocturnal oxygenation. The M1 device collects trunk actigraphy and the electrocardiogram (ECG); sleep stability indices were computed using cardiopulmonary coupling using the ECG. Two nights of recording were averaged. RESULTS Stable sleep was positively associated with WM health. Actigraphic periods of wake during the sleep period were associated with increased mean diffusivity. One marker of sleep fragmentation which covaries with respiratory chemoreflex activation was associated with reduced fractional anisotropy and increased mean diffusivity. Both oxygen desaturation index and oxygen saturation time under 90% were associated with pathological directions of diffusion tensor imaging signals. Gender differences were noted across most variables, with female sex showing the larger and significant impact. CONCLUSIONS Sleep quality assessed by a novel digital analysis and sleep hypoxia was associated with WM injury, especially in women.
Collapse
Affiliation(s)
- Robert Joseph Thomas
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Hyun Kim
- Department of Anatomy & Neurobiology, and Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Pauline Maillard
- Department of Neurology, University of California Davis Health, Sacramento, CA 95817, USA
| | - Charles S. DeCarli
- Department of Neurology, University of California Davis Health, Sacramento, CA 95817, USA
| | - Eric James Heckman
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Cody Karjadi
- Department of Anatomy & Neurobiology, and Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ting Fang Alvin Ang
- Department of Anatomy & Neurobiology, and Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Rhoda Au
- Department of Anatomy & Neurobiology, and Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Neurology and Epidemiology, Boston University School of Medicine and Public Health, Boston, MA 02118, USA
| |
Collapse
|
41
|
Fernandes M, Placidi F, Mercuri NB, Liguori C. The Importance of Diagnosing and the Clinical Potential of Treating Obstructive Sleep Apnea to Delay Mild Cognitive Impairment and Alzheimer's Disease: A Special Focus on Cognitive Performance. J Alzheimers Dis Rep 2021; 5:515-533. [PMID: 34368635 PMCID: PMC8293664 DOI: 10.3233/adr-210004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 12/23/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a highly frequent sleep disorder in the middle-aged and older population, and it has been associated with an increased risk of developing cognitive decline and dementia, including mild cognitive impairment (MCI) and Alzheimer's disease (AD). In more recent years, a growing number of studies have focused on: 1) the presence of OSA in patients with MCI or AD, 2) the link between OSA and markers of AD pathology, and 3) the role of OSA in accelerating cognitive deterioration in patients with MCI or AD. Moreover, some studies have also assessed the effects of continuous positive airway pressure (CPAP) treatment on the cognitive trajectory in MCI and AD patients with comorbid OSA. This narrative review summarizes the findings of studies that analyzed OSA as a risk factor for developing MCI and/or AD in the middle-aged and older populations with a special focus on cognition. In addition, it describes the results regarding the effects of CPAP treatment in hampering the progressive cognitive decline in AD and delaying the conversion to AD in MCI patients. Considering the importance of identifying and treating OSA in patients with MCI or AD in order to prevent or reduce the progression of cognitive decline, further larger and adequately powered studies are needed both to support these findings and to set programs for the early recognition of OSA in patients with cognitive impairment.
Collapse
Affiliation(s)
- Mariana Fernandes
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Fabio Placidi
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Neurology Unit, University Hospital of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, University Hospital of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Claudio Liguori
- Sleep Medicine Centre, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Neurology Unit, University Hospital of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
42
|
Yoon JE, Oh D, Hwang I, Park JA, Im HJ, Lee SK, Jung KY, Park SH, Thomas RJ, Shin C, Yun CH. Sleep structure and electroencephalographic spectral power of middle-aged or older adults: Normative values by age and sex in the Korean population. J Sleep Res 2021; 30:e13358. [PMID: 33949014 DOI: 10.1111/jsr.13358] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 11/28/2022]
Abstract
The fine structure of sleep electrocortical activity reflects health and disease. The current study provides normative data for sleep structure and electroencephalography (EEG) spectral power measures derived from overnight polysomnography (PSG) and examines the effect of age and sex among Korean middle-aged and older adults with or without obstructive sleep apnea (OSA). We analysed home PSG data from 1,153 adult participants of an ongoing population-based cohort study, the Korean Genome and Epidemiology Study. Sleep stages were visually scored and spectral power was measured on a single-channel EEG (C4-A1). We computed spectral power for five frequency ranges. The EEG power was reported in relative (%) and log-transformed absolute values (µV2 ). With ageing, the proportion of N1 sleep increased, whereas N3 decreased, which is more noticeable in men than in women. The amount of N3 was relatively low in this cohort. With ageing, relative delta power decreased and alpha and sigma power increased for the whole sleep period, which was more pronounced during REM sleep in non-OSA. For men compared with women, relative theta power was lower during REM and sigma and beta were higher during N1 sleep. The differences of relative powers by age and sex in OSA were comparable to those in non-OSA. In a community-based Korean population, we present normative data of sleep structure and spectral power for middle-aged or older adults of a non-Caucasian ethnicity. The values varied with age and sex and were not influenced by sleep apnea.
Collapse
Affiliation(s)
- Jee-Eun Yoon
- Department of Neurology, Uijeongbu Eulji Medical Center, Uijeongbu, Korea
| | - Dana Oh
- Department of Neurology, Seoul Medical Center, Seoul, Korea
| | - Inha Hwang
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jung Ah Park
- Department of Neurology, School of Medicine, Catholic University of Daegu, Daegu, Korea
| | - Hee-Jin Im
- Department of Neurology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Seung Ku Lee
- Institute of Human Genomic Study, Korea University Ansan Hospital, Ansan, Korea
| | - Ki-Young Jung
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seong-Ho Park
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Robert J Thomas
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, USA
| | - Chol Shin
- Institute of Human Genomic Study, Korea University Ansan Hospital, Ansan, Korea.,Division of Pulmonary, Sleep and Critical Care Medicine, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Chang-Ho Yun
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
43
|
Betta M, Handjaras G, Leo A, Federici A, Farinelli V, Ricciardi E, Siclari F, Meletti S, Ballotta D, Benuzzi F, Bernardi G. Cortical and subcortical hemodynamic changes during sleep slow waves in human light sleep. Neuroimage 2021; 236:118117. [PMID: 33940148 DOI: 10.1016/j.neuroimage.2021.118117] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 04/09/2021] [Accepted: 04/18/2021] [Indexed: 12/22/2022] Open
Abstract
EEG slow waves, the hallmarks of NREM sleep are thought to be crucial for the regulation of several important processes, including learning, sensory disconnection and the removal of brain metabolic wastes. Animal research indicates that slow waves may involve complex interactions within and between cortical and subcortical structures. Conventional EEG in humans, however, has a low spatial resolution and is unable to accurately describe changes in the activity of subcortical and deep cortical structures. To overcome these limitations, here we took advantage of simultaneous EEG-fMRI recordings to map cortical and subcortical hemodynamic (BOLD) fluctuations time-locked to slow waves of light sleep. Recordings were performed in twenty healthy adults during an afternoon nap. Slow waves were associated with BOLD-signal increases in the posterior brainstem and in portions of thalamus and cerebellum characterized by preferential functional connectivity with limbic and somatomotor areas, respectively. At the cortical level, significant BOLD-signal decreases were instead found in several areas, including insula and somatomotor cortex. Specifically, a slow signal increase preceded slow-wave onset and was followed by a delayed, stronger signal decrease. Similar hemodynamic changes were found to occur at different delays across most cortical brain areas, mirroring the propagation of electrophysiological slow waves, from centro-frontal to inferior temporo-occipital cortices. Finally, we found that the amplitude of electrophysiological slow waves was positively related to the magnitude and inversely related to the delay of cortical and subcortical BOLD-signal changes. These regional patterns of brain activity are consistent with theoretical accounts of the functions of sleep slow waves.
Collapse
Affiliation(s)
- Monica Betta
- MoMiLab Research Unit, IMT School for Advanced Studies Lucca, Piazza San Francesco, 19, Lucca 55100, Italy
| | - Giacomo Handjaras
- MoMiLab Research Unit, IMT School for Advanced Studies Lucca, Piazza San Francesco, 19, Lucca 55100, Italy
| | - Andrea Leo
- MoMiLab Research Unit, IMT School for Advanced Studies Lucca, Piazza San Francesco, 19, Lucca 55100, Italy
| | - Alessandra Federici
- MoMiLab Research Unit, IMT School for Advanced Studies Lucca, Piazza San Francesco, 19, Lucca 55100, Italy
| | - Valentina Farinelli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Emiliano Ricciardi
- MoMiLab Research Unit, IMT School for Advanced Studies Lucca, Piazza San Francesco, 19, Lucca 55100, Italy
| | - Francesca Siclari
- Center for Investigation and Research on Sleep, Lausanne University Hospital, Lausanne, Switzerland
| | - Stefano Meletti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Neurology Dept., Azienda Ospedaliera Universitaria di Modena, Modena, Italy
| | - Daniela Ballotta
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Benuzzi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulio Bernardi
- MoMiLab Research Unit, IMT School for Advanced Studies Lucca, Piazza San Francesco, 19, Lucca 55100, Italy.
| |
Collapse
|
44
|
D'Atri A, Scarpelli S, Gorgoni M, Truglia I, Lauri G, Cordone S, Ferrara M, Marra C, Rossini PM, De Gennaro L. EEG alterations during wake and sleep in mild cognitive impairment and Alzheimer's disease. iScience 2021; 24:102386. [PMID: 33981973 PMCID: PMC8086022 DOI: 10.1016/j.isci.2021.102386] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/03/2021] [Accepted: 03/30/2021] [Indexed: 02/08/2023] Open
Abstract
Patients with Alzheimer's disease (AD) undergo a slowing of waking electroencephalographic (EEG) rhythms since prodromal stages, which could be ascribed to poor sleep quality. We examined the relationship between wake and sleep alterations by assessing EEG activity during sleep and (pre-sleep/post-sleep) wakefulness in AD, mild cognitive impairment (MCI) and healthy controls. AD and MCI show high sleep latency and less slow-wave sleep. Reduced sigma activity characterizes non-rapid eye movement (NREM) sleep, reflecting sleep spindles loss. The EEG slowing characterizes REM sleep and wakefulness of AD and MCI, with strong correlations among the two phenomena suggesting common neuropathological mechanisms. Evening-to-morning variations in waking EEG revealed the gradual disappearance in MCI and AD of overnight changes in delta activity, indicating a progressive decay of sleep restorative functions on diurnal activity that correlates with the impairment of sleep high-frequency activity in AD. Our findings support a linkage between wake and sleep alterations, and the importance of sleep-related processes in Alzheimer's disease progression.
Collapse
Affiliation(s)
- Aurora D'Atri
- Department of Psychology, University of Rome “Sapienza”, Via dei Marsi, 78, Rome 00185, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Coppito (L'Aquila) 67100, Italy
| | | | - Maurizio Gorgoni
- Department of Psychology, University of Rome “Sapienza”, Via dei Marsi, 78, Rome 00185, Italy
| | - Ilaria Truglia
- Department of Psychology, University of Rome “Sapienza”, Via dei Marsi, 78, Rome 00185, Italy
| | - Giulia Lauri
- Department of Psychology, University of Rome “Sapienza”, Via dei Marsi, 78, Rome 00185, Italy
| | - Susanna Cordone
- Department of Psychology, University of Rome “Sapienza”, Via dei Marsi, 78, Rome 00185, Italy
| | - Michele Ferrara
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Coppito (L'Aquila) 67100, Italy
| | - Camillo Marra
- Foundation Policlinico Universitario Agostino Gemelli IRCCS – Department of aging, neuroscience, orthopaedic and head-neck, Rome 00168, Italy
| | - Paolo Maria Rossini
- Department of Neuroscience & Neurorehabil., IRCCS San Raffaele-Pisana, Rome, 00163, Italy
| | - Luigi De Gennaro
- Department of Psychology, University of Rome “Sapienza”, Via dei Marsi, 78, Rome 00185, Italy
| |
Collapse
|
45
|
Cordone S, Scarpelli S, Alfonsi V, De Gennaro L, Gorgoni M. Sleep-Based Interventions in Alzheimer's Disease: Promising Approaches from Prevention to Treatment along the Disease Trajectory. Pharmaceuticals (Basel) 2021; 14:383. [PMID: 33921870 PMCID: PMC8073746 DOI: 10.3390/ph14040383] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/10/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
The multifactorial nature of Alzheimer's disease (AD) has led scientific researchers to focus on the modifiable and treatable risk factors of AD. Sleep fits into this context, given the bidirectional relationship with AD confirmed by several studies over the last years. Sleep disorders appear at an early stage of AD and continue throughout the entire course of the pathology. Specifically, sleep abnormalities, such as more fragmented sleep, increase in time of awakenings, worsening of sleep quality and primary sleep disorders raise with the severity and progression of AD. Intervening on sleep, therefore, means acting both with prevention strategies in the pre-clinical phase and with treatments during the course of the disease. This review explores sleep disturbances in the different stages of AD, starting from the pre-clinical stage. Particular attention is given to the empirical evidence investigating obstructive sleep apnea (OSA) disorder and the mechanisms overlapping and sharing with AD. Next, we discuss sleep-based intervention strategies in the healthy elderly population, mild cognitive impairment (MCI) and AD patients. We mention interventions related to behavioral strategies, combination therapies, and bright light therapy, leaving extensive space for new and raising evidence on continuous positive air pressure (CPAP) treatment effectiveness. Finally, we clarify the role of NREM sleep across the AD trajectory and consider the most recent studies based on the promising results of NREM sleep enhancement, which use innovative experimental designs and techniques.
Collapse
Affiliation(s)
- Susanna Cordone
- UniCamillus, Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
| | - Serena Scarpelli
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
| | | | - Luigi De Gennaro
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | - Maurizio Gorgoni
- Department of Psychology, University of Rome “Sapienza”, 00185 Rome, Italy; (S.S.); (M.G.)
| |
Collapse
|
46
|
Zhao X, Chen C, Zhou W, Wang Y, Fan J, Wang Z, Akbarzadeh S, Chen W. An energy screening and morphology characterization-based hybrid expert scheme for automatic identification of micro-sleep event K-complex. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 201:105955. [PMID: 33556760 DOI: 10.1016/j.cmpb.2021.105955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/24/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND AND OBJECTIVE K-complexes, as a significant indicator in sleep staging and sleep protection, are an important micro-event in sleep analysis. Clinically, K-complexes are recognized through the expert visual inspection of electroencephalogram (EEG) during sleep. Since this process is laborious and has high inter-observer variability, developing automated K-complex detection methods can alleviate the burden on clinicians while providing reliable recognition results. However, existing methods face the following issues. First, most work only identifies the K-complexes in stage 2, which requires distinguishing the sleep stages as the prerequisite for further events' identification. Second, most approaches can only detect the occurrence of events without the ability to predict their location and duration, which are also essential to sleep analysis. METHODS In this work, a novel hybrid expert scheme for K-complex detection is proposed by integrating signal morphology with expert knowledge into the decision-making process. To eliminate artifacts, and to minimize the individual variability in raw sleep EEG signals, the potential K-complex candidates are first screened by combining Teager energy operator (TEO) and personalized thresholds. Then, to distinguish signal shapes from background activity, a novel frame of filtering based on morphological filtering (MF) is devised to differentiate morphological components of K-complex waveforms from EEG series. Finally, K-complex waveforms are identified from the extracted morphological information by judgment rules, which are inspired by expert knowledge of micro-sleep events. RESULTS Detection performance is evaluated by its application on the public database MASS-C1 (Montreal archives of sleep studies cohort one) which includes the recordings of 19 healthy adults. The detection performance demonstrates an F-measure of 0.63 with a recall of 0.81 and a precision of 0.53 on average. The duration error between events and detections is 0.10 s. CONCLUSIONS The presented scheme has detected the occurrence of events. Meanwhile, it has recognized their locations and durations. The favorable results exhibit that the proposed scheme outperforms the state-of-the-art studies and has great potential to help release the burden of experts in sleep EEG analysis.
Collapse
Affiliation(s)
- Xian Zhao
- Center for Intelligent Medical Electronics (CIME), School of Information Science and Engineering, Fudan University, Shanghai 200433, China.
| | - Chen Chen
- Center for Intelligent Medical Electronics (CIME), School of Information Science and Engineering, Fudan University, Shanghai 200433, China; Human Phenome Institute, Fudan University, Shanghai 201203, China.
| | - Wei Zhou
- Center for Intelligent Medical Electronics (CIME), School of Information Science and Engineering, Fudan University, Shanghai 200433, China.
| | - Yalin Wang
- Center for Intelligent Medical Electronics (CIME), School of Information Science and Engineering, Fudan University, Shanghai 200433, China; Human Phenome Institute, Fudan University, Shanghai 201203, China.
| | - Jiahao Fan
- Center for Intelligent Medical Electronics (CIME), School of Information Science and Engineering, Fudan University, Shanghai 200433, China; Human Phenome Institute, Fudan University, Shanghai 201203, China.
| | - Zeyu Wang
- Center for Intelligent Medical Electronics (CIME), School of Information Science and Engineering, Fudan University, Shanghai 200433, China.
| | - Saeed Akbarzadeh
- Center for Intelligent Medical Electronics (CIME), School of Information Science and Engineering, Fudan University, Shanghai 200433, China.
| | - Wei Chen
- Center for Intelligent Medical Electronics (CIME), School of Information Science and Engineering, Fudan University, Shanghai 200433, China; Human Phenome Institute, Fudan University, Shanghai 201203, China.
| |
Collapse
|
47
|
Berdina O, Madaeva I, Bolshakova S, Polyakov V, Bugun O, Rychkova L. Alteration of sleep homeostasis and cognitive impairment in apneic obese adolescents. Sleep Biol Rhythms 2021. [DOI: 10.1007/s41105-021-00317-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
48
|
Gorgoni M, De Gennaro L. Sleep in the Aging Brain. Brain Sci 2021; 11:229. [PMID: 33673285 PMCID: PMC7918041 DOI: 10.3390/brainsci11020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 02/05/2023] Open
Abstract
We have entered an era of a steep increase in the absolute and relative number of older people. This well-come phenomenon represents a major challenge for health care. However, maturational changes in sleep associated with aging do not easily appear as main factors, even though sleep alterations in the aging process lead to many detrimental consequences. In this editorial paper, we summarize the present knowledge about the main aging-related sleep modifications and their relevance for health problems and cognitive decline. Then, we present the papers published in the Special Issue "Disturbances of Sleep Among Older People".
Collapse
Affiliation(s)
- Maurizio Gorgoni
- Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy;
| | - Luigi De Gennaro
- Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy;
- IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| |
Collapse
|
49
|
Affiliation(s)
- Sandra Doria Xavier
- Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,Corresponding author: Sandra Doria Xavier. E-mail:
| |
Collapse
|
50
|
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: 32] [Impact Index Per Article: 8.0] [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
|