1
|
Mitchell V, Frenguelli BG, Bakker S, Ngomba RT, Richardson M, Hill E, Wall MJ. Soluble tau aggregates decrease the threshold for thalamic oscillations and increase the excitability of thalamic neurons. Neuropharmacology 2025; 273:110455. [PMID: 40189019 DOI: 10.1016/j.neuropharm.2025.110455] [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: 02/28/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Sleep disturbances frequently occur early in dementias such as Alzheimer's disease (AD) and potentially arise from many factors including cortico-thalamo-cortical (CTC) loop dysfunction. It has been reported that tau filament deposition occurs in the thalamus and there is thalamic atrophy in symptomatic AD patients which could contribute to CTC loop disturbance. Here we have investigated whether human recombinant tau soluble aggregates can induce dysfunction in thalamic circuits. Electrophysiological measurements were made from acutely isolated male and female rat corticothalamic slices following incubation with tau aggregates. Tau aggregates markedly reduced the threshold for inducing spindle-like oscillations and increased the excitability of thalamic neurons. Tau aggregates also significantly enhanced the frequency of miniature excitatory postsynaptic currents recorded in ventrobasal thalamic neurons, suggesting possible changes in terminal Ca2+ influx. These pro-excitatory effects of tau aggregates could contribute to the aberrant CTC loop dysfunction observed in AD models and patients, which manifests as sleep disturbances and absence seizures.
Collapse
Affiliation(s)
| | | | | | - Richard T Ngomba
- School of Life Sciences, University of Warwick, UK; College of Health and Science, School of Health and Care Sciences, Pharmacy, University of Lincoln, Joseph Banks Laboratories, Room JBL2W25, Green Lane, Lincoln, Lincolnshire LN6 7DL, UK
| | - Magnus Richardson
- School of Life Sciences, University of Warwick, UK; Institute of Mathematics, University of Warwick, UK
| | - Emily Hill
- School of Life Sciences, University of Warwick, UK
| | - Mark J Wall
- School of Life Sciences, University of Warwick, UK.
| |
Collapse
|
2
|
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
|
3
|
Kaur S, Kumari D, Dandekar MP. Importance of Gut Microbiota Dysbiosis and Circadian Disruption-Associated Biomarkers in Emergence of Alzheimer's Disease. Mol Neurobiol 2025; 62:6308-6316. [PMID: 39775480 DOI: 10.1007/s12035-024-04685-5] [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: 07/19/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Alzheimer's disease (AD) is a major devastating neurodegenerative disorder afflicting majorly the geriatric population. Emerging studies augur the connection of gut dysbiosis and circadian disruption with the early onset of AD. Gut dysbiosis is characterized by dysregulated gut microbiota signature and compromised intestinal integrity, which provokes the translocation of bacterial metabolites into the systemic circulation. Noteworthy, gut-derived metabolites like calprotectin, trimethylamine-N-oxide, kynurenine, isoamylamine, and short-chain fatty acids play a key role in AD pathogenesis. Circadian dysregulation also corresponds with the exacerbated AD pathogenesis by accumulating Aβ and tau proteins. Moreover, circadian dysregulation is one of the causative factors for gut dysbiosis. This review discusses the complex interplay between the microbiota-gut-brain axis, circadian rhythmicity, and the emergence of AD. We reviewed preclinical and clinical studies on AD describing potential biomarkers of gut dysbiosis and circadian dysregulation. The identification of new biomarkers associated with the microbiota-gut-brain axis and circadian rhythmicity may help in early diagnosis and development of targeted therapies for mitigating neurodegenerative AD.
Collapse
Affiliation(s)
- Simranjit Kaur
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India, 500037
| | - Deepali Kumari
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India, 500037
| | - Manoj P Dandekar
- Department of Biological Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India, 500037.
| |
Collapse
|
4
|
Tian ZY, Jiang B, Jin M, Yu XK, Chen QL, Wang JH. Alzheimer's disease and insomnia: a bibliometric study and visualization analysis. Front Aging Neurosci 2025; 17:1542607. [PMID: 40264463 PMCID: PMC12011777 DOI: 10.3389/fnagi.2025.1542607] [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: 12/10/2024] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Background Alzheimer's disease (AD) is the fastest-growing neurodegenerative disorder globally, with patient numbers expected to rise to 130 million by 2050. Insomnia, a prevalent comorbidity, exhibits a bidirectional relationship with AD: insomnia accelerates AD pathology, while AD worsens sleep disorders. This relationship has emerged as a key area of research. Current mechanisms involve oxidative stress, inflammatory responses, and glymphatic system dysfunction, yet a comprehensive review of these processes remains absent. Objective To conduct a visual analysis of the relationship between Alzheimer's disease and insomnia using CiteSpace. Methods Literature on "insomnia" and "Alzheimer's disease" published between January 1, 2000, and October 31, 2024, was retrieved from the Web of Science Core Collection. CiteSpace and VOSviewer software were used to analyze institutions, authors, and keywords. Results A total of 1,907 articles were analyzed, revealing a consistent upward trend in publication volume. The United States and the Mayo Clinic were identified as leading contributors, producing 704 and 57 publications, respectively. Boeve Bradley F the most prolific author contributed 30 publications. Collaboration was actively observed among countries, institutions, and authors. High-frequency keywords identified were "Parkinson's disease," "cognitive impairment," and "sleep behavior disorder." Emerging research areas are likely to focus on "sleep quality" and the "glymphatic system." Conclusion This study is the first to apply bibliometric analysis to identify three key trends in AD and insomnia research: the dominance of the United States and Mayo Clinic, strong international collaboration, and a focus on critical areas such as cognitive impairment, the glymphatic system, and sleep interventions. Insomnia may accelerate AD progression via multiple pathways, indicating that enhancing sleep quality could provide new strategies for early intervention. Future research should prioritize advancing the clinical translation of sleep interventions and investigating the mechanisms of the glymphatic system.
Collapse
Affiliation(s)
- Zi-Yue Tian
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Meng Jin
- The Third Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Xiao-Kun Yu
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Qi-Lin Chen
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- KweiChow Moutai Hospital, Zunyi, Guizhou, China
| | - Jia-Hui Wang
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
5
|
Canet G, Da Gama Monteiro F, Rocaboy E, Diego-Diaz S, Khelaifia B, Godbout K, Lachhab A, Kim J, Valencia DI, Yin A, Wu HT, Howell J, Blank E, Laliberté F, Fortin N, Boscher E, Fereydouni-Forouzandeh P, Champagne S, Guisle I, Hébert SS, Pernet V, Liu H, Lu W, Debure L, Rapoport DM, Ayappa I, Varga AW, Parekh A, Osorio RS, Lacroix S, Burns MP, Lucey BP, Blessing EM, Planel E. Sleep-wake variation in body temperature regulates tau secretion and correlates with CSF and plasma tau. J Clin Invest 2025; 135:e182931. [PMID: 39903530 PMCID: PMC11957704 DOI: 10.1172/jci182931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/31/2025] [Indexed: 02/06/2025] Open
Abstract
Sleep disturbance is bidirectionally associated with an increased risk of Alzheimer's disease and other tauopathies. While the sleep-wake cycle regulates interstitial and cerebrospinal fluid (CSF) tau levels, the underlying mechanisms remain unknown. Understanding these mechanisms is crucial, given the evidence that tau pathology spreads through neuron-to-neuron transfer, involving the secretion and internalization of pathological tau forms. Here, we combined in vitro, in vivo, and clinical methods to reveal a pathway by which changes in body temperature (BT) over the sleep-wake cycle modulate extracellular tau levels. In mice, a higher BT during wakefulness and sleep deprivation increased CSF and plasma tau levels, while also upregulating unconventional protein secretion pathway I (UPS-I) events including (a) intracellular tau dephosphorylation, (b) caspase 3-mediated cleavage of tau (TauC3), and (c) membrane translocation of tau through binding to phosphatidylinositol 4,5-bisphosphate (PIP2) and syndecan 3. In humans, the increase in CSF and plasma tau levels observed after wakefulness correlated with BT increases during wakefulness. By demonstrating that sleep-wake variation in BT regulates extracellular tau levels, our findings highlight the importance of thermoregulation in linking sleep disturbances to tau-mediated neurodegeneration and the preventative potential of thermal interventions.
Collapse
Affiliation(s)
- Geoffrey Canet
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Felipe Da Gama Monteiro
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Médecine Moléculaire, Québec, Québec City, Canada
| | - Emma Rocaboy
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Sofia Diego-Diaz
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Boutheyna Khelaifia
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Kelly Godbout
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Aymane Lachhab
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Jessica Kim
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Daphne I. Valencia
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Audrey Yin
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Hau-Tieng Wu
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Jordan Howell
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Emily Blank
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Francis Laliberté
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
| | - Nadia Fortin
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
| | - Emmanuelle Boscher
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | | | - Stéphanie Champagne
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Isabelle Guisle
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Sébastien S. Hébert
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| | - Vincent Pernet
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Médecine Moléculaire, Québec, Québec City, Canada
- Department of Neurology, Inselspital, and
- Center for Experimental Neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Haiyan Liu
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - William Lu
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ludovic Debure
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - David M. Rapoport
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Indu Ayappa
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew W. Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ankit Parekh
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ricardo S. Osorio
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Steve Lacroix
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Médecine Moléculaire, Québec, Québec City, Canada
| | - Mark P. Burns
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Brendan P. Lucey
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Esther M. Blessing
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Emmanuel Planel
- Centre de Recherche du CHU de Québec – Université Laval, Axe Neurosciences, Québec, Québec City, Canada
- Université Laval, Faculté de Médecine, Département de Psychiatrie et Neurosciences, Québec, Québec City, Canada
| |
Collapse
|
6
|
Páez A, Gillman SO, Dogaheh SB, Carnes A, Dakterzada F, Barbé F, Dang‐Vu TT, Ripoll GP. Sleep spindles and slow oscillations predict cognition and biomarkers of neurodegeneration in mild to moderate Alzheimer's disease. Alzheimers Dement 2025; 21:e14424. [PMID: 39878233 PMCID: PMC11848347 DOI: 10.1002/alz.14424] [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: 09/09/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 01/31/2025]
Abstract
INTRODUCTION Changes in sleep physiology can predate cognitive symptoms by decades in persons with Alzheimer's disease (AD), but it remains unclear which sleep characteristics predict cognitive and neurodegenerative changes after AD onset. METHODS Using data from a prospective cohort of mild to moderate AD (n = 60), we analyzed non-rapid eye movement sleep spindles and slow oscillations (SOs) at baseline and their associations with baseline amyloid beta (Aβ) and tau and with cognition from baseline to 3-year follow-up. RESULTS Higher spindle and SO activity predicted significant changes in Aβ and tau at baseline, lower Alzheimer's Disease Assessment Scale Cognitive Subscale (better cognitive performance) score, and higher Mini-Mental State Examination score from baseline to 36 months. Spindles and SOs mediated the effect of phosphorylated tau 181 (pTau181)/Aβ42 on cognition, while pTau181/aβ42 moderated the effect of spindles and SOs on cognition. DISCUSSION Our findings demonstrate that spindle and SO activity during sleep constitute predictive and non-invasive biomarkers of neurodegeneration and cognition in AD patients. HIGHLIGHTS Sleep spindles predict long-term cognitive performance in AD. Sleep spindle and SOs can be predictive, non-invasive biomarkers for AD. Sleep may be one of the most important modifiable risk factors for AD progression. Sleep microarchitecture is a novel therapeutic target for preserving brain heath. Sleep physiology can provide novel therapeutic targets to slow AD progression.
Collapse
Affiliation(s)
- Arsenio Páez
- Sleep, Cognition and Neuroimaging LaboratoryConcordia UniversityMontrealCanada
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM)Montréal (Québec)Canada
- Nuffield Department for Primary Care Health SciencesUniversity of OxfordOxfordUK
| | - Sam O. Gillman
- Sleep, Cognition and Neuroimaging LaboratoryConcordia UniversityMontrealCanada
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM)Montréal (Québec)Canada
| | | | - Anna Carnes
- Unitat de Trastorns CognitiusCognition and Behavior Study GroupHospital Universitari Santa Maria Universitat de LleidaLleidaSpain
| | - Faride Dakterzada
- Unitat de Trastorns CognitiusCognition and Behavior Study GroupHospital Universitari Santa Maria Universitat de LleidaLleidaSpain
| | - Ferran Barbé
- Translational Research in Respiratory Medicine (TRRM)Hospital Universitari Arnau de Vilanova‐Santa MariaBiomedical Research Institute of Lleida (IRBLleida)LleidaSpain
| | - Thien Thanh Dang‐Vu
- Sleep, Cognition and Neuroimaging LaboratoryConcordia UniversityMontrealCanada
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM)Montréal (Québec)Canada
| | - Gerard Piñol Ripoll
- Unitat de Trastorns CognitiusCognition and Behavior Study GroupHospital Universitari Santa Maria Universitat de LleidaLleidaSpain
- Alzheimer's Disease and Other Cognitive Disorders UnitNeurology ServiceHospital Clínic de BarcelonaFundació de Recerca Clínic ‐ Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| |
Collapse
|
7
|
Overton M, Sindi S, Basna R, Elmståhl S. Excessive sleep is associated with worse cognition, cognitive decline, and dementia in mild cognitive impairment. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70093. [PMID: 39996036 PMCID: PMC11848587 DOI: 10.1002/dad2.70093] [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: 10/05/2024] [Revised: 01/16/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION This study examines the link between daytime and nighttime excessive sleep and cognition, cognitive decline, and dementia in individuals with existing mild cognitive impairment (MCI). METHODS Using data from the Swedish longitudinal study Good Aging in Skåne, participants aged 60-102 years were retrospectively classified as MCI based on cognitive testing. The average follow-up time was 6.59 years. Mixed linear models assessed cross-sectional and longitudinal associations between excessive sleep patterns (napping ≥2 h or nighttime sleep ≥9 h) and multiple cognitive domains. Cox regressions estimated dementia risk for excessive sleep. RESULTS Of 4930 participants, 2052 (41%) had MCI. Excessive daytime napping and nighttime sleep were associated with worse cognition and cognitive decline. Excessive napping and nighttime sleep were also linked to higher dementia risk (hazard ratios: 1.75 and 1.86, respectively). DISCUSSION These findings suggest that excessive sleep in MCI is associated with further cognitive decline and dementia. Highlights Excessive daytime napping and nighttime sleep are linked cognitive decline for those with MCI.Excessive sleep during the day or at night heighten dementia risk.Worse test scores across multiple cognitive domains were observed for excessive daytime nappers.Excessive sleep in MCI may be a warning sign for further cognitive decline.
Collapse
Affiliation(s)
- Marieclaire Overton
- Division of Geriatric MedicineDepartment of Clinical Sciences in MalmöSkåne University HospitalLund UniversityMalmöSweden
| | - Shireen Sindi
- Division of Clinical GeriatricsCenter for Alzheimer ResearchKarolinska Institutet and Karolinska University HospitalMalmöSweden
- Ageing Epidemiology (AGE) Research UnitSchool of Public HealthImperial College LondonLondonUK
| | - Rani Basna
- Division of Geriatric MedicineDepartment of Clinical Sciences in MalmöSkåne University HospitalLund UniversityMalmöSweden
| | - Sölve Elmståhl
- Division of Geriatric MedicineDepartment of Clinical Sciences in MalmöSkåne University HospitalLund UniversityMalmöSweden
| |
Collapse
|
8
|
Hopkins MA, Tabuchi M. The power of the rocking cradle: improving sleep function by gentle vibration. Sleep 2024; 47:zsae245. [PMID: 39441991 DOI: 10.1093/sleep/zsae245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Indexed: 10/25/2024] Open
Affiliation(s)
- Makenzie A Hopkins
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Masashi Tabuchi
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
9
|
Valencia-Sanchez S, Davis M, Martensen J, Hoeffer C, Link C, Opp MR. Sleep-wake behavior and responses to sleep deprivation and immune challenge of protein kinase RNA-activated knockout mice. Brain Behav Immun 2024; 121:74-86. [PMID: 39043346 PMCID: PMC11563030 DOI: 10.1016/j.bbi.2024.07.027] [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: 04/19/2024] [Revised: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024] Open
Abstract
Protein Kinase RNA-activated (PKR) is an enzyme that plays a role in many systemic processes, including modulation of inflammation, and is implicated in neurodegenerative diseases, such as Alzheimer's disease (AD). PKR phosphorylation results in the production of several cytokines involved in the regulation / modulation of sleep, including interleukin-1β, tumor necrosis factor-α and interferon-γ. We hypothesized targeting PKR would alter spontaneous sleep of mice, attenuate responses to sleep deprivation, and inhibit responses to immune challenge. To test these hypotheses, we determined the sleep-wake phenotype of mice lacking PKR (knockout; PKR-/-) during undisturbed baseline conditions; in responses to six hours of sleep deprivation; and after immune challenge with lipopolysaccharide (LPS). Adult male mice (C57BL/6J, n = 7; PKR-/-, n = 7) were surgically instrumented with EEG recording electrodes and an intraperitoneal microchip to record core body temperature. During undisturbed baseline conditions, PKR -/- mice spent more time in non-rapid eye movement sleep (NREMS) and rapid-eye movement sleep (REMS), and less time awake at the beginning of the dark period of the light:dark cycle. Delta power during NREMS, a measure of sleep depth, was less in PKR-/- mice during the dark period, and core body temperatures were lower during the light period. Both mouse strains responded to sleep deprivation with increased NREMS and REMS, although these changes did not differ substantively between strains. The initial increase in delta power during NREMS after sleep deprivation was greater in PKR-/- mice, suggesting a faster buildup of sleep pressure with prolonged waking. Immune challenge with LPS increased NREMS and inhibited REMS to the same extent in both mouse strains, whereas the initial LPS-induced suppression of delta power during NREMS was greater in PKR-/- mice. Because sleep regulatory and immune responsive systems in brain are redundant and overlapping, other mediators and signaling pathways in addition to PKR are involved in the responses to acute sleep deprivation and LPS immune challenge.
Collapse
Affiliation(s)
- S Valencia-Sanchez
- Department of Integrative Physiology, University of Colorado Boulder, USA
| | - M Davis
- Department of Integrative Physiology, University of Colorado Boulder, USA
| | - J Martensen
- Department of Integrative Physiology, University of Colorado Boulder, USA
| | - C Hoeffer
- Institute for Behavioral Genetics, University of Colorado Boulder, USA
| | - C Link
- Department of Integrative Physiology, University of Colorado Boulder, USA
| | - M R Opp
- Department of Integrative Physiology, University of Colorado Boulder, USA.
| |
Collapse
|
10
|
Carrero L, Antequera D, Municio C, Carro E. Circadian rhythm disruption and retinal dysfunction: a bidirectional link in Alzheimer's disease? Neural Regen Res 2024; 19:1967-1972. [PMID: 38227523 DOI: 10.4103/1673-5374.390962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/07/2023] [Indexed: 01/17/2024] Open
Abstract
Dysfunction in circadian rhythms is a common occurrence in patients with Alzheimer's disease. A predominant function of the retina is circadian synchronization, carrying information to the brain through the retinohypothalamic tract, which projects to the suprachiasmatic nucleus. Notably, Alzheimer's disease hallmarks, including amyloid-β, are present in the retinas of Alzheimer's disease patients, followed/associated by structural and functional disturbances. However, the mechanistic link between circadian dysfunction and the pathological changes affecting the retina in Alzheimer's disease is not fully understood, although some studies point to the possibility that retinal dysfunction could be considered an early pathological process that directly modulates the circadian rhythm.
Collapse
Affiliation(s)
- Laura Carrero
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
| | - Desireé Antequera
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Cristina Municio
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Eva Carro
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| |
Collapse
|
11
|
Lozano-Tovar S, Nuccetelli M, Placidi F, Izzi F, Sancesario G, Bernardini S, Biagio Mercuri N, Liguori C. CSF dynamics of orexin and β-amyloid 42 levels in narcolepsy and Alzheimer's disease patients: a controlled study. Neurosci Lett 2024; 837:137914. [PMID: 39032802 DOI: 10.1016/j.neulet.2024.137914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/12/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
β-amyloid42 (Aβ42) in Alzheimer's disease (AD) and orexin in narcolepsy are considered crucial biomarkers for diagnosis and therapeutic targets. Recently, orexin and Aβ cerebral dynamics have been studied in both pathologies, but how they interact with each other remains further to be known. In this study, we investigated the reliability of using the correlation between orexin-A and Aβ42 CSF levels as a candidate marker to explain the chain of events leading to narcolepsy or AD pathology. In order to test the correlation between these biomarkers, patients diagnosed with AD (n = 76), narcolepsy type 1 (NT1, n = 17), narcolepsy type 2 (NT2, n = 23) and healthy subjects (n = 91) were examined. Patients and healthy subjects underwent lumbar puncture between 8:00 and 10:00 am at the Neurology Unit of the University Hospital of Rome "Tor Vergata". CSF levels of Aβ42, total-tau, phosphorylated-tau, and orexin-A were assessed. The results showed that CSF levels of Aβ42 were significantly lower (p < 0.001) in AD (332.28 ± 237.36 pg/mL) compared to NT1 (569.88 ± 187.00 pg/mL), NT2 (691.00 ± 292.63 pg/mL) and healthy subjects (943.68 ± 198.12 pg/mL). CSF orexin-A levels were statistically different (p < 0.001) between AD (148.01 ± 29.49 pg/mL), NT1 (45.94 ± 13.63 pg/mL), NT2 (104.92 ± 25.55 pg/mL) and healthy subjects (145.18 ± 27.01 pg/mL). Moderate-severe AD patients (mini mental state examination < 21) showed the highest CSF orexin-A levels, whereas NT1 patients showed the lowest CSF orexin-A levels. Correlation between CSF levels of Aβ42 and orexin-A was found only in healthy subjects (r = 0.26; p = 0.01), and not in narcolepsy or AD patients. This lack of correlation in both diseases may be explained by the pathology itself since the correlation between these two biomarkers is evident only in the healthy subjects. This study adds to the present literature by further documenting the interplay between orexinergic neurotransmission and cerebral Aβ dynamics, possibly sustained by sleep.
Collapse
Affiliation(s)
- Susana Lozano-Tovar
- Facultad de Psicología, Universidad Nacional Autónoma de México (UNAM), Circuito Ciudad Universitaria Avenida, C.U., Mexico City 04510, Mexico
| | - Marzia Nuccetelli
- Department of Clinical Biochemistry and Molecular Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Fabio Placidi
- 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
| | - Francesca Izzi
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome "Tor Vergata", 00133 Rome, Italy
| | - Giuseppe Sancesario
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Sergio Bernardini
- Department of Clinical Biochemistry and Molecular Biology, University of Rome "Tor Vergata", 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
| | - 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
|
12
|
Stankeviciute L, Chhatwal JP, Levin R, Pinilla V, Schultz AP, Redline S, Johnson KA, Sperling RA, Kozhemiako N, Purcell S, Djonlagic I. Amyloid beta-independent sleep markers associated with early regional tau burden and cortical thinning. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12616. [PMID: 39077684 PMCID: PMC11284643 DOI: 10.1002/dad2.12616] [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: 01/26/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Sleep is crucial for memory consolidation and the clearance of toxic proteins associated with Alzheimer's disease (AD). We examined the association between sleep characteristics and imaging biomarkers of early amyloid beta (Aβ) and tau pathology as well as neurodegeneration in brain regions known to be affected in the incipient stages of AD. METHODS Thirty-nine cognitively unimpaired (CU) participants of the Harvard Aging Brain Study underwent at-home polysomnography as well as tau positron emission tomography (flortaucipir-PET), amyloid PET (Pittsburgh compound B [PiB]-PET), and magnetic resonance imaging-derived assessment of cortical thickness (CT). RESULTS Increased N1 sleep was associated with a higher tau PET signal (β = 0.009, p = 0.001) and lower CT in the temporal composite region of interest (β = -0.017, p = 0.007). Decreased slow-wave sleep (SWS) was associated with higher tau burden in the temporal composite (β = -0.008, p = 0.005) and lower CT (β = 0.008, p = 0.002), even after controlling for global PiB-PET. DISCUSSION In CU older adults, lower SWS and higher N1 sleep were associated with higher tau burden and lower CT in brain regions associated with early tau deposition and vulnerable to AD-related neurodegeneration through mechanisms dissociable from amyloid deposition. Highlights We report the results of an observational study, which leveraged -a well-characterized cohort of healthy aging (Harvard Aging Brain Study) by adding in-home full polysomnograms.By adding at-home polysomnograms to this unique and deeply phenotyped cohort, we examined variations in sleep architecture that are associated with Alzheimer's disease (AD) pathologic changes.Our results confirmed the association of sleep changes with early tau and cortical neurodegenerative changes that were independent of amyloid.The results will be of importance in monitoring sleep-related variations in relation to the natural history of AD pathology and in designing sleep-focused clinical trials.
Collapse
Affiliation(s)
- Laura Stankeviciute
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - Jasmeer P. Chhatwal
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Raina Levin
- Massachusetts General HospitalBostonMassachusettsUSA
| | | | - Aaron P. Schultz
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Susan Redline
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Keith A. Johnson
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Reisa A. Sperling
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Nataliia Kozhemiako
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Shaun Purcell
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Ina Djonlagic
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| |
Collapse
|
13
|
Cushing SD, Moseley SC, Stimmell AC, Schatschneider C, Wilber AA. Rescuing impaired hippocampal-cortical interactions and spatial reorientation learning and memory during sleep in a mouse model of Alzheimer's disease using hippocampal 40 Hz stimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599921. [PMID: 38979221 PMCID: PMC11230253 DOI: 10.1101/2024.06.20.599921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
In preclinical Alzheimer's disease (AD), spatial learning and memory is impaired. We reported similar impairments in 3xTg-AD mice on a virtual maze (VM) spatial-reorientation-task that requires using landmarks to navigate. Hippocampal (HPC)-cortical dysfunction during sleep (important for memory consolidation) is a potential mechanism for memory impairments in AD. We previously found deficits in HPC-cortical coordination during sleep coinciding with VM impairments the next day. Some forms of 40 Hz stimulation seem to clear AD pathology in mice, and improve functional connectivity in AD patients. Thus, we implanted a recording array targeting parietal cortex (PC) and HPC to assess HPC-PC coordination, and an optical fiber targeting HPC for 40 Hz or sham optogenetic stimulation in 3xTg/PV cre mice. We assessed PC delta waves (DW) and HPC sharp wave ripples (SWRs). In sham mice, SWR-DW cross-correlations were reduced, similar to 3xTg-AD mice. In 40 Hz mice, this phase-locking was rescued, as was performance on the VM. However, rescued HPC-PC coupling no longer predicted performance as in NonTg animals. Instead, DWs and SWRs independently predicted performance in 40 Hz mice. Thus, 40 Hz stimulation of HPC rescued functional interactions in the HPC-PC network, and rescued impairments in spatial navigation, but did not rescue the correlation between HPC-PC coordination during sleep and learning and memory. Together this pattern of results could inform AD treatment timing by suggesting that despite applying 40 Hz stimulation before significant tau and amyloid aggregation, pathophysiological processes led to brain changes that were not fully reversed even though cognition was recovered. Significance Statement One of the earliest symptoms of Alzheimer's disease (AD) is getting lost in space or experiencing deficits in spatial navigation, which involve navigation computations as well as learning and memory. We investigated cross brain region interactions supporting memory formation as a potential causative factor of impaired spatial learning and memory in AD. To assess this relationship between AD pathophysiology, brain changes, and behavioral alterations, we used a targeted approach for clearing amyloid beta and tau to rescue functional interactions in the brain. This research strongly connects brain activity patterns during sleep to tau and amyloid accumulation, and will aid in understanding the mechanisms underlying cognitive dysfunction in AD. Furthermore, the results offer insight for improving early identification and treatment strategies.
Collapse
|
14
|
Canet G, Monteiro FDG, Rocaboy E, Diego-Diaz S, Khelaifia B, Kim J, Valencia D, Yin A, Wu HT, Howell J, Blank E, Laliberté F, Fortin N, Boscher E, Fereydouni-Forouzandeh P, Champagne S, Guisle I, Hébert S, Pernet V, Liu H, Lu W, Debure L, Rapoport D, Ayappa I, Varga A, Parekh A, Osorio R, Lacroix S, Lucey B, Blessing E, Planel E. Sleep-wake body temperature regulates tau secretion in mice and correlates with CSF and plasma tau in humans. RESEARCH SQUARE 2024:rs.3.rs-4384494. [PMID: 38798432 PMCID: PMC11118695 DOI: 10.21203/rs.3.rs-4384494/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The sleep-wake cycle regulates interstitial fluid and cerebrospinal fluid (CSF) tau levels in both mouse and human by mechanisms that remain unestablished. Here, we reveal a novel pathway by which wakefulness increases extracellular tau levels in mouse and humans. In mice, higher body temperature (BT) associated with wakefulness and sleep deprivation increased CSF tau. In vitro, wakefulness temperatures upregulated tau secretion via a temperature-dependent increase in activity and expression of unconventional protein secretion pathway-1 components, namely caspase-3-mediated C-terminal cleavage of tau (TauC3), and membrane expression of PIP2 and syndecan-3. In humans, the increase in both CSF and plasma tau levels observed post-wakefulness correlated with BT increase during wakefulness. Our findings suggest sleep-wake variation in BT may contribute to regulating extracellular tau levels, highlighting the importance of thermoregulation in pathways linking sleep disturbance to neurodegeneration, and the potential for thermal intervention to prevent or delay tau-mediated neurodegeneration.
Collapse
Affiliation(s)
| | | | - Emma Rocaboy
- Research Center of CHU de Quebec - Laval University
| | | | | | - Jessica Kim
- Department of Psychiatry, NYU Grossman School of Medicine
| | - Daphne Valencia
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai
| | - Audrey Yin
- Department of Psychiatry, NYU Grossman School of Medicine
| | - Hau-Tieng Wu
- Department of Psychiatry, NYU Grossman School of Medicine
| | - Jordan Howell
- Department of Psychiatry, NYU Grossman School of Medicine
| | - Emily Blank
- Department of Psychiatry, NYU Grossman School of Medicine
| | | | - Nadia Fortin
- Research Center of CHU de Quebec - Laval University
| | - Emmanuelle Boscher
- Centre de recherche du CHU de Québec-Université Laval, CHUL, Axe Neurosciences, Faculté de médecine, Département de psychiatrie et de neurosciences, Québec, C
| | | | | | | | - Sébastien Hébert
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, Québec
| | | | | | - William Lu
- Department of Neurology, Washington University School of Medicine
| | | | - David Rapoport
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai
| | - Indu Ayappa
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai
| | - Andrew Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai
| | - Ankit Parekh
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai
| | | | | | - Brendan Lucey
- Department of Neurology, Washington University School of Medicine
| | | | - Emmanuel Planel
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, Québec
| |
Collapse
|
15
|
Sato T, Ochiishi T, Higo-Yamamoto S, Oishi K. Circadian and sleep phenotypes in a mouse model of Alzheimer's disease characterized by intracellular accumulation of amyloid β oligomers. Exp Anim 2024; 73:186-192. [PMID: 38092387 PMCID: PMC11091359 DOI: 10.1538/expanim.23-0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/06/2023] [Indexed: 05/08/2024] Open
Abstract
Disturbances in sleep-wake and circadian rhythms may reportedly precede the onset of cognitive symptoms in the early stages of Alzheimer's disease (AD); however, the underlying mechanisms of these AD-induced sleep disturbances remain unelucidated. To specifically evaluate the involvement of amyloid beta (Aβ) oligomers in AD-induced sleep disturbances, we examined circadian and sleep phenotypes using an Aβ-GFP transgenic (Aβ-GFP Tg) mouse characterized by intracellular accumulation of Aβ oligomers. The circadian rhythm and free-running period of wheel running activity were identical between Aβ-GFP Tg and littermate wild-type mice. The durations of rapid eye movement (REM) sleep were elongated in Aβ-GFP Tg mice; however, the durations of non-REM sleep and wakefulness were unaffected. The Aβ-GFP Tg mice exhibited shifts in the electroencephalogram (EEG) power spectra toward higher frequencies in the inactive light phase. These findings suggest that the intracellular accumulation of Aβ oligomers might be associated with sleep quality; however, its impact on circadian systems is limited.
Collapse
Affiliation(s)
- Tomoyuki Sato
- Healthy Food Science Research Group, Cellular, and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Tomoyo Ochiishi
- Molecular Neurobiology Research Group, Biomedical Research Institute (BMRI), National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Sayaka Higo-Yamamoto
- Healthy Food Science Research Group, Cellular, and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Katsutaka Oishi
- Healthy Food Science Research Group, Cellular, and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-0882, Japan
- Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
16
|
Mergenthaler P, Balami JS, Neuhaus AA, Mottahedin A, Albers GW, Rothwell PM, Saver JL, Young ME, Buchan AM. Stroke in the Time of Circadian Medicine. Circ Res 2024; 134:770-790. [PMID: 38484031 DOI: 10.1161/circresaha.124.323508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/15/2024] [Indexed: 03/19/2024]
Abstract
Time-of-day significantly influences the severity and incidence of stroke. Evidence has emerged not only for circadian governance over stroke risk factors, but also for important determinants of clinical outcome. In this review, we provide a comprehensive overview of the interplay between chronobiology and cerebrovascular disease. We discuss circadian regulation of pathophysiological mechanisms underlying stroke onset or tolerance as well as in vascular dementia. This includes cell death mechanisms, metabolism, mitochondrial function, and inflammation/immunity. Furthermore, we present clinical evidence supporting the link between disrupted circadian rhythms and increased susceptibility to stroke and dementia. We propose that circadian regulation of biochemical and physiological pathways in the brain increase susceptibility to damage after stroke in sleep and attenuate treatment effectiveness during the active phase. This review underscores the importance of considering circadian biology for understanding the pathology and treatment choice for stroke and vascular dementia and speculates that considering a patient's chronotype may be an important factor in developing precision treatment following stroke.
Collapse
Affiliation(s)
- Philipp Mergenthaler
- Center for Stroke Research Berlin (P.M., A.M.B.), Charité - Universitätsmedizin Berlin, Germany
- Department of Neurology with Experimental Neurology (P.M.), Charité - Universitätsmedizin Berlin, Germany
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Joyce S Balami
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Ain A Neuhaus
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, United Kingdom (A.A.N.)
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Amin Mottahedin
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Nuffield Department of Clinical Neurosciences (A.M., P.M.R.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Gregory W Albers
- Department of Neurology, Stanford Hospital, Palo Alto, CA (G.W.A.)
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Peter M Rothwell
- Nuffield Department of Clinical Neurosciences (A.M., P.M.R.), University of Oxford, United Kingdom
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences (P.M.R.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Jeffrey L Saver
- Department of Neurology and Comprehensive Stroke Center, Geffen School of Medicine, University of Los Angeles, CA (J.L.S.)
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham (M.E.Y.)
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| | - Alastair M Buchan
- Center for Stroke Research Berlin (P.M., A.M.B.), Charité - Universitätsmedizin Berlin, Germany
- Stroke Research, Radcliffe Department of Medicine (P.M., J.S.B., A.A.N., A.M., A.M.B.), University of Oxford, United Kingdom
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA) (P.M., J.S.B., A.A.N., A.M., G.W.A., P.M.R., J.L.S., M.E.Y., A.M.B.)
| |
Collapse
|
17
|
Alghanimy A, Work LM, Holmes WM. The glymphatic system and multiple sclerosis: An evolving connection. Mult Scler Relat Disord 2024; 83:105456. [PMID: 38266608 DOI: 10.1016/j.msard.2024.105456] [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: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder that affects the central nervous system, resulting in demyelination and an array of neurological manifestations. Recently, there has been significant scientific interest in the glymphatic system, which operates as a waste-clearance system for the brain. This article reviews the existing literature, and explores potential links between the glymphatic system and MS, shedding light on its evolving significance in the context of MS pathogenesis. The authors consider the pathophysiological implications of glymphatic dysfunction in MS, the impact of disrupted sleep on glymphatic function, and the bidirectional relationship between MS and sleep disturbances. By offering an understanding of the intricate interplay between the glymphatic system and MS, this review provides valuable insights which may lead to improved diagnostic techniques and more effective therapeutic interventions.
Collapse
Affiliation(s)
- Alaa Alghanimy
- School of Psychology and Neuroscience, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow G61 1QH, United Kingdom; Radiological Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Lorraine M Work
- School of Cardiovascular and Metabolic Health, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - William M Holmes
- School of Psychology and Neuroscience, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow G61 1QH, United Kingdom
| |
Collapse
|
18
|
Martin SC, Joyce KK, Harper KM, Harp SJ, Cohen TJ, Moy SS, Diering GH. Evaluating Fatty Acid Amide Hydrolase as a Suitable Target for Sleep Promotion in a Transgenic TauP301S Mouse Model of Neurodegeneration. Pharmaceuticals (Basel) 2024; 17:319. [PMID: 38543105 PMCID: PMC10975243 DOI: 10.3390/ph17030319] [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: 01/22/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 04/01/2024] Open
Abstract
Sleep disruption is an expected component of aging and neurodegenerative conditions, including Alzheimer's disease (AD). Sleep disruption has been demonstrated as a driver of AD pathology and cognitive decline. Therefore, treatments designed to maintain sleep may be effective in slowing or halting AD progression. However, commonly used sleep aid medications are associated with an increased risk of AD, highlighting the need for sleep aids with novel mechanisms of action. The endocannabinoid system holds promise as a potentially effective and novel sleep-enhancing target. By using pharmacology and genetic knockout strategies, we evaluated fatty acid amide hydrolase (FAAH) as a therapeutic target to improve sleep and halt disease progression in a transgenic Tau P301S (PS19) model of Tauopathy and AD. We have recently shown that PS19 mice exhibit sleep disruption in the form of dark phase hyperarousal as an early symptom that precedes robust Tau pathology and cognitive decline. Acute FAAH inhibition with PF3845 resulted in immediate improvements in sleep behaviors in male and female PS19 mice, supporting FAAH as a potentially suitable sleep-promoting target. Moreover, sustained drug dosing for 5-10 days resulted in maintained improvements in sleep. To evaluate the effect of chronic FAAH inhibition as a possible therapeutic strategy, we generated FAAH-/- PS19 mice models. Counter to our expectations, FAAH knockout did not protect PS19 mice from progressive sleep loss, neuroinflammation, or cognitive decline. Our results provide support for FAAH as a novel target for sleep-promoting therapies but further indicate that the complete loss of FAAH activity may be detrimental.
Collapse
Affiliation(s)
- Shenée C. Martin
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathryn K. Joyce
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathryn M. Harper
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Samuel J. Harp
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Todd J. Cohen
- Department of Neurology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sheryl S. Moy
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, Carrboro, NC 27510, USA
| | - Graham H. Diering
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, Carrboro, NC 27510, USA
| |
Collapse
|
19
|
Cortes-Flores H, Torrandell-Haro G, Brinton RD. Association between CNS-active drugs and risk of Alzheimer's and age-related neurodegenerative diseases. Front Psychiatry 2024; 15:1358568. [PMID: 38487578 PMCID: PMC10937406 DOI: 10.3389/fpsyt.2024.1358568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/08/2024] [Indexed: 03/17/2024] Open
Abstract
Objective As neuropsychiatric conditions can increase the risk of age-related neurodegenerative diseases (NDDs), the impact of CNS-active drugs on the risk of developing Alzheimer's Disease (AD), non-AD dementia, Multiple Sclerosis (MS), Parkinson's Disease (PD) and Amyotrophic Lateral Sclerosis (ALS) was investigated. Research design and methods A retrospective cohort analysis of a medical claims dataset over a 10 year span was conducted in patients aged 60 years or older. Participants were propensity score matched for comorbidity severity and demographic parameters. Relative risk (RR) ratios and 95% confidence intervals (CI) were determined for age-related NDDs. Cumulative hazard ratios and treatment duration were determined to assess the association between CNS-active drugs and NDDs at different ages and treatment duration intervals. Results In 309,128 patients who met inclusion criteria, exposure to CNS-active drugs was associated with a decreased risk of AD (0.86% vs 1.73%, RR: 0.50; 95% CI: 0.47-0.53; p <.0001) and all NDDs (3.13% vs 5.76%, RR: 0.54; 95% CI: 0.53-0.56; p <.0001). Analysis of impact of drug class on risk of AD indicated that antidepressant, sedative, anticonvulsant, and stimulant medications were associated with significantly reduced risk of AD whereas atypical antipsychotics were associated with increased AD risk. The greatest risk reduction for AD and NDDs occurred in patients aged 70 years or older with a protective effect only in patients with long-term therapy (>3 years). Furthermore, responders to these therapeutics were characterized by diagnosed obesity and higher prescriptions of anti-inflammatory drugs and menopausal hormonal therapy, compared to patients with a diagnosis of AD (non-responders). Addition of a second CNS-active drug was associated with greater reduction in AD risk compared to monotherapy, with the combination of a Z-drug and an SNRI associated with greatest AD risk reduction. Conclusion Collectively, these findings indicate that CNS-active drugs were associated with reduced risk of developing AD and other age-related NDDs. The exception was atypical antipsychotics, which increased risk. Potential use of combination therapy with atypical antipsychotics could mitigate the risk conferred by these drugs. Evidence from these analyses advance precision prevention strategies to reduce the risk of age-related NDDs in persons with neuropsychiatric disorders.
Collapse
Affiliation(s)
- Helena Cortes-Flores
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Georgina Torrandell-Haro
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- Department of Neurology, University of Arizona College of Medicine, Tucson, AZ, United States
| |
Collapse
|
20
|
Samudra N, Lerner H, Yack L, Walsh CM, Kirsch HE, Kudo K, Yballa C, La Joie R, Gorno‐Tempini ML, Spina S, Seeley WW, Neylan TC, Miller BL, Rabinovici GD, Boxer A, Grinberg LT, Rankin KP, Nagarajan SS, Ranasinghe KG. Spatiotemporal characteristics of neurophysiological changes in patients with four-repeat tauopathies. Ann Clin Transl Neurol 2024; 11:525-535. [PMID: 38226843 PMCID: PMC10863921 DOI: 10.1002/acn3.51974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 01/17/2024] Open
Abstract
INTRODUCTION Progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD), are the most common four-repeat tauopathies (4RT), and both frequently occur with varying degree of Alzheimer's disease (AD) copathology. Intriguingly, patients with 4RT and patients with AD are at opposite ends of the wakefulness spectrum-AD showing reduced wakefulness and excessive sleepiness whereas 4RT showing decreased homeostatic sleep. The neural mechanisms underlying these distinct phenotypes in the comorbid condition of 4RT and AD are unknown. The objective of the current study was to define the alpha oscillatory spectrum, which is prominent in the awake resting-state in the human brain, in patients with primary 4RT, and how it is modified in comorbid AD-pathology. METHOD In an autopsy-confirmed case series of 4R-tauopathy patients (n = 10), whose primary neuropathological diagnosis was either PSP (n = 7) or CBD (n = 3), using high spatiotemporal resolution magnetoencephalography (MEG), we quantified the spectral power density within alpha-band (8-12 Hz) and examined how this pattern was modified in increasing AD-copathology. For each patient, their regional alpha power was compared to an age-matched normative control cohort (n = 35). RESULT Patients with 4RT showed increased alpha power but in the presence of AD-copathology alpha power was reduced. CONCLUSIONS Alpha power increase in PSP-tauopathy and reduction in the presence of AD-tauopathy is consistent with the observation that neurons activating wakefulness-promoting systems are preserved in PSP but degenerated in AD. These results highlight the selectively vulnerable impacts in 4RT versus AD-tauopathy that may have translational significance on disease-modifying therapies for specific proteinopathies.
Collapse
Affiliation(s)
- Niyatee Samudra
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Hannah Lerner
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Leslie Yack
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
- Department of PsychiatrySan Francisco Veterans Affairs, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Christine M. Walsh
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Heidi E. Kirsch
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCalifornia94143USA
- Epilepsy Center, Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Kiwamu Kudo
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCalifornia94143USA
- Medical Imaging Business CenterRicoh CompanyKanazawaJapan
| | - Claire Yballa
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Renaud La Joie
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Maria L. Gorno‐Tempini
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Salvatore Spina
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - William W. Seeley
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Thomas C. Neylan
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
- Department of PsychiatrySan Francisco Veterans Affairs, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Bruce L. Miller
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Gil D. Rabinovici
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCalifornia94143USA
| | - Adam Boxer
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Lea T. Grinberg
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
- Department of PathologyUniversity of CaliforniaSan FranciscoCalifornia94158USA
- Department of PathologyUniversity of Sao Paulo Medical SchoolSao PauloBrazil
| | - Katherine P. Rankin
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| | - Srikantan S. Nagarajan
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCalifornia94143USA
| | - Kamalini G. Ranasinghe
- Memory and Aging Center, Department of NeurologyWeill Institute for Neurosciences, University of California San FranciscoSan FranciscoCalifornia94158USA
| |
Collapse
|
21
|
Rapaport P, Amador S, Adeleke M, Banerjee S, Barber J, Charlesworth G, Clarke C, Connell C, Espie C, Gonzalez L, Horsley R, Hunter R, Kyle SD, Manela M, Morris S, Pikett L, Raczek M, Thornton E, Walker Z, Webster L, Livingston G. Clinical and cost-effectiveness of DREAMS START (Dementia RElAted Manual for Sleep; STrAtegies for RelaTives) for people living with dementia and their carers: a study protocol for a parallel multicentre randomised controlled trial. BMJ Open 2024; 14:e075273. [PMID: 38307536 PMCID: PMC10836385 DOI: 10.1136/bmjopen-2023-075273] [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: 05/02/2023] [Accepted: 01/19/2024] [Indexed: 02/04/2024] Open
Abstract
INTRODUCTION Many people living with dementia experience sleep disturbance and there are no known effective treatments. Non-pharmacological treatment options should be the first-line sleep management. For family carers, relatives' sleep disturbance leads to interruption of their sleep, low mood and breakdown of care. Our team developed and delivered DREAMS START (Dementia RElAted Manual for Sleep; STrAtegies for RelaTives), a multimodal non-pharmacological intervention, showing it to be feasible and acceptable. The aim of this randomised controlled trial is to establish whether DREAMS START is clinically cost-effective in reducing sleep disturbances in people living with dementia living at home compared with usual care. METHODS AND ANALYSIS We will recruit 370 participant dyads (people living with dementia and family carers) from memory services, community mental health teams and the Join Dementia Research Website in England. Those meeting inclusion criteria will be randomised (1:1) either to DREAMS START or to usual treatment. DREAMS START is a six-session (1 hour/session), manualised intervention delivered every 1-2 weeks by supervised, non-clinically trained graduates. Outcomes will be collected at baseline, 4 months and 8 months with the primary outcome being the Sleep Disorders Inventory score at 8 months. Secondary outcomes for the person with dementia (all proxy) include quality of life, daytime sleepiness, neuropsychiatric symptoms and cost-effectiveness. Secondary outcomes for the family carer include quality of life, sleep disturbance, mood, burden and service use and caring/work activity. Analyses will be intention-to-treat and we will conduct a process evaluation. ETHICS AND DISSEMINATION London-Camden & Kings Cross Ethics Committee (20/LO/0894) approved the study. We will disseminate our findings in high-impact peer-reviewed journals and at national and international conferences. This research has the potential to improve sleep and quality of life for people living with dementia and their carers, in a feasible and scalable intervention. TRIAL REGISTRATION NUMBER ISRCTN13072268.
Collapse
Affiliation(s)
- Penny Rapaport
- Division of Psychiatry, University College London, London, UK
| | - Sarah Amador
- Division of Psychiatry, University College London, London, UK
| | - Mariam Adeleke
- Department of Statistical Science, University College London, London, UK
| | - Sube Banerjee
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Julie Barber
- Department of Statistical Science, University College London, London, UK
| | - Georgina Charlesworth
- Division of Psychology and Language Sciences, University College London, London, UK
- North East London NHS Foundation Trust, Rainham, UK
| | | | | | - Colin Espie
- Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, UK
| | - Lina Gonzalez
- Research Department of Primary Care and Population Health, University College London, London, UK
| | | | - Rachael Hunter
- Research Department of Primary Care and Population Health, University College London, London, UK
| | - Simon D Kyle
- Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, UK
| | - Monica Manela
- Division of Psychiatry, University College London, London, UK
| | - Sarah Morris
- Tees Esk and Wear Valleys NHS Foundation Trust, Darlington, UK
| | - Liam Pikett
- Division of Psychiatry, University College London, London, UK
| | - Malgorzata Raczek
- Centre for Dementia Studies, Brighton and Sussex Medical School, Brighton, UK
| | - Emma Thornton
- Tees Esk and Wear Valleys NHS Foundation Trust, Darlington, UK
| | - Zuzana Walker
- Division of Psychiatry, University College London, London, UK
| | - Lucy Webster
- Division of Psychiatry, University College London, London, UK
| | - Gill Livingston
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
22
|
Kron JOZJ, Keenan RJ, Hoyer D, Jacobson LH. Orexin Receptor Antagonism: Normalizing Sleep Architecture in Old Age and Disease. Annu Rev Pharmacol Toxicol 2024; 64:359-386. [PMID: 37708433 DOI: 10.1146/annurev-pharmtox-040323-031929] [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: 09/16/2023]
Abstract
Sleep is essential for human well-being, yet the quality and quantity of sleep reduce as age advances. Older persons (>65 years old) are more at risk of disorders accompanied and/or exacerbated by poor sleep. Furthermore, evidence supports a bidirectional relationship between disrupted sleep and Alzheimer's disease (AD) or related dementias. Orexin/hypocretin neuropeptides stabilize wakefulness, and several orexin receptor antagonists (ORAs) are approved for the treatment of insomnia in adults. Dysregulation of the orexin system occurs in aging and AD, positioning ORAs as advantageous for these populations. Indeed, several clinical studies indicate that ORAs are efficacious hypnotics in older persons and dementia patients and, as in adults, are generally well tolerated. ORAs are likely to be more effective when administered early in sleep/wake dysregulation to reestablish good sleep/wake-related behaviors and reduce the accumulation of dementia-associated proteinopathic substrates. Improving sleep in aging and dementia represents a tremendous opportunity to benefit patients, caregivers, and health systems.
Collapse
Affiliation(s)
- Jarrah O-Z J Kron
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
| | - Ryan J Keenan
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Daniel Hoyer
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia;
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Laura H Jacobson
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia;
| |
Collapse
|
23
|
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
|
24
|
Picard K, Dolhan K, Watters JJ, Tremblay MÈ. Microglia and Sleep Disorders. ADVANCES IN NEUROBIOLOGY 2024; 37:357-377. [PMID: 39207702 DOI: 10.1007/978-3-031-55529-9_20] [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: 09/04/2024]
Abstract
Sleep is a physiological state that is essential for maintaining physical and mental health. Sleep disorders and sleep deprivation therefore have many adverse effects, including an increased risk of metabolic diseases and a decline in cognitive function that may be implicated in the long-term development of neurodegenerative diseases. There is increasing evidence that microglia, the resident immune cells of the central nervous system (CNS), are involved in regulating the sleep-wake cycle and the CNS response to sleep alteration and deprivation. In this chapter, we will discuss the involvement of microglia in various sleep disorders, including sleep-disordered breathing, insomnia, narcolepsy, myalgic encephalomyelitis/chronic fatigue syndrome, and idiopathic rapid-eye-movement sleep behavior disorder. We will also explore the impact of acute and chronic sleep deprivation on microglial functions. Moreover, we will look into the potential involvement of microglia in sleep disorders as a comorbidity to Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Katherine Picard
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Kira Dolhan
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Department of Psychology, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | - Jyoti J Watters
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI, USA
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
25
|
Johnson CE, Duncan MJ, Murphy MP. Sex and Sleep Disruption as Contributing Factors in Alzheimer's Disease. J Alzheimers Dis 2024; 97:31-74. [PMID: 38007653 PMCID: PMC10842753 DOI: 10.3233/jad-230527] [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] [Indexed: 11/27/2023]
Abstract
Alzheimer's disease (AD) affects more women than men, with women throughout the menopausal transition potentially being the most under researched and at-risk group. Sleep disruptions, which are an established risk factor for AD, increase in prevalence with normal aging and are exacerbated in women during menopause. Sex differences showing more disrupted sleep patterns and increased AD pathology in women and female animal models have been established in literature, with much emphasis placed on loss of circulating gonadal hormones with age. Interestingly, increases in gonadotropins such as follicle stimulating hormone are emerging to be a major contributor to AD pathogenesis and may also play a role in sleep disruption, perhaps in combination with other lesser studied hormones. Several sleep influencing regions of the brain appear to be affected early in AD progression and some may exhibit sexual dimorphisms that may contribute to increased sleep disruptions in women with age. Additionally, some of the most common sleep disorders, as well as multiple health conditions that impair sleep quality, are more prevalent and more severe in women. These conditions are often comorbid with AD and have bi-directional relationships that contribute synergistically to cognitive decline and neuropathology. The association during aging of increased sleep disruption and sleep disorders, dramatic hormonal changes during and after menopause, and increased AD pathology may be interacting and contributing factors that lead to the increased number of women living with AD.
Collapse
Affiliation(s)
- Carrie E. Johnson
- University of Kentucky, College of Medicine, Department of Molecular and Cellular Biochemistry, Lexington, KY, USA
| | - Marilyn J. Duncan
- University of Kentucky, College of Medicine, Department of Neuroscience, Lexington, KY, USA
| | - M. Paul Murphy
- University of Kentucky, College of Medicine, Department of Molecular and Cellular Biochemistry, Lexington, KY, USA
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY, USA
| |
Collapse
|
26
|
Keil SA, Schindler AG, Wang MX, Piantino J, Silbert LC, Elliott JE, Werhane ML, Thomas RG, Willis S, Lim MM, Iliff JJ. Longitudinal Sleep Patterns and Cognitive Impairment in Older Adults. JAMA Netw Open 2023; 6:e2346006. [PMID: 38048131 PMCID: PMC10696486 DOI: 10.1001/jamanetworkopen.2023.46006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/22/2023] [Indexed: 12/05/2023] Open
Abstract
Importance Sleep disturbances and clinical sleep disorders are associated with all-cause dementia and neurodegenerative conditions, but it remains unclear how longitudinal changes in sleep impact the incidence of cognitive impairment. Objective To evaluate the association of longitudinal sleep patterns with age-related changes in cognitive function in healthy older adults. Design, Setting, and Participants This cross-sectional study is a retrospective longitudinal analyses of the Seattle Longitudinal Study (SLS), which evaluated self-reported sleep duration (1993-2012) and cognitive performance (1997-2020) in older adults. Participants within the SLS were enrolled as part of a community-based cohort from the Group Health Cooperative of Puget Sound and Health Maintenance Organization of Washington between 1956 and 2020. Data analysis was performed from September 2020 to May 2023. Main Outcomes and Measures The main outcome for this study was cognitive impairment, as defined by subthreshold performance on both the Mini-Mental State Examination and the Mattis Dementia Rating Scale. Sleep duration was defined by self-report of median nightly sleep duration over the last week and was assessed longitudinally over multiple time points. Median sleep duration, sleep phenotype (short sleep, median ≤7 hours; medium sleep, median = 7 hour; long sleep, median ≥7 hours), change in sleep duration (slope), and variability in sleep duration (SD of median sleep duration, or sleep variability) were evaluated. Results Of the participants enrolled in SLS, only 1104 participants who were administered both the Health Behavior Questionnaire and the neuropsychologic battery were included for analysis in this study. A total of 826 individuals (mean [SD] age, 76.3 [11.8] years; 468 women [56.7%]; 217 apolipoprotein E ε4 allele carriers [26.3%]) had complete demographic information and were included in the study. Analysis using a Cox proportional hazard regression model (concordance, 0.76) showed that status as a short sleeper (hazard ratio, 3.67; 95% CI, 1.59-8.50) and higher sleep variability (hazard ratio, 3.06; 95% CI, 1.14-5.49) were significantly associated with the incidence of cognitive impairment. Conclusions and Relevance In this community-based longitudinal study of the association between sleep patterns and cognitive performance, the short sleep phenotype was significantly associated with impaired cognitive performance. Furthermore, high sleep variability in longitudinal sleep duration was significantly associated with the incidence of cognitive impairment, highlighting the possibility that instability in sleep duration over long periods of time may impact cognitive decline in older adults.
Collapse
Affiliation(s)
- Samantha A Keil
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, New York
| | - Abigail G Schindler
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle
- Geriatric Research Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, Washington
- Gerontology Division, Department of Medicine, University of Washington School of Medicine, Seattle
| | - Marie X Wang
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle
- Now with Seagen, Inc, Bothell, Washington
| | - Juan Piantino
- Department of Pediatrics, Oregon Health & Science University, Portland
- Department of Neurology, Oregon Health & Science University, Portland
| | - Lisa C Silbert
- Department of Neurology, Oregon Health & Science University, Portland
- Neurology Service, VA Portland Health Care System, Portland, Oregon
- Oregon Alzheimer's Disease Research Center, Oregon Health & Science University, Portland
| | - Jonathan E Elliott
- Department of Neurology, Oregon Health & Science University, Portland
- Research Service, VA Portland Health Care System, Portland, Oregon
| | - Madeleine L Werhane
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington
| | - Ronald G Thomas
- School of Public Health, University of California, San Diego
| | - Sherry Willis
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle
| | - Miranda M Lim
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington
- Department of Neurology, Oregon Health & Science University, Portland
- Neurology Service, VA Portland Health Care System, Portland, Oregon
- Oregon Alzheimer's Disease Research Center, Oregon Health & Science University, Portland
- Oregon Institute of Occupational Health Sciences, Portland
| | - Jeffrey J Iliff
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle
- Department of Neurology, University of Washington School of Medicine, Seattle
| |
Collapse
|
27
|
Devulder A, Macea J, Kalkanis A, De Winter F, Vandenbulcke M, Vandenberghe R, Testelmans D, Van Den Bossche MJA, Van Paesschen W. Subclinical epileptiform activity and sleep disturbances in Alzheimer's disease. Brain Behav 2023; 13:e3306. [PMID: 37950422 PMCID: PMC10726840 DOI: 10.1002/brb3.3306] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 11/12/2023] Open
Abstract
INTRODUCTION Subclinical epileptiform activity (SEA) and sleep disturbances are frequent in Alzheimer's disease (AD). Both have an important relation to cognition and potential therapeutic implications. We aimed to study a possible relationship between SEA and sleep disturbances in AD. METHODS In this cross-sectional study, we performed a 24-h ambulatory EEG and polysomnography in 48 AD patients without diagnosis of epilepsy and 34 control subjects. RESULTS SEA, mainly detected in frontotemporal brain regions during N2 with a median of three spikes/night [IQR1-17], was three times more prevalent in AD. AD patients had lower sleep efficacy, longer wake after sleep onset, more awakenings, more N1%, less REM sleep and a higher apnea-hypopnea index (AHI) and oxygen desaturation index (ODI). Sleep was not different between AD subgroup with SEA (AD-Epi+) and without SEA (AD-Epi-); however, compared to controls, REM% was decreased and AHI and ODI were increased in the AD-Epi+ subgroup. DISCUSSION Decreased REM sleep and more severe sleep-disordered breathing might be related to SEA in AD. These results could have diagnostic and therapeutic implications and warrant further study at the intersection between sleep and epileptiform activity in AD.
Collapse
Affiliation(s)
- Astrid Devulder
- Laboratory for Epilepsy Research, KU Leuven and Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Jaiver Macea
- Laboratory for Epilepsy Research, KU Leuven and Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Alexandros Kalkanis
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven and Department of Pulmonary DiseasesUniversity Hospitals LeuvenLeuvenBelgium
| | - François‐Laurent De Winter
- Division of Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven and Department of Geriatric PsychiatryUniversity Psychiatric Center (UPC) KU LeuvenLeuvenBelgium
| | - Mathieu Vandenbulcke
- Division of Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven and Department of Geriatric PsychiatryUniversity Psychiatric Center (UPC) KU LeuvenLeuvenBelgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, KU Leuven and Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Dries Testelmans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven and Department of Pulmonary DiseasesUniversity Hospitals LeuvenLeuvenBelgium
| | - Maarten J. A. Van Den Bossche
- Division of Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven and Department of Geriatric PsychiatryUniversity Psychiatric Center (UPC) KU LeuvenLeuvenBelgium
| | - Wim Van Paesschen
- Laboratory for Epilepsy Research, KU Leuven and Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| |
Collapse
|
28
|
Levendowski DJ, Neylan TC, Walsh CM, Tsuang D, Salat D, Hamilton JM, Lee-Iannotti JK, Berka C, Mazeika G, Boeve BF, St. Louis EK. Proof-of-concept for characterization of neurodegenerative disorders utilizing two non-REM sleep biomarkers. Front Neurol 2023; 14:1272369. [PMID: 37928153 PMCID: PMC10623683 DOI: 10.3389/fneur.2023.1272369] [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: 08/03/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Study objective This proof-of-concept study aimed to determine whether the combined features of two non-rapid eye movement (NREM) sleep biomarkers acquired predominantly in-home could characterize different neurodegenerative disorders. Methods Sleep spindle duration and non-REM hypertonia (NRH) were evaluated in seven groups including a control group (CG = 61), and participants with isolated REM sleep behavior disorder (iRBD = 19), mild cognitive impairment (MCI = 41), Parkinson disease (PD = 16), Alzheimer disease dementia (ADem = 29), dementia with Lewy Bodies or Parkinson disease dementia (LBD = 19) and progressive supranuclear palsy (PSP = 13). One-way analysis of variance (ANOVA), Mann-Whitney U, intra-class (ICC) and Spearman ranked correlations, Bland-Altman plots and Kappa scores, Chi-square and Fisher exact probability test, and multiple-logistic regression were focused primarily on spindle duration and NRH and the frequencies assigned to the four normal/abnormal spindle duration/NRH combinations. Results ANOVA identified group differences in age, sleep efficiency, REM, NRH (p < 0.0001) and sleep time (p = 0.015), Spindle duration and NRH each demonstrated good night-to-night reliabilities (ICC = 0.95 and 0.75, Kappa = 0.93 and 0.66, respectively) and together exhibited an association in the PD and LBD groups only (p < 0.01). Abnormal spindle duration was greater in records of PSP (85%) and LBD (84%) patients compared to CG, MCI, PD and ADem (p < 0.025). Abnormal NRH was greater in PSP = 92%, LBD = 79%, and iRBD = 74% compared to MCI = 32%, ADem = 17%, and CG = 16% (p < 0.005).The combination biomarker normal spindle duration/normal NRH was observed most frequently in CG (56%) and MCI (41%). ADem most frequently demonstrated normal spindle duration/normal NRH (45%) and abnormal spindle duration/normal NRH (38%). Normal spindle duration/abnormal NRH was greatest in iRBD = 47%, while abnormal spindle duration/abnormal NRH was predominant in PSP = 85% and LBD = 74%. Conclusion The NREM sleep biomarkers spindle duration and NRH may be useful in distinguishing patients with different neurodegenerative disorders. Larger prospective cohort studies are needed to determine whether spindle duration and NRH can be combined for prodromal assessment and/or monitoring disease progression.
Collapse
Affiliation(s)
| | - Thomas C. Neylan
- UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Christine M. Walsh
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Debby Tsuang
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States
| | - David Salat
- Massachusetts General Hospital, Charlestown, MA, United States
| | | | | | - Chris Berka
- Advanced Brain Monitoring, Inc., Carlsbad, CA, United States
| | - Gandis Mazeika
- Advanced Brain Monitoring, Inc., Carlsbad, CA, United States
| | - Bradley F. Boeve
- Department of Neurology and Center for Sleep Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Erik K. St. Louis
- Department of Neurology and Center for Sleep Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
29
|
Whittaker DS, Akhmetova L, Carlin D, Romero H, Welsh DK, Colwell CS, Desplats P. Circadian modulation by time-restricted feeding rescues brain pathology and improves memory in mouse models of Alzheimer's disease. Cell Metab 2023; 35:1704-1721.e6. [PMID: 37607543 PMCID: PMC10591997 DOI: 10.1016/j.cmet.2023.07.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 06/12/2023] [Accepted: 07/27/2023] [Indexed: 08/24/2023]
Abstract
Circadian disruptions impact nearly all people with Alzheimer's disease (AD), emphasizing both their potential role in pathology and the critical need to investigate the therapeutic potential of circadian-modulating interventions. Here, we show that time-restricted feeding (TRF) without caloric restriction improved key disease components including behavioral timing, disease pathology, hippocampal transcription, and memory in two transgenic (TG) mouse models of AD. We found that TRF had the remarkable capability of simultaneously reducing amyloid deposition, increasing Aβ42 clearance, improving sleep and memory, and normalizing daily transcription patterns of multiple genes, including those associated with AD and neuroinflammation. Thus, our study unveils for the first time the pleiotropic nature of timed feeding on AD, which has far-reaching effects beyond metabolism, ameliorating neurodegeneration and the misalignment of circadian rhythmicity. Since TRF can substantially modify disease trajectory, this intervention has immediate translational potential, addressing the urgent demand for accessible approaches to reduce or halt AD progression.
Collapse
Affiliation(s)
- Daniel S Whittaker
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Laila Akhmetova
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Daniel Carlin
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - Haylie Romero
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA
| | - David K Welsh
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Paula Desplats
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA, USA; Department of Pathology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
30
|
Keil SA, Schindler AG, Wang MX, Piantino J, Silbert LC, Elliott JE, Thomas RG, Willis S, Lim MM, Iliff JJ. Instability in longitudinal sleep duration predicts cognitive impairment in aged participants of the Seattle Longitudinal Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.07.23291098. [PMID: 37398317 PMCID: PMC10312848 DOI: 10.1101/2023.06.07.23291098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Importance Sleep disturbances and clinical sleep disorders are associated with all-cause dementia and neurodegenerative conditions. It remains unclear how longitudinal changes in sleep impact the incidence of cognitive impairment. Objective To evaluate how longitudinal sleep patterns contribute to age-related changes in cognitive function in healthy adults. Design Setting Participants This study utilizes retrospective longitudinal analyses of a community-based study within Seattle, evaluating self-reported sleep (1993-2012) and cognitive performance (1997-2020) in aged adults. Main Outcomes and Measures The main outcome is cognitive impairment as defined by sub-threshold performance on 2 of 4 neuropsychological batteries: Mini-Mental State Examination (MMSE), Mattis Dementia Rating Scale, Trail Making Test, and Wechsler Adult Intelligent Scale (Revised). Sleep duration was defined through self-report of 'average nightly sleep duration over the last week' and assessed longitudinally. Median sleep duration, change in sleep duration (slope), variability in sleep duration (standard deviation, Sleep Variability), and sleep phenotype ("Short Sleep" median ≤7hrs.; "Medium Sleep" median = 7hrs; "Long Sleep" median ≥7hrs.). Results A total of 822 individuals (mean age of 76.2 years [11.8]; 466 women [56.7%]; 216 APOE allele positive [26.3%]) were included in the study. Analysis using a Cox Proportional Hazard Regression model (concordance 0.70) showed that increased Sleep Variability (95% CI [1.27,3.86]) was significantly associated with the incidence of cognitive impairment. Further analysis using linear regression prediction analysis (R2=0.201, F (10, 168)=6.010, p=2.67E-07) showed that high Sleep Variability (β=0.3491; p=0.048) was a significant predictor of cognitive impairment over a 10-year period. Conclusions and Relevance High variability in longitudinal sleep duration was significantly associated with the incidence of cognitive impairment and predictive of decline in cognitive performance ten years later. These data highlight that instability in longitudinal sleep duration may contribute to age-related cognitive decline.
Collapse
Affiliation(s)
- Samantha A Keil
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA
| | - Abigail G Schindler
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA
- Geriatric Research Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA
- Gerontology Division Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Marie X Wang
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA
| | - Juan Piantino
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Lisa C Silbert
- Neurology Service VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Jonathan E Elliott
- Research Service VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Ronald G Thomas
- School of Public Health, University of California, San Diego, San Diego, CA
| | - Sherry Willis
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA
| | - Miranda M Lim
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA
- Neurology Service VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Department of Medicine, Oregon Health & Science University, Portland, OR
- Oregon Institute of Occupational Health Sciences, Portland, OR
| | - Jeffrey J Iliff
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
31
|
Turrini S, Wong B, Eldaief M, Press DZ, Sinclair DA, Koch G, Avenanti A, Santarnecchi E. The multifactorial nature of healthy brain ageing: Brain changes, functional decline and protective factors. Ageing Res Rev 2023; 88:101939. [PMID: 37116664 DOI: 10.1016/j.arr.2023.101939] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023]
Abstract
As the global population faces a progressive shift towards a higher median age, understanding the mechanisms underlying healthy brain ageing has become of paramount importance for the preservation of cognitive abilities. The first part of the present review aims to provide a comprehensive look at the anatomical changes the healthy brain endures with advanced age, while also summarizing up to date findings on modifiable risk factors to support a healthy ageing process. Subsequently, we describe the typical cognitive profile displayed by healthy older adults, conceptualizing the well-established age-related decline as an impairment of four main cognitive factors and relating them to their neural substrate previously described; different cognitive trajectories displayed by typical Alzheimer's Disease patients and successful agers with a high cognitive reserve are discussed. Finally, potential effective interventions and protective strategies to promote cognitive reserve and defer cognitive decline are reviewed and proposed.
Collapse
Affiliation(s)
- Sonia Turrini
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Centro studi e ricerche in Neuroscienze Cognitive, Dipartimento di Psicologia "Renzo Canestrari", Alma Mater Studiorum Università di Bologna, Campus di Cesena, Cesena, Italy
| | - Bonnie Wong
- Neuropsychology Program, Frontotemporal Disorders Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA , USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark Eldaief
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel Z Press
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David A Sinclair
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of ageing Research, Harvard Medical School, Boston, MA, USA
| | - Giacomo Koch
- Stroke Unit, Department of Systems Medicine, University of Tor Vergata, Rome, Italy; Department of Clinical and Behavioural Neurology, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Alessio Avenanti
- Centro studi e ricerche in Neuroscienze Cognitive, Dipartimento di Psicologia "Renzo Canestrari", Alma Mater Studiorum Università di Bologna, Campus di Cesena, Cesena, Italy; Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Universidad Católica del Maule, Talca, Chile
| | - Emiliano Santarnecchi
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Sugiura S, Yokoyama S, Inoue K, Okada S. Dementia Scale Classification with Sequential Model from Sleep Activity Data. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-5. [PMID: 38082717 DOI: 10.1109/embc40787.2023.10340400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Dementia, a disorder caused by brain diseases, has been found to influence the sleep patterns of patients. The finding indicates that monitoring sleep activity is helpful to detect the change in cognitive status. With this in mind, the aim of this study is to explore the possibility to develop a machine learning model for classifying the scores of dementia screening tests based on sleep activity data which could be recorded with less burden for participants. In this study, We collected sleep activity data from 124 elderly patients with varying cognitive states, including heart rate, respiratory rate and depth of sleep, using a single sensor. The score of Mini Mental State Estimation (MMSE) cognitive test is used to determine the level of cognitive states. First, we conducted a statistical analysis of the measured sleep activity data to find specific features observed in people with low-MMSE scores. Second, we utilized an efficient sequence model for capturing time-series changes in sleep activity for binary classification of the dementia scale to detect such low-MMSE people. Our findings revealed significant distinctions in sleep patterns between high and low cognitive status groups, and in the classification task, a maximum macro F1 score of 0.67 was achieved using LSTM models. Our results suggest the validity of using sleep activity data for the prediction of dementia classification.
Collapse
|
33
|
Drew VJ, Park M, Kim T. GABA-Positive Astrogliosis in Sleep-Promoting Areas Associated with Sleep Disturbance in 5XFAD Mice. Int J Mol Sci 2023; 24:9695. [PMID: 37298646 PMCID: PMC10253883 DOI: 10.3390/ijms24119695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Sleep disturbances, a debilitating symptom of Alzheimer's disease (AD), are associated with neuropathological changes. However, the relationship between these disturbances and regional neuron and astrocyte pathologies remains unclear. This study examined whether sleep disturbances in AD result from pathological changes in sleep-promoting brain areas. Male 5XFAD mice underwent electroencephalography (EEG) recordings at 3, 6, and 10 months, followed by an immunohistochemical analysis of three brain regions associated with sleep promotion. The findings showed that 5XFAD mice demonstrated reduced duration and bout counts of nonrapid eye movement (NREM) sleep by 6 months and reduced duration and bout counts of rapid eye movement (REM) sleep by 10 months. Additionally, peak theta EEG power frequency during REM sleep decreased by 10 months. Sleep disturbances correlated with the total number of GFAP-positive astrocytes and the ratio of GFAP- and GABA-positive astrocytes across all three sleep-associated regions corresponding to their roles in sleep promotion. The presence of GABRD in sleep-promoting neurons indicated their susceptibility to inhibition by extrasynaptic GABA. This study reveals that neurotoxic reactive astrogliosis in NREM and REM sleep-promoting areas is linked to sleep disturbances in 5XFAD mice, which suggests a potential target for the treatment of sleep disorders in AD.
Collapse
Affiliation(s)
| | | | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea; (V.J.D.); (M.P.)
| |
Collapse
|
34
|
Drew VJ, Wang C, Kim T. Progressive sleep disturbance in various transgenic mouse models of Alzheimer's disease. Front Aging Neurosci 2023; 15:1119810. [PMID: 37273656 PMCID: PMC10235623 DOI: 10.3389/fnagi.2023.1119810] [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: 12/09/2022] [Accepted: 04/24/2023] [Indexed: 06/06/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia. The relationship between AD and sleep dysfunction has received increased attention over the past decade. The use of genetically engineered mouse models with enhanced production of amyloid beta (Aβ) or hyperphosphorylated tau has played a critical role in the understanding of the pathophysiology of AD. However, their revelations regarding the progression of sleep impairment in AD have been highly dependent on the mouse model used and the specific techniques employed to examine sleep. Here, we discuss the sleep disturbances and general pathology of 15 mouse models of AD. Sleep disturbances covered in this review include changes to NREM and REM sleep duration, bout lengths, bout counts and power spectra. Our aim is to describe in detail the severity and chronology of sleep disturbances within individual mouse models of AD, as well as reveal broader trends of sleep deterioration that are shared among most models. This review also explores a variety of potential mechanisms relating Aβ accumulation and tau neurofibrillary tangles to the progressive deterioration of sleep observed in AD. Lastly, this review offers perspective on how study design might impact our current understanding of sleep disturbances in AD and provides strategies for future research.
Collapse
Affiliation(s)
- Victor J. Drew
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Chanung Wang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
35
|
Brady ES, Griffiths J, Andrianova L, Bielska M, Saito T, Saido TC, Randall AD, Tamagnini F, Witton J, Craig MT. Alterations to parvalbumin-expressing interneuron function and associated network oscillations in the hippocampal - medial prefrontal cortex circuit during natural sleep in App NL-G-F/NL-G-F mice. Neurobiol Dis 2023; 182:106151. [PMID: 37172910 DOI: 10.1016/j.nbd.2023.106151] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023] Open
Abstract
In the early stages of Alzheimer's disease (AD), the accumulation of the peptide amyloid-β (Aβ) damages synapses and disrupts neuronal activity, leading to the disruption of neuronal oscillations associated with cognition. This is thought to be largely due to impairments in CNS synaptic inhibition, particularly via parvalbumin (PV)-expressing interneurons that are essential for generating several key oscillations. Research in this field has largely been conducted in mouse models that over-express humanised, mutated forms of AD-associated genes that produce exaggerated pathology. This has prompted the development and use of knock-in mouse lines that express these genes at an endogenous level, such as the AppNL-G-F/NL-G-F mouse model used in the present study. These mice appear to model the early stages of Aβ-induced network impairments, yet an in-depth characterisation of these impairments in currently lacking. Therefore, using 16 month-old AppNL-G-F/NL-G-F mice, we analysed neuronal oscillations found in the hippocampus and medial prefrontal cortex (mPFC) during awake behaviour, rapid eye movement (REM) and non-REM (NREM) sleep to assess the extent of network dysfunction. No alterations to gamma oscillations were found to occur in the hippocampus or mPFC during either awake behaviour, REM or NREM sleep. However, during NREM sleep an increase in the power of mPFC spindles and decrease in the power of hippocampal sharp-wave ripples was identified. The latter was accompanied by an increase in the synchronisation of PV-expressing interneuron activity, as measured using two-photon Ca2+ imaging, as well as a decrease in PV-expressing interneuron density. Furthermore, although changes were detected in local network function of mPFC and hippocampus, long-range communication between these regions appeared intact. Altogether, our results suggest that these NREM sleep-specific impairments represent the early stages of circuit breakdown in response to amyloidopathy.
Collapse
Affiliation(s)
- Erica S Brady
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; Gladstone Institute for Neurological Disease, 1650 Owens Street, San Francisco, CA 91458, United States of America
| | - Jessica Griffiths
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK
| | - Lilya Andrianova
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Monika Bielska
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Andrew D Randall
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - Francesco Tamagnini
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK
| | - Jonathan Witton
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK.
| | - Michael T Craig
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK.
| |
Collapse
|
36
|
Ahmad F, Sachdeva P, Sarkar J, Izhaar R. Circadian dysfunction and Alzheimer's disease - An updated review. Aging Med (Milton) 2023; 6:71-81. [PMID: 36911088 PMCID: PMC10000289 DOI: 10.1002/agm2.12221] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/15/2022] [Accepted: 08/01/2022] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease (AD) is considered to be the most typical form of dementia that provokes irreversible cognitive impairment. Along with cognitive impairment, circadian rhythm dysfunction is a fundamental factor in aggravating AD. A link among circadian rhythms, sleep, and AD has been well-documented. The etiopathogenesis of circadian system disruptions and AD serves some general characteristics that also open up the possibility of viewing them as a mutually reliant path. In this review, we have focused on different factors that are related to circadian rhythm dysfunction. The various pathogenic factors, such as amyloid-beta, neurofibrillary tangles, oxidative stress, neuroinflammation, and circadian rhythm dysfunction may all contribute to AD. In this review, we also tried to focus on melatonin which is produced from the pineal gland and can be used to treat circadian dysfunction in AD. Aside from amyloid beta, tau pathology may have a notable influence on sleep. Conclusively, the center of this review is primarily based on the principal mechanistic complexities associated with circadian rhythm disruption, sleep deprivation, and AD, and it also emphasizes the potential therapeutic strategies to treat and prevent the progression of AD.
Collapse
Affiliation(s)
- Faizan Ahmad
- Department of Medical Elementology and ToxicologyJamia Hamdard UniversityDelhiIndia
| | - Punya Sachdeva
- Amity Institute of Neuropsychology and NeurosciencesAmity UniversityNoidaUttar PradeshIndia
| | - Jasmine Sarkar
- Amity Institute of Neuropsychology and NeurosciencesAmity UniversityNoidaUttar PradeshIndia
| | | |
Collapse
|
37
|
Si Y, Chen J, Shen Y, Kubra S, Mei B, Qin ZS, Pan B, Meng B. Circadian rhythm sleep disorders and time-of-day-dependent memory deficiency in Presenilin1/2 conditional knockout mice with long noncoding RNA expression profiling changes. Sleep Med 2023; 103:146-158. [PMID: 36805914 DOI: 10.1016/j.sleep.2023.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Alzheimer's disease (AD) patients exhibit sleep and circadian disturbances prior to the onset of cognitive decline, and these disruptions worsen with disease severity. However, the molecular mechanisms behind sleep and circadian disruptions in AD patients are poorly understood. In this study, we investigated sleep pattern and circadian rhythms in Presenilin-1/2 conditional knockout (DKO) mice. Assessment of EEG and EMG recordings showed that DKO mice displayed increased NREM sleep time but not REM sleep during the dark phase compared to WT mice at the age of two months; at the age of six months, the DKO mice showed increased wakefulness periods and decreased total time spent in both NREM and REM sleep. WT exhibited time-of-day dependent modulation of contextual and cued memory. Compared with WT mice, 4-month-old DKO mice exhibited the deficiency regardless trained and tested in the same light/night phase or not. Particularly interesting was that DKO showed circadian modulation deficiency when trained in the resting period but not in the active period. Long noncoding RNAs (lncRNAs) are typically defined as transcripts longer than 200 nucleotides, and they have rhythmic expression in mammals. To date no study has investigated rhythmic lncRNA expression in Alzheimer's disease. We applied RNA-seq technology to profile hippocampus expression of lncRNAs in DKO mice during the light (/resting) and dark (/active) phases and performed gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses of the cis lncRNA targets. Expression alteration of lncRNAs associated with immune response and metallodipeptidase activity may contribute to the circadian disruptions of DKO mice. Especially we identified some LncRNAs which expression change oppositely between day and light in DKO mice compared to WT mice and are worthy to be studied further. Our results exhibited the circadian rhythm sleep disorders and a noteworthy time-of-day-dependent memory deficiency in AD model mice and provide a useful resource for studying the expression and function of lncRNAs during circadian disruptions in Alzheimer's disease.
Collapse
Affiliation(s)
- Youwen Si
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Jing Chen
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Yang Shen
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, United States.
| | - Syeda Kubra
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Bing Mei
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Zhaohui S Qin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, United States.
| | - Boxi Pan
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China.
| | - Bo Meng
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
38
|
Abulafia C, Vidal MF, Olivar N, Odzak A, Brusco I, Guinjoan SM, Cardinali DP, Vigo DE. An Exploratory Study of Sleep-Wake Differences of Autonomic Activity in Patients with Mild Cognitive Impairment: The Role of Melatonin as a Modulating Factor. Clin Interv Aging 2023; 18:771-781. [PMID: 37200894 PMCID: PMC10187579 DOI: 10.2147/cia.s394749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 05/08/2023] [Indexed: 05/20/2023] Open
Abstract
Purpose The objective of the present study was to assess sleep-wake differences of autonomic activity in patients with mild cognitive impairment (MCI) compared to control subjects. As a post-hoc objective, we sought to evaluate the mediating effect of melatonin on this association. Patients and Methods A total of 22 MCI patients (13 under melatonin treatment) and 12 control subjects were included in this study. Sleep-wake periods were identified by actigraphy and 24hr-heart rate variability measures were obtained to study sleep-wake autonomic activity. Results MCI patients did not show any significant differences in sleep-wake autonomic activity when compared to control subjects. Post-hoc analyses revealed that MCI patients not taking melatonin displayed lower parasympathetic sleep-wake amplitude than controls not taking melatonin (RMSSD -7 ± 1 vs 4 ± 4, p = 0.004). In addition, we observed that melatonin treatment was associated with greater parasympathetic activity during sleep (VLF 15.5 ± 0.1 vs 15.1 ± 0.1, p = 0.010) and in sleep-wake differences in MCI patients (VLF 0.5 ± 0.1 vs 0.2 ± 0.0, p = 0.004). Conclusion These preliminary findings hint at a possible sleep-related parasympathetic vulnerability in patients at prodromal stages of dementia as well as a potential protective effect of exogenous melatonin in this population.
Collapse
Affiliation(s)
- Carolina Abulafia
- Laboratory of Chronophysiology, Institute for Biomedical Research (BIOMED), Pontifical Catholic University of Argentina (UCA) and CONICET, Buenos Aires, Argentina
- Facultad de Psicología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María F Vidal
- Servicio de Psiquiatría, Departamento de Neurología, Fleni, Buenos Aires, Argentina
| | - Natividad Olivar
- Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Andrea Odzak
- Servicio de Clínica Médica, Hospital Argerich, Buenos Aires, Argentina
| | - Ignacio Brusco
- Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Buenos Aires, Argentina
- Servicio de Clínica Médica, Hospital Argerich, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | | | - Daniel P Cardinali
- Facultad de Ciencias Médicas, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Daniel E Vigo
- Laboratory of Chronophysiology, Institute for Biomedical Research (BIOMED), Pontifical Catholic University of Argentina (UCA) and CONICET, Buenos Aires, Argentina
- Faculty of Psychology and Educational Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
- Correspondence: Daniel E Vigo, Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Alicia Moreau de Justo 1500, 4° piso, Buenos Aires, C1107AAZ, Argentina, Tel +54 0810-2200-822 ext 1152, Email ;
| |
Collapse
|
39
|
Prediction of Cognitive Decline by Behavioral Symptoms in Neuropsychiatric Disorders. IRANIAN JOURNAL OF PSYCHIATRY AND BEHAVIORAL SCIENCES 2022. [DOI: 10.5812/ijpbs-126596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background: Neuropsychiatric disorders are described by their neurological, behavioral, and cognitive symptoms. However, behavioral symptoms may often be overlooked due to the current approach in neurology. Objectives: This study investigated the relationship between behavioral symptoms and cognitive functioning in neurological disorders. The second aim was to predict neurocognitive patterns by behavioral symptoms as independent variables. Methods: Behavioral symptoms were collected based on semi-structured neuropsychiatric interviews with 211 patients admitted to the neuropsychiatry department of Ayatollah Kashani hospital in Isfahan by both a neuropsychiatry fellow and an attending neuropsychiatrist. A neuropsychiatry fellow assessed all patients using the neuropsychiatry unit cognitive (NUCog) assessment tool. We used a generalized linear model (GLM) to indicate the effect of behavioral symptoms on the risk of decline in cognitive domains. Due to the use of all available samples, this study had no age limit, and the patients were 15 to 92 years old. Results: The regression coefficient of NUCog subscale scores for behavioral symptoms using GLM revealed that education level had a positive relationship with the scores of attention (P < 0.001), visuoconstruction (P < 0.001), memory (P < 0.001), executive function (P < 0.001), language (P < 0.001), and the total score of NUCog (P < 0.001). Patients with apathy had lower scores on the memory subscale (P = 0.002) and total NUCog (P = 0.021). Similarly, patients with delusion had lower scores on memory (P = 0.006) and executive function (P = 0.026). There was a negative relationship between agitation and attention (P = 0.049), visuoconstruction (P = 0.015), memory (P = 0.018), executive function (P = 0.005), and total score of NUCog (P = 0.007). Sleep disturbances were accompanied by lower memory scores (P = 0.056) and lower mean NUCog scores (P = 0.052). Visual hallucination was associated with declined performance in attention (P = 0.057). Conclusions: Behavioral assessment can help predict cognitive patterns in patients with neurobehavioral syndromes.
Collapse
|
40
|
Iacobelli P. Circadian dysregulation and Alzheimer’s disease: A comprehensive review. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Alzheimer’s disease (AD), the foremost variant of dementia, has been associated with a menagerie of risk factors, many of which are considered to be modifiable. Among these modifiable risk factors is circadian rhythm, the chronobiological system that regulates sleep‐wake cycles, food consumption timing, hydration timing, and immune responses amongst many other necessary physiological processes. Circadian rhythm at the level of the suprachiasmatic nucleus (SCN), is tightly regulated in the human body by a host of biomolecular substances, principally the hormones melatonin, cortisol, and serotonin. In addition, photic information projected along afferent pathways to the SCN and peripheral oscillators regulates the synthesis of these hormones and mediates the manner in which they act on the SCN and its substructures. Dysregulation of this cycle, whether induced by environmental changes involving irregular exposure to light, or through endogenous pathology, will have a negative impact on immune system optimization and will heighten the deposition of Aβ and the hyperphosphorylation of the tau protein. Given these correlations, it appears that there is a physiologic association between circadian rhythm dysregulation and AD. This review will explore the physiology of circadian dysregulation in the AD brain, and will propose a basic model for its role in AD‐typical pathology, derived from the literature compiled and referenced throughout.
Collapse
Affiliation(s)
- Peter Iacobelli
- Department of Arts and Sciences, University of South Carolina, Columbia, USA
| |
Collapse
|
41
|
Chen WC, Wang XY. Longitudinal associations between sleep duration and cognitive impairment in Chinese elderly. Front Aging Neurosci 2022; 14:1037650. [PMID: 36466606 PMCID: PMC9714471 DOI: 10.3389/fnagi.2022.1037650] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/25/2022] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Age-associated cognitive decline has become a major threat to both personal welfare and public health and can further develop into Dementia/Alzheimer's disease. Sleep is significantly correlated with cognitive function, but both cognitive impairment and sleep problems increase with normal aging. This study explored how sleep duration affects cognitive performance among older adults in China. METHODS Using data from the Chinese Longitudinal Healthy Longevity Survey (CLHLS) in 2014 and 2018, cognitive function was assessed via the Mini-Mental State Examination (MMSE), which included five domains: orientation, registration, attention or calculation, recall, and language. Logistic regression was used to examine whether the change in sleep duration was a risk factor for cognitive impairment. We also used multinomial logistic regression to study the impact of sleep duration and the changes in sleep duration on cognitive changes during the follow-up period. RESULTS The empirical study showed a U-shaped relationship between sleep duration and increased risk of cognitive impairment. Short (< 6 hours) and long (> 8 hours) sleep durations were positively associated with cognitive impairment. Tests of interactions between sleep duration and sleep quality showed that short sleep durations with fair sleep quality had an increased risk of cognitive impairment. Further, the participants were divided into three groups: normal cognition (MMSE > 24), mild cognitive impairment (MCI, 18 ≤ MMSE score ≤ 24), and severe cognitive impairment (MMSE < 18). First, of the participants with normal cognition at baseline, those who sleeping > 7 h at follow-up and > 7 h at both baseline and 4-year follow-up assessments could increase the risk of cognitive impairment. Second, for individuals with MCI at baseline, those who transitioned to sleeping > 7 h at follow-up period and > 7 h at both baseline and 4-year follow-up assessments had a lower chance of reverting to normal cognition. CONCLUSION Excessive sleep may be a major risk for cognitive impairment among older adults. Furthermore, a moderate amount of sleep could be a possible strategy to prevent cognitive impairment.
Collapse
Affiliation(s)
- Wei-chao Chen
- School of Journalism and Communication, Hunan Normal University, Changsha, China
| | - Xiao-yan Wang
- College of Finance and Statistics, Hunan University, Changsha, China
| |
Collapse
|
42
|
Liu C, Lee SH, Loewenstein DA, Galvin JE, Camargo CJ, Alperin N. Poor sleep accelerates hippocampal and posterior cingulate volume loss in cognitively normal healthy older adults. J Sleep Res 2022; 31:e13538. [PMID: 34927298 PMCID: PMC10731580 DOI: 10.1111/jsr.13538] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 11/12/2021] [Accepted: 12/03/2021] [Indexed: 01/05/2023]
Abstract
Poor sleep quality is a known risk factor for Alzheimer's disease. This longitudinal imaging study aimed to determine the acceleration in the rates of tissue loss in cognitively critical brain regions due to poor sleep in healthy elderly individuals. Cognitively-normal healthy individuals, aged ≥60 years, reported Pittsburgh Sleep Quality Index (PSQI) and underwent baseline and 2-year follow-up magnetic resonance imaging brain scans. The links between self-reported sleep quality, rates of tissue loss in cognitively-critical brain regions, and white matter hyperintensity load were assessed. A total of 48 subjects were classified into normal (n = 23; PSQI score <5) and poor sleepers (n = 25; PSQI score ≥5). The two groups were not significantly different in terms of age, gender, years of education, ethnicity, handedness, body mass index, and cognitive performance. Compared to normal sleepers, poor sleepers exhibited much faster rates of volume loss, over threefold in the right hippocampus and fivefold in the right posterior cingulate over 2 years. In contrast, there were no significant differences in the rates of volume loss in the cerebral and cerebellar grey and white matter between the two groups. Rates of volume loss in the right posterior cingulate were negatively associated with global PSQI scores. Poor sleep significantly accelerates volume loss in the right hippocampus and the right posterior cingulate cortex. These findings demonstrate that self-reported sleep quality explains inter-individual differences in the rates of volume loss in cognitively-critical brain regions in healthy older adults and provide a strong impetus to offer sleep interventions to cognitively normal older adults who are poor sleepers.
Collapse
Affiliation(s)
- Che Liu
- Department of Radiology, University of Miami Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Sang H. Lee
- Department of Radiology, University of Miami Miller School of Medicine, University of Miami, Miami, FL, USA
| | - David A. Loewenstein
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - James E. Galvin
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christian J. Camargo
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Noam Alperin
- Department of Radiology, University of Miami Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| |
Collapse
|
43
|
Roberts R, Wall MJ, Braren I, Dhillon K, Evans A, Dunne J, Nyakupinda S, Huckstepp RTR. An Improved Model of Moderate Sleep Apnoea for Investigating Its Effect as a Comorbidity on Neurodegenerative Disease. Front Aging Neurosci 2022; 14:861344. [PMID: 35847678 PMCID: PMC9278434 DOI: 10.3389/fnagi.2022.861344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
Sleep apnoea is a highly prevalent disease that often goes undetected and is associated with poor clinical prognosis, especially as it exacerbates many different disease states. However, most animal models of sleep apnoea (e.g., intermittent hypoxia) have recently been dispelled as physiologically unrealistic and are often unduly severe. Owing to a lack of appropriate models, little is known about the causative link between sleep apnoea and its comorbidities. To overcome these problems, we have created a more realistic animal model of moderate sleep apnoea by reducing the excitability of the respiratory network. This has been achieved through controlled genetically mediated lesions of the preBötzinger complex (preBötC), the inspiratory oscillator. This novel model shows increases in sleep disordered breathing with alterations in breathing during wakefulness (decreased frequency and increased tidal volume) as observed clinically. The increase in dyspnoeic episodes leads to reduction in REM sleep, with all lost active sleep being spent in the awake state. The increase in hypoxic and hypercapnic insults induces both systemic and neural inflammation. Alterations in neurophysiology, an inhibition of hippocampal long-term potentiation (LTP), is reflected in deficits in both long- and short-term spatial memory. This improved model of moderate sleep apnoea may be the key to understanding why this disorder has such far-reaching and often fatal effects on end-organ function.
Collapse
Affiliation(s)
- Reno Roberts
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Mark J. Wall
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Ingke Braren
- University Medical Center Eppendorf, Vector Facility, Institute for Experimental Pharmacology and Toxikology, Hamburg, Germany
| | - Karendeep Dhillon
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Amy Evans
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Jack Dunne
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | | | - Robert T. R. Huckstepp
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
- *Correspondence: Robert T. R. Huckstepp
| |
Collapse
|
44
|
Depressive history as a major predictor of postmenopausal depressive symptoms. Menopause 2022; 29:763-764. [PMID: 35728014 DOI: 10.1097/gme.0000000000002027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Law AD, Cassar M, Long DM, Chow ES, Giebultowicz JM, Venkataramanan A, Strauss R, Kretzschmar D. FTD-associated mutations in Tau result in a combination of dominant and recessive phenotypes. Neurobiol Dis 2022; 170:105770. [PMID: 35588988 PMCID: PMC9261467 DOI: 10.1016/j.nbd.2022.105770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 11/26/2022] Open
Abstract
Although mutations in the microtubules-associated protein Tau have long been connected with several neurodegenerative diseases, the underlying molecular mechanisms causing these tauopathies are still not fully understood. Studies in various models suggested that dominant gain-of-function effects underlie the pathogenicity of these mutants; however, there is also evidence that the loss of normal physiological functions of Tau plays a role in tauopathies. Previous studies on Tau in Drosophila involved expressing the human Tau protein in the background of the endogenous Tau gene in addition to inducing high expression levels. To study Tau pathology in more physiological conditions, we recently created Drosophila knock-in models that express either wildtype human Tau (hTauWT) or disease-associated mutant hTau (hTauV337M and hTauK369I) in place of the endogenous Drosophila Tau (dTau). Analyzing these flies as homozygotes, we could therefore detect recessive effects of the mutations while identifying dominant effects in heterozygotes. Using memory, locomotion and sleep assays, we found that homozygous mutant hTau flies showed deficits already when quite young whereas in heterozygous flies, disease phenotypes developed with aging. Homozygotes also revealed an increase in microtubule diameter, suggesting that changes in the cytoskeleton underlie the axonal degeneration we observed in these flies. In contrast, heterozygous mutant hTau flies showed abnormal axonal targeting and no detectable changes in microtubules. However, we previously showed that heterozygosity for hTauV337M interfered with synaptic homeostasis in central pacemaker neurons and we now show that heterozygous hTauK369I flies have decreased levels of proteins involved in the release of synaptic vesicles. Taken together, our results demonstrate that both mutations induce a combination of dominant and recessive disease-related phenotypes that provide behavioral and molecular insights into the etiology of Tauopathies.
Collapse
Affiliation(s)
- Alexander D Law
- Oregon Institute of Occupational Health Sciences, 3181 S.W. Sam Jackson Park Road, Portland, OR 97219, USA
| | - Marlène Cassar
- Oregon Institute of Occupational Health Sciences, 3181 S.W. Sam Jackson Park Road, Portland, OR 97219, USA
| | - Dani M Long
- Oregon Institute of Occupational Health Sciences, 3181 S.W. Sam Jackson Park Road, Portland, OR 97219, USA
| | - Eileen S Chow
- Department of Integrative Biology, Oregon State University, Corvallis, OR 97331, USA
| | | | - Anjana Venkataramanan
- Institut für Entwicklungsbiologie und Neurobiologie, Johannes Gutenberg-Universität Mainz, Hanns-Dieter-Hüsch Weg 15, 55128 Mainz, Germany
| | - Roland Strauss
- Institut für Entwicklungsbiologie und Neurobiologie, Johannes Gutenberg-Universität Mainz, Hanns-Dieter-Hüsch Weg 15, 55128 Mainz, Germany
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, 3181 S.W. Sam Jackson Park Road, Portland, OR 97219, USA.
| |
Collapse
|
46
|
Qian D, Wong J. Sleep disturbances, sleep-disordered breathing, and cognitive impairment in older adults: perioperative implications. Int Anesthesiol Clin 2022; 60:20-26. [PMID: 35261342 DOI: 10.1097/aia.0000000000000359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Dorothy Qian
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jean Wong
- University Health Network, Women's College Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Ge X, Qiao Y, Choi J, Raman R, Ringman JM, Shiand Y. Enhanced Association of Tau Pathology and Cognitive Impairment in Mild Cognitive Impairment Subjects with Behavior Symptoms. J Alzheimers Dis 2022; 87:557-568. [PMID: 35342088 DOI: 10.3233/jad-215555] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mild cognitive impairment (MCI) individuals with neuropsychiatric symptoms (NPS) are more likely to develop dementia. OBJECTIVE We sought to understand the relationship between neuroimaging markers such as tau pathology and cognitive symptoms both with and without the presence of NPS during the prodromal period of Alzheimer's disease. METHODS A total of 151 MCI subjects with tau positron emission tomographic (PET) scanning with 18F AV-1451, amyloid-β (Aβ) PET scanning with florbetapir or florbetaben, magnetic resonance imaging, and cognitive and behavioral evaluations were selected from the Alzheimer's Disease Neuroimaging Initiative. A 4-group division approach was proposed using amyloid (A-/A+) and behavior (B-/B+) status: A-B-, A-B+, A+B-, and A+B+. Pearson's correlation test was conducted for each group to examine the association between tau deposition and cognitive performance. RESULTS No statistically significant association between tau deposition and cognitive impairment was found for subjects without behavior symptoms in either the A-B-or A+B-groups after correction for false discovery rate. In contrast, tau deposition was found to be significantly associated with cognitive impairment in entorhinal cortex and temporal pole for the A-B+ group and nearly the whole cerebrum for the A+B+ group. CONCLUSION Enhanced associations between tauopathy and cognitive impairment are present in MCI subjects with behavior symptoms, which is more prominent in the presence of elevated amyloid pathology. MCI individuals with NPS may thus be at greater risk for further cognitive decline with the increase of tau deposition in comparison to those without NPS.
Collapse
Affiliation(s)
- Xinting Ge
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, China.,School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuchuan Qiao
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jiyoon Choi
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, CA, USA
| | - Rema Raman
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, CA, USA
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yonggang Shiand
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
48
|
Reynolds C, Mattek N, Lim MM, Beattie Z, Dodge HH, Kaye J. Association Between Mild Cognitive Impairment and Seasonal Rest-Activity Patterns of Older Adults. Front Digit Health 2022; 4:809370. [PMID: 35281221 PMCID: PMC8904352 DOI: 10.3389/fdgth.2022.809370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Seasonal variation in rest-activity patterns has been observed in healthy adult populations. This study examined seasonal variation in total time spent overnight in the bedroom by cognitively intact older adults and older adults with mild cognitive impairment (MCI). We hypothesize that seasonal variation in rest-activity patterns is observed in the cognitively intact group and that this variation is disturbed in those with MCI. Study participants were 128 older adults; mean age 85.2 years. Ninety-eight were cognitively intact, and 30 had been diagnosed with MCI. All were enrolled in an ongoing longitudinal study using in-home passive monitoring technology. Infrared presence sensors were placed throughout each participant's home to monitor movement and presence in each room of the home. Activity data was collected from the sensors over a period of up to 527 days. Overnight time in bedroom was found to vary seasonally for the cognitively intact group, with longer times spent overnight in the bedroom during the winter months. This seasonal variation was not observed for those with non-amnestic MCI. MCI is associated with an attenuation of seasonal variation in total time spent in the bedroom at night. Detection of changes in infradian sleep patterns may be an early marker of cognitive decline. Which key determinants are driving these disturbed rhythms, such as features intrinsic to changes in the brain or to environmental factors or external cues, remains an important question for ongoing and future studies.
Collapse
Affiliation(s)
- Christina Reynolds
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- *Correspondence: Christina Reynolds
| | - Nora Mattek
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Oregon Center for Aging and Technology, Oregon Health and Science University, Portland, OR, United States
- National Institute on Aging (NIA)-Layton Aging and Alzheimer's Disease Center, Portland, OR, United States
| | - Miranda M. Lim
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Veterans Affairs (VA) Portland Health Care System, Portland, OR, United States
| | - Zachary Beattie
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Oregon Center for Aging and Technology, Oregon Health and Science University, Portland, OR, United States
- National Institute on Aging (NIA)-Layton Aging and Alzheimer's Disease Center, Portland, OR, United States
| | - Hiroko H. Dodge
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Oregon Center for Aging and Technology, Oregon Health and Science University, Portland, OR, United States
- National Institute on Aging (NIA)-Layton Aging and Alzheimer's Disease Center, Portland, OR, United States
| | - Jeffrey Kaye
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Oregon Center for Aging and Technology, Oregon Health and Science University, Portland, OR, United States
- National Institute on Aging (NIA)-Layton Aging and Alzheimer's Disease Center, Portland, OR, United States
| |
Collapse
|
49
|
Austad SN, Ballinger S, Buford TW, Carter CS, Smith DL, Darley-Usmar V, Zhang J. Targeting whole body metabolism and mitochondrial bioenergetics in the drug development for Alzheimer's disease. Acta Pharm Sin B 2022; 12:511-531. [PMID: 35256932 PMCID: PMC8897048 DOI: 10.1016/j.apsb.2021.06.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Aging is by far the most prominent risk factor for Alzheimer's disease (AD), and both aging and AD are associated with apparent metabolic alterations. As developing effective therapeutic interventions to treat AD is clearly in urgent need, the impact of modulating whole-body and intracellular metabolism in preclinical models and in human patients, on disease pathogenesis, have been explored. There is also an increasing awareness of differential risk and potential targeting strategies related to biological sex, microbiome, and circadian regulation. As a major part of intracellular metabolism, mitochondrial bioenergetics, mitochondrial quality-control mechanisms, and mitochondria-linked inflammatory responses have been considered for AD therapeutic interventions. This review summarizes and highlights these efforts.
Collapse
Key Words
- ACE2, angiotensin I converting enzyme (peptidyl-dipeptidase A) 2
- AD, Alzheimer's disease
- ADP, adenosine diphosphate
- ADRD, AD-related dementias
- Aβ, amyloid β
- CSF, cerebrospinal fluid
- Circadian regulation
- DAMPs
- DAMPs, damage-associated molecular patterns
- Diabetes
- ER, estrogen receptor
- ETC, electron transport chain
- FCCP, trifluoromethoxy carbonylcyanide phenylhydrazone
- FPR-1, formyl peptide receptor 1
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide-1
- HBP, hexoamine biosynthesis pathway
- HTRA, high temperature requirement A
- Hexokinase biosynthesis pathway
- I3A, indole-3-carboxaldehyde
- IRF-3, interferon regulatory factor 3
- LC3, microtubule associated protein light chain 3
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- MAVS, mitochondrial anti-viral signaling
- MCI, mild cognitive impairment
- MRI, magnetic resonance imaging
- MRS, magnetic resonance spectroscopy
- Mdivi-1, mitochondrial division inhibitor 1
- Microbiome
- Mitochondrial DNA
- Mitochondrial electron transport chain
- Mitochondrial quality control
- NLRP3, leucine-rich repeat (LRR)-containing protein (NLR)-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- NeuN, neuronal nuclear protein
- PET, fluorodeoxyglucose (FDG)-positron emission tomography
- PKA, protein kinase A
- POLβ, the base-excision repair enzyme DNA polymerase β
- ROS, reactive oxygen species
- Reactive species
- SAMP8, senescence-accelerated mice
- SCFAs, short-chain fatty acids
- SIRT3, NAD-dependent deacetylase sirtuin-3
- STING, stimulator of interferon genes
- STZ, streptozotocin
- SkQ1, plastoquinonyldecyltriphenylphosphonium
- T2D, type 2 diabetes
- TCA, Tricarboxylic acid
- TLR9, toll-like receptor 9
- TMAO, trimethylamine N-oxide
- TP, tricyclic pyrone
- TRF, time-restricted feeding
- cAMP, cyclic adenosine monophosphate
- cGAS, cyclic GMP/AMP synthase
- hAPP, human amyloid precursor protein
- hPREP, human presequence protease
- i.p., intraperitoneal
- mTOR, mechanistic target of rapamycin
- mtDNA, mitochondrial DNA
- αkG, alpha-ketoglutarate
Collapse
Affiliation(s)
- Steven N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christy S. Carter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
50
|
Shin C, Kim REY, Thomas RJ, Yun CH, Lee SK, Abbott RD. Severity of Daytime Sleepiness and Parkinsonian-Like Symptoms in Korean Adults Aged 50-64 Years. J Clin Neurol 2022; 18:33-40. [PMID: 35021274 PMCID: PMC8762500 DOI: 10.3988/jcn.2022.18.1.33] [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: 02/08/2021] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 11/17/2022] Open
Abstract
Background and Purpose While excessive daytime sleepiness can predate Parkinson’s disease in late-life, its association with parkinsonian-like (P-L) symptoms in middle age are unknown. Since neurodegeneration can appear decades before a diagnosis of Parkinson’s disease, identifying clinical features associated with this early progression is important. The purpose of this study was to determine the association of daytime sleepiness with P-L symptoms in a population-based sample of middle-aged Korean adults. Methods During 2013 and 2014, daytime sleepiness and P-L symptoms were assessed in 2,063 males and females aged 50–64 years who were participating in the Korean Genome and Epidemiology Study. The severity of daytime sleepiness was quantified by the score on the Epworth Sleepiness Scale (ESS). Self-reported P-L symptoms included nine motor disorders commonly associated with Parkinson’s disease. Participants with parkinsonism and related conditions are excluded. Results The prevalence of excessive daytime sleepiness (ESS score >10) was 7.0%. The frequencies of P-L symptoms ranged from 0.5% (for “trouble buttoning buttons”) to 18.4% (for “handwriting smaller than it once was”). After adjustment for covariates and multiple testing, the relative odds of P-L symptoms comparing the 80th and 20th percentiles of ESS scores was 1.6 (p=0.001) for “voice is softer than it once was,” 2.1 (p<0.001) for “balance when walking is poor,” and 1.5 (p=0.002) for “loss of facial expression.” The prevalence of excessive daytime sleepiness increased from 6.3% to 19.8% when the number of symptoms increased from zero to three (p=0.004). Conclusions In Korean adults aged 50–64 years, daytime sleepiness is significantly associated with P-L symptoms. Whether coexisting daytime sleepiness and P-L symptoms predate extrapyramidal and other impairments in later life warrants further investigation.
Collapse
Affiliation(s)
- Chol Shin
- Institute of Human Genomic Study, Korea University College of Medicine, Ansan, Korea.
| | - Regina E Y Kim
- Institute of Human Genomic Study, Korea University College of Medicine, Ansan, Korea
| | - Robert J Thomas
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center and Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Chang-Ho Yun
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seung Ku Lee
- Institute of Human Genomic Study, Korea University College of Medicine, Ansan, Korea
| | - Robert D Abbott
- Institute of Human Genomic Study, Korea University College of Medicine, Ansan, Korea
| |
Collapse
|