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
|
Satomi E, Apolinário D, Magaldi RM, Busse AL, Vieira Gomes GC, Ribeiro E, Genta PR, Piovezan RD, Poyares D, Jacob-Filho W, Suemoto CK. Beyond sleep: Rest and activity rhythm as a marker of preclinical and mild dementia in older adults with less education. Neurobiol Sleep Circadian Rhythms 2025; 18:100110. [PMID: 39834590 PMCID: PMC11745811 DOI: 10.1016/j.nbscr.2024.100110] [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: 09/24/2024] [Revised: 12/22/2024] [Accepted: 12/22/2024] [Indexed: 01/22/2025] Open
Abstract
Background Although sleep duration and sleep-related breathing disorders were associated with dementia previously, few studies examined the association between circadian rhythm association and cognitive status. Objective We aimed to investigate the association of rest and activity rhythm with cognitive performance in older people with cognitive complaints and less education. Methods Activity rhythm was evaluated with wrist actigraphy in 109 community-dwelling older people with cognitive complaints without diagnosed dementia. Each participant completed a neuropsychological battery and was classified as having cognitive impairment (MCI), dementia, or normal cognition. We used adjusted multinomial logistic regression and linear regression models to compare sleep and circadian non-parametric measures with cognitive groups and cognitive z-scores, respectively. Results The mean age of the 109 participants was 79.3 ± 6.3 years old, 74% were women, 68% were white, and the mean education was 5.6 ± 5.2 years. Daytime activity intensity was associated with better language (β = 0.178; 95% CI = 0.022, 0.334; p = 0.03) and visuospatial performance (β = 0.158; 95%CI = 0.008, 0.308; p = 0.04). Also, less fragmented rhythm was associated with better visuospatial (β = 0.172; 95%CI = 0.025, 0.320; p = 0.02) and global cognitive scores (β = 0.134; 95%CI = 0.005, 0.263; p = 0.04). More interdaily stability was associated with a lower risk of MCI and dementia (RR = 0.54; 95%CI = 0.29-0.99; p = 0.04, and RR = 0.44; 95%CI = 0.21-0.94; p = 0.03, respectively). Moreover, more daytime activity (RR = 0.40; 95%CI = 0.18-0.89; p = 0.02) and less rhythm fragmentation (RR = 0.31; 95%CI = 0.14-0.73; p = 0.007) were associated with lower risk for dementia. Conclusion Daytime activity intensity and fragmented rhythm during the day and night may play an important role as markers for cognitive impairment in less educated populations. Future studies with larger samples should confirm these findings.
Collapse
Affiliation(s)
- Erika Satomi
- Division of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Daniel Apolinário
- Division of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
- Hospital Do Coração (HCOR), São Paulo, Brazil
| | | | | | - Gisele Cristina Vieira Gomes
- Department of Physical Therapy, Speech Therapy and Occupational Therapy, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Elyse Ribeiro
- Department of Psychology, University of São Paulo Medical School, São Paulo, Brazil
| | - Pedro Rodrigues Genta
- Sleep Laboratory, Pulmonary Division, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Ronaldo Delmonte Piovezan
- Division of Sleep Medicine, Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
- Adelaide Geriatrics Training and Research with Aged Care (G-TRAC) Centre, Adelaide Medical School, Faculty of Health and Medical Sciences, Adelaide, SA, Australia
| | - Dalva Poyares
- Division of Sleep Medicine, Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Wilson Jacob-Filho
- Division of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | | |
Collapse
|
3
|
Jouvencel A, Dilharreguy B, Baillet M, Pérès K, Dartigues JF, Amieva H, Mayo W, Catheline G. Age-related disturbances in rest-activity rhythms and integrity of the hippocampal network: An exploratory study. Neurobiol Sleep Circadian Rhythms 2025; 18:100111. [PMID: 39834591 PMCID: PMC11743803 DOI: 10.1016/j.nbscr.2024.100111] [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: 10/20/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
To better understand the relationship between the rest-activity rhythms and cognitive impairments during aging, we assessed the longitudinal changes in the rest-activity rhythms in an elderly population and their possible detrimental effect on the hippocampal network. This was done longitudinally in a rural cohort with two actigraphic assessments and brain imaging examinations, seven years apart. A segmentation of the hippocampus and its related structures was used to assess volumes and functional connectivity in this network based on anatomical and resting state functional data. Regression models were carried out to investigate the potential association of the evolution of sleep and rest-activity rhythms parameters with the structural and functional integrity of the hippocampal network. Our sample was composed of 33 subjects aged 75.2 ± 2.4 years old at the first time point with 40% of women. After seven years, the sleep of our participants did not change but their rest-activity rhythms did (p < 0.05), with a decrease in relative amplitude (∂RA = -0.021) and stability (∂IS = -0.044) as well as an increase in fragmentation (∂IV = +0.072). The deterioration of rest-activity rhythms was correlated with a lower anterior hippocampal volume (p corrected <0.05) while no correlation with functional connectivity was observed. These findings suggest that a degradation of rest-activity rhythms in people over 70 years old could constitute a factor of hippocampal vulnerability. Preventive interventions should consider rest-activity rhythms in the oldest-old population.
Collapse
Affiliation(s)
- Aurore Jouvencel
- INCIA, EPHE, Université PSL, Univ Bordeaux, CNRS, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Bixente Dilharreguy
- INCIA, EPHE, Université PSL, Univ Bordeaux, CNRS, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Marion Baillet
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Karine Pérès
- INSERM, Bordeaux Population Health Research Center, University of Bordeaux, UMR U1219, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Jean-François Dartigues
- INSERM, Bordeaux Population Health Research Center, University of Bordeaux, UMR U1219, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Hélène Amieva
- INSERM, Bordeaux Population Health Research Center, University of Bordeaux, UMR U1219, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Willy Mayo
- INCIA, EPHE, Université PSL, Univ Bordeaux, CNRS, 146, Rue Léo Saignat, 33076, Bordeaux, France
| | - Gwenaëlle Catheline
- INCIA, EPHE, Université PSL, Univ Bordeaux, CNRS, 146, Rue Léo Saignat, 33076, Bordeaux, France
| |
Collapse
|
4
|
Pundir M, Lobanova L, Papagerakis P, Chen X, Papagerakis S. Competitive enzyme linked aptamer based assay for salivary melatonin detection. Sci Rep 2025; 15:14276. [PMID: 40274844 PMCID: PMC12022177 DOI: 10.1038/s41598-025-94304-7] [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: 12/02/2024] [Accepted: 03/12/2025] [Indexed: 04/26/2025] Open
Abstract
Melatonin is a key hormone that regulates the sleep-wake cycle and plays an important role in maintaining circadian rhythm and sleep onset. The daily rise in melatonin secretion is associated with an increased tendency to sleep, occurring approximately 2 h before bedtime. This correlation between melatonin levels and sleep onset makes it a reliable biomarker for circadian rhythm sleep-wake disorders. An accurate assessment of dim light melatonin onset (DLMO) is vital for understanding circadian timing and diagnosing sleep-wake cycle disruptions. However, the traditional methods for detecting melatonin in saliva are either complex or lack the sensitivity required for the accurate assessment of DLMO, especially at low concentrations. Here, we present a novel competitive enzyme-linked aptamer-based assay developed to detect melatonin in saliva. Unlike conventional assays, this technique utilizes chemically synthesized single-stranded DNA or RNA aptamers, which bind to melatonin with high specificity and sensitivity. The assay measures melatonin, attaining a linear dynamic range from 8.62 × 10‒6 M to 3.9 × 10‒11 M, with a detection limit of 2.5 × 10‒12 M (~ 0.57 pg/mL). Additionally, the aptamer showed small binding to its counter targets and acceptable recovery of melatonin when spiked in four times diluted saliva in assay buffer. Overall, the assay portrayed the potential of aptamers to detect low melatonin levels in saliva that could be beneficial in accurately determining DLMO, particularly in individuals with very low melatonin levels, such as the elderly or those with neurodegenerative conditions. Determining precise measurement of DLMO will facilitate the accurate diagnosis of circadian rhythm disruption, enabling healthcare providers to optimize the timing and selection of therapeutic and behavioural interventions tailored to an individual's unique circadian rhythm.
Collapse
Affiliation(s)
- Meenakshi Pundir
- Centre interprofessionnel pour la recherche clinique et appliquée (CIRCA), Faculty of Dentistry, Université Laval, Dental Medicine Pavilion, 2420, rue de la Terrasse, Quebec City, G1V0A6, Canada
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, S7N 5E4, Canada
- Division of Biomedical Engineering, University of Saskatchewan, 57 Campus Dr, Saskatoon, S7K 5A9, Canada
| | - Liubov Lobanova
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, S7N 5E4, Canada
| | - Petros Papagerakis
- Centre interprofessionnel pour la recherche clinique et appliquée (CIRCA), Faculty of Dentistry, Université Laval, Dental Medicine Pavilion, 2420, rue de la Terrasse, Quebec City, G1V0A6, Canada.
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, S7N 5E4, Canada.
- Division of Biomedical Engineering, University of Saskatchewan, 57 Campus Dr, Saskatoon, S7K 5A9, Canada.
| | - Xiongbiao Chen
- Division of Biomedical Engineering, University of Saskatchewan, 57 Campus Dr, Saskatoon, S7K 5A9, Canada
| | - Silvana Papagerakis
- Centre interprofessionnel pour la recherche clinique et appliquée (CIRCA), Faculty of Dentistry, Université Laval, Dental Medicine Pavilion, 2420, rue de la Terrasse, Quebec City, G1V0A6, Canada.
- Département d'oto-rhino-laryngologie - chirurgie cervico-faciale, Faculté de médecine, Université Laval, 1050, Avenue de la Médecine, Quebec City, G1V0A6, Canada.
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
5
|
Milton S, Cavaillès C, Ancoli-Israel S, Stone KL, Yaffe K, Leng Y. Five-Year Changes in 24-Hour Sleep-Wake Activity and Dementia Risk in Oldest Old Women. Neurology 2025; 104:e213403. [PMID: 40106755 PMCID: PMC11919274 DOI: 10.1212/wnl.0000000000213403] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/02/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Sleep disruptions are associated with cognitive aging in older adults. However, little is known about longitudinal sleep changes in the oldest old and whether these changes are linked to cognitive impairment. We aimed to determine whether changes in 24-hour multidimensional sleep-wake activity are associated with mild cognitive impairment (MCI) and dementia in oldest old women. METHODS We studied cognitively unimpaired women enrolled in the Study of Osteoporotic Fractures who completed wrist actigraphy twice (baseline and follow-up) and had cognitive status evaluated at follow-up using a neuropsychological battery and adjudication. To identify multidimensional sleep-wake change profiles, we performed hierarchical clustering on principal components on the 5-year changes (median 5.0 [range 3.5-6.3] years) in nighttime sleep (sleep duration, sleep efficiency [SE], and wake after sleep onset [WASO]), napping (duration and frequency), and circadian rest-activity rhythms (RARs; acrophase, amplitude, mesor, and robustness). Using multinomial logistic regression, we evaluated the associations between these profiles-and individual parameter changes-and MCI and dementia risk at follow-up. RESULTS Of 733 participants (mean age 82.5 ± 2.9 years), 164 (22.4%) developed MCI and 93 (12.7%) developed dementia by the follow-up visit. We identified 3 sleep-wake change profiles: stable sleep (SS; n = 321 [43.8%]) was characterized by stability or small improvements; declining nighttime sleep (n = 256 [34.9%]) showed decreases in nighttime sleep quality and duration, moderate napping increases, and worsening circadian RARs; and increasing sleepiness (IS; n = 156 [21.3%]) exhibited large increases in daytime and nighttime sleep duration and quality, and worsening circadian RARs. After adjustment for age, education, race, body mass index, diabetes, hypertension, myocardial infarction, antidepressant use, and baseline cognition, women with IS had approximately double the risk of dementia (odds ratio 2.21, 95% CI 1.14-4.26) compared with those with SS. SE, WASO, nap duration, and nap frequency were individually associated with dementia. Neither sleep-wake change profiles nor individual parameters were associated with MCI. DISCUSSION Among community-dwelling women in their 80s, those with increasing 24-hour sleepiness over 5 years had doubled dementia risk during that time. Change in multidimensional 24-hour sleep-wake activity may serve as an early marker or risk factor for dementia in oldest old women.
Collapse
Affiliation(s)
- Sasha Milton
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| | - Clémence Cavaillès
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| | | | - Katie L Stone
- Research Institute, California Pacific Medical Center, San Francisco
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
- Department of Epidemiology and Biostatistics, University of California, San Francisco
- Department of Neurology, University of California, San Francisco; and
- San Francisco Veterans Affairs Health Care System, CA
| | - Yue Leng
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| |
Collapse
|
6
|
Bianchin MM, Zimmer ER. Wake-Up Call: The Association Between Sleep Disturbances and Dementia. Neurology 2025; 104:e213516. [PMID: 40106754 DOI: 10.1212/wnl.0000000000213516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/28/2025] [Indexed: 03/22/2025] Open
Affiliation(s)
- Marino Muxfeldt Bianchin
- Division of Neurology, Hospital de Clínicas de Porto Alegre, Brazil
- Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Brazil
- BRAIN - Basic Research and Advanced Investigations in Neurosciences, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Brazil
| | - Eduardo Rigon Zimmer
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica (PPGbioq) e Farmacologia e Terapêutica (PPGFT), Universidade Federal do Rio Grande do Sul, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Brazil; and
- McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Turton SM, Padgett S, Maisel MT, Johnson CE, Buzinova VA, Barth SE, Kohler K, Spearman HM, Macheda T, Manauis EC, Guo LZ, Whitlock HR, Bachstetter AD, Sunderam S, O'Hara BF, Duncan MJ, Murphy MP. Interactions between daily sleep-wake rhythms, γ-secretase, and amyloid-β peptide pathology point to complex underlying relationships. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167840. [PMID: 40222459 DOI: 10.1016/j.bbadis.2025.167840] [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: 01/23/2025] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Disrupted or insufficient sleep is a well-documented risk factor for Alzheimer's disease (AD) and related dementias. Previous studies in our lab and others have shown that chronic fragmentation of the daily sleep-wake rhythm in mice can accelerate the development of AD-related neuropathology in the brain, including increases in the levels of amyloid-β (Aβ). Although sleep is known to increase clearance of Aβ via the glymphatic system, little is known about the effect of sleep on Aβ production and the role this might play in amyloid deposition. To examine the relationship of Aβ production and its interaction with sleep and sleep dysfunction, we treated mice from an APP × PS1 mutant knock-in line (APPΔNLh/ΔNLh × PS1P264L/P264L) with an inhibitor of γ-secretase (LY-450,139; Semagacestat®) during a protocol of mild sleep fragmentation (SF). Compared to the male mice, the female mice slept less, and had more Aβ pathology. Semagacestat treatment reduced Aβ, but only in the most soluble extractable fraction. Although the female mice showed an increase in the amount of Aβ following SF, this effect was blocked by Semagacestat, an effect that was not seen in the male mice. SF also led to a significant, sex-dependent changes in the relative amounts of C-terminal fragments of the amyloid precursor protein, the immediate substrate of the γ-secretase enzyme. These findings indicate that the relationship between disruption of the daily sleep-wake rhythm and the development of AD-related pathology is complex, and may involve unappreciated interactions with biological sex. Consideration of these factors is necessary for a better understanding of AD risk, especially the elevated risk in women.
Collapse
Affiliation(s)
| | | | | | - Carrie E Johnson
- The Sanders-Brown Center on Aging, USA; Department of Molecular and Cellular Biochemistry, USA
| | - Valeria A Buzinova
- The Sanders-Brown Center on Aging, USA; Department of Molecular and Cellular Biochemistry, USA
| | | | | | | | | | | | | | | | - Adam D Bachstetter
- The Sanders-Brown Center on Aging, USA; The Spinal Cord and Brain Injury Research Center, USA; Department of Neuroscience, USA
| | | | | | | | - M Paul Murphy
- The Sanders-Brown Center on Aging, USA; Department of Molecular and Cellular Biochemistry, USA.
| |
Collapse
|
8
|
Yang H, Niu L, Tian L, Hu Y, Cheng C, Li S, Le W. Circadian rhythm disturbances in Alzheimer's disease: insights from plaque-free and plaque-burdened stages in APP SWE/PS1 dE9 mice. Alzheimers Res Ther 2025; 17:76. [PMID: 40188157 PMCID: PMC11971749 DOI: 10.1186/s13195-025-01724-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Disruptions in circadian rhythms are commonly observed in patients with Alzheimer's disease (AD) and could potentially accelerate the progression of the condition. However, the relationship between circadian rhythm disruptions and AD development, as well as the mechanisms involved, remain poorly understood. METHODS This study investigated the circadian behavior, rhythmic gene expression in multiple brain regions, and its correlation with sleep architecture of AD mice at two disease stages: plaque-free stage (2-month-old) and plaque-burdened stage (10-month-old) as compared to age-matched wild-type (WT) mice. RESULTS Two-month-old AD mice already displayed alteration in the activity patterns compared to WT mice, showing increased activity during the light phase and decreased activity during the dark phase, and the change in the activity pattern of 10-month-old AD mice was more significant. Further, electroencephalogram (EEG) examination showed increased wakefulness and reduced non-rapid eye movement (NREM) sleep in 2- and 10-month-old AD mice. In addition, we documented a significant change in circadian core clock genes in the suprachiasmatic nucleus (SCN), hippocampus, and cortex of 2- and 10-month-old AD mice. Correlation analyses demonstrated the close relationship between circadian clock gene expression level and specific sleep-wake parameters, especially within the SCN and hippocampus. CONCLUSIONS These findings revealed that circadian rhythm disturbances in AD mice preceded Aβ deposition. The circadian rhythm disturbances observed in the early AD might be attributed to the abnormal expression of core clock genes in the brain regions involved in circadian rhythm regulation.
Collapse
Affiliation(s)
- Huijia Yang
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Long Niu
- Department of Neurology, Heping Hospital affiliated to Changzhi Medical College, Changzhi, China
| | - Lulu Tian
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Yiying Hu
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Cheng Cheng
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Weidong Le
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China.
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
9
|
Mortimer T, Smith JG, Muñoz-Cánoves P, Benitah SA. Circadian clock communication during homeostasis and ageing. Nat Rev Mol Cell Biol 2025; 26:314-331. [PMID: 39753699 DOI: 10.1038/s41580-024-00802-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 03/28/2025]
Abstract
Maintaining homeostasis is essential for continued health, and the progressive decay of homeostatic processes is a hallmark of ageing. Daily environmental rhythms threaten homeostasis, and circadian clocks have evolved to execute physiological processes in a manner that anticipates, and thus mitigates, their effects on the organism. Clocks are active in almost all cell types; their rhythmicity and functional output are determined by a combination of tissue-intrinsic and systemic inputs. Numerous inputs for a specific tissue are produced by the activity of circadian clocks of other tissues or cell types, generating a form of crosstalk known as clock communication. In mammals, the central clock in the hypothalamus integrates signals from external light-dark cycles to align peripheral clocks elsewhere in the body. This regulation is complemented by a tissue-specific milieu of external, systemic and niche inputs that modulate and cooperate with the cellular circadian clock machinery of a tissue to tailor its functional output. These mechanisms of clock communication decay during ageing, and growing evidence suggests that this decline might drive ageing-related morbidities. Dietary, behavioural and pharmacological interventions may offer the possibility to overcome these changes and in turn improve healthspan.
Collapse
Affiliation(s)
- Thomas Mortimer
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Jacob G Smith
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain.
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain.
| | - Pura Muñoz-Cánoves
- Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
- Altos Labs Inc., San Diego Institute of Science, San Diego, CA, USA.
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
10
|
Paget‐Blanc A, Thurston RC, Smagula SF, Chang Y, Maki PM. Rest-activity rhythm characteristics associated with lower cognitive performance and Alzheimer's disease biomarkers in midlife women. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70105. [PMID: 40242837 PMCID: PMC12000222 DOI: 10.1002/dad2.70105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/26/2025] [Accepted: 03/07/2025] [Indexed: 04/18/2025]
Abstract
INTRODUCTION Disrupted rest-activity rhythms (RARs) have been linked to poorer cognitive function and Alzheimer's disease (AD) biomarkers. Here we extend this work to midlife women, who commonly experience menopause-related sleep and cognitive problems. METHODS One hundred ninety-four postmenopausal participants underwent a neuropsychological evaluation, 72 h of wrist actigraphy generating RAR variables, and a blood draw to measure AD biomarkers: phosphorylated tau (p-tau181, p-tau231) and amyloid beta (Aβ40, Aβ42). RESULTS Lower interdaily stability (IS) and relative amplitude (RA) and higher interdaily variability (IV) and least active 5 h (L5) were associated with worse processing speed, independent of sleep. Adjustment for sleep significantly attenuated the associations of RA with memory. Lower RA was associated with higher p-tau231 level, independent of sleep. Further adjustment for menopause-related factors modestly accounted for the associations between RAR, cognitive measures, and AD biomarkers. DISCUSSION Weaker RAR, particularly RA, was associated with worse cognitive functions, and higher AD biomarkers levels, possibly linking RAR with AD pathology in women. Highlights Lower rhythm stability and robustness and higher fragmentation were associated with worse processing speed.Lower robustness was associated with higher levels of phosphorylated tau-231.Menopause factors did not attenuate the association between rest-activity rhythms and cognitive function.
Collapse
Affiliation(s)
| | - Rebecca C. Thurston
- Department of Psychiatry, School of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Epidemiology, Graduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Stephen F. Smagula
- Department of Psychiatry, School of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Epidemiology, Graduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Yuefang Chang
- Department of Epidemiology, Graduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Pauline M. Maki
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of PsychologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of Obstetrics and GynecologyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
11
|
Liberale L, Tual-Chalot S, Sedej S, Ministrini S, Georgiopoulos G, Grunewald M, Bäck M, Bochaton-Piallat ML, Boon RA, Ramos GC, de Winther MPJ, Drosatos K, Evans PC, Ferguson JF, Forslund-Startceva SK, Goettsch C, Giacca M, Haendeler J, Kallikourdis M, Ketelhuth DFJ, Koenen RR, Lacolley P, Lutgens E, Maffia P, Miwa S, Monaco C, Montecucco F, Norata GD, Osto E, Richardson GD, Riksen NP, Soehnlein O, Spyridopoulos I, Van Linthout S, Vilahur G, Wentzel JJ, Andrés V, Badimon L, Benetos A, Binder CJ, Brandes RP, Crea F, Furman D, Gorbunova V, Guzik TJ, Hill JA, Lüscher TF, Mittelbrunn M, Nencioni A, Netea MG, Passos JF, Stamatelopoulos KS, Tavernarakis N, Ungvari Z, Wu JC, Kirkland JL, Camici GG, Dimmeler S, Kroemer G, Abdellatif M, Stellos K. Roadmap for alleviating the manifestations of ageing in the cardiovascular system. Nat Rev Cardiol 2025:10.1038/s41569-025-01130-5. [PMID: 39972009 DOI: 10.1038/s41569-025-01130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Ageing of the cardiovascular system is associated with frailty and various life-threatening diseases. As global populations grow older, age-related conditions increasingly determine healthspan and lifespan. The circulatory system not only supplies nutrients and oxygen to all tissues of the human body and removes by-products but also builds the largest interorgan communication network, thereby serving as a gatekeeper for healthy ageing. Therefore, elucidating organ-specific and cell-specific ageing mechanisms that compromise circulatory system functions could have the potential to prevent or ameliorate age-related cardiovascular diseases. In support of this concept, emerging evidence suggests that targeting the circulatory system might restore organ function. In this Roadmap, we delve into the organ-specific and cell-specific mechanisms that underlie ageing-related changes in the cardiovascular system. We raise unanswered questions regarding the optimal design of clinical trials, in which markers of biological ageing in humans could be assessed. We provide guidance for the development of gerotherapeutics, which will rely on the technological progress of the diagnostic toolbox to measure residual risk in elderly individuals. A major challenge in the quest to discover interventions that delay age-related conditions in humans is to identify molecular switches that can delay the onset of ageing changes. To overcome this roadblock, future clinical trials need to provide evidence that gerotherapeutics directly affect one or several hallmarks of ageing in such a manner as to delay, prevent, alleviate or treat age-associated dysfunction and diseases.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Myriam Grunewald
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Magnus Bäck
- Translational Cardiology, Centre for Molecular Medicine, Department of Medicine Solna, and Department of Cardiology, Heart and Vascular Centre, Karolinska Institutet, Stockholm, Sweden
- Inserm, DCAC, Université de Lorraine, Nancy, France
| | | | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC location VUmc, Amsterdam, Netherlands
| | - Gustavo Campos Ramos
- Department of Internal Medicine I/Comprehensive Heart Failure Centre, University Hospital Würzburg, Würzburg, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences: Atherosclerosis and Ischaemic Syndromes; Amsterdam Infection and Immunity: Inflammatory Diseases, Amsterdam UMC location AMC, Amsterdam, Netherlands
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul C Evans
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mauro Giacca
- British Heart foundation Centre of Reseach Excellence, King's College London, London, UK
| | - Judith Haendeler
- Cardiovascular Degeneration, Medical Faculty, University Hospital and Heinrich-Heine University, Düsseldorf, Germany
| | - Marinos Kallikourdis
- Adaptive Immunity Lab, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Daniel F J Ketelhuth
- Cardiovascular and Renal Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rory R Koenen
- CARIM-School for Cardiovascular Diseases, Department of Biochemistry, Maastricht University, Maastricht, Netherlands
| | | | - Esther Lutgens
- Department of Cardiovascular Medicine & Immunology, Mayo Clinic, Rochester, MN, USA
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Satomi Miwa
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Claudia Monaco
- Kennedy Institute, NDORMS, University of Oxford, Oxford, UK
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Münster, Germany
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin, Germany
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu y Sant Pau l, IIB-Sant Pau, Barcelona, Spain
| | - Jolanda J Wentzel
- Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, Netherlands
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBERCV, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Health and Innovation Research Foundation (FICSI) and Cardiovascular Health and Network Medicine Department, University of Vic (UVIC-UCC), Barcelona, Spain
| | - Athanase Benetos
- Department of Geriatrics, University Hospital of Nancy and Inserm DCAC, Université de Lorraine, Nancy, France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Roma, Italy
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Joseph A Hill
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas F Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche-DIMI, Università degli Studi di Genova, Genova, Italy
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Kimon S Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Tavernarakis
- Medical School, University of Crete, and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Zoltan Ungvari
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm, Institut Universitaire de France, Paris, France
| | | | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
12
|
Chan JWY, Li CT, Chau SWH, Chan NY, Li TMH, Huang B, Tsoh J, Li SX, Chong KKL, Roecklein KA, Wing YK. Attenuated melanopsin-mediated post-illumination pupillary response is associated with reduced actigraphic amplitude and mesor in older adults. Sleep 2025; 48:zsae239. [PMID: 39383299 DOI: 10.1093/sleep/zsae239] [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: 06/24/2024] [Revised: 09/03/2024] [Indexed: 10/11/2024] Open
Abstract
STUDY OBJECTIVES This study aimed to explore the relationship between post-illumination pupillary response (PIPR) with sleep and circadian measures in a community sample of healthy older adults. METHODS Eligible participants were invited to complete a 1 week sleep diary and actigraphy, and provide an overnight urine sample to measure urinary 6-sulfatoxymelatonin (aMT6s). PIPR was defined as the (1) pupil constriction at 6 second poststimulus (PIPR-6s) and (2) for -30s beginning 10 seconds after stimulus (PIPR-30s), normalized as a percentage to the baseline pupil diameter, after 1 second of blue and 1 second of red light stimulus, respectively. The Net-PIPRs were reported by subtracting the PIPR to red stimulus from the PIPR to blue stimulus. The relationship between PIPR metrics to aMT6s and actigraphic rest-activity rhythm parameters was examined by generalized linear models. RESULTS A total of 48 participants were recruited (mean age: 62.6 ± 7.1 years, male: 44%). Both Net PIPR-6s and Net PIPR-30s were significantly associated with actigraphic rest-activity amplitude (B = 0.03, p = .001 and B = 0.03, p = .01, respectively) and actigraphic rest-activity mesor (B = 0.02, p = .001 and B = 0.03, p = .004, respectively). Additionally, the Net PIPR-30s were positively associated with overnight aMT6s level (B = 0.04, p = .03) and negatively associated with actigraphic rest-activity acrophase (B = -0.01, p = .004) in the fully adjusted models. CONCLUSIONS Attenuated PIPR is associated with a reduced actigraphic amplitude and mesor. The reduced retinal light responsivity may be a potential pathway contributing to impaired photic input to the circadian clock and resulted in age-related circadian changes in older adults.
Collapse
Affiliation(s)
- Joey W Y Chan
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chun-Tung Li
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Steven Wai Ho Chau
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ngan Yin Chan
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tim Man-Ho Li
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bei Huang
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joshua Tsoh
- Department of Psychiatry, Prince of Wales Hospital, Hong Kong SAR, China
| | - Shirley X Li
- Department of Psychology, The University of Hong Kong, Hong Kong SAR, China
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Kelvin K L Chong
- Department of Ophthalmology and Visual Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Yun Kwok Wing
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
13
|
Samson DR, McKinnon L. Are humans facing a sleep epidemic or enlightenment? Large-scale, industrial societies exhibit long, efficient sleep yet weak circadian function. Proc Biol Sci 2025; 292:20242319. [PMID: 39999887 PMCID: PMC11858753 DOI: 10.1098/rspb.2024.2319] [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: 09/26/2024] [Revised: 11/04/2024] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
The Centers for Disease Control and Prevention declared sleep-related problems to be a public health epidemic. With the advent of biometric sleep tracking technology taking the sleep lab into the field, the study of human sleep is now global, and these new datasets show contrasting findings. Previous reports suggest sleep in small-scale, non-industrial societies to be short and fragmented yet characterized by greater circadian rhythmicity. However, the role of circadian rhythm indicators in understanding global variations in human sleep patterns remains unclear. We examine population-level sleep studies (n = 54) using polysomnography and actigraphy to test the sleep restriction epidemic hypothesis, which posits that labour demands and technological disruption in large-scale, industrial societies have reduced sleep duration. We used an actigraphy-generated circadian function index from both non-industrial and industrial societies (n = 866) to test the circadian mismatch hypothesis, which suggests that poor chronohygiene in regulated environments misaligns circadian rhythms in industrial societies. In rejection of the sleep restriction epidemic hypothesis, our results show that industrial societies experience the longest, most efficient sleep, whereas in support of the circadian mismatch hypothesis, sleepers in non-industrial societies are characterized by the greatest circadian function.
Collapse
Affiliation(s)
- David Ryan Samson
- Department of Anthropology, University of Toronto Mississauga, Mississauga, OntarioL5L 1C6, Canada
| | - Leela McKinnon
- Department of Anthropology, University of Toronto Mississauga, Mississauga, OntarioL5L 1C6, Canada
| |
Collapse
|
14
|
Gubin D, Weinert D, Stefani O, Otsuka K, Borisenkov M, Cornelissen G. Wearables in Chronomedicine and Interpretation of Circadian Health. Diagnostics (Basel) 2025; 15:327. [PMID: 39941257 PMCID: PMC11816745 DOI: 10.3390/diagnostics15030327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/14/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Wearable devices have gained increasing attention for use in multifunctional applications related to health monitoring, particularly in research of the circadian rhythms of cognitive functions and metabolic processes. In this comprehensive review, we encompass how wearables can be used to study circadian rhythms in health and disease. We highlight the importance of these rhythms as markers of health and well-being and as potential predictors for health outcomes. We focus on the use of wearable technologies in sleep research, circadian medicine, and chronomedicine beyond the circadian domain and emphasize actigraphy as a validated tool for monitoring sleep, activity, and light exposure. We discuss various mathematical methods currently used to analyze actigraphic data, such as parametric and non-parametric approaches, linear, non-linear, and neural network-based methods applied to quantify circadian and non-circadian variability. We also introduce novel actigraphy-derived markers, which can be used as personalized proxies of health status, assisting in discriminating between health and disease, offering insights into neurobehavioral and metabolic status. We discuss how lifestyle factors such as physical activity and light exposure can modulate brain functions and metabolic health. We emphasize the importance of establishing reference standards for actigraphic measures to further refine data interpretation and improve clinical and research outcomes. The review calls for further research to refine existing tools and methods, deepen our understanding of circadian health, and develop personalized healthcare strategies.
Collapse
Affiliation(s)
- Denis Gubin
- Department of Biology, Tyumen Medical University, 625023 Tyumen, Russia
- Laboratory for Chronobiology and Chronomedicine, Research Institute of Biomedicine and Biomedical Technologies, Tyumen Medical University, 625023 Tyumen, Russia
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Dietmar Weinert
- Institute of Biology/Zoology, Martin Luther University, 06108 Halle-Wittenberg, Germany;
| | - Oliver Stefani
- Department Engineering and Architecture, Institute of Building Technology and Energy, Lucerne University of Applied Sciences and Arts, 6048 Horw, Switzerland;
| | - Kuniaki Otsuka
- Tokyo Women’s Medical University, Tokyo 162-8666, Japan;
- Halberg Chronobiology Center, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Mikhail Borisenkov
- Department of Molecular Immunology and Biotechnology, Institute of Physiology of Komi Science Centre, Ural Branch of the Russian Academy of Sciences, 167982 Syktyvkar, Russia;
| | - Germaine Cornelissen
- Halberg Chronobiology Center, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
15
|
DU S, Fang L, Li Y, Liu S, Luo X, Zeng S, Zheng S, Yang H, Xu Y, Li D, Zhang B. Association between post-COVID-19 sleep disturbance and neurocognitive function: a comparative study based on propensity score matching. J Zhejiang Univ Sci B 2025; 26:172-184. [PMID: 40015936 PMCID: PMC11867782 DOI: 10.1631/jzus.b2300831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/19/2024] [Indexed: 03/01/2025]
Abstract
Despite that sleep disturbance and poor neurocognitive performance are common complaints among coronavirus disease 2019 (COVID-19) survivors, few studies have focused on the effect of post-COVID-19 sleep disturbance (PCSD) on cognitive function. This study aimed to identify the impact of PCSD on neurocognitive function and explore the associated risk factors for the worsening of this condition. This cross-sectional study was conducted via the web-based assessment in Chinese mainland. Neurocognitive function was evaluated by the modified online Integrated Cognitive Assessment (ICA) and the Number Ordering Test (NOT). Propensity score matching (PSM) was utilized to match the confounding factors between individuals with and without PCSD. Univariate analyses were performed to evaluate the effect of PCSD on neurocognitive function. The risk factors associated with worsened neurocognitive performance in PCSD individuals were explored using binary logistic regression. A total of 8692 individuals with COVID-19 diagnosis were selected for this study. Nearly half (48.80%) of the COVID-19 survivors reported sleep disturbance. After matching by PSM, a total of 3977 pairs (7954 individuals in total) were obtained. Univariate analyses revealed that PCSD was related to worse ICA and NOT performance (P<0.05). Underlying disease, upper respiratory infection, loss of smell or taste, severe pneumonia, and self-reported cognitive complaints were associated with worsened neurocognitive performance among PCSD individuals (P<0.05). Furthermore, aging, ethnicity (minority), and lower education level were found to be independent risk factors for worsened neurocognitive performance in PCSD individuals (P<0.05). PCSD was related to impaired neurocognitive performance. Therefore, appropriate prevention and intervention measures should be taken to minimize or prevent PCSD and eliminate its potential adverse effect on neurocognitive function.
Collapse
Affiliation(s)
- Shixu DU
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China
| | - Leqin Fang
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China
| | - Yuanhui Li
- Adai Technology (Beijing) Company Limited, Beijing 100083, China
| | - Shuai Liu
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China
| | - Xue Luo
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China
| | - Shufei Zeng
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China
| | - Shuqiong Zheng
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China
| | - Hangyi Yang
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China
| | - Yan Xu
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China
| | - Dai Li
- Adai Technology (Beijing) Company Limited, Beijing 100083, China
| | - Bin Zhang
- Department of Psychiatry, Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou 510515, China.
| |
Collapse
|
16
|
Santisteban MM, Iadecola C. The pathobiology of neurovascular aging. Neuron 2025; 113:49-70. [PMID: 39788087 DOI: 10.1016/j.neuron.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025]
Abstract
As global life expectancy increases, age-related brain diseases such as stroke and dementia have become leading causes of death and disability. The aging of the neurovasculature is a critical determinant of brain aging and disease risk. Neurovascular cells are particularly vulnerable to aging, which induces significant structural and functional changes in arterial, venous, and lymphatic vessels. Consequently, neurovascular aging impairs oxygen and glucose delivery to active brain regions, disrupts endothelial transport mechanisms essential for blood-brain exchange, compromises proteostasis by reducing the clearance of potentially toxic proteins, weakens immune surveillance and privilege, and deprives the brain of key growth factors required for repair and renewal. In this review, we examine the effects of neurovascular aging on brain function and its role in stroke, vascular cognitive impairment, and Alzheimer's disease. Finally, we discuss key unanswered questions that must be addressed to develop neurovascular strategies aimed at promoting healthy brain aging.
Collapse
Affiliation(s)
- Monica M Santisteban
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
17
|
Zhang M, Chi NC, Gardner SE, Moon C. Rest-activity rhythm and cognitive function in older adults: A scoping review and integrative framework. Geriatr Nurs 2025; 61:80-90. [PMID: 39546912 DOI: 10.1016/j.gerinurse.2024.10.074] [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: 04/28/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Rest-activity rhythm (RAR) encompasses sleep, activity, and rest across a 24-hour cycle and over time (i.e., peak activity timing, mean activity level, activity amplitude, and rhythmicity). While the link between sleep, exercise, and cognitive function is established, the specific relationship between RAR and cognitive function in older adults remains unclear. This review synthesizes the literature on RAR and cognition, identifying 26 eligible studies from databases including PubMed, CINAHL, PsycINFO, and Web of Science. Although it was difficult to draw definitive conclusions due to the limited number of available studies focusing on each of cognitive domain and methodological heterogeneity among them, the collective findings broadly indicate that later peak activity timing, higher mean activity levels, higher activity amplitude, and more stable rhythmicity were associated with better global cognition. Future research with larger, diverse samples, more accurate RAR and cognition measurements, and incorporating biomarkers will provide deeper insights into these relationships.
Collapse
Affiliation(s)
- Meina Zhang
- College of Nursing, University of Iowa, Iowa City, IA, USA.
| | - Nai-Ching Chi
- College of Nursing, University of Iowa, Iowa City, IA, USA
| | - Sue E Gardner
- College of Nursing, University of Iowa, Iowa City, IA, USA
| | - Chooza Moon
- College of Nursing, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
18
|
Silvestri R, Guarnieri B. Advanced sleep phase syndrome: Role of genetics and aging. HANDBOOK OF CLINICAL NEUROLOGY 2025; 206:61-70. [PMID: 39864932 DOI: 10.1016/b978-0-323-90918-1.00005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Advanced sleep phase (ASP) is seldom brought to medical attention because many individuals easily adapt to their early chronotype, especially if it emerges before the age of 30 and is present in a first-degree relative. In this case, the disorder is considered familial (FASP) and is mostly discovered coincidentally in the presence of other sleep disorders, mainly obstructive sleep apnea syndrome (OSAS). The prevalence of FASP is currently estimated to be between 0.21% and 0.5%. Autosomal dominant mutations in circadian clock genes like PER2, CK1, PER3, CRY2, TIMELESS, and DEC2 have been linked to FASP, some with pleiotropic effects influencing other health aspects like migraine and depression. Early morning awakening is, instead, more common among older individuals, occurring in almost 4% of cases, without considering associated comorbidities. Advanced sleep-wake phase disorder (ASWPD) is characterized by a consistent and distressing anticipation of sleep-wake timing, affecting almost 1% of middle-aged individuals. On average, women have a shorter circadian period than men, making them more susceptible to ASWPD, albeit no significant gender discrepancies have been observed. Age-related alterations in circadian rhythms are exacerbated and compounded by neurodegenerative disorders, impacting the suprachiasmatic nucleus (SCN), sensitivity to light, and light responsiveness in those affected. Conflicting data has surfaced regarding the protective or detrimental effects of ASWPD in studies on aging, mild cognitive impairment (MCI), and diverse dementia conditions.
Collapse
Affiliation(s)
- Rosalia Silvestri
- Sleep Medicine Center, Department of Clinical and Experimental Medicine, AOU G. Martino, Messina, Italy
| | - Biancamaria Guarnieri
- Sleep Medicine Center, Department of Neurology, Villa Serena Hospital, Città S. Angelo, Pescara, Italy; Villaserena Research Foundation, Città S. Angelo, Pescara, Italy
| |
Collapse
|
19
|
Safiri S, Ghaffari Jolfayi A, Fazlollahi A, Morsali S, Sarkesh A, Daei Sorkhabi A, Golabi B, Aletaha R, Motlagh Asghari K, Hamidi S, Mousavi SE, Jamalkhani S, Karamzad N, Shamekh A, Mohammadinasab R, Sullman MJM, Şahin F, Kolahi AA. Alzheimer's disease: a comprehensive review of epidemiology, risk factors, symptoms diagnosis, management, caregiving, advanced treatments and associated challenges. Front Med (Lausanne) 2024; 11:1474043. [PMID: 39736972 PMCID: PMC11682909 DOI: 10.3389/fmed.2024.1474043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 01/01/2025] Open
Abstract
Background Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired reasoning. It is the leading cause of dementia in older adults, marked by the pathological accumulation of amyloid-beta plaques and neurofibrillary tangles. These pathological changes lead to widespread neuronal damage, significantly impacting daily functioning and quality of life. Objective This comprehensive review aims to explore various aspects of Alzheimer's disease, including its epidemiology, risk factors, clinical presentation, diagnostic advancements, management strategies, caregiving challenges, and emerging therapeutic interventions. Methods A systematic literature review was conducted across multiple electronic databases, including PubMed, MEDLINE, Cochrane Library, and Scopus, from their inception to May 2024. The search strategy incorporated a combination of keywords and Medical Subject Headings (MeSH) terms such as "Alzheimer's disease," "epidemiology," "risk factors," "symptoms," "diagnosis," "management," "caregiving," "treatment," and "novel therapies." Boolean operators (AND, OR) were used to refine the search, ensuring a comprehensive analysis of the existing literature on Alzheimer's disease. Results AD is significantly influenced by genetic predispositions, such as the apolipoprotein E (APOE) ε4 allele, along with modifiable environmental factors like diet, physical activity, and cognitive engagement. Diagnostic approaches have evolved with advances in neuroimaging techniques (MRI, PET), and biomarker analysis, allowing for earlier detection and intervention. The National Institute on Aging and the Alzheimer's Association have updated diagnostic criteria to include biomarker data, enhancing early diagnosis. Conclusion The management of AD includes pharmacological treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, which provide symptomatic relief but do not slow disease progression. Emerging therapies, including amyloid-beta and tau-targeting treatments, gene therapy, and immunotherapy, offer potential for disease modification. The critical role of caregivers is underscored, as they face considerable emotional, physical, and financial burdens. Support programs, communication strategies, and educational interventions are essential for improving caregiving outcomes. While significant advancements have been made in understanding and managing AD, ongoing research is necessary to identify new therapeutic targets and enhance diagnostic and treatment strategies. A holistic approach, integrating clinical, genetic, and environmental factors, is essential for addressing the multifaceted challenges of Alzheimer's disease and improving outcomes for both patients and caregivers.
Collapse
Affiliation(s)
- Saeid Safiri
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghaffari Jolfayi
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Asra Fazlollahi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Morsali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnam Golabi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Aletaha
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kimia Motlagh Asghari
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sana Hamidi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Seyed Ehsan Mousavi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepehr Jamalkhani
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Karamzad
- Department of Persian Medicine, School of Traditional, Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mark J. M. Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Landvreugd A, Nivard MG, Bartels M. The Effect of Light on Wellbeing: A Systematic Review and Meta-analysis. JOURNAL OF HAPPINESS STUDIES 2024; 26:1. [PMID: 39664799 PMCID: PMC11628446 DOI: 10.1007/s10902-024-00838-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/31/2024] [Indexed: 12/13/2024]
Abstract
Due to the dominant presence of studies and reviews exploring the impact of light on physical and mental illness, studies specifically investigating the effect of light on wellbeing are often overshadowed. The aim of this review is to give an overview of specifically these studies conducted on light and wellbeing, and to summarize the reported effects. After a literature search in PubMed, PsycInfo, and Web of Science, 74 studies were found eligible to be included in this systematic review, i.e. they included surveys assessing wellbeing, happiness, life satisfaction, positive affect, or quality of life. Of these 74 studies, 30 were included in the meta-analysis and assessed for their risk of bias. The meta-analysis showed a pooled effect size of 0.46 (CI = 0.29-0.62), indicating that light has a small-to-moderate positive effect on wellbeing. After removing outliers and studies with a high risk of bias, the sensitivity analysis showed the pooled effect size to be robust (0.53, CI = 0.35-0.72). Although the sensitivity analysis indicated a robust effect, the results might still be biased due to the relatively small sample sizes, risk of bias in the designs (due to e.g. difficulties handling confounders and the reporting of the outcomes), and publication bias. We encourage future studies to replicate these positive results in larger samples, and to give extensive details about the light design and statistical outcomes, to increase the number of studies that can be included in these types of systematic reviews. Supplementary Information The online version contains supplementary material available at 10.1007/s10902-024-00838-4.
Collapse
Affiliation(s)
- A. Landvreugd
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Van Der Boechorstraat 7-9, 1081 BT Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam University Medical Centres, Van Der Boechorstraat 7-9, 1081 BT Amsterdam, The Netherlands
| | - M. G. Nivard
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Van Der Boechorstraat 7-9, 1081 BT Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam University Medical Centres, Van Der Boechorstraat 7-9, 1081 BT Amsterdam, The Netherlands
| | - M. Bartels
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Van Der Boechorstraat 7-9, 1081 BT Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam University Medical Centres, Van Der Boechorstraat 7-9, 1081 BT Amsterdam, The Netherlands
| |
Collapse
|
21
|
Pappas JA, Miner B. Sleep Deficiency in the Elderly. Sleep Med Clin 2024; 19:593-606. [PMID: 39455180 DOI: 10.1016/j.jsmc.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
With aging, there are normative changes to sleep physiology and circadian rhythmicity that may predispose older adults to sleep deficiency, whereas many health-related and psychosocial/behavioral factors may precipitate sleep deficiency. In this article, we describe age-related changes to sleep and describe how the health-related and psychosocial/behavioral factors typical of aging may converge in older adults to increase the risk for sleep deficiency. Next, we review the consequences of sleep deficiency in older adults, focusing specifically on important age-related outcomes, including mortality, cognition, depression, and physical function. Finally, we review treatments for sleep deficiency, highlighting safe and effective nonpharmacologic interventions.
Collapse
Affiliation(s)
- Jane Alexandra Pappas
- San Juan Bautista School of Medicine, Salida 21 Carr. 172 Urb. Turabo Gardens, Caguas 00726, Puerto Rico
| | - Brienne Miner
- Section of Geriatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
22
|
Jin Z, Xing Y, Duan P, Bi Y, Li X, Feng W, Zhang B. Revealing the molecular links between coronary heart disease and cognitive impairment: the role of aging-related genes and therapeutic potential of stellate ganglion block. Biogerontology 2024; 26:16. [PMID: 39609308 PMCID: PMC11604741 DOI: 10.1007/s10522-024-10159-x] [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: 09/29/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
Coronary heart disease (CHD) and cognitive impairment frequently co-occur in aging populations, yet the molecular mechanisms linking these conditions remain unclear. This study aims to elucidate the roles of key aging-related genes (ARGs), specifically FKBP5 and DDIT3, in the pathophysiology of CHD and cognitive impairment, and to evaluate the therapeutic potential of stellate ganglion block (SGB). Using single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) data, we identified FKBP5 and DDIT3 as pivotal genes upregulated in both conditions. Experimental findings show that SGB effectively modulates these ARG-related pathways through autonomic regulation, specifically suppressing estrogen and NF-κB signaling pathways, thereby reducing the expression of pro-inflammatory cytokines such as SRC, MMP2, FKBP5, IRAK1, and MYD88, while upregulating the vasodilation-related gene NOS3. This modulation improved endothelial and cardiac function and enhanced cerebral blood flow (CBF), leading to cognitive improvement. Behavioral assessments, including novel object recognition (NOR) and Morris water maze (MWM) tests, demonstrated that SGB-treated rats outperformed untreated MI rats, with significant cognitive recovery over time. Further support from laser Doppler flowmetry (LDF) and electroencephalogram (EEG) analyses revealed increased left frontal blood flow and stabilized neural activity, indicating a favorable neurophysiological environment for cognitive rehabilitation. Our findings suggest that left stellate ganglion block (LSGB) provides both cardiac and cognitive benefits through targeted gene modulation, establishing its therapeutic potential for addressing the intersecting pathologies of CHD and cognitive impairment.
Collapse
Affiliation(s)
- Zhehao Jin
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yuling Xing
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
| | - Pengyu Duan
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China
| | - Yonghong Bi
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Xiaoyan Li
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
| | - Weiyu Feng
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
| | - Bing Zhang
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China.
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
23
|
Fernandes M, Antonucci M, Capecci F, Mercuri NB, Della-Morte D, Liguori C. Prevalence of sleep disorders in geriatrics: an exploratory study using sleep questionnaires. Geriatr Nurs 2024; 60:107-113. [PMID: 39236368 DOI: 10.1016/j.gerinurse.2024.08.032] [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: 02/23/2024] [Revised: 07/31/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVES This study aimed to investigate the prevalence of sleep problems in older subjects, considering sex and age differences. METHODS Subjects admitted to a geriatrics clinic underwent a medical visit and completed a battery of questionnaires assessing sleep quality, insomnia, sleep apnea risk, excessive daytime sleepiness (EDS), restless legs syndrome (RLS), chronotype, depression and global cognition. RESULTS Fifty-eight subjects (58.6 % women, mean age 77.36±6.07) were included. The most predominant sleep-related complaint was poor sleep quality (36.2 %), followed by sleep apnea risk (34.5 %), insomnia symptoms (25.9 %), EDS (15.5 %) and RLS (12.1 %). Older women reported more insomnia, poorer sleep quality and depressive symptoms than males. Patients aged ≥ 75 years old had more comorbidities and higher sleep apnea risk compared to those under 75 years old. CONCLUSIONS Sleep problems are frequent in older adults, requiring their screening and treatment for possibly improving well-being and reduce the burden of neuropsychiatric and medical comorbidities.
Collapse
Affiliation(s)
- Mariana Fernandes
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Matteo Antonucci
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Francesca Capecci
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy; Sleep Medicine Centre, Neurology Unit, University Hospital of Rome "Tor Vergata", 00133 Rome, Italy
| | - David Della-Morte
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy; Sleep Medicine Centre, Neurology Unit, University Hospital of Rome "Tor Vergata", 00133 Rome, Italy.
| |
Collapse
|
24
|
Mazurek KA, Li L, Klein RJ, Rong S, Mullan AF, Jones DT, St Louis EK, Worrell GA, Chen CY. Investigating the effects of indoor lighting on measures of brain health in older adults: protocol for a cross-over randomized controlled trial. BMC Geriatr 2024; 24:816. [PMID: 39394603 PMCID: PMC11468298 DOI: 10.1186/s12877-023-04594-7] [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: 12/09/2022] [Accepted: 12/13/2023] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND The worldwide number of adults aged 60 years and older is expected to double from 1 billion in 2019 to 2.1 billion by 2050. As the population lives longer, the rising incidence of chronic diseases, cognitive disorders, and behavioral health issues threaten older adults' health span. Exercising, getting sufficient sleep, and staying mentally and socially active can improve quality of life, increase independence, and potentially lower the risk for Alzheimer's disease or other dementias. Nonpharmacological approaches might help promote such behaviors. Indoor lighting may impact sleep quality, physical activity, and cognitive function. Dynamically changing indoor lighting brightness and color throughout the day has positive effects on sleep, cognitive function, and physical activity of its occupants. The aim of this study is to investigate how different indoor lighting conditions affect such health measures to promote healthier aging. METHODS This protocol is a randomized, cross-over, single-site trial followed by an exploratory third intervention. Up to 70 older adults in independent living residences at a senior living facility will be recruited. During this 16-week study, participants will experience three lighting conditions. Two cohorts will first experience a static and a dynamic lighting condition in a cluster-randomized cross-over design. The static condition lighting will have fixed brightness and color to match lighting typically provided in the facility. For the dynamic condition, brightness and color will change throughout the day with increased brightness in the morning. After the cross-over, both cohorts will experience another dynamic lighting condition with increased morning brightness to determine if there is a saturation effect between light exposure and health-related measures. Light intake, sleep quality, and physical activity will be measured using wearable devices. Sleep, cognitive function, mood, and social engagement will be assessed using surveys and cognitive assessments. DISCUSSION We hypothesize participants will have better sleep quality and greater physical activity during the dynamic lighting compared to the static lighting condition. Additionally, we hypothesize there is a maximal threshold at which health-outcomes improve based on light exposure. Study findings may identify optimal indoor lighting solutions to promote healthy aging for older adults. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05978934.
Collapse
Affiliation(s)
- Kevin A Mazurek
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Neuroscience, University of Rochester, Rochester, NY, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Linhao Li
- Well Living Lab, Rochester, MN, USA.
- Delos Living LLC, New York, NY, USA.
| | - Robert J Klein
- Well Living Lab, Rochester, MN, USA
- Delos Living LLC, New York, NY, USA
| | | | - Aidan F Mullan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Erik K St Louis
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Center for Sleep Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gregory A Worrell
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Christina Y Chen
- Department of Community Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
25
|
Yang Q, Li S, Yang Y, Lin X, Yang M, Tian C, Mao J. Prolonged sleep duration as a predictor of cognitive decline: A meta-analysis encompassing 49 cohort studies. Neurosci Biobehav Rev 2024; 164:105817. [PMID: 39032844 DOI: 10.1016/j.neubiorev.2024.105817] [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: 04/28/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Despite numerous studies have explored the association between sleep duration and cognition, the link between sleep duration trajectories and cognition remains underexplored. This systematic review aims to elucidate this correlation. We analyzed 55 studies from 14 countries, comprising 36 studies focusing on sleep duration, 20 on insomnia, and 13 on hypersomnia. A total of 10,767,085 participants were included in 49 cohort studies with a mean follow-up duration of 9.1 years. A non-linear association between sleep duration and cognitive decline was identified. Both long (risk ratio (RR):1.35, 95 % confidence intervals (CIs):1.23-1.48) and short sleep durations (RR: 1.12, 95 % CIs:1.03-1.22) were associated with an elevated risk of cognitive decline compared to moderate sleep duration. Additionally, hypersomnia (RR:1.26, 95 % CIs: 1.15-1.39) and insomnia (RR: 1.16, 95 % CIs: 1.002-1.34) were also linked to an increased risk. Moreover, prolonged sleep duration posed a higher risk of cognitive decline than stable sleep duration (RR:1.42, 95 % CIs:1.27-1.59). Importantly, transitioning from short or moderate to long sleep duration, as well as persistent long sleep duration, exhibited higher RRs for cognitive decline (RRs: 1.94, 1.40, and 1.28, respectively) compared to persistent moderate sleep duration. Our findings underscore the significance of prolonged sleep duration, alongside short and long sleep durations, with an elevated risk of cognitive decline. The association is tied to the degree of sleep duration changes. Our study highlights the importance of considering changes in sleep patterns over time, not just static sleep durations.
Collapse
Affiliation(s)
- Qing Yang
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Suya Li
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Yang
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuechun Lin
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengshu Yang
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chong Tian
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jing Mao
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
26
|
Cavaillès C, Wallace M, Leng Y, Stone KL, Ancoli-Israel S, Yaffe K. Multidimensional Sleep Profiles via Machine learning and Risk of Dementia and Cardiovascular Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.19.24312248. [PMID: 39228701 PMCID: PMC11370502 DOI: 10.1101/2024.08.19.24312248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Importance Sleep health comprises several dimensions such as duration and fragmentation of sleep, circadian activity, and daytime behavior. Yet, most research has focused on individual sleep characteristics. Studies are needed to identify sleep profiles incorporating multiple dimensions and to assess how different profiles may be linked to adverse health outcomes. Objective To identify actigraphy-based 24-hour sleep/circadian profiles in older men and to investigate whether these profiles are associated with the incidence of dementia and cardiovascular disease (CVD) events over 12 years. Design Data came from a prospective sleep study with participants recruited between 20032005 and followed until 2015-2016. Setting Multicenter population-based cohort study. Participants Among the 3,135 men enrolled, we excluded 331 men with missing or invalid actigraphy data and 137 with significant cognitive impairment at baseline, leading to a sample of 2,667 participants. Exposures Leveraging 20 actigraphy-derived sleep and circadian activity rhythm variables, we determined sleep/circadian profiles using an unsupervised machine learning technique based on multiple coalesced generalized hyperbolic mixture modeling. Main Outcomes and Measures Incidence of dementia and CVD events. Results We identified three distinct sleep/circadian profiles: active healthy sleepers (AHS; n=1,707 (64.0%); characterized by normal sleep duration, higher sleep quality, stronger circadian rhythmicity, and higher activity during wake periods), fragmented poor sleepers (FPS; n=376 (14.1%); lower sleep quality, higher sleep fragmentation, shorter sleep duration, and weaker circadian rhythmicity), and long and frequent nappers (LFN; n=584 (21.9%); longer and more frequent naps, higher sleep quality, normal sleep duration, and more fragmented circadian rhythmicity). Over the 12-year follow-up, compared to AHS, FPS had increased risks of dementia and CVD events (Hazard Ratio (HR)=1.35, 95% confidence interval (CI)=1.02-1.78 and HR=1.32, 95% CI=1.08-1.60, respectively) after multivariable adjustment, whereas LFN showed a marginal association with increased CVD events risk (HR=1.16, 95% CI=0.98-1.37) but not with dementia (HR=1.09, 95%CI=0.86-1.38). Conclusion and Relevance We identified three distinct multidimensional profiles of sleep health. Compared to healthy sleepers, older men with overall poor sleep and circadian activity rhythms exhibited worse incident cognitive and cardiovascular health. These results highlight potential targets for sleep interventions and the need for more comprehensive screening of poor sleepers for adverse outcomes.
Collapse
Affiliation(s)
- Clémence Cavaillès
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, California, USA
| | - Meredith Wallace
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yue Leng
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, California, USA
| | - Katie L. Stone
- Research Institute, California Pacific Medical Center, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Sonia Ancoli-Israel
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, California, USA
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
- Department of Epidemiology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
27
|
He CYY, Zhou Z, Kan MMP, Chan DHY, Wong ACT, Mok KHY, Lam FMH, Chan SCC, Cheung CKC, Yeung MKC, Wong AYL. Modifiable risk factors for mild cognitive impairment among cognitively normal community-dwelling older adults: A systematic review and meta-analysis. Ageing Res Rev 2024; 99:102350. [PMID: 38942197 DOI: 10.1016/j.arr.2024.102350] [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: 08/21/2023] [Revised: 05/16/2024] [Accepted: 05/26/2024] [Indexed: 06/30/2024]
Abstract
Although numerous studies have investigated modifiable risk factors for mild cognitive impairment (MCI) among community-dwelling seniors, no meta-analysis has summarized these findings. Five databases were searched from January 1, 2000, to December 30, 2023. The protocol was registered with PROSPERO. Data were extracted and reported following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Relevant meta-analyses of modifiable risk factors were performed. The evidence of each factor was assessed by the GRADE for cohort studies. Of 16,651 citations, 87 studies involving 225,584 community-dwelling seniors were included. Fourteen meta-analyses involving 20 studies with 44,199 participants were performed. The analyses revealed low-to-moderate-quality evidence supporting that diabetes, 2 or more comorbidities, anxiety, apathy, depressive symptoms, and physical frailty were risk factors for incident MCI in older adults. Conversely, hypertension, agitation, and irritability might not be risk factors. Additionally, moderate-quality evidence supports the protective effect of engaging in cognitive-demanding activities on the onset of MCI. Collectively, this study constitutes the first extensive compilation of evidence regarding the various risk factors for the development of MCI in older adults. Our findings hold significant potential to guide the formulation of prevention and management strategies to either prevent or potentially reverse the onset of MCI.
Collapse
Affiliation(s)
- Christo Y Y He
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Zhixing Zhou
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Mandy M P Kan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Dorothy H Y Chan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Athena C T Wong
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Kenny H Y Mok
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Freddy M H Lam
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Sam C C Chan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Chelsia K C Cheung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China.
| | - Michael K C Yeung
- Department of Psychology, The Education University of Hong Kong, 999077, Hong Kong Special Administrative Region of China.
| | - Arnold Y L Wong
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong Special Administrative Region of China; Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China.
| |
Collapse
|
28
|
Tucker A, Goldberg TE, Kim H. Biomarkers of sleep-wake disturbance as predictors of cognitive decline and accelerated disease progression. Expert Rev Mol Diagn 2024; 24:649-657. [PMID: 39129222 DOI: 10.1080/14737159.2024.2389307] [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: 04/29/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION In older adults, where sleep disturbances and cognitive impairment are common, mounting evidence suggests a potential connection between sleep and cognitive function, highlighting the significance of utilizing sleep as a biomarker for early detection of cognitive impairment to improve clinical outcomes in a noninvasive, cost-effective manner. AREAS COVERED This review describes the relationship between sleep and cognitive function in older adults, encompassing both subjective and objective measures of sleep quality, duration, architecture, and sleep-disordered breathing. The authors consider the directionality of the associations observed in prospective and cross-sectional studies, exploring whether sleep disturbances precede cognitive decline or vice versa. Furthermore, they discuss the potential bidirectional relationships between sleep and Alzheimer's disease (AD) risks in older adults while also examining the neurodegenerative pathways of this relationship. EXPERT OPINION Routine sleep monitoring in primary care settings has the potential to bolster early detection and treatment of sleep disturbance, and by extension, reduce the risk of dementia. Improving sleep assessment tools, such as wearables, provide scalable alternatives to traditional methods like polysomnography, potentially enabling widespread monitoring of sleep characteristics. Standardized measurement and inclusive participant recruitment are needed to enhance generalizability, while longitudinal studies are essential to understand the interaction between sleep and AD pathology.
Collapse
Affiliation(s)
- Aren Tucker
- Brain Aging and Mental Health Division, New York State Psychiatric Institute, New York, NY, USA
| | - Terry E Goldberg
- Brain Aging and Mental Health Division, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Psychology, New York, NY, USA
- Department of Anesthisiology, Columbia University Irving Medical Psychology, New York, NY, USA
| | - Hyun Kim
- Brain Aging and Mental Health Division, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Psychology, New York, NY, USA
| |
Collapse
|
29
|
Weng W, Fu J, Cheng F, Wang Y, Zhang J. Integrated Bulk and Single-Cell RNA-Sequencing Reveals the Effects of Circadian Rhythm Disruption on the Metabolic Reprogramming of CD4+ T Cells in Alzheimer's Disease. Mol Neurobiol 2024; 61:6013-6030. [PMID: 38265551 DOI: 10.1007/s12035-023-03907-6] [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: 03/14/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024]
Abstract
Although growing evidence suggests close correlations between Alzheimer's disease (AD) and circadian rhythm disruption (CRD), few studies have focused on the influence of circadian rhythm on levels of immune cells in AD. We aimed to delineate the mechanism underlying the effects of circadian related genes on T cell immune function in AD. A total of 112 brain samples were used to construct the CRD-related model by performing weighted gene co-expression network analysis and machine learning algorithms (LASSO, SVM-RFE, and RF). The ssGSEA method was used to calculate the CRDscore in order to quantify CRD status. Using single-cell transcriptome data of CSF cells, we investigated the CD4+ T cell metabolism and cell-cell communication in high- and low-risk CRD groups. Connectivity map (CMap) was applied to explore small molecule drugs targeting CRD, and the expression of the signature gene GPR4 was further validated in AD. The CRDscore algorithm, which is based on 23 circadian-related genes, can effectively classify the CRD status in AD datasets. The single-cell analysis revealed that the CD4+ T cells with high CRDscore were characterized by hypometabolism. Cell communication analysis revealed that CD4+ T cells might be involved in promoting CD8+ T cell adhesion under CRD, which may facilitate T cell infiltration into the brain parenchyma. Overall, this study indicates the potential connotation of circadian rhythm in AD, providing insights into understanding T cell metabolic reprogramming under CRD.
Collapse
Affiliation(s)
- Weipin Weng
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianhan Fu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fan Cheng
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yixuan Wang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie Zhang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China.
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China.
- Department of Neurology, Turpan City People's Hospital, Tulufan, China.
| |
Collapse
|
30
|
Smagula SF, Zhang G, Krafty RT, Ramos A, Sotres-Alvarez D, Rodakowski J, Gallo LC, Lamar M, Gujral S, Fischer D, Tarraf W, Mossavar-Rahmani Y, Redline S, Stone KL, Gonzalez HM, Patel SR. Sleep-wake behaviors associated with cognitive performance in middle-aged participants of the Hispanic Community Health Study/Study of Latinos. Sleep Health 2024; 10:500-507. [PMID: 38693044 PMCID: PMC11309910 DOI: 10.1016/j.sleh.2024.02.002] [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: 10/02/2023] [Revised: 01/26/2024] [Accepted: 02/20/2024] [Indexed: 05/03/2024]
Abstract
OBJECTIVES Many sleep-wake behaviors have been associated with cognition. We examined a panel of sleep-wake/activity characteristics to determine which are most robustly related to having low cognitive performance in midlife. Secondarily, we evaluate the predictive utility of sleep-wake measures to screen for low cognitive performance. METHODS The outcome was low cognitive performance defined as being >1 standard deviation below average age/sex/education internally normalized composite cognitive performance levels assessed in the Hispanic Community Health Study/Study of Latinos. Analyses included 1006 individuals who had sufficient sleep-wake measurements about 2years later (mean age=54.9, standard deviation= 5.1; 68.82% female). We evaluated associations of 31 sleep-wake variables with low cognitive performance using separate logistic regressions. RESULTS In individual models, the strongest sleep-wake correlates of low cognitive performance were measures of weaker and unstable 24-hour rhythms; greater 24-hour fragmentation; longer time-in-bed; and lower rhythm amplitude. One standard deviation worse on these sleep-wake factors was associated with ∼20%-30% greater odds of having low cognitive performance. In an internally cross-validated prediction model, the independent correlates of low cognitive performance were: lower Sleep Regularity Index scores; lower pseudo-F statistics (modellability of 24-hour rhythms); lower activity rhythm amplitude; and greater time in bed. Area under the curve was low/moderate (64%) indicating poor predictive utility. CONCLUSION The strongest sleep-wake behavioral correlates of low cognitive performance were measures of longer time-in-bed and irregular/weak rhythms. These sleep-wake assessments were not useful to identify previous low cognitive performance. Given their potential modifiability, experimental trials could test if targeting midlife time-in-bed and/or irregular rhythms influences cognition.
Collapse
Affiliation(s)
- Stephen F Smagula
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | - Gehui Zhang
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert T Krafty
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Alberto Ramos
- Department of Neurology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Daniela Sotres-Alvarez
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Juleen Rodakowski
- Department of Occupational Therapy, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Linda C Gallo
- Department of Psychology, University of California San Diego, San Diego, California, USA
| | - Melissa Lamar
- Institute of Minority Health Research, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Swathi Gujral
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dorothee Fischer
- Department of Sleep and Human Factors Research, Institute for Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Wassim Tarraf
- Institute of Gerontology, Wayne State University, Detroit, Michigan, USA
| | - Yasmin Mossavar-Rahmani
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, New York, New York, USA
| | - Susan Redline
- Division of Sleep Medicine, Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Katie L Stone
- California Pacific Medical Center Research Institute, San Francisco, California, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Hector M Gonzalez
- Department of Neurosciences and the Shiley-Marcos Alzheimer's Disease Research Center, UC San Diego, San Diego, California, USA
| | - Sanjay R Patel
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Campbell KJ, Jiang P, Olker C, Lin X, Kim SY, Lee CJ, Song EJ, Turek FW, Vitaterna MH. The impacts of sex and the 5xFAD model of Alzheimer's disease on the sleep and spatial learning responses to feeding time. Front Neurol 2024; 15:1430989. [PMID: 39144714 PMCID: PMC11322461 DOI: 10.3389/fneur.2024.1430989] [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: 05/10/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
Introduction The relationships between the feeding rhythm, sleep and cognition in Alzheimer's disease (AD) are incompletely understood, but meal time could provide an easy-to-implement method of curtailing disease-associated disruptions in sleep and cognition. Furthermore, known sex differences in AD incidence could relate to sex differences in circadian rhythm/sleep/cognition interactions. Methods The 5xFAD transgenic mouse model of AD and non-transgenic wild-type controls were studied. Both female and male mice were used. Food access was restricted each day to either the 12-h light phase (light-fed groups) or the 12-h dark phase (dark-fed groups). Sleep (electroencephalographic/electromyographic) recording and cognitive behavior measures were collected. Results The 5xFAD genotype reduces NREM and REM as well as the number of sleep spindles. In wild-type mice, light-fed groups had disrupted vigilance state amounts, characteristics, and rhythms relative to dark-fed groups. These feeding time differences were reduced in 5xFAD mice. Sex modulates these effects. 5xFAD mice display poorer spatial memory that, in female mice, is curtailed by dark phase feeding. Similarly, female 5xFAD mice have decreased anxiety-associated behavior. These emotional and cognitive measures are correlated with REM amount. Discussion Our study demonstrates that the timing of feeding can alter many aspects of wake, NREM and REM. Unexpectedly, 5xFAD mice are less sensitive to these feeding time effects. 5xFAD mice demonstrate deficits in cognition which are correlated with REM, suggesting that this circadian-timed aspect of sleep may link feeding time and cognition. Sex plays an important role in regulating the impact of feeding time on sleep and cognition in both wild-type and 5xFAD mice, with females showing a greater cognitive response to feeding time than males.
Collapse
Affiliation(s)
- Katrina J. Campbell
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Peng Jiang
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Christopher Olker
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Xuanyi Lin
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Sarah Y. Kim
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Christopher J. Lee
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Eun Joo Song
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Fred W. Turek
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| | - Martha Hotz Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, United States
| |
Collapse
|
32
|
Milton S, Cavaillès C, Ancoli-Israel S, Stone KL, Yaffe K, Leng Y. Five-year changes in 24-hour sleep-wake activity and dementia risk in oldest old women. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.23.24310882. [PMID: 39211875 PMCID: PMC11361246 DOI: 10.1101/2024.07.23.24310882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Sleep disruptions are associated with cognitive aging in older adults. However, it is unclear whether longitudinal changes in 24-hour multidimensional sleep-wake activity are linked to cognitive impairment in the oldest old. METHODS We studied 733 cognitively unimpaired women (mean age=82.5±2.9 years) who completed two actigraphy assessments over five years. We performed hierarchical clustering on principal components in nine sleep, napping, and circadian rest-activity rhythm parameters to identify multidimensional sleep-wake change profiles and multinomial logistic regression to evaluate the associations between sleep-wake changes and risk of cognitive impairment at follow-up. RESULTS We identified three sleep-wake change profiles: Stable Sleep (43.8%), Declining Nighttime Sleep (34.9%), and Increasing Sleepiness (21.3%). After adjustment for demographics and comorbidities, women with Increasing Sleepiness had approximately doubled (odds ratio=2.21, p=0.018) risk of dementia compared to those with Stable Sleep. DISCUSSION Increasing sleepiness may be an independent marker or risk factor for dementia in oldest old women.
Collapse
|
33
|
Janssen Daalen JM, van den Bergh R, Prins EM, Moghadam MSC, van den Heuvel R, Veen J, Mathur S, Meijerink H, Mirelman A, Darweesh SKL, Evers LJW, Bloem BR. Digital biomarkers for non-motor symptoms in Parkinson's disease: the state of the art. NPJ Digit Med 2024; 7:186. [PMID: 38992186 PMCID: PMC11239921 DOI: 10.1038/s41746-024-01144-2] [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: 01/05/2024] [Accepted: 05/22/2024] [Indexed: 07/13/2024] Open
Abstract
Digital biomarkers that remotely monitor symptoms have the potential to revolutionize outcome assessments in future disease-modifying trials in Parkinson's disease (PD), by allowing objective and recurrent measurement of symptoms and signs collected in the participant's own living environment. This biomarker field is developing rapidly for assessing the motor features of PD, but the non-motor domain lags behind. Here, we systematically review and assess digital biomarkers under development for measuring non-motor symptoms of PD. We also consider relevant developments outside the PD field. We focus on technological readiness level and evaluate whether the identified digital non-motor biomarkers have potential for measuring disease progression, covering the spectrum from prodromal to advanced disease stages. Furthermore, we provide perspectives for future deployment of these biomarkers in trials. We found that various wearables show high promise for measuring autonomic function, constipation and sleep characteristics, including REM sleep behavior disorder. Biomarkers for neuropsychiatric symptoms are less well-developed, but show increasing accuracy in non-PD populations. Most biomarkers have not been validated for specific use in PD, and their sensitivity to capture disease progression remains untested for prodromal PD where the need for digital progression biomarkers is greatest. External validation in real-world environments and large longitudinal cohorts remains necessary for integrating non-motor biomarkers into research, and ultimately also into daily clinical practice.
Collapse
Affiliation(s)
- Jules M Janssen Daalen
- Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands.
| | - Robin van den Bergh
- Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| | - Eva M Prins
- Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| | - Mahshid Sadat Chenarani Moghadam
- Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| | - Rudie van den Heuvel
- HAN University of Applied Sciences, School of Engineering and Automotive, Health Concept Lab, Arnhem, The Netherlands
| | - Jeroen Veen
- HAN University of Applied Sciences, School of Engineering and Automotive, Health Concept Lab, Arnhem, The Netherlands
| | | | - Hannie Meijerink
- ParkinsonNL, Parkinson Patient Association, Bunnik, The Netherlands
| | - Anat Mirelman
- Tel Aviv University, Sagol School of Neuroscience, Department of Neurology, Faculty of Medicine, Laboratory for Early Markers of Neurodegeneration (LEMON), Center for the Study of Movement, Cognition, and Mobility (CMCM), Tel Aviv, Israel
| | - Sirwan K L Darweesh
- Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| | - Luc J W Evers
- Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
- Radboud University, Institute for Computing and Information Sciences, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands.
| |
Collapse
|
34
|
Wang X, Yan X, Li M, Cheng L, Qi X, Zhang J, Pan S, Xu X, Wei W, Li Y. U-shaped association between sleep duration and biological aging: Evidence from the UK Biobank study. Aging Cell 2024; 23:e14159. [PMID: 38556842 PMCID: PMC11258478 DOI: 10.1111/acel.14159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024] Open
Abstract
Previous research on sleep and aging largely has failed to illustrate the optimal dose-response curve of this relationship. We aimed to analyze the associations between sleep duration and measures of predicted age. In total, 241,713 participants from the UK Biobank were included. Habitual sleep duration was collected from the baseline questionnaire. Four indicators, homeostatic dysregulation (HD), phenoAge (PA), Klemera-Doubal method (KDM), and allostatic load (AL), were chosen to assess predicted age. Multivariate linear regression models were utilized. The association of sleep duration and predicted age followed a U-shape (All p for nonlinear <0.05). Compared with individuals who sleep for 7 h/day, the multivariable-adjusted beta of ≤5 and ≥9 h/day were 0.05 (95% CI 0.03, 0.07) and 0.03 (95% CI 0.02, 0.05) for HD, 0.08 (95% CI 0.01, 0.14) and 0.36 (95% CI 0.31, 0.41) for PA, and 0.21 (95% CI 0.12, 0.30) and 0.30 (95% CI 0.23, 0.37) for KDM. Significant independent and joint effects of sleep and cystatin C (CysC) and gamma glutamyltransferase (GGT) on predicted age metrics were future found. Similar results were observed when conducting stratification analyses. Short and long sleep duration were associated with accelerated predicted age metrics mediated by CysC and GGT.
Collapse
Affiliation(s)
- Xuanyang Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of EducationHarbin Medical UniversityHarbinHeilongjiangChina
| | - Xuemin Yan
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of EducationHarbin Medical UniversityHarbinHeilongjiangChina
| | - Mengdi Li
- Department of Endodontics, The First HospitalHarbin Medical UniversityHarbinChina
| | - Licheng Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of EducationHarbin Medical UniversityHarbinHeilongjiangChina
| | - Xiang Qi
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of EducationHarbin Medical UniversityHarbinHeilongjiangChina
| | - Jia Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of EducationHarbin Medical UniversityHarbinHeilongjiangChina
| | - Sijia Pan
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of EducationHarbin Medical UniversityHarbinHeilongjiangChina
| | - Xiaoqing Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of EducationHarbin Medical UniversityHarbinHeilongjiangChina
| | - Wei Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of EducationHarbin Medical UniversityHarbinHeilongjiangChina
- Department of Pharmacology, College of Pharmacy, Key Laboratory of Cardiovascular Research, Ministry of EducationHarbin Medical UniversityHarbinChina
| | - Ying Li
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of EducationHarbin Medical UniversityHarbinHeilongjiangChina
| |
Collapse
|
35
|
Liu Y, Feng H, Du J, Yang L, Xue H, Zhang J, Liang YY, Liu Y. Associations between accelerometer-measured circadian rest-activity rhythm, brain structural and genetic mechanisms, and dementia. Psychiatry Clin Neurosci 2024; 78:393-404. [PMID: 38676558 PMCID: PMC11498105 DOI: 10.1111/pcn.13671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 04/29/2024]
Abstract
AIM Knowledge of how circadian rhythm influences brain health remains limited. We aimed to investigate the associations of accelerometer-measured circadian rest-activity rhythm (CRAR) with incident dementia, cognitive dysfunction, and structural brain abnormalities in the general population and underlying biological mechanisms. METHODS Fifty-seven thousand five hundred and two participants aged over 60 years with accelerometer data were included to investigate the association of CRAR with incidental dementia. Non-parametric CRAR parameters were utilized, including activity level during active periods of the day (M10), activity level during rest periods of the day (L5), and the relative difference between the M10 and L5 (relative amplitude, RA). Associations of CRAR with cognitive dysfunction and brain structure were studied in a subset of participants. Neuroimaging-transcriptomics analysis was utilized to identify the underlying molecular mechanisms. RESULTS Over 6.86 (4.94-8.78) years of follow-up, 494 participants developed dementia. The risk of incident dementia was associated with decreasing M10 (hazard ratio [HR] 1.45; 95% conference interval [CI], 1.28-1.64) and RA (HR 1.37; 95% CI, 1.28-1.64), increasing L5 (HR 1.14, 95% CI 1.07-1.21) and advanced L5 onset time (HR 1.12; 95% CI, 1.02-1.23). The detrimental associations were exacerbated by APOE ε4 status and age (>65 years). Decreased RA was associated with lower processing speed (Beta -0.04; SE 0.011), predominantly mediated by abnormalities in subcortical regions and white matter microstructure. The genes underlying CRAR-related brain regional structure variation were enriched for synaptic function. CONCLUSIONS Our study underscores the potential of intervention targeting at maintaining a healthy CRAR pattern to prevent dementia risk.
Collapse
Affiliation(s)
- Yue Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalSouthern Medical UniversityGuangzhouChina
- Center for Sleep and Circadian MedicineThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Hongliang Feng
- Center for Sleep and Circadian MedicineThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaGuangzhou Medical UniversityGuangzhouChina
| | - Jing Du
- Center for Sleep and Circadian MedicineThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaGuangzhou Medical UniversityGuangzhouChina
| | - Lulu Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalSouthern Medical UniversityGuangzhouChina
| | - Huachen Xue
- Center for Sleep and Circadian MedicineThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaGuangzhou Medical UniversityGuangzhouChina
| | - Jihui Zhang
- Center for Sleep and Circadian MedicineThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaGuangzhou Medical UniversityGuangzhouChina
| | - Yannis Yan Liang
- Center for Sleep and Circadian MedicineThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaGuangzhou Medical UniversityGuangzhouChina
- Institute of Psycho‐neuroscienceThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Yaping Liu
- Center for Sleep and Circadian MedicineThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaGuangzhou Medical UniversityGuangzhouChina
- Department of Psychiatry, Faculty of MedicineThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
36
|
Spira AP, Liu F, Zipunnikov V, Bilgel M, Rabinowitz JA, An Y, Di J, Bai J, Wanigatunga SK, Wu MN, Lucey BP, Schrack JA, Wanigatunga AA, Rosenberg PB, Simonsick EM, Walker KA, Ferrucci L, Resnick SM. Evaluating a novel 24-hour rest/activity rhythm marker of preclinical β-amyloid deposition. Sleep 2024; 47:zsae037. [PMID: 38381532 PMCID: PMC11082462 DOI: 10.1093/sleep/zsae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/10/2024] [Indexed: 02/23/2024] Open
Abstract
STUDY OBJECTIVES To compare sleep and 24-hour rest/activity rhythms (RARs) between cognitively normal older adults who are β-amyloid-positive (Aβ+) or Aβ- and replicate a novel time-of-day-specific difference between these groups identified in a previous exploratory study. METHODS We studied 82 cognitively normal participants from the Baltimore Longitudinal Study of Aging (aged 75.7 ± 8.5 years, 55% female, 76% white) with wrist actigraphy data and Aβ+ versus Aβ- status measured by [11C] Pittsburgh compound B positron emission tomography. RARs were calculated using epoch-level activity count data from actigraphy. We used novel, data-driven function-on-scalar regression analyses and standard RAR metrics to cross-sectionally compare RARs between 25 Aβ+ and 57 Aβ- participants. RESULTS Compared to Aβ- participants, Aβ+ participants had higher mean activity from 1:00 p.m. to 3:30 p.m. when using less conservative pointwise confidence intervals (CIs) and from 1:30 p.m. to 2:30 p.m. using more conservative, simultaneous CIs. Furthermore, Aβ+ participants had higher day-to-day variability in activity from 9:00 a.m. to 11:30 a.m. and lower variability from 1:30 p.m. to 4:00 p.m. and 7:30 p.m. to 10:30 p.m. according to pointwise CIs, and lower variability from 8:30 p.m. to 10:00 p.m. using simultaneous CIs. There were no Aβ-related differences in standard sleep or RAR metrics. CONCLUSIONS Findings suggest Aβ+ older adults have higher, more stable day-to-day afternoon/evening activity than Aβ- older adults, potentially reflecting circadian dysfunction. Studies are needed to replicate our findings and determine whether these or other time-of-day-specific RAR features have utility as markers of preclinical Aβ deposition and if they predict clinical dementia and agitation in the afternoon/evening (i.e. "sundowning").
Collapse
Affiliation(s)
- Adam P Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Johns Hopkins Center on Aging and Health, Baltimore, MD, USA
| | - Fangyu Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Vadim Zipunnikov
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Murat Bilgel
- National Institute on Aging Intramural Research Program, Baltimore MD, USA
| | - Jill A Rabinowitz
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Yang An
- National Institute on Aging Intramural Research Program, Baltimore MD, USA
| | - Junrui Di
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jiawei Bai
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sarah K Wanigatunga
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Mark N Wu
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Jennifer A Schrack
- Johns Hopkins Center on Aging and Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Amal A Wanigatunga
- Johns Hopkins Center on Aging and Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Paul B Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Keenan A Walker
- National Institute on Aging Intramural Research Program, Baltimore MD, USA
| | - Luigi Ferrucci
- National Institute on Aging Intramural Research Program, Baltimore MD, USA
| | - Susan M Resnick
- National Institute on Aging Intramural Research Program, Baltimore MD, USA
| |
Collapse
|
37
|
Haghayegh S, Gao C, Sugg E, Zheng X, Yang HW, Saxena R, Rutter MK, Weedon M, Ibanez A, Bennett DA, Li P, Gao L, Hu K. Association of Rest-Activity Rhythm and Risk of Developing Dementia or Mild Cognitive Impairment in the Middle-Aged and Older Population: Prospective Cohort Study. JMIR Public Health Surveill 2024; 10:e55211. [PMID: 38713911 PMCID: PMC11109857 DOI: 10.2196/55211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/21/2024] [Accepted: 03/16/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND The relationship between 24-hour rest-activity rhythms (RARs) and risk for dementia or mild cognitive impairment (MCI) remains an area of growing interest. Previous studies were often limited by small sample sizes, short follow-ups, and older participants. More studies are required to fully explore the link between disrupted RARs and dementia or MCI in middle-aged and older adults. OBJECTIVE We leveraged the UK Biobank data to examine how RAR disturbances correlate with the risk of developing dementia and MCI in middle-aged and older adults. METHODS We analyzed the data of 91,517 UK Biobank participants aged between 43 and 79 years. Wrist actigraphy recordings were used to derive nonparametric RAR metrics, including the activity level of the most active 10-hour period (M10) and its midpoint, the activity level of the least active 5-hour period (L5) and its midpoint, relative amplitude (RA) of the 24-hour cycle [RA=(M10-L5)/(M10+L5)], interdaily stability, and intradaily variability, as well as the amplitude and acrophase of 24-hour rhythms (cosinor analysis). We used Cox proportional hazards models to examine the associations between baseline RAR and subsequent incidence of dementia or MCI, adjusting for demographic characteristics, comorbidities, lifestyle factors, shiftwork status, and genetic risk for Alzheimer's disease. RESULTS During the follow-up of up to 7.5 years, 555 participants developed MCI or dementia. The dementia or MCI risk increased for those with lower M10 activity (hazard ratio [HR] 1.28, 95% CI 1.14-1.44, per 1-SD decrease), higher L5 activity (HR 1.15, 95% CI 1.10-1.21, per 1-SD increase), lower RA (HR 1.23, 95% CI 1.16-1.29, per 1-SD decrease), lower amplitude (HR 1.32, 95% CI 1.17-1.49, per 1-SD decrease), and higher intradaily variability (HR 1.14, 95% CI 1.05-1.24, per 1-SD increase) as well as advanced L5 midpoint (HR 0.92, 95% CI 0.85-0.99, per 1-SD advance). These associations were similar in people aged <70 and >70 years, and in non-shift workers, and they were independent of genetic and cardiovascular risk factors. No significant associations were observed for M10 midpoint, interdaily stability, or acrophase. CONCLUSIONS Based on findings from a large sample of middle-to-older adults with objective RAR assessment and almost 8-years of follow-up, we suggest that suppressed and fragmented daily activity rhythms precede the onset of dementia or MCI and may serve as risk biomarkers for preclinical dementia in middle-aged and older adults.
Collapse
Affiliation(s)
- Shahab Haghayegh
- Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute, Cambridge, MA, United States
- Brigham and Women's Hospital, Boston, MA, United States
| | - Chenlu Gao
- Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute, Cambridge, MA, United States
- Brigham and Women's Hospital, Boston, MA, United States
| | - Elizabeth Sugg
- Massachusetts General Hospital, Boston, MA, United States
| | - Xi Zheng
- Brigham and Women's Hospital, Boston, MA, United States
| | - Hui-Wen Yang
- Brigham and Women's Hospital, Boston, MA, United States
| | - Richa Saxena
- Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute, Cambridge, MA, United States
| | - Martin K Rutter
- Faculty of Medicine, Biology and Health, University of Manchester, Manchester, United Kingdom
- Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, NIHR Manchester Biomedical Research Centre, Manchester, United Kingdom
| | | | | | | | - Peng Li
- Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute, Cambridge, MA, United States
- Brigham and Women's Hospital, Boston, MA, United States
| | - Lei Gao
- Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Kun Hu
- Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute, Cambridge, MA, United States
- Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
38
|
Liddie JM, Vieira CLZ, Coull BA, Sparrow D, Koutrakis P, Weisskopf MG. Associations between solar and geomagnetic activity and cognitive function in the Normative Aging study. ENVIRONMENT INTERNATIONAL 2024; 187:108666. [PMID: 38648690 PMCID: PMC11146138 DOI: 10.1016/j.envint.2024.108666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 03/22/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Studies show that changes in solar and geomagnetic activity (SGA) influence melatonin secretion and the autonomic nervous system. We evaluated associations between solar and geomagnetic activity and cognitive function in the Normative Aging Study from 1992 to 2013. METHODS We used logistic and linear generalized estimating equations and regressions to evaluate the associations between moving averages of sunspot number (SSN) and Kp index (a measure of geomagnetic activity) and a binary measure for Mini-Mental State Examination (MMSE) scores (≤25 or > 25) and six other cognitive tests as continuous measures, combined into one global composite score and considered separately. RESULTS A one-IQR increase in same-day SSN and Kp index were associated with 17% (95% CI: 3%, 34%) and 19% (95% CI: 4%, 36%) increases in the odds of low MMSE score. We observed small increases in the global cognitive score with increasing SSN, although we observed decreases specifically in relation to the backwards digit span test. CONCLUSIONS Periods of high SGA were associated with cognitive function. SGA may not equally impact all aspects of cognitive function, as evidenced by differences in associations observed for the MMSE, global cognitive score, and individual cognitive tests. Given that much of the pathology of cognitive decline in the elderly remains unexplained, studies specifically targeting decline and with longer follow-up periods are warranted.
Collapse
Affiliation(s)
- Jahred M Liddie
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Carolina L Z Vieira
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Brent A Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - David Sparrow
- VA Normative Aging Study, Veterans Affairs Boston Healthcare System, Boston, MA, USA; Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marc G Weisskopf
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
39
|
Zhu P, Peek CB. Circadian timing of satellite cell function and muscle regeneration. Curr Top Dev Biol 2024; 158:307-339. [PMID: 38670711 DOI: 10.1016/bs.ctdb.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Recent research has highlighted an important role for the molecular circadian machinery in the regulation of tissue-specific function and stress responses. Indeed, disruption of circadian function, which is pervasive in modern society, is linked to accelerated aging, obesity, and type 2 diabetes. Furthermore, evidence supporting the importance of the circadian clock within both the mature muscle tissue and satellite cells to regulate the maintenance of muscle mass and repair capacity in response injury has recently emerged. Here, we review the discovery of circadian clocks within the satellite cell (a.k.a. adult muscle stem cell) and how they act to regulate metabolism, epigenetics, and myogenesis during both healthy and diseased states.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States; Department of Medicine-Endocrinology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.
| | - Clara B Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States; Department of Medicine-Endocrinology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.
| |
Collapse
|
40
|
Mayer G, Frohnhofen H, Jokisch M, Hermann DM, Gronewold J. Associations of sleep disorders with all-cause MCI/dementia and different types of dementia - clinical evidence, potential pathomechanisms and treatment options: A narrative review. Front Neurosci 2024; 18:1372326. [PMID: 38586191 PMCID: PMC10995403 DOI: 10.3389/fnins.2024.1372326] [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: 01/17/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Due to worldwide demographic change, the number of older persons in the population is increasing. Aging is accompanied by changes of sleep structure, deposition of beta-amyloid (Aß) and tau proteins and vascular changes and can turn into mild cognitive impairment (MCI) as well as dementia. Sleep disorders are discussed both as a risk factor for and as a consequence of MCI/dementia. Cross-sectional and longitudinal population-based as well as case-control studies revealed sleep disorders, especially sleep-disorderded breathing (SDB) and excessive or insufficient sleep durations, as risk factors for all-cause MCI/dementia. Regarding different dementia types, SDB was especially associated with vascular dementia while insomnia/insufficient sleep was related to an increased risk of Alzheimer's disease (AD). Scarce and still inconsistent evidence suggests that therapy of sleep disorders, especially continuous positive airway pressure (CPAP) in SDB, can improve cognition in patients with sleep disorders with and without comorbid dementia and delay onset of MCI/dementia in patients with sleep disorders without previous cognitive impairment. Regarding potential pathomechanisms via which sleep disorders lead to MCI/dementia, disturbed sleep, chronic sleep deficit and SDB can impair glymphatic clearance of beta-amyloid (Aß) and tau which lead to amyloid deposition and tau aggregation resulting in changes of brain structures responsible for cognition. Orexins are discussed to modulate sleep and Aß pathology. Their diurnal fluctuation is suppressed by sleep fragmentation and the expression suppressed at the point of hippocampal atrophy, contributing to the progression of dementia. Additionally, sleep disorders can lead to an increased vascular risk profile and vascular changes such as inflammation, endothelial dysfunction and atherosclerosis which can foster neurodegenerative pathology. There is ample evidence indicating that changes of sleep structure in aging persons can lead to dementia and also evidence that therapy of sleep disorder can improve cognition. Therefore, sleep disorders should be identified and treated early.
Collapse
Affiliation(s)
- Geert Mayer
- Department of Neurology, Philipps-Universität Marburg, Marburg, Germany
| | - Helmut Frohnhofen
- Department of Orthopedics and Trauma Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Department of Medicine, Geriatrics, Faculty of Health, University Witten-Herdecke, Witten, Germany
| | - Martha Jokisch
- Department of Neurology and Center for Translational Neuro-and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Dirk M. Hermann
- Department of Neurology and Center for Translational Neuro-and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Janine Gronewold
- Department of Neurology and Center for Translational Neuro-and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
41
|
Gottesman RF, Lutsey PL, Benveniste H, Brown DL, Full KM, Lee JM, Osorio RS, Pase MP, Redeker NS, Redline S, Spira AP. Impact of Sleep Disorders and Disturbed Sleep on Brain Health: A Scientific Statement From the American Heart Association. Stroke 2024; 55:e61-e76. [PMID: 38235581 DOI: 10.1161/str.0000000000000453] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Accumulating evidence supports a link between sleep disorders, disturbed sleep, and adverse brain health, ranging from stroke to subclinical cerebrovascular disease to cognitive outcomes, including the development of Alzheimer disease and Alzheimer disease-related dementias. Sleep disorders such as sleep-disordered breathing (eg, obstructive sleep apnea), and other sleep disturbances, as well, some of which are also considered sleep disorders (eg, insomnia, sleep fragmentation, circadian rhythm disorders, and extreme sleep duration), have been associated with adverse brain health. Understanding the causal role of sleep disorders and disturbances in the development of adverse brain health is complicated by the common development of sleep disorders among individuals with neurodegenerative disease. In addition to the role of sleep disorders in stroke and cerebrovascular injury, mechanistic hypotheses linking sleep with brain health and biomarker data (blood-based, cerebrospinal fluid-based, and imaging) suggest direct links to Alzheimer disease-specific pathology. These potential mechanisms and the increasing understanding of the "glymphatic system," and the recognition of the importance of sleep in poststroke recovery, as well, support a biological basis for the indirect (through the worsening of vascular disease) and direct (through specific effects on neuropathology) connections between sleep disorders and brain health. Given promising evidence for the benefits of treatment and prevention, sleep disorders and disturbances represent potential targets for early treatment that may improve brain health more broadly. In this scientific statement, we discuss the evidence supporting an association between sleep disorders and disturbances and poor brain health ranging from stroke to dementia and opportunities for prevention and early treatment.
Collapse
|
42
|
Winer JR, Lok R, Weed L, He Z, Poston KL, Mormino EC, Zeitzer JM. Impaired 24-h activity patterns are associated with an increased risk of Alzheimer's disease, Parkinson's disease, and cognitive decline. Alzheimers Res Ther 2024; 16:35. [PMID: 38355598 PMCID: PMC10865579 DOI: 10.1186/s13195-024-01411-0] [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: 09/12/2023] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Sleep-wake regulating circuits are affected during prodromal stages in the pathological progression of both Alzheimer's disease (AD) and Parkinson's disease (PD), and this disturbance can be measured passively using wearable devices. Our objective was to determine whether accelerometer-based measures of 24-h activity are associated with subsequent development of AD, PD, and cognitive decline. METHODS This study obtained UK Biobank data from 82,829 individuals with wrist-worn accelerometer data aged 40 to 79 years with a mean (± SD) follow-up of 6.8 (± 0.9) years. Outcomes were accelerometer-derived measures of 24-h activity (derived by cosinor, nonparametric, and functional principal component methods), incident AD and PD diagnosis (obtained through hospitalization or primary care records), and prospective longitudinal cognitive testing. RESULTS One hundred eighty-seven individuals progressed to AD and 265 to PD. Interdaily stability (a measure of regularity, hazard ratio [HR] per SD increase 1.25, 95% confidence interval [CI] 1.05-1.48), diurnal amplitude (HR 0.79, CI 0.65-0.96), mesor (mean activity; HR 0.77, CI 0.59-0.998), and activity during most active 10 h (HR 0.75, CI 0.61-0.94), were associated with risk of AD. Diurnal amplitude (HR 0.28, CI 0.23-0.34), mesor (HR 0.13, CI 0.10-0.16), activity during least active 5 h (HR 0.24, CI 0.08-0.69), and activity during most active 10 h (HR 0.20, CI 0.16-0.25) were associated with risk of PD. Several measures were additionally predictive of longitudinal cognitive test performance. CONCLUSIONS In this community-based longitudinal study, accelerometer-derived metrics were associated with elevated risk of AD, PD, and accelerated cognitive decline. These findings suggest 24-h rhythm integrity, as measured by affordable, non-invasive wearable devices, may serve as a scalable early marker of neurodegenerative disease.
Collapse
Affiliation(s)
- Joseph R Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA, 94304, USA.
| | - Renske Lok
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Lara Weed
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Zihuai He
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA, 94304, USA
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA, 94304, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA, 94304, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Jamie M Zeitzer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Sierra-Pacific Mental Illness Research, Education, and Clinical Center (MIRECC), Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
43
|
Ferini-Strambi L, Liguori C, Lucey BP, Mander BA, Spira AP, Videnovic A, Baumann C, Franco O, Fernandes M, Gnarra O, Krack P, Manconi M, Noain D, Saxena S, Kallweit U, Randerath W, Trenkwalder C, Rosenzweig I, Iranzo A, Bradicich M, Bassetti C. Role of sleep in neurodegeneration: the consensus report of the 5th Think Tank World Sleep Forum. Neurol Sci 2024; 45:749-767. [PMID: 38087143 DOI: 10.1007/s10072-023-07232-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/26/2023] [Indexed: 01/18/2024]
Abstract
Sleep abnormalities may represent an independent risk factor for neurodegeneration. An international expert group convened in 2021 to discuss the state-of-the-science in this domain. The present article summarizes the presentations and discussions concerning the importance of a strategy for studying sleep- and circadian-related interventions for early detection and prevention of neurodegenerative diseases. An international expert group considered the current state of knowledge based on the most relevant publications in the previous 5 years; discussed the current challenges in the field of relationships among sleep, sleep disorders, and neurodegeneration; and identified future priorities. Sleep efficiency and slow wave activity during non-rapid eye movement (NREM) sleep are decreased in cognitively normal middle-aged and older adults with Alzheimer's disease (AD) pathology. Sleep deprivation increases amyloid-β (Aβ) concentrations in the interstitial fluid of experimental animal models and in cerebrospinal fluid in humans, while increased sleep decreases Aβ. Obstructive sleep apnea (OSA) is a risk factor for dementia. Studies indicate that positive airway pressure (PAP) treatment should be started in patients with mild cognitive impairment or AD and comorbid OSA. Identification of other measures of nocturnal hypoxia and sleep fragmentation could better clarify the role of OSA as a risk factor for neurodegeneration. Concerning REM sleep behavior disorder (RBD), it will be crucial to identify the subset of RBD patients who will convert to a specific neurodegenerative disorder. Circadian sleep-wake rhythm disorders (CSWRD) are strong predictors of caregiver stress and institutionalization, but the absence of recommendations or consensus statements must be considered. Future priorities include to develop and validate existing and novel comprehensive assessments of CSWRD in patients with/at risk for dementia. Strategies for studying sleep-circadian-related interventions for early detection/prevention of neurodegenerative diseases are required. CSWRD evaluation may help to identify additional biomarkers for phenotyping and personalizing treatment of neurodegeneration.
Collapse
Affiliation(s)
- Luigi Ferini-Strambi
- Sleep Disorders Center, Division of Neuroscience, Università Vita-Salute San Raffaele, Milan, Italy.
| | - Claudio Liguori
- Sleep Medicine Center, University of Rome Tor Vergata, Rome, Italy
| | - Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Adam P Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Aleksandar Videnovic
- Department of Neurology, Division of Sleep Medicine, Massachussets General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christian Baumann
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Oscar Franco
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | | | - Oriella Gnarra
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Paul Krack
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Mauro Manconi
- Sleep Medicine Unit, Faculty of Biomedical Sciences, Neurocenter of the Southern Switzerland, Regional Hospital of Lugano, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Daniela Noain
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Smita Saxena
- Department of Neurology, University of Bern, Bern, Switzerland
| | - Ulf Kallweit
- Clinical Sleep and Neuroimmunology, University Witten/Herdecke, Witten, Germany
| | | | - C Trenkwalder
- Department of Neurosurgery, Paracelsus-Elena Klinik, University Medical Center, KasselGoettingen, Germany
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, King's College London, London, UK
| | - Alex Iranzo
- Sleep Center, Neurology Service, Hospital Clinic de Barcelona, Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Matteo Bradicich
- Department of Pulmonology and Sleep Disorders Centre, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
44
|
Yu X, Zhou X, He Z, He B, Wan K, Wei M, Guo T, Han Y. Sleep and APOE-ε4 have a synergistic effect on plasma biomarkers and longitudinal cognitive decline in older adults. CNS Neurosci Ther 2024; 30:e14558. [PMID: 38421124 PMCID: PMC10850800 DOI: 10.1111/cns.14558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/31/2023] [Accepted: 11/25/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Sleep disorders are prevalent among patients with Alzheimer's disease (AD), and the APOE ε4 genotype is a key genetic risk factor for sporadic AD. However, the combined effect of the genotype and sleep disorders on cognitive decline remains uncertain. METHODS A total of 972 participants were drawn from the SILCODE cohort, comprising 655 without the ε4 allele (APOE-) and 317 with ε4 allele (APOE+). Data were collected, including neuropsychological assessments, sleep measurements, plasma biomarkers, and PET imaging. A Sleep Composite Index (SCI) was created, categorizing participants into high risk (Sleep+) and low risk (Sleep-). RESULTS Significant predictions of dementia risk associated with plasma p-tau181, neurofilament light chain (NfL), and SCI. Individuals with both Sleep+ and APOE+ had a higher risk of dementia compared to those with Sleep-. The Sleep+/APOE+ group had higher plasma NfL levels than the Sleep-/APOE- group. Similar trends emerged in plasma NfL levels among the Aβ PET-positive subgroup. Plasma NfL levels explained 23% of the relationship between SCI and cognitive impairment. CONCLUSION Our study highlights sleep disorder was associated with cognitive decline, with plasma NfL playing a partial mediating role. These findings explain how sleep disorders affect cognitive function and emphasize the importance of healthy sleep for older adults.
Collapse
Affiliation(s)
- Xianfeng Yu
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Xia Zhou
- Department of NeurologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Zhengbo He
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Beiqi He
- School of Information and Communication EngineeringHainan UniversityHaikouChina
| | - Ke Wan
- Department of NeurologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Min Wei
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Tengfei Guo
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Ying Han
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
- School of Information and Communication EngineeringHainan UniversityHaikouChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- National Clinical Research Center for Geriatric DisordersBeijingChina
| |
Collapse
|
45
|
Rykov YG, Patterson MD, Gangwar BA, Jabar SB, Leonardo J, Ng KP, Kandiah N. Predicting cognitive scores from wearable-based digital physiological features using machine learning: data from a clinical trial in mild cognitive impairment. BMC Med 2024; 22:36. [PMID: 38273340 PMCID: PMC10809621 DOI: 10.1186/s12916-024-03252-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Continuous assessment and remote monitoring of cognitive function in individuals with mild cognitive impairment (MCI) enables tracking therapeutic effects and modifying treatment to achieve better clinical outcomes. While standardized neuropsychological tests are inconvenient for this purpose, wearable sensor technology collecting physiological and behavioral data looks promising to provide proxy measures of cognitive function. The objective of this study was to evaluate the predictive ability of digital physiological features, based on sensor data from wrist-worn wearables, in determining neuropsychological test scores in individuals with MCI. METHODS We used the dataset collected from a 10-week single-arm clinical trial in older adults (50-70 years old) diagnosed with amnestic MCI (N = 30) who received a digitally delivered multidomain therapeutic intervention. Cognitive performance was assessed before and after the intervention using the Neuropsychological Test Battery (NTB) from which composite scores were calculated (executive function, processing speed, immediate memory, delayed memory and global cognition). The Empatica E4, a wrist-wearable medical-grade device, was used to collect physiological data including blood volume pulse, electrodermal activity, and skin temperature. We processed sensors' data and extracted a range of physiological features. We used interpolated NTB scores for 10-day intervals to test predictability of scores over short periods and to leverage the maximum of wearable data available. In addition, we used individually centered data which represents deviations from personal baselines. Supervised machine learning was used to train models predicting NTB scores from digital physiological features and demographics. Performance was evaluated using "leave-one-subject-out" and "leave-one-interval-out" cross-validation. RESULTS The final sample included 96 aggregated data intervals from 17 individuals. In total, 106 digital physiological features were extracted. We found that physiological features, especially measures of heart rate variability, correlated most strongly to the executive function compared to other cognitive composites. The model predicted the actual executive function scores with correlation r = 0.69 and intra-individual changes in executive function scores with r = 0.61. CONCLUSIONS Our findings demonstrated that wearable-based physiological measures, primarily HRV, have potential to be used for the continuous assessments of cognitive function in individuals with MCI.
Collapse
Affiliation(s)
| | | | | | | | - Jacklyn Leonardo
- Dementia Research Centre, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Kok Pin Ng
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Nagaendran Kandiah
- Dementia Research Centre, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
46
|
Yiallourou SR, Cribb L, Cavuoto MG, Rowsthorn E, Nicolazzo J, Gibson M, Baril AA, Pase MP. Association of the Sleep Regularity Index With Incident Dementia and Brain Volume. Neurology 2024; 102:e208029. [PMID: 38165323 DOI: 10.1212/wnl.0000000000208029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Irregular sleep may increase the risk of cardiometabolic conditions, but its association with incident dementia is unclear. The aim of this study was to assess the association between sleep regularity, that is, the day-to-day consistency in sleep-wake patterns and the risk of incident dementia and related brain MRI endophenotypes. METHODS We used Cox proportional hazard models to investigate the relationships between sleep regularity and incident dementia in 88,094 UK Biobank participants. The sleep regularity index (SRI) was calculated as the probability of being in the same state (asleep/awake) at any 2 time points 24 hours apart, averaged over 7 days of accelerometry. RESULTS The mean age of the sample was 62 years (SD = 8), 56% were women, and the median SRI was 60 (SD = 10). There were 480 cases of incident dementia over a median 7.2 years of follow-up. Following adjustments for demographic, clinical, and genetic confounders (APOE ε4), there was a nonlinear association between the SRI and dementia hazard (p [global test of spline term] < 0.001) with hazard ratios (HRs) following a U-shape pattern. HRs, relative to the median SRI, were 1.53 (95% CI 1.24-1.89) for participants with SRI at the 5th percentile (SRI = 41) and 1.16 (95% CI 0.89-1.50) for those with SRI at the 95th percentile (SRI = 71). In a subset with brain MRI (n = 15,263), gray matter and hippocampal volume tended to be lowest at the extremes of the SRI. DISCUSSION Sleep regularity displayed a U-shaped association with risk of incident dementia. Irregular sleep may represent a novel dementia risk factor.
Collapse
Affiliation(s)
- Stephanie R Yiallourou
- From the Turner Institute for Brain and Mental Health (S.R.Y., L.C., M.G.C., E.R., J.N., M.G., M.P.P.), School of Psychological Science, Monash University; National Ageing Research Institute (M.G.C.), Melbourne, Australia; Douglas Mental Health University Institute (A.-A.B.), McGill University, Montreal, Quebec, Canada; and Harvard T.H. Chan School of Public Health (M.P.P.), Harvard University, Boston, MA
| | - Lachlan Cribb
- From the Turner Institute for Brain and Mental Health (S.R.Y., L.C., M.G.C., E.R., J.N., M.G., M.P.P.), School of Psychological Science, Monash University; National Ageing Research Institute (M.G.C.), Melbourne, Australia; Douglas Mental Health University Institute (A.-A.B.), McGill University, Montreal, Quebec, Canada; and Harvard T.H. Chan School of Public Health (M.P.P.), Harvard University, Boston, MA
| | - Marina G Cavuoto
- From the Turner Institute for Brain and Mental Health (S.R.Y., L.C., M.G.C., E.R., J.N., M.G., M.P.P.), School of Psychological Science, Monash University; National Ageing Research Institute (M.G.C.), Melbourne, Australia; Douglas Mental Health University Institute (A.-A.B.), McGill University, Montreal, Quebec, Canada; and Harvard T.H. Chan School of Public Health (M.P.P.), Harvard University, Boston, MA
| | - Ella Rowsthorn
- From the Turner Institute for Brain and Mental Health (S.R.Y., L.C., M.G.C., E.R., J.N., M.G., M.P.P.), School of Psychological Science, Monash University; National Ageing Research Institute (M.G.C.), Melbourne, Australia; Douglas Mental Health University Institute (A.-A.B.), McGill University, Montreal, Quebec, Canada; and Harvard T.H. Chan School of Public Health (M.P.P.), Harvard University, Boston, MA
| | - Jessica Nicolazzo
- From the Turner Institute for Brain and Mental Health (S.R.Y., L.C., M.G.C., E.R., J.N., M.G., M.P.P.), School of Psychological Science, Monash University; National Ageing Research Institute (M.G.C.), Melbourne, Australia; Douglas Mental Health University Institute (A.-A.B.), McGill University, Montreal, Quebec, Canada; and Harvard T.H. Chan School of Public Health (M.P.P.), Harvard University, Boston, MA
| | - Madeline Gibson
- From the Turner Institute for Brain and Mental Health (S.R.Y., L.C., M.G.C., E.R., J.N., M.G., M.P.P.), School of Psychological Science, Monash University; National Ageing Research Institute (M.G.C.), Melbourne, Australia; Douglas Mental Health University Institute (A.-A.B.), McGill University, Montreal, Quebec, Canada; and Harvard T.H. Chan School of Public Health (M.P.P.), Harvard University, Boston, MA
| | - Andrée-Ann Baril
- From the Turner Institute for Brain and Mental Health (S.R.Y., L.C., M.G.C., E.R., J.N., M.G., M.P.P.), School of Psychological Science, Monash University; National Ageing Research Institute (M.G.C.), Melbourne, Australia; Douglas Mental Health University Institute (A.-A.B.), McGill University, Montreal, Quebec, Canada; and Harvard T.H. Chan School of Public Health (M.P.P.), Harvard University, Boston, MA
| | - Matthew P Pase
- From the Turner Institute for Brain and Mental Health (S.R.Y., L.C., M.G.C., E.R., J.N., M.G., M.P.P.), School of Psychological Science, Monash University; National Ageing Research Institute (M.G.C.), Melbourne, Australia; Douglas Mental Health University Institute (A.-A.B.), McGill University, Montreal, Quebec, Canada; and Harvard T.H. Chan School of Public Health (M.P.P.), Harvard University, Boston, MA
| |
Collapse
|
47
|
Leng Y, Knutson K, Carnethon MR, Yaffe K. Association Between Sleep Quantity and Quality in Early Adulthood With Cognitive Function in Midlife. Neurology 2024; 102:e208056. [PMID: 38170947 PMCID: PMC10870739 DOI: 10.1212/wnl.0000000000208056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Growing evidence supports an association between sleep quality and risk of dementia. However, little is known about whether objectively measured sleep duration and quality influence cognition in midlife, a period of importance for understanding the direction of the association between sleep and dementia. We examined the association between sleep duration and quality, measured when participants were in their mid-30s to late 40s, and midlife cognition assessed 11 years later among Black and White adults. METHODS As part of the Coronary Artery Risk Development in Young Adults cohort study, sleep duration and quality were assessed objectively using wrist actigraphy and subjectively by Pittsburgh Sleep Quality Index (PSQI) at 2003-2005. During 2015-2016, we evaluated midlife cognition using the Digit Symbol Substitution Test (DSST), Stroop test, Rey Auditory Verbal Learning Test, Montreal Cognitive Assessment (MoCA), and Letter Fluency and Category Fluency tests. We used multivariable logistic regression to examine the association between sleep parameters and poor cognitive performance, which was defined as a score that was >1 SD below the mean score. RESULTS The 526 participants (58% women and 44% Black) had a mean age of 40.1 ± 3.6 years at baseline, a mean sleep duration of 6.1 ± 1.1 hours, and mean sleep fragmentation index (calculated as the sum of the percentage of time spent moving and the percentage of immobile periods ≤1 minute) of 19.2 ± 8.1%, and 239 (45.6%) participants reported poor sleep as defined by a PSQI global score of >5. After adjustment for demographics, education, smoking, body mass index, depression, physical activity, hypertension, and diabetes, those in the highest vs lowest tertile of sleep fragmentation index had over twice the odds of having poor cognitive performance (>1 SD below the mean) on the DSST (odds ratio [OR] = 2.97; 95% CI 1.34-6.56), fluency (OR = 2.42; 95% CI 1.17-5.02), and MoCA test (OR = 2.29; 95% CI 1.06-4.94). The association between sleep fragmentation and cognitive performance did not differ by race or sex. Objective sleep duration or subjective sleep quality was not associated with cognition in midlife. DISCUSSION Actigraphy-measured high sleep fragmentation rather than sleep duration was associated with worse cognition among middle-aged Black and White men and women. Sleep quality is important for cognitive health even as early as midlife.
Collapse
Affiliation(s)
- Yue Leng
- From the Department of Psychiatry and Behavioral Sciences (Y.L., K.Y.), Neurology (K.Y.), and Epidemiology (K.Y.), University of California, San Francisco; Departments of Neurology (K.K.) and Preventive Medicine (M.R.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; and VA Medical Center (K.Y.), San Francisco, CA
| | - Kristen Knutson
- From the Department of Psychiatry and Behavioral Sciences (Y.L., K.Y.), Neurology (K.Y.), and Epidemiology (K.Y.), University of California, San Francisco; Departments of Neurology (K.K.) and Preventive Medicine (M.R.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; and VA Medical Center (K.Y.), San Francisco, CA
| | - Mercedes R Carnethon
- From the Department of Psychiatry and Behavioral Sciences (Y.L., K.Y.), Neurology (K.Y.), and Epidemiology (K.Y.), University of California, San Francisco; Departments of Neurology (K.K.) and Preventive Medicine (M.R.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; and VA Medical Center (K.Y.), San Francisco, CA
| | - Kristine Yaffe
- From the Department of Psychiatry and Behavioral Sciences (Y.L., K.Y.), Neurology (K.Y.), and Epidemiology (K.Y.), University of California, San Francisco; Departments of Neurology (K.K.) and Preventive Medicine (M.R.C.), Northwestern University Feinberg School of Medicine, Chicago, IL; and VA Medical Center (K.Y.), San Francisco, CA
| |
Collapse
|
48
|
Altunkaya A, Deichsel C, Kreuzer M, Nguyen DM, Wintergerst AM, Rammes G, Schneider G, Fenzl T. Altered sleep behavior strengthens face validity in the ArcAβ mouse model for Alzheimer's disease. Sci Rep 2024; 14:951. [PMID: 38200079 PMCID: PMC10781983 DOI: 10.1038/s41598-024-51560-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024] Open
Abstract
Demographic changes will expand the number of senior citizens suffering from Alzheimer's disease (AD). Key aspects of AD pathology are sleep impairments, associated with onset and progression of AD. AD mouse models may provide insights into mechanisms of AD-related sleep impairments. Such models may also help to establish new biomarkers predicting AD onset and monitoring AD progression. The present study aimed to establish sleep-related face validity of a widely used mouse model of AD (ArcAβ model) by comprehensively characterizing its baseline sleep/wake behavior. Chronic EEG recordings were performed continuously on four consecutive days in freely behaving mice. Spectral and temporal sleep/wake parameters were assessed and analyzed. EEG recordings showed decreased non-rapid eye movement sleep (NREMS) and increased wakefulness in transgenic mice (TG). Vigilance state transitions were different in TG mice when compared to wildtype littermates (WT). During NREMS, TG mice had lower power between 1 and 5 Hz and increased power between 5 and 30 Hz. Sleep spindle amplitudes in TG mice were lower. Our study strongly provides sleep-linked face validity for the ArcAβ model. These findings extend the potential of the mouse model to investigate mechanisms of AD-related sleep impairments and the impact of sleep impairments on the development of AD.
Collapse
Affiliation(s)
- Alp Altunkaya
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Cassandra Deichsel
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Matthias Kreuzer
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Duy-Minh Nguyen
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ann-Marie Wintergerst
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gerhard Schneider
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Fenzl
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.
| |
Collapse
|
49
|
Carpi M, Fernandes M, Mercuri NB, Liguori C. Sleep Biomarkers for Predicting Cognitive Decline and Alzheimer's Disease: A Systematic Review of Longitudinal Studies. J Alzheimers Dis 2024; 97:121-143. [PMID: 38043016 DOI: 10.3233/jad-230933] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2023]
Abstract
BACKGROUND Sleep disturbances are considered a hallmark of dementia, and strong evidence supports the association between alterations in sleep parameters and cognitive decline in patients with mild cognitive impairment and Alzheimer's disease (AD). OBJECTIVE This systematic review aims to summarize the existing evidence on the longitudinal association between sleep parameters and cognitive decline, with the goal of identifying potential sleep biomarkers of AD-related neurodegeneration. METHODS Literature search was conducted in PubMed, Web of Science, and Scopus databases from inception to 28 March 2023. Longitudinal studies investigating the association between baseline objectively-measured sleep parameters and cognitive decline were assessed for eligibility. RESULTS Seventeen studies were included in the qualitative synthesis. Sleep fragmentation, reduced sleep efficiency, reduced REM sleep, increased light sleep, and sleep-disordered breathing were identified as predictors of cognitive decline. Sleep duration exhibited a U-shaped relation with subsequent neurodegeneration. Additionally, several sleep microstructural parameters were associated with cognitive decline, although inconsistencies were observed across studies. CONCLUSIONS These findings suggest that sleep alterations hold promise as early biomarker of cognitive decline, but the current evidence is limited due to substantial methodological heterogeneity among studies. Further research is necessary to identify the most reliable sleep parameters for predicting cognitive impairment and AD, and to investigate interventions targeting sleep that can assist clinicians in the early recognition and treatment of cognitive decline. Standardized procedures for longitudinal studies evaluating sleep and cognition should be developed and the use of continuous sleep monitoring techniques, such as actigraphy or EEG headband, might be encouraged.
Collapse
Affiliation(s)
- Matteo Carpi
- Sleep Medicine Centre, Neurology Unit, University Hospital Tor Vergata, Rome, Italy
| | - Mariana Fernandes
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Sleep Medicine Centre, Neurology Unit, University Hospital Tor Vergata, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Claudio Liguori
- Sleep Medicine Centre, Neurology Unit, University Hospital Tor Vergata, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
50
|
Okuda M, Noda A, Iwamoto K, Hishikawa N, Miyata S, Yasuma F, Taoka T, Ozaki N, Suhr JA, Miyazaki S. Assessment of cognitive function and sleep-wake rhythms in community-dwelling older adults. Sleep Biol Rhythms 2024; 22:137-145. [PMID: 38476850 PMCID: PMC10899937 DOI: 10.1007/s41105-023-00491-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: 05/03/2023] [Accepted: 09/06/2023] [Indexed: 03/14/2024]
Abstract
Disruption of the circadian rhythm and sleep-wake cycles is a consequence of aging and is associated with the cognitive decline and many neurodegenerative conditions. We investigated the bedtime, wake-up time, sleep timing (midpoint between bedtime and wake-up time), and sleep timing standard deviation (SD) using the actigraphy among 80 consecutive volunteers aged ≥ 60 years. Global cognitive function and executive function of detailed cognitive domains were evaluated using the mini-mental state examination (MMSE) and Wisconsin card sorting test (WCST) and subjective daytime sleepiness was assessed using the Epworth Sleepiness Scale (ESS). The category achievement (CA), total errors (TE), perseverative errors of Nelson (PEN), non-perseverative errors (NPE), and difficulties in maintaining set (DMS) on the WCST were significantly correlated with sleep timing SD (CA: r = - 0.276, p = 0.013, TE: r = 0.311, p = 0.005, PEN: r = 0.241, p = 0.032, NPE: r = 0.250, p = 0.025, DMS: r = 0.235, p = 0.036), but not with the MMSE score. Multiple regression analyses with the stepwise forward selection method including age, ESS score, bedtime, sleep timing, and sleep timing SD, revealed that the ESS score, and sleep timing SD were significant factors related to CA on the WCST (ESS score: β = - 0.322, p = 0.004; sleep timing SD: β = - 0.250, p = 0.022). Assessment of sleep-wake rhythms, daytime sleepiness, and cognitive function using the MMSE and WCST is valuable for the prediction of cognitive decline in the geriatric population.
Collapse
Affiliation(s)
- Masato Okuda
- Department of Biomedical Sciences, Chubu University Graduate School of Life and Health Sciences, 1200, Matsumoto-cho, Kasugai, Aichi 487-8501 Japan
| | - Akiko Noda
- Department of Biomedical Sciences, Chubu University Graduate School of Life and Health Sciences, 1200, Matsumoto-cho, Kasugai, Aichi 487-8501 Japan
| | - Kunihiro Iwamoto
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nozomi Hishikawa
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Seiko Miyata
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumihiko Yasuma
- Department of Biomedical Sciences, Chubu University Collage of Life and Health Sciences, Kasugai, Japan
| | - Toshiaki Taoka
- Department of Innovative Biomedical Visualization, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Julie A. Suhr
- Department of Psychology, Ohio University, Athens, OH USA
| | - Soichiro Miyazaki
- Research Institute of Life and Health Sciences, Chubu University, Kasugai, Japan
| |
Collapse
|