1
|
Khaing K, Dolja-Gore X, Nair BR, Byles J, Attia J. The Effect of Sleep Duration and Excessive Daytime Sleepiness on All-Cause Dementia: A Longitudinal Analysis from the Hunter Community Study. J Am Med Dir Assoc 2024; 25:105299. [PMID: 39395812 DOI: 10.1016/j.jamda.2024.105299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 10/14/2024]
Abstract
OBJECTIVES It has been proposed that abnormal sleep duration and excessive daytime sleepiness might be risk factors for dementia. This study assessed the interaction between sleep duration and excessive daytime sleepiness, and the effect of sleep duration in the presence or absence of excessive daytime sleepiness on dementia risk in community-dwelling older adults. DESIGN A longitudinal study. SETTING AND PARTICIPANTS Data from 2187 community-dwelling participants with mean age 70 years from the Hunter Community Study were included in this study. METHODS Participants were classified as participants with long sleep duration (slept >8 hours per night), recommended sleep duration (7-8 hours) as per the National Sleep Foundation, or short sleep duration (slept <7 hours per night). The Berlin Questionnaire was used to identify excessive daytime sleepiness. Dementia was defined as per International Classification of Diseases, 10th Revision codes. To calculate all-cause dementia risk, the Fine-Gray sub-distribution hazard model was computed with death as a competing risk. RESULTS Over a mean follow-up of 6 years, 64 participants developed dementia and 154 deaths were identified. The average onset of dementia was 5.4 years. Long sleep duration was associated with increased dementia risk only in the presence of excessive daytime sleepiness (adjusted hazard ratio, 2.86; 95% confidence interval 1.03-7.91). A statistically significant interaction was found between excessive daytime sleepiness and sleep duration for all-cause dementia. CONCLUSIONS AND IMPLICATIONS Long sleep duration with excessive daytime sleepiness was associated with increased risk of all-cause dementia. This suggests the importance of promoting awareness of healthy sleep and the possible role of nurturing good quantity and quality sleep in reducing the risk of dementia.
Collapse
Affiliation(s)
- Kay Khaing
- University of Newcastle, New Lambton, New South Wales, Australia.
| | - Xenia Dolja-Gore
- University of Newcastle, New Lambton, New South Wales, Australia
| | | | - Julie Byles
- University of Newcastle, New Lambton, New South Wales, Australia
| | - John Attia
- University of Newcastle, New Lambton, New South Wales, Australia
| |
Collapse
|
2
|
Shraim MA, Massé-Alarie H, Farrell MJ, Cavaleri R, Loggia ML, Hodges PW. Neuroinflammatory activation in sensory and motor regions of the cortex is related to sensorimotor function in individuals with low back pain maintained by nociplastic mechanisms: A preliminary proof-of-concept study. Eur J Pain 2024; 28:1607-1626. [PMID: 39007713 DOI: 10.1002/ejp.2313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 06/26/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Chronic pain involves communication between neural and immune systems. Recent data suggest localization of glial (brain immune cells) activation to the sensorimotor regions of the brain cortex (S1/M1) in chronic low back pain (LBP). As glia perform diverse functions that impact neural function, activation might contribute to sensorimotor changes, particularly in LBP maintained by increased nervous system sensitivity (i.e., nociplastic pain). This preliminary proof-of-concept study aimed to: (i) compare evidence of neuroinflammatory activation in S1/M1 between individuals with and without LBP (and between nociceptive and nociplastic LBP phenotypes), and (ii) evaluate relationships between neuroinflammatory activation and sensorimotor function. METHODS Simultaneous PET-fMRI measured neuroinflammatory activation in functionally defined S1/M1 in pain-free individuals (n = 8) and individuals with chronic LBP (n = 9; nociceptive: n = 4, nociplastic: n = 5). Regions of S1/M1 related to the back were identified using fMRI during motor tasks and thermal stimuli. Sensorimotor measures included single and paired-pulse transcranial magnetic stimulation (TMS) and quantitative sensory testing (QST). Sleep, depression, disability and pain questionnaires were administered. RESULTS Neuroinflammatory activation was greater in the lower back cortical representation of S1/M1 of the nociplastic LBP group than both nociceptive LBP and pain-free groups. Neuroinflammatory activation in S1/M1 was positively correlated with sensitivity to hot (r = 0.52) and cold (r = 0.55) pain stimuli, poor sleep, depression, disability and BMI, and negatively correlated with intracortical facilitation (r = -0.41). CONCLUSION This preliminary proof-of-concept study suggests that neuroinflammation in back regions of S1/M1 in individuals with nociplastic LBP could plausibly explain some characteristic features of this LBP phenotype. SIGNIFICANCE STATEMENT Neuroinflammatory activation localized to sensorimotor areas of the brain in individuals with nociplastic pain might contribute to changes in sensory and motor function and aspects of central sensitization. If cause-effect relationships are established in longitudinal studies, this may direct development of therapies that target neuroinflammatory activation.
Collapse
Affiliation(s)
- Muath A Shraim
- The University of Queensland, School of Health & Rehabilitation Sciences, St Lucia, Queensland, Australia
| | - Hugo Massé-Alarie
- The University of Queensland, School of Health & Rehabilitation Sciences, St Lucia, Queensland, Australia
- Centre Interdisciplinaire de Recherche en réadaptation et Integration Sociale (CIRRIS), Université Laval, Québec City, Québec, Canada
| | - Michael J Farrell
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Rocco Cavaleri
- Brain Stimulation and Rehabilitation Lab, Western Sydney University, School of Health Sciences, Sydney, New South Wales, Australia
| | - Marco L Loggia
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul W Hodges
- The University of Queensland, School of Health & Rehabilitation Sciences, St Lucia, Queensland, Australia
| |
Collapse
|
3
|
Pérez-González AP, García-Kroepfly AL, Pérez-Fuentes KA, García-Reyes RI, Solis-Roldan FF, Alba-González JA, Hernández-Lemus E, de Anda-Jáuregui G. The ROSMAP project: aging and neurodegenerative diseases through omic sciences. Front Neuroinform 2024; 18:1443865. [PMID: 39351424 PMCID: PMC11439699 DOI: 10.3389/fninf.2024.1443865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
The Religious Order Study and Memory and Aging Project (ROSMAP) is an initiative that integrates two longitudinal cohort studies, which have been collecting clinicopathological and molecular data since the early 1990s. This extensive dataset includes a wide array of omic data, revealing the complex interactions between molecular levels in neurodegenerative diseases (ND) and aging. Neurodegenerative diseases (ND) are frequently associated with morbidity and cognitive decline in older adults. Omics research, in conjunction with clinical variables, is crucial for advancing our understanding of the diagnosis and treatment of neurodegenerative diseases. This summary reviews the extensive omics research-encompassing genomics, transcriptomics, proteomics, metabolomics, epigenomics, and multiomics-conducted through the ROSMAP study. It highlights the significant advancements in understanding the mechanisms underlying neurodegenerative diseases, with a particular focus on Alzheimer's disease.
Collapse
Affiliation(s)
- Alejandra P Pérez-González
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomedicas, Unidad de Posgrado Edificio B Primer Piso, Ciudad Universitaria, Mexico City, Mexico
- Facultad de Estudios Superiores Iztacala UNAM, Mexico City, Mexico
| | | | | | | | | | | | - Enrique Hernández-Lemus
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Guillermo de Anda-Jáuregui
- División de Genómica Computacional, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Programa de Investigadoras e Investigadores por México Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City, Mexico
| |
Collapse
|
4
|
Dias I, Kollarik S, Siegel M, Baumann CR, Moreira CG, Noain D. Novel murine closed-loop auditory stimulation paradigm elicits macrostructural sleep benefits in neurodegeneration. J Sleep Res 2024:e14316. [PMID: 39223830 DOI: 10.1111/jsr.14316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/05/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Boosting slow-wave activity (SWA) by modulating slow waves through closed-loop auditory stimulation (CLAS) might provide a powerful non-pharmacological tool to investigate the link between sleep and neurodegeneration. Here, we established mouse CLAS (mCLAS)-mediated SWA enhancement and explored its effects on sleep deficits in neurodegeneration, by targeting the up-phase of slow waves in mouse models of Alzheimer's disease (AD, Tg2576) and Parkinson's disease (PD, M83). We found that tracking a 2 Hz component of slow waves leads to highest precision of non-rapid eye movement (NREM) sleep detection in mice, and that its combination with a 30° up-phase target produces a significant 15-30% SWA increase from baseline in wild-type (WTAD) and transgenic (TGAD) mice versus a mock stimulation group. Conversely, combining 2 Hz with a 40° phase target yields a significant increase ranging 30-35% in WTPD and TGPD mice. Interestingly, these phase-target-triggered SWA increases are not genotype dependent but strain specific. Sleep alterations that may contribute to disease progression and burden were described in AD and PD lines. Notably, pathological sleep traits were rescued by mCLAS, which elicited a 14% decrease of pathologically heightened NREM sleep fragmentation in TGAD mice, accompanied by a steep decrease in microarousal events during both light and dark periods. Overall, our results indicate that model-tailored phase targeting is key to modulate SWA through mCLAS, prompting the acute alleviation of key neurodegeneration-associated sleep phenotypes and potentiating sleep regulation and consolidation. Further experiments assessing the long-term effect of mCLAS in neurodegeneration may majorly impact the establishment of sleep-based therapies.
Collapse
Affiliation(s)
- Inês Dias
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich (UZH), Zurich, Switzerland
| | - Sedef Kollarik
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
| | - Michelle Siegel
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
| | - Christian R Baumann
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich (UZH), Zurich, Switzerland
- Center of Competence Sleep and Health, University of Zurich (UZH), Zurich, Switzerland
| | - Carlos G Moreira
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
| | - Daniela Noain
- Department of Neurology, University Hospital Zurich (USZ), Schlieren, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich (UZH), Zurich, Switzerland
- Center of Competence Sleep and Health, University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
5
|
Wang M, Zhang L, Yang H, Lu H. Translatome and transcriptome profiling of neonatal mice hippocampus exposed to sevoflurane anesthesia. Heliyon 2024; 10:e28876. [PMID: 38707353 PMCID: PMC11066607 DOI: 10.1016/j.heliyon.2024.e28876] [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: 03/21/2023] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 05/07/2024] Open
Abstract
Exposure to anesthesia in early life may cause severe damage to the brain and lead to cognitive impairment. The underlying mechanisms, which have only been investigated in a limited scale, remains largely elusive. We performed translatome and transcriptome sequencing together for the first time in hippocampus of neonatal mice that were exposed to sevoflurane. We treated a group of neonatal mice with 2.5 % sevoflurane for 2 h on day 6, 7, 8, 9 and treated another group on day 6, 7. We performed behavioral study after day 30 for both groups and the control to evaluate the cognitive impairment. On day 36, we collected translatome and transcriptome from the hippocampus in the two groups, compared the gene expression levels between the groups and the control, and validated the results with RT-qPCR. We identified 1750 differentially expressed genes (DEGs) from translatome comparison and 1109 DEGs from transcriptome comparison. As expected, translatome-based DEGs significantly overlapped with transcriptome-based DEGs, and functional enrichment analysis generated similar enriched cognition-related GO terms and KEGG pathways. However, for many genes like Hspa5, their alterations in translatome differed remarkably from those in transcriptome, and Western blot results were largely concordant with the former, suggesting that translational regulation plays a significant role in cellular response to sevoflurane. Our study revealed global alterations in translatome and transcriptome of mice hippocampus after neonatal exposure to sevoflurane anesthesia and highlighted the importance of translatome analysis in understanding the mechanisms responsible for anesthesia-induced cognitive impairment.
Collapse
Affiliation(s)
- Menghan Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Limin Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hecheng Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
6
|
Kapasi A, Yu L, Leurgans SE, Agrawal S, Boyle PA, Bennett DA, Schneider JA. Association between hippocampal microglia, AD and LATE-NC, and cognitive decline in older adults. Alzheimers Dement 2024; 20:3193-3202. [PMID: 38494787 PMCID: PMC11095444 DOI: 10.1002/alz.13780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/29/2024] [Indexed: 03/19/2024]
Abstract
INTRODUCTION This study investigates the relationship between microglia inflammation in the hippocampus, brain pathologies, and cognitive decline. METHODS Participants underwent annual clinical evaluations and agreed to brain donation. Neuropathologic evaluations quantified microglial burden in the hippocampus, amyloid beta (Aβ), tau tangles, and limbic age-related transactive response DNA-binding protein 43 (TDP-43) encephalopathy neuropathologic changes (LATE-NC), and other common brain pathologies. Mixed-effect and linear regression models examined the association of microglia with a decline in global and domain-specific cognitive measures, and separately with brain pathologies. Path analyses estimated direct and indirect effects of microglia on global cognition. RESULT Hippocampal microglia were associated with a faster decline in global cognition, specifically in episodic memory, semantic memory, and perceptual speed. Tau tangles and LATE-NC were independently associated with microglia. Other pathologies, including Aβ, were not related. Regional hippocampal burden of tau tangles and TDP-43 accounted for half of the association of microglia with cognitive decline. DISCUSSION Microglia inflammation in the hippocampus contributes to cognitive decline. Tau tangles and LATE-NC partially mediate this association.
Collapse
Affiliation(s)
- Alifiya Kapasi
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
| | - Lei Yu
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Sue E Leurgans
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Sonal Agrawal
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
| | - Patricia A Boyle
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Psychiatry and Behavioral SciencesRush University Medical CenterChicagoIllinoisUSA
| | - David A Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Julie A Schneider
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
7
|
Pierson SR, Kolling LJ, James TD, Pushpavathi SG, Marcinkiewcz CA. Serotonergic dysfunction may mediate the relationship between alcohol consumption and Alzheimer's disease. Pharmacol Res 2024; 203:107171. [PMID: 38599469 PMCID: PMC11088857 DOI: 10.1016/j.phrs.2024.107171] [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: 12/05/2023] [Revised: 03/14/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024]
Abstract
The impact of Alzheimer's disease (AD) and its related dementias is rapidly expanding, and its mitigation remains an urgent social and technical challenge. To date there are no effective treatments or interventions for AD, but recent studies suggest that alcohol consumption is correlated with the risk of developing dementia. In this review, we synthesize data from preclinical, clinical, and epidemiological models to evaluate the combined role of alcohol consumption and serotonergic dysfunction in AD, underscoring the need for further research on this topic. We first discuss the limitations inherent to current data-collection methods, and how neuropsychiatric symptoms common among AD, alcohol use disorder, and serotonergic dysfunction may mask their co-occurrence. We additionally describe how excess alcohol consumption may accelerate the development of AD via direct effects on serotonergic function, and we explore the roles of neuroinflammation and proteostasis in mediating the relationship between serotonin, alcohol consumption, and AD. Lastly, we argue for a shift in current research to disentangle the pathogenic effects of alcohol on early-affected brainstem structures in AD.
Collapse
Affiliation(s)
- Samantha R Pierson
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | - Louis J Kolling
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | - Thomas D James
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | | | | |
Collapse
|
8
|
Liu X, Lu B, Huang H. Investigation of the shared biological mechanisms and common biomarker APTAF1 of sleep deprivation and mild cognitive impairment using integrated bioinformatics analysis. Front Pharmacol 2024; 15:1387569. [PMID: 38694919 PMCID: PMC11061425 DOI: 10.3389/fphar.2024.1387569] [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: 02/18/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction: The relationship between sleep loss and cognitive impairment has long been widely recognized, but there is still a lack of complete understanding of the underlying mechanisms and potential biomarkers. The purpose of this study is to further explore the shared biological mechanisms and common biomarkers between sleep loss and cognitive impairment. Methods: The mitochondria-related genes and gene expression data were downloaded from the MitoCarta3.0 and Gene Expression Omnibus (GEO) databases. We identified the differentially expressed mitochondrial-related genes by combing the differentially expressed genes (DEGs) in sleep deprivation (SD) and mild cognitive impairment (MCI) datasets with mitochondria-related gene lists. Shared DEGs were then further analyzed for enrichment analysis. Next, the common biomarker was identified using two machine learning techniques and further validated using two independent GEO datasets. Then GSEA and GSVA were conducted to analyze the functional categories and pathways enriched for the common biomarker. Finally, immune infiltration analysis was used to investigate the correlation of immune cell infiltration with the common biomarker in SD and MCI. Results: A total of 32 mitochondrial-related differentially expressed genes were identified in SD and MCI. GO analysis indicated that these genes were significantly enriched for mitochondrial transport, and KEGG analysis showed they were mainly involved in pathways of neurodegenerative diseases. In addition, ATPAF1, which was significantly down-regulated in both SD and MCI, was identified through machine learning algorithms as the common biomarker with favorable diagnostic performance. GSEA and GSVA revealed that ATPAF1 was mainly involved in metabolic pathways, such as oxidative phosphorylation, acetylcholine metabolic process, valine, leucine and isoleucine degradation. Immune infiltration analysis showed that the expression of ATPAF1 was correlated with changes in immune cells, especially those key immune cell types associated with SD and MCI. Discussion: This study firstly revealed that mitochondrial dysfunction may be the common pathogenesis of sleep loss and mild cognitive impairment and identified ATPAF1 as a possible biomarker and therapeutic target involved in SD and MCI.
Collapse
Affiliation(s)
- Xiaolan Liu
- Wuhan Mental Health Center, Wuhan, Hubei, China
- Wuhan Hospital for Psychotherapy, Wuhan, Hubei, China
| | - Baili Lu
- Wuhan Mental Health Center, Wuhan, Hubei, China
- Wuhan Hospital for Psychotherapy, Wuhan, Hubei, China
| | - Hui Huang
- Wuhan Mental Health Center, Wuhan, Hubei, China
- Wuhan Hospital for Psychotherapy, Wuhan, Hubei, China
| |
Collapse
|
9
|
Fu X, Wan XJ, Liu JY, Sun Q, Shen Y, Li J, Mao CJ, Ma QH, Wang F, Liu CF. Effects of sleep fragmentation on white matter pathology in a rat model of cerebral small vessel disease. Sleep 2024; 47:zsad225. [PMID: 37638817 DOI: 10.1093/sleep/zsad225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/10/2023] [Indexed: 08/29/2023] Open
Abstract
STUDY OBJECTIVES Mounting evidence indicated the correlation between sleep and cerebral small vessel disease (CSVD). However, little is known about the exact causality between poor sleep and white matter injury, a typical signature of CSVD, as well as the underlying mechanisms. METHODS Spontaneously hypertensive rats (SHR) and control Wistar Kyoto rats were subjected to sleep fragmentation (SF) for 16 weeks. The effects of chronic sleep disruption on the deep white matter and cognitive performance were observed. RESULTS SHR were validated as a rat model for CSVD. Fragmented sleep induced strain-dependent white matter abnormalities, characterized by reduced myelin integrity, impaired oligodendrocytes precursor cells (OPC) maturation and pro-inflammatory microglial polarization. Partially reversible phenotypes of OPC and microglia were observed in parallel following sleep recovery. CONCLUSIONS Long-term SF-induced pathological effects on the deep white matter in a rat model of CSVD. The pro-inflammatory microglial activation and the block of OPC maturation may be involved in the mechanisms linking sleep to white matter injury.
Collapse
Affiliation(s)
- Xiang Fu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xiao-Jie Wan
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jun-Yi Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Qian Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yun Shen
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Li
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Quan-Hong Ma
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
10
|
Kim RT, Zhou L, Li Y, Krieger AC, Nordvig AS, Butler T, de Leon MJ, Chiang GC. Impaired sleep is associated with tau deposition on 18F-flortaucipir PET and accelerated cognitive decline, accounting for medications that affect sleep. J Neurol Sci 2024; 458:122927. [PMID: 38341949 PMCID: PMC10947806 DOI: 10.1016/j.jns.2024.122927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Impaired sleep is commonly associated with Alzheimer's disease (AD), although the underlying mechanisms remain unclear. Furthermore, the moderating effects of sleep-affecting medications, which have been linked to AD pathology, are incompletely characterized. Using data from the Alzheimer's Disease Neuroimaging Initiative, we investigated whether a medical history of impaired sleep, informant-reported nighttime behaviors, and sleep-affecting medications are associated with beta-amyloid and tau deposition on PET and cognitive change, cross-sectionally and longitudinally. METHODS We included 964 subjects with 18F-florbetapir PET scans. Measures of sleep impairment and medication use were obtained from medical histories and the Neuropsychiatric Inventory Questionnaire. Multivariate models, adjusted for covariates, were used to assess associations among sleep-related features, beta-amyloid and tau, and cognition. Cortical tau deposition, categorized by Braak stage, was assessed using the standardized uptake value peak alignment (SUVP) method on 18F-flortaucipir PET. RESULTS Medical history of sleep impairment was associated with greater baseline tau in the meta-temporal, Braak 1, and Braak 4 regions (p = 0.04, p < 0.001, p = 0.025, respectively). Abnormal nighttime behaviors were also associated with greater baseline tau in the meta-temporal region (p = 0.024), and greater cognitive impairment, cross-sectionally (p = 0.007) and longitudinally (p < 0.001). Impaired sleep was not associated with baseline beta-amyloid (p > 0.05). Short-term use of selective serotonin reuptake inhibitors and benzodiazepines slightly weakened the sleep-tau relationship. CONCLUSIONS Sleep impairment was associated with tauopathy and cognitive decline, which could be linked to increased tau secretion from neuronal hyperactivity. Clinically, our results help identify high-risk individuals who could benefit from sleep-related interventions aimed to delay cognitive decline and AD.
Collapse
Affiliation(s)
- Ryan T Kim
- From the Department of Stem Cell and Regenerative Biology, Harvard University, Bauer-Sherman Fairchild Complex 7 Divinity Avenue, Cambridge, MA 02138, United States of America.
| | - Liangdong Zhou
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Yi Li
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Ana C Krieger
- From the Departments of Medicine and Neurology, Division of Sleep Neurology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 425 E 61st St., 5th Floor, New York, NY 10065, United States of America.
| | - Anna S Nordvig
- From the Department of Neurology, Alzheimer's Disease and Memory Disorders Program, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 428 East 72(nd) Street Suite 500, New York, NY 10021, United States of America.
| | - Tracy Butler
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Mony J de Leon
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Gloria C Chiang
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America; From the Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, United States of America.
| |
Collapse
|
11
|
Xiao X, Rui Y, Jin Y, Chen M. Relationship of Sleep Disorder with Neurodegenerative and Psychiatric Diseases: An Updated Review. Neurochem Res 2024; 49:568-582. [PMID: 38108952 DOI: 10.1007/s11064-023-04086-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/19/2023]
Abstract
Sleep disorders affect many people worldwide and can accompany neurodegenerative and psychiatric diseases. Sleep may be altered before the clinical manifestations of some of these diseases appear. Moreover, some sleep disorders affect the physiological organization and function of the brain by influencing gene expression, accelerating the accumulation of abnormal proteins, interfering with the clearance of abnormal proteins, or altering the levels of related hormones and neurotransmitters, which can cause or may be associated with the development of neurodegenerative and psychiatric diseases. However, the detailed mechanisms of these effects are unclear. This review mainly focuses on the relationship between and mechanisms of action of sleep in Alzheimer's disease, depression, and anxiety, as well as the relationships between sleep and Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. This summary of current research hotspots may provide researchers with better clues and ideas to develop treatment solutions for neurodegenerative and psychiatric diseases associated with sleep disorders.
Collapse
Affiliation(s)
- Xiao Xiao
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yimin Rui
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yu Jin
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
12
|
Kou L, Chi X, Sun Y, Yin S, Wu J, Zou W, Wang Y, Jin Z, Huang J, Xiong N, Xia Y, Wang T. Circadian regulation of microglia function: Potential targets for treatment of Parkinson's Disease. Ageing Res Rev 2024; 95:102232. [PMID: 38364915 DOI: 10.1016/j.arr.2024.102232] [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: 07/17/2023] [Revised: 02/11/2024] [Accepted: 02/11/2024] [Indexed: 02/18/2024]
Abstract
Circadian rhythms are involved in the regulation of many aspects of the body, including cell function, physical activity and disease. Circadian disturbance often predates the typical symptoms of neurodegenerative diseases and is not only a non-motor symptom, but also one of the causes of their occurrence and progression. Glial cells possess circadian clocks that regulate their function to maintain brain development and homeostasis. Emerging evidence suggests that the microglial circadian clock is involved in the regulation of many physiological processes, such as cytokine release, phagocytosis, and nutritional and metabolic support, and that disruption of the microglia clock may affect multiple aspects of Parkinson's disease, especially neuroinflammation and α-synuclein processes. Herein, we review recent advances in the circadian control of microglia function in health and disease, and discuss novel pharmacological interventions for microglial clocks in neurodegenerative disorders.
Collapse
Affiliation(s)
- Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
13
|
Jaiswal SJ, Gadaleta M, Quer G, Radin JM, Waalen J, Ramos E, Pandit J, Owens RL. Objectively measured peri-vaccination sleep does not predict COVID-19 breakthrough infection. Sci Rep 2024; 14:4655. [PMID: 38409137 PMCID: PMC10897487 DOI: 10.1038/s41598-024-53743-4] [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: 04/07/2023] [Accepted: 02/04/2024] [Indexed: 02/28/2024] Open
Abstract
Prior studies have shown that sleep duration peri-vaccination influences an individual's antibody response. However, whether peri-vaccination sleep affects real-world vaccine effectiveness is unknown. Here, we tested whether objectively measured sleep around COVID-19 vaccination affected breakthrough infection rates. DETECT is a study of digitally recruited participants who report COVID-19-related information, including vaccination and illness data. Objective sleep data are also recorded through activity trackers. We compared the impact of sleep duration, sleep efficiency, and frequency of awakenings on reported breakthrough infection after the 2nd vaccination and 1st COVID-19 booster. Logistic regression models were created to examine if sleep metrics predicted COVID-19 breakthrough infection independent of age and gender. Self-reported breakthrough COVID-19 infection following 2nd COVID-19 vaccination and 1st booster. 256 out of 5265 individuals reported a breakthrough infection after the 2nd vaccine, and 581 out of 2583 individuals reported a breakthrough after the 1st booster. There was no difference in sleep duration between those with and without breakthrough infection. Increased awakening frequency was associated with breakthrough infection after the 1st booster with 3.01 ± 0.65 awakenings/hour in the breakthrough group compared to 2.82 ± 0.65 awakenings/hour in those without breakthrough (P < 0.001). Cox proportional hazards modeling showed that age < 60 years (hazard ratio 2.15, P < 0.001) and frequency of awakenings (hazard ratio 1.17, P = 0.019) were associated with breakthrough infection after the 1st booster. Sleep duration was not associated with breakthrough infection after COVID vaccination. While increased awakening frequency during sleep was associated with breakthrough infection beyond traditional risk factors, the clinical implications of this finding are unclear.
Collapse
Affiliation(s)
| | | | - Giorgio Quer
- The Scripps Research Institute, La Jolla, CA, USA
| | | | - Jill Waalen
- The Scripps Research Institute, La Jolla, CA, USA
| | - Edward Ramos
- The Scripps Research Institute, La Jolla, CA, USA
| | - Jay Pandit
- The Scripps Research Institute, La Jolla, CA, USA
| | - Robert L Owens
- University of California San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
14
|
Curtis AF, Costa AN, Musich M, Schmiedeler A, Jagannathan S, Connell M, Atkinson A, Miller MB, McCrae CS. Sex as a moderator of the sleep and cognition relationship in middle-aged and older adults: A preliminary investigation. Behav Sleep Med 2024; 22:14-27. [PMID: 36809223 DOI: 10.1080/15402002.2023.2177293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
OBJECTIVES Despite known sex differences in the prevalence of sleep disturbance and cognitive impairment, research investigating sex differences in sleep/cognition associations is limited. We examined sex as a moderator of associations between self-reported sleep and objective cognition in middle-aged/older adults. METHODS Adults aged 50+ (32 men/31 women, Mage = 63.6 ± 7.7) completed the Pittsburgh Sleep Quality Index (PSQI) and cognitive tasks: Stroop (processing speed, inhibition), Posner (spatial attentional orienting) and Sternberg (working memory). Multiple regressions examined whether PSQI metrics (global score, sleep quality ratings, sleep duration, sleep efficiency) were independently or interactively (with sex) associated with cognition, controlling for age and education. RESULTS Sex interacted with sleep quality ratings in its association with endogenous spatial attentional orienting (∆R2 = .10, p = .01). Worse ratings of sleep quality were associated with worse orienting in women (B = 22.73, SE = 9.53, p = .02), not men (p = .24). Sex interacted with sleep efficiency in its associations with processing speed (∆R2 = .06, p = .04). Lower sleep efficiency was associated with slower Stroop control trial performance in women (B = -15.91, SE = 7.57, p = .04), not men (p = .48). CONCLUSIONS Preliminary findings suggest middle-aged/older women are more vulnerable to associations between poor sleep quality and low sleep efficiency on spatial attentional orienting and processing speed, respectively. Future studies in larger samples investigating sex-specific prospective sleep and cognition associations are warranted.
Collapse
Affiliation(s)
- Ashley F Curtis
- College of Nursing, University of South Florida, Tampa, Florida, USA
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri, USA
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Amy N Costa
- Department of Psychological Sciences, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Madison Musich
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Anthony Schmiedeler
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri, USA
- Department of Educational, School, and Counseling Psychology, University of Missouri-Columbia, Columbia, Missouri, USA
| | | | - Maggie Connell
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Angela Atkinson
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Mary Beth Miller
- Department of Psychiatry, University of Missouri-Columbia, Columbia, Missouri, USA
| | | |
Collapse
|
15
|
Malvaso A, Gatti A, Negro G, Calatozzolo C, Medici V, Poloni TE. Microglial Senescence and Activation in Healthy Aging and Alzheimer's Disease: Systematic Review and Neuropathological Scoring. Cells 2023; 12:2824. [PMID: 38132144 PMCID: PMC10742050 DOI: 10.3390/cells12242824] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
The greatest risk factor for neurodegeneration is the aging of the multiple cell types of human CNS, among which microglia are important because they are the "sentinels" of internal and external perturbations and have long lifespans. We aim to emphasize microglial signatures in physiologic brain aging and Alzheimer's disease (AD). A systematic literature search of all published articles about microglial senescence in human healthy aging and AD was performed, searching for PubMed and Scopus online databases. Among 1947 articles screened, a total of 289 articles were assessed for full-text eligibility. Microglial transcriptomic, phenotypic, and neuropathological profiles were analyzed comprising healthy aging and AD. Our review highlights that studies on animal models only partially clarify what happens in humans. Human and mice microglia are hugely heterogeneous. Like a two-sided coin, microglia can be protective or harmful, depending on the context. Brain health depends upon a balance between the actions and reactions of microglia maintaining brain homeostasis in cooperation with other cell types (especially astrocytes and oligodendrocytes). During aging, accumulating oxidative stress and mitochondrial dysfunction weaken microglia leading to dystrophic/senescent, otherwise over-reactive, phenotype-enhancing neurodegenerative phenomena. Microglia are crucial for managing Aβ, pTAU, and damaged synapses, being pivotal in AD pathogenesis.
Collapse
Affiliation(s)
- Antonio Malvaso
- IRCCS “C. Mondino” Foundation, National Neurological Institute, Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (A.M.); (A.G.)
| | - Alberto Gatti
- IRCCS “C. Mondino” Foundation, National Neurological Institute, Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (A.M.); (A.G.)
| | - Giulia Negro
- Department of Neurology, University of Milano Bicocca, 20126 Milan, Italy;
| | - Chiara Calatozzolo
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation, Abbiategrasso, 20081 Milan, Italy;
| | - Valentina Medici
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy;
| | - Tino Emanuele Poloni
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation, Abbiategrasso, 20081 Milan, Italy;
| |
Collapse
|
16
|
Ramasubbu K, Ramanathan G, Venkatraman G, Rajeswari VD. Sleep-associated insulin resistance promotes neurodegeneration. Mol Biol Rep 2023; 50:8665-8681. [PMID: 37580496 DOI: 10.1007/s11033-023-08710-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/25/2023] [Indexed: 08/16/2023]
Abstract
Lifestyle modification can lead to numerous health issues closely associated with sleep. Sleep deprivation and disturbances significantly affect inflammation, immunity, neurodegeneration, cognitive depletion, memory impairment, neuroplasticity, and insulin resistance. Sleep significantly impacts brain and memory formation, toxin excretion, hormonal function, metabolism, and motor and cognitive functions. Sleep restriction associated with insulin resistance affects these functions by interfering with the insulin signalling pathway, neurotransmission, inflammatory pathways, and plasticity of neurons. So, in this review, We discuss the evidence that suggests that neurodegeneration occurs via sleep and is associated with insulin resistance, along with the insulin signalling pathways involved in neurodegeneration and neuroplasticity, while exploring the role of hormones in these conditions.
Collapse
Affiliation(s)
- Kanagavalli Ramasubbu
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Gnanasambandan Ramanathan
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Ganesh Venkatraman
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - V Devi Rajeswari
- Department of Bio-Medical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
17
|
Roh SE, Xiao M, Delgado A, Kwak C, Savonenko A, Bakker A, Kwon HB, Worley P. Sleep and circadian rhythm disruption by NPTX2 loss of function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559408. [PMID: 37808783 PMCID: PMC10557648 DOI: 10.1101/2023.09.26.559408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Sleep and circadian rhythm disruption (SCRD) is commonly observed in aging, especially in individuals who experience progressive cognitive decline to mild cognitive impairment (MCI) and Alzheimer's disease (AD). However, precise molecular mechanisms underlying the association between SCRD and aging are not fully understood. Orexin A is a well-characterized "sleep neuropeptide" that is expressed in hypothalamic neurons and evokes wake behavior. The importance of Orexin is exemplified in narcolepsy where it is profoundly down-regulated. Interestingly, the synaptic immediate early gene NPTX2 is co-expressed in Orexin neurons and is similarly reduced in narcolepsy. NPTX2 is also down-regulated in CSF of some cognitively normal older individuals and predicts the time of transition from normal cognition to MCI. The association between Orexin and NPTX2 is further evinced here where we observe that Orexin A and NPTX2 are highly correlated in CSF of cognitively normal aged individuals and raises the question of whether SCRD that are typically attributed to Orexin A loss of function may be modified by concomitant NPTX2 down-regulation. Is NPTX2 an effector of sleep or simply a reporter of orexin-dependent SCRD? To address this question, we examined NPTX2 KO mice and found they retain Orexin expression in the brain and so provide an opportunity to examine the specific contribution of NPTX2 to SCRD. Our results reveal that NPTX2 KO mice exhibit a disrupted circadian onset time, coupled with increased activity during the sleep phase, suggesting difficulties in maintaining states. Sleep EEG indicates distinct temporal allocation shifts across vigilance states, characterized by reduced wake and increased NREM time. Evident sleep fragmentation manifests through alterations of event occurrences during Wake and NREM, notably during light transition periods, in conjunction with an increased frequency of sleep transitions in NPTX2 KO mice, particularly between Wake and NREM. EEG spectral analysis indicated significant shifts in power across various frequency bands in the wake, NREM, and REM states, suggestive of disrupted neuronal synchronicity. An intriguing observation is the diminished occurrence of sleep spindles, one of the earliest measures of human sleep disruption, in NPTX2 KO mice. These findings highlight the effector role of NPTX2 loss of function as an instigator of SCRD and a potential mediator of sleep disruption in aging.
Collapse
Affiliation(s)
- Seung-Eon Roh
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Meifang Xiao
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ana Delgado
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chuljung Kwak
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alena Savonenko
- Department of Neuroanatomy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hyung-Bae Kwon
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul Worley
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
18
|
He D, Chen J, Du X, Xu L. Summary of drug therapy to treat cognitive impairment-induced obstructive sleep apnea. Front Cell Neurosci 2023; 17:1222626. [PMID: 37731463 PMCID: PMC10507626 DOI: 10.3389/fncel.2023.1222626] [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: 05/15/2023] [Accepted: 08/01/2023] [Indexed: 09/22/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a severe sleep disorder associated with intermittent hypoxia and sleep fragmentation. Cognitive impairment is a signifi- cant and common OSA complication often described in such patients. The most commonly utilized methods in clinical OSA treatment are oral appliances and continuous positive airway pressure (CPAP). However, the current therapeutic methods for improving cognitive function could not achieve the expected efficacy in same patients. Therefore, further understanding the molecular mechanism behind cognitive dysfunction in OSA disease will provide new treatment methods and targets. This review briefly summarized the clinical manifestations of cognitive impairment in OSA disease. Moreover, the pathophysiological molecular mechanism of OSA was outlined. Our study concluded that both SF and IH could induce cognitive impairment by multiple signaling pathways, such as oxidative stress activation, inflammation, and apoptosis. However, there is a lack of effective drug therapy for cognitive impairment in OSA. Finally, the therapeutic potential of some novel compounds and herbal medicine was evaluated on attenuating cognitive impairment based on certain preclinical studies.
Collapse
Affiliation(s)
- Daqiang He
- Department of Laboratory Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jian Chen
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoxue Du
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Translational Medicine Research Center, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linhao Xu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Translational Medicine Research Center, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
Li Y, Zhao L, Zhang K, Shen M, Li Y, Yu Y, Yu J, Feng J, Xie K, Yu Y. Neurometabolic and structural alterations of medial septum and hippocampal CA1 in a model of post-operative sleep fragmentation in aged mice: a study combining 1H-MRS and DTI. Front Cell Neurosci 2023; 17:1160761. [PMID: 37333891 PMCID: PMC10272368 DOI: 10.3389/fncel.2023.1160761] [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: 02/07/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Post-operative sleep disturbance is a common feature of elderly surgical patients, and sleep fragmentation (SF) is closely related to post-operative cognitive dysfunction (POCD). SF is characterized by sleep interruption, increased number of awakenings and sleep structure destruction, similar to obstructive sleep apnea (OSA). Research shows that sleep interruption can change neurotransmitter metabolism and structural connectivity in sleep and cognitive brain regions, of which the medial septum and hippocampal CA1 are key brain regions connecting sleep and cognitive processes. Proton magnetic resonance spectroscopy (1H-MRS) is a non-invasive method for the evaluation of neurometabolic abnormalities. Diffusion tensor imaging (DTI) realizes the observation of structural integrity and connectivity of brain regions of interest in vivo. However, it is unclear whether post-operative SF induces harmful changes in neurotransmitters and structures of the key brain regions and their contribution to POCD. In this study, we evaluated the effects of post-operative SF on neurotransmitter metabolism and structural integrity of medial septum and hippocampal CA1 in aged C57BL/6J male mice. The animals received a 24-h SF procedure after isoflurane anesthesia and right carotid artery exposure surgery. 1H-MRS results showed after post-operative SF, the glutamate (Glu)/creatine (Cr) and glutamate + glutamine (Glx)/Cr ratios increased in the medial septum and hippocampal CA1, while the NAA/Cr ratio decreased in the hippocampal CA1. DTI results showed post-operative SF decreased the fractional anisotropy (FA) of white matter fibers in the hippocampal CA1, while the medial septum was not affected. Moreover, post-operative SF aggravated subsequent Y-maze and novel object recognition performances accompanied by abnormal enhancement of glutamatergic metabolism signal. This study suggests that 24-h SF induces hyperglutamate metabolism level and microstructural connectivity damage in sleep and cognitive brain regions in aged mice, which may be involved in the pathophysiological process of POCD.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Lina Zhao
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Kai Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Mengxi Shen
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Jiafeng Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Jingyu Feng
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Research Institute of Anesthesiology, Tianjin, China
| |
Collapse
|
20
|
Wu R, Tripathy S, Menon V, Yu L, Buchman AS, Bennett DA, De Jager PL, Lim ASP. Fragmentation of rest periods, astrocyte activation, and cognitive decline in older adults with and without Alzheimer's disease. Alzheimers Dement 2023; 19:1888-1900. [PMID: 36335579 PMCID: PMC10697074 DOI: 10.1002/alz.12817] [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: 04/25/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Sleep disruption is associated with astrocyte activation and impaired cognition in model organisms. However, the relationship among sleep, astrocyte activation, and cognition in humans is uncertain. METHODS We used RNA-seq to quantify the prefrontal cortex expression of a panel of human activated astrocyte marker genes in 1076 older adults in the Religious Orders Study and Rush Memory and Aging Project, 411 of whom had multi-day actigraphy prior to death. We related this to rest fragmentation, a proxy for sleep fragmentation, and to longitudinal cognitive function. RESULTS Fragmentation of rest periods was associated with higher expression of activated astrocyte marker genes, which was associated with a lower level and faster decline of cognitive function. DISCUSSION Astrocyte activation and fragmented rest are associated with each other and with accelerated cognitive decline. If experimental studies confirm a causal relationship, targeting sleep fragmentation and astrocyte activation may benefit cognition in older adults. HIGHLIGHTS Greater fragmentation of rest periods, a proxy for sleep fragmentation, is associated with higher composite expression of a panel of genes characteristic of activated astrocytes. Increased expression of genes characteristic of activated astrocytes was associated with a lower level and more rapid decline of cognitive function, beyond that accounted for by the burden of amyloid and neurofibrillary tangle pathology. Longitudinal and experimental studies are needed to delineate the causal relationships among sleep, astrocyte activation, and cognition.
Collapse
Affiliation(s)
- Rebecca Wu
- Division of Neurology, Department of Medicine, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Shreejoy Tripathy
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, New York, USA
| | - Lei Yu
- Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, Rush University, Chicago, Illinois, USA
| | - Aron S Buchman
- Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, Rush University, Chicago, Illinois, USA
| | - David A Bennett
- Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, Rush University, Chicago, Illinois, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, New York, USA
| | - Andrew S P Lim
- Division of Neurology, Department of Medicine, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Parhizkar S, Gent G, Chen Y, Rensing N, Gratuze M, Strout G, Sviben S, Tycksen E, Zhang Q, Gilmore PE, Sprung R, Malone J, Chen W, Remolina Serrano J, Bao X, Lee C, Wang C, Landsness E, Fitzpatrick J, Wong M, Townsend R, Colonna M, Schmidt RE, Holtzman DM. Sleep deprivation exacerbates microglial reactivity and Aβ deposition in a TREM2-dependent manner in mice. Sci Transl Med 2023; 15:eade6285. [PMID: 37099634 PMCID: PMC10449561 DOI: 10.1126/scitranslmed.ade6285] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 04/07/2023] [Indexed: 04/28/2023]
Abstract
Sleep loss is associated with cognitive decline in the aging population and is a risk factor for Alzheimer's disease (AD). Considering the crucial role of immunomodulating genes such as that encoding the triggering receptor expressed on myeloid cells type 2 (TREM2) in removing pathogenic amyloid-β (Aβ) plaques and regulating neurodegeneration in the brain, our aim was to investigate whether and how sleep loss influences microglial function in mice. We chronically sleep-deprived wild-type mice and the 5xFAD mouse model of cerebral amyloidosis, expressing either the humanized TREM2 common variant, the loss-of-function R47H AD-associated risk variant, or without TREM2 expression. Sleep deprivation not only enhanced TREM2-dependent Aβ plaque deposition compared with 5xFAD mice with normal sleeping patterns but also induced microglial reactivity that was independent of the presence of parenchymal Aβ plaques. We investigated lysosomal morphology using transmission electron microscopy and found abnormalities particularly in mice without Aβ plaques and also observed lysosomal maturation impairments in a TREM2-dependent manner in both microglia and neurons, suggesting that changes in sleep modified neuro-immune cross-talk. Unbiased transcriptome and proteome profiling provided mechanistic insights into functional pathways triggered by sleep deprivation that were unique to TREM2 and Aβ pathology and that converged on metabolic dyshomeostasis. Our findings highlight that sleep deprivation directly affects microglial reactivity, for which TREM2 is required, by altering the metabolic ability to cope with the energy demands of prolonged wakefulness, leading to further Aβ deposition, and underlines the importance of sleep modulation as a promising future therapeutic approach.
Collapse
Affiliation(s)
- Samira Parhizkar
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Grace Gent
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yun Chen
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - Nicholas Rensing
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Maud Gratuze
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory Strout
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO, USA
| | - Sanja Sviben
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric Tycksen
- Genome Technology Access Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Qiang Zhang
- Department of Medicine, Washington University Medical School, St. Louis, MO, USA
| | | | - Robert Sprung
- Department of Medicine, Washington University Medical School, St. Louis, MO, USA
| | - Jim Malone
- Department of Medicine, Washington University Medical School, St. Louis, MO, USA
| | - Wei Chen
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Javier Remolina Serrano
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Xin Bao
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Choonghee Lee
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Chanung Wang
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric Landsness
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - James Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael Wong
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Reid Townsend
- Department of Medicine, Washington University Medical School, St. Louis, MO, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - Robert E Schmidt
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
22
|
Carvalhas-Almeida C, Serra J, Moita J, Cavadas C, Álvaro AR. Understanding neuron-glia crosstalk and biological clocks in insomnia. Neurosci Biobehav Rev 2023; 147:105100. [PMID: 36804265 DOI: 10.1016/j.neubiorev.2023.105100] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
According to the World Health Organization, about one-third of the population experiences insomnia symptoms, and about 10-15% suffer from chronic insomnia, the most common sleep disorder. Sleeping difficulties associated with insomnia are often linked to chronic sleep deprivation, which has a negative health impact partly due to disruption in the internal synchronisation of biological clocks. These are regulated by clock genes and modulate most biological processes. Most studies addressing circadian rhythm regulation have focused on the role of neurons, yet glial cells also impact circadian rhythms and sleep regulation. Chronic insomnia and sleep loss have been associated with glial cell activation, exacerbated neuroinflammation, oxidative stress, altered neuronal metabolism and synaptic plasticity, accelerated age-related processes and decreased lifespan. It is, therefore, essential to highlight the importance of glia-neuron interplay on sleep/circadian regulation and overall healthy brain function. Hence, in this review, we aim to address the main neurobiological mechanisms involved in neuron-glia crosstalk, with an emphasis on microglia and astrocytes, in both healthy sleep, chronic sleep deprivation and chronic insomnia.
Collapse
Affiliation(s)
- Catarina Carvalhas-Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Joana Serra
- Sleep Medicine Unit, Coimbra Hospital and University Center (CHUC), Coimbra, Portugal
| | - Joaquim Moita
- Sleep Medicine Unit, Coimbra Hospital and University Center (CHUC), Coimbra, Portugal
| | - Cláudia Cavadas
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Ana Rita Álvaro
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
23
|
Sleep-Related Changes Prior to Cognitive Dysfunction. Curr Neurol Neurosci Rep 2023; 23:177-183. [PMID: 36881255 DOI: 10.1007/s11910-023-01258-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 03/08/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to summarize the current evidence on the relationship between sleep and cognition and present available data reporting the impact that sleep alterations may have on cognitive functions. RECENT FINDINGS Research findings support the idea that sleep is involved in cognitive processes and that altered sleep homeostasis or circadian rhythms may lead to clinical and biochemical changes associated with cognitive impairment. Evidence is particularly solid for the association between specific sleep architecture and circadian alterations and Alzheimer's disease. Sleep changes, as early manifestations or possible risk factors for neurodegeneration and cognitive decline, may be appropriate targets for interventions aiming to reduce the likelihood of dementia.
Collapse
|
24
|
Sleep Fragmentation Accelerates Carcinogenesis in a Chemical-Induced Colon Cancer Model. Int J Mol Sci 2023; 24:ijms24054547. [PMID: 36901981 PMCID: PMC10003038 DOI: 10.3390/ijms24054547] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Aims of this study were to test whether sleep fragmentation (SF) increased carcinogenesis and to investigate the possible mechanisms of carcinogenesis in a chemical-induced colon cancer model. In this study, eight-week-old C57BL/6 mice were divided into Home cage (HC) and SF groups. After the azoxymethane (AOM) injection, the mice in the SF group were subjected to SF for 77 days. SF was accomplished in a sleep fragmentation chamber. In the second protocol, mice were divided into 2% dextran sodium sulfate (DSS)-treated, HC, and SF groups and were exposed to the HC or SF procedures. Immunohistochemical and immunofluorescent stainings were conducted to determine the level of 8-OHdG and reactive oxygen species (ROS), respectively. Quantitative real-time polymerase chain reaction was used to assess the relative expression of inflammatory and ROS-generating genes. The number of tumors and average tumor size were significantly higher in the SF group than in the HC group. The intensity (%) of the 8-OHdG stained area was significantly higher in the SF group than in the HC group. The fluorescence intensity of ROS was significantly higher in the SF group than the HC group. SF accelerated cancer development in a murine AOM/DSS-induced model of colon cancer, and the increased carcinogenesis was associated with ROS- and oxidative stress-induced DNA damage.
Collapse
|
25
|
Iweka CA, Seigneur E, Hernandez AL, Paredes SH, Cabrera M, Blacher E, Pasternak CT, Longo FM, de Lecea L, Andreasson KI. Myeloid deficiency of the intrinsic clock protein BMAL1 accelerates cognitive aging by disrupting microglial synaptic pruning. J Neuroinflammation 2023; 20:48. [PMID: 36829230 PMCID: PMC9951430 DOI: 10.1186/s12974-023-02727-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/10/2023] [Indexed: 02/26/2023] Open
Abstract
Aging is associated with loss of circadian immune responses and circadian gene transcription in peripheral macrophages. Microglia, the resident macrophages of the brain, also show diurnal rhythmicity in regulating local immune responses and synaptic remodeling. To investigate the interaction between aging and microglial circadian rhythmicity, we examined mice deficient in the core clock transcription factor, BMAL1. Aging Cd11bcre;Bmallox/lox mice demonstrated accelerated cognitive decline in association with suppressed hippocampal long-term potentiation and increases in immature dendritic spines. C1q deposition at synapses and synaptic engulfment were significantly decreased in aging Bmal1-deficient microglia, suggesting that BMAL1 plays a role in regulating synaptic pruning in aging. In addition to accelerated age-associated hippocampal deficits, Cd11bcre;Bmallox/lox mice also showed deficits in the sleep-wake cycle with increased wakefulness across light and dark phases. These results highlight an essential role of microglial BMAL1 in maintenance of synapse homeostasis in the aging brain.
Collapse
Affiliation(s)
- Chinyere Agbaegbu Iweka
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Erica Seigneur
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Amira Latif Hernandez
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | | | - Mica Cabrera
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Eran Blacher
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus Givat-Ram, 9190401, Jerusalem, Israel
| | - Connie Tsai Pasternak
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Stanford Immunology Program, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| |
Collapse
|
26
|
Associations between objectively measured sleep parameters and cognition in healthy older adults: A meta-analysis. Sleep Med Rev 2023; 67:101734. [PMID: 36577339 DOI: 10.1016/j.smrv.2022.101734] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/03/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Multiple studies have examined associations between sleep and cognition in older adults, but a majority of these depend on self-reports on sleep and utilize cognitive tests that assess overall cognitive function. The current meta-analysis involved 72 independent studies and sought to quantify associations between objectively measured sleep parameters and cognitive performance in healthy older adults. Both sleep macrostructure (e.g., sleep duration, continuity, and stages) and microstructure (e.g., slow wave activity and spindle activity) were evaluated. For macrostructure, lower restlessness at night was associated with better memory performance (r = 0.43, p = 0.02), while lower sleep onset latency was associated with better executive functioning (r = 0.28, p = 0.03). Greater relative amount of N2 and REM sleep, but not N3, positively correlated with cognitive performance. The association between microstructure and cognition in older adults was marginally significant. This relationship was moderated by age (z = 0.07, p < 0.01), education (z = 0.26, p = 0.03), and percentage of female participants (z = 0.01, p < 0.01). The current meta-analysis emphasizes the importance of considering objective sleep measures to understand the relationship between sleep and cognition in healthy older adults. These results also form a base from which researchers using wearable sleep technology and measuring behavior through computerized testing tools can evaluate their findings.
Collapse
|
27
|
Puech C, Badran M, Runion AR, Barrow MB, Qiao Z, Khalyfa A, Gozal D. Explicit memory, anxiety and depressive like behavior in mice exposed to chronic intermittent hypoxia, sleep fragmentation, or both during the daylight period. Neurobiol Sleep Circadian Rhythms 2022; 13:100084. [PMID: 36254342 PMCID: PMC9568859 DOI: 10.1016/j.nbscr.2022.100084] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/01/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic and highly prevalent condition characterized by chronic intermittent hypoxia (IH) and sleep fragmentation (SF), and can lead to a vast array of end-organ morbidities, particularly affecting cardiovascular, metabolic and neurobehavioral functioning. OSA can induce cognitive and behavioral and mood deficits. Male C57Bl/6J 8-week-old mice were housed in custom-designed cages with a silent motorized mechanical sweeper traversing the cage floor at 2-min intervals (SF) during daylight for four weeks. Sleep control (SC) consisted of keeping sweeper immobile. IH consisted of cycling FiO2 21% 90 seconds-6.3% 90s or room air (RA; FiO2 21%) for sixteen weeks and combined SF-IH was conducted for nine weeks. Open field novel object recognition (NOR) testing, elevated-plus maze test (EPMT), and forced swimming test (FST) were performed. SF induced cognitive NOR performance impairments in mice along with reduced anxiety behaviors while IH induced deficits in NOR performance, but increased anxiety behaviors. SF-IH induced impaired performance in NOR test of similar magnitude to IH or SF alone. Combined SF-IH exposures did not affect anxiety behaviors. Thus, both SF an IH altered cognitive function while imposing opposite effects on anxiety behaviors. SF-IH did not magnify the detrimental effects of isolated SF or IH and canceled out the effects on anxiety. Based on these findings, the underlying pathophysiologic processes underlying IH and SF adverse effects on cognitive function appear to differ, while those affecting anxiety counteract each other.
Collapse
Affiliation(s)
- Clementine Puech
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mohammad Badran
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Alexandra R Runion
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - Max B Barrow
- Undergraduate Student Research Program, University of Missouri, Columbia, MO, USA
| | - Zhuanhong Qiao
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Abdelnaby Khalyfa
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - David Gozal
- Child Health Research Institute, Department of Child Health, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
28
|
Stevenson AJ, McCartney DL, Gadd DA, Shireby G, Hillary RF, King D, Tzioras M, Wrobel N, McCafferty S, Murphy L, McColl BW, Redmond P, Taylor AM, Harris SE, Russ TC, McIntosh AM, Mill J, Smith C, Deary IJ, Cox SR, Marioni RE, Spires‐Jones TL. A comparison of blood and brain-derived ageing and inflammation-related DNA methylation signatures and their association with microglial burdens. Eur J Neurosci 2022; 56:5637-5649. [PMID: 35362642 PMCID: PMC9525452 DOI: 10.1111/ejn.15661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/18/2022] [Accepted: 03/29/2022] [Indexed: 12/31/2022]
Abstract
Inflammation and ageing-related DNA methylation patterns in the blood have been linked to a variety of morbidities, including cognitive decline and neurodegenerative disease. However, it is unclear how these blood-based patterns relate to patterns within the brain and how each associates with central cellular profiles. In this study, we profiled DNA methylation in both the blood and in five post mortem brain regions (BA17, BA20/21, BA24, BA46 and hippocampus) in 14 individuals from the Lothian Birth Cohort 1936. Microglial burdens were additionally quantified in the same brain regions. DNA methylation signatures of five epigenetic ageing biomarkers ('epigenetic clocks'), and two inflammatory biomarkers (methylation proxies for C-reactive protein and interleukin-6) were compared across tissues and regions. Divergent associations between the inflammation and ageing signatures in the blood and brain were identified, depending on region assessed. Four out of the five assessed epigenetic age acceleration measures were found to be highest in the hippocampus (β range = 0.83-1.14, p ≤ 0.02). The inflammation-related DNA methylation signatures showed no clear variation across brain regions. Reactive microglial burdens were found to be highest in the hippocampus (β = 1.32, p = 5 × 10-4 ); however, the only association identified between the blood- and brain-based methylation signatures and microglia was a significant positive association with acceleration of one epigenetic clock (termed DNAm PhenoAge) averaged over all five brain regions (β = 0.40, p = 0.002). This work highlights a potential vulnerability of the hippocampus to epigenetic ageing and provides preliminary evidence of a relationship between DNA methylation signatures in the brain and differences in microglial burdens.
Collapse
Affiliation(s)
- Anna J. Stevenson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Daniel L. McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Danni A. Gadd
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Gemma Shireby
- University of Exeter Medical SchoolUniversity of ExeterExeterUK
| | - Robert F. Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
| | - Declan King
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Makis Tzioras
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Nicola Wrobel
- Edinburgh Clinical Research FacilityWestern General HospitalEdinburghUK
| | - Sarah McCafferty
- Edinburgh Clinical Research FacilityWestern General HospitalEdinburghUK
| | - Lee Murphy
- Edinburgh Clinical Research FacilityWestern General HospitalEdinburghUK
| | - Barry W. McColl
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Paul Redmond
- Lothian Birth CohortsUniversity of EdinburghEdinburghUK
| | | | - Sarah E. Harris
- Lothian Birth CohortsUniversity of EdinburghEdinburghUK
- Department of PsychologyUniversity of EdinburghEdinburghUK
| | - Tom C. Russ
- Lothian Birth CohortsUniversity of EdinburghEdinburghUK
- Alzheimer Scotland Dementia Research Centre, 7 George SquareUniversity of EdinburghEdinburghUK
- Division of PsychiatryUniversity of Edinburgh, Royal Edinburgh HospitalEdinburghUK
| | - Andrew M. McIntosh
- Division of PsychiatryUniversity of Edinburgh, Royal Edinburgh HospitalEdinburghUK
| | - Jonathan Mill
- University of Exeter Medical SchoolUniversity of ExeterExeterUK
| | - Colin Smith
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Ian J. Deary
- Lothian Birth CohortsUniversity of EdinburghEdinburghUK
- Department of PsychologyUniversity of EdinburghEdinburghUK
| | - Simon R. Cox
- Lothian Birth CohortsUniversity of EdinburghEdinburghUK
- Department of PsychologyUniversity of EdinburghEdinburghUK
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular MedicineUniversity of EdinburghEdinburghUK
- Lothian Birth CohortsUniversity of EdinburghEdinburghUK
| | - Tara L. Spires‐Jones
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| |
Collapse
|
29
|
Associations of environmental and lifestyle factors with spatial navigation in younger and older adults. J Int Neuropsychol Soc 2022; 29:377-387. [PMID: 36039948 PMCID: PMC9971349 DOI: 10.1017/s1355617722000303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Advanced age is associated with prominent impairment in allocentric navigation dependent on the hippocampus. This study examined whether age-related impairment in allocentric navigation and strategy selection was associated with sleep disruption or circadian rest-activity fragmentation. Further, we examined whether associations with navigation were moderated by perceived stress and physical activity. METHOD Sleep fragmentation and total sleep time over the course of 1 week were assayed in younger (n = 42) and older (n = 37) adults via wrist actigraphy. Subsequently, participants completed cognitive mapping and route learning tasks, as well a measure of spontaneous navigation strategy selection. Measurements of perceived stress and an actigraphy-based index of physical activity were also obtained. Circadian rest-activity fragmentation was estimated via actigraphy post-hoc. RESULTS Age was associated with reduced cognitive mapping, route learning, allocentric strategy use, and total sleep time (ps < .01), replicating prior findings. Novel findings included that sleep fragmentation increased with advancing age (p = .009) and was associated with lower cognitive mapping (p = .022) within the older adult cohort. Total sleep time was not linearly associated with the navigation tasks (ps > .087). Post-hoc analyses revealed that circadian rest-activity fragmentation increased with advancing age within the older adults (p = .026) and was associated with lower cognitive mapping across the lifespan (p = .001) and within older adults (p = .005). Neither stress nor physical activity were robust moderators of sleep fragmentation associations with the navigation tasks (ps > .113). CONCLUSION Sleep fragmentation and circadian rest-activity fragmentation are potential contributing factors to age effects on cognitive mapping within older adults.
Collapse
|
30
|
Boa Sorte Silva NC, Falck RS, Chan PCY, Tai D, Backhouse D, Stein R, Liu-Ambrose T. The association of sleep and cortical thickness in mild cognitive impairment. Exp Gerontol 2022; 167:111923. [PMID: 35963454 DOI: 10.1016/j.exger.2022.111923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 07/12/2022] [Accepted: 08/07/2022] [Indexed: 11/24/2022]
Abstract
We investigated whether device-measured sleep parameters are associated with cortical thickness in older adults with probable mild cognitive impairment (MCI). We performed a cross-sectional, exploratory analysis of sleep and structural MRI data. Sleep data were collected with MotionWatch8© actigraphy over 7 days. We computed average and variability for sleep duration, sleep efficiency, and fragmentation index. T1-weighted MRI scans were used to measure cortical thickness in FreeSurfer. We employed surface-based analysis to determine the association between sleep measures and cortical thickness, adjusting for age, sex, Montreal Cognitive Assessment (MoCA) score, and sleep medication use. Our sample included 113 participants (age = 73.1 [5.7], female = 72 [63.7 %]). Higher fragmentation index variability predicted lower cortical thickness in the left superior frontal gyrus (cluster size = 970.9 mm2, cluster-wise p = 0.017, cortical thickness range = 2.1 mm2 to 3.0 mm2), adjusting for age, sex, MoCA, and sleep medication. Our results suggest that higher variability in sleep fragmentation, an indicator of irregular sleep pattern, is linked to lower cortical thickness. Future longitudinal studies are needed to determine the directionality of these associations.
Collapse
Affiliation(s)
- Nárlon C Boa Sorte Silva
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Ryan S Falck
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Patrick C Y Chan
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Daria Tai
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Daniel Backhouse
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Ryan Stein
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Teresa Liu-Ambrose
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.
| |
Collapse
|
31
|
Kumari A, Rahaman A, Zeng XA, Farooq MA, Huang Y, Yao R, Ali M, Ishrat R, Ali R. Temporal Cortex Microarray Analysis Revealed Impaired Ribosomal Biogenesis and Hyperactivity of the Glutamatergic System: An Early Signature of Asymptomatic Alzheimer's Disease. Front Neurosci 2022; 16:966877. [PMID: 35958988 PMCID: PMC9359077 DOI: 10.3389/fnins.2022.966877] [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: 06/11/2022] [Accepted: 06/23/2022] [Indexed: 11/21/2022] Open
Abstract
Pathogenic aging is regarded as asymptomatic AD when there is no cognitive deficit except for neuropathology consistent with Alzheimer's disease. These individuals are highly susceptible to developing AD. Braak and Braak's theory specific to tau pathology illustrates that the brain's temporal cortex region is an initiation site for early AD progression. So, the hub gene analysis of this region may reveal early altered biological cascades that may be helpful to alleviate AD in an early stage. Meanwhile, cognitive processing also drags its attention because cognitive impairment is the ultimate result of AD. Therefore, this study aimed to explore changes in gene expression of aged control, asymptomatic AD (AsymAD), and symptomatic AD (symAD) in the temporal cortex region. We used microarray data sets to identify differentially expressed genes (DEGs) with the help of the R programming interface. Further, we constructed the protein-protein interaction (PPI) network by performing the STRING plugin in Cytoscape and determined the hub genes via the CytoHubba plugin. Furthermore, we conducted Gene Ontology (GO) enrichment analysis via Bioconductor's cluster profile package. Resultant, the AsymAD transcriptome revealed the early-stage changes of glutamatergic hyperexcitability. Whereas the connectivity of major hub genes in this network indicates a shift from initially reduced rRNA biosynthesis in the AsymAD group to impaired protein synthesis in the symAD group. Both share the phenomenon of breaking tight junctions and others. In conclusion, this study offers new understandings of the early biological vicissitudes that occur in the brain before the manifestation of symAD and gives new promising therapeutic targets for early AD intervention.
Collapse
Affiliation(s)
- Ankita Kumari
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
| | - Abdul Rahaman
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
- Abdul Rahaman
| | - Xin-An Zeng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
- *Correspondence: Xin-An Zeng
| | - Muhammad Adil Farooq
- Institute of Food Science and Technology, Khwaja Fareed University of Engineering and Information Technology, Rahim Yar Khan, Pakistan
| | - Yanyan Huang
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
| | - Runyu Yao
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
| | - Murtaza Ali
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, China
- Overseas Expertise Introduction Centre for Discipline Innovation of Food Nutrition and Human Health (111 Centre), Guangzhou, China
| | - Romana Ishrat
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
- Romana Ishrat
| | - Rafat Ali
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
32
|
de Oliveira P, Cella C, Locker N, Ravindran KKG, Mendis A, Wafford K, Gilmour G, Dijk DJ, Winsky-Sommerer R. Improved Sleep, Memory, and Cellular Pathological Features of Tauopathy, Including the NLRP3 Inflammasome, after Chronic Administration of Trazodone in rTg4510 Mice. J Neurosci 2022; 42:3494-3509. [PMID: 35273086 PMCID: PMC9034788 DOI: 10.1523/jneurosci.2162-21.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 12/02/2022] Open
Abstract
Several cellular pathways contribute to neurodegenerative tauopathy-related disorders. Microglial activation, a major component of neuroinflammation, is an early pathologic hallmark that correlates with cognitive decline, while the unfolded protein response (UPR) contributes to synaptic pathology. Sleep disturbances are prevalent in tauopathies and may also contribute to disease progression. Few studies have investigated whether manipulations of sleep influence cellular pathologic and behavioral features of tauopathy. We investigated whether trazodone, a licensed antidepressant with hypnotic efficacy in dementia, can reduce disease-related cellular pathways and improve memory and sleep in male rTg4510 mice with a tauopathy-like phenotype. In a 9 week dosing regimen, trazodone decreased microglial NLRP3 inflammasome expression and phosphorylated p38 mitogen-activated protein kinase levels, which correlated with the NLRP3 inflammasome, the UPR effector ATF4, and total tau levels. Trazodone reduced theta oscillations during rapid eye movement (REM) sleep and enhanced REM sleep duration. Olfactory memory transiently improved, and memory performance correlated with REM sleep duration and theta oscillations. These findings on the effects of trazodone on the NLRP3 inflammasome, the unfolded protein response and behavioral hallmarks of dementia warrant further studies on the therapeutic value of sleep-modulating compounds for tauopathies.SIGNIFICANCE STATEMENT Dementia and associated behavioral symptoms such as memory loss and sleep disturbance are debilitating. Identifying treatments that alleviate symptoms and concurrently target cellular pathways contributing to disease progression is paramount for the patients and their caregivers. Here we show that a chronic treatment with trazodone, an antidepressant with positive effects on sleep, has beneficial effects on several cellular pathways contributing to neuroinflammation and tau pathology, in tauopathy-like rTg4510 mice. Trazodone also improved rapid eye movement (REM) sleep, the slowing of brain oscillations, and olfactory memory disturbances, which are all early symptoms observed in Alzheimer's disease. Thus, trazodone and compounds with REM sleep-promoting properties may represent a promising treatment approach to reduce the early symptoms of tauopathy and slow down disease progression.
Collapse
Affiliation(s)
- Paula de Oliveira
- Surrey Sleep Research Centre, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
- Lilly Research Centre, Eli Lilly and Company, Windlesham GU20 6PH, United Kingdom
| | - Claire Cella
- Lilly Research Centre, Eli Lilly and Company, Windlesham GU20 6PH, United Kingdom
| | - Nicolas Locker
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7TE, United Kingdom
| | - Kiran K G Ravindran
- Surrey Sleep Research Centre, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
- UK Dementia Research Institute Care Research and Technology Centre, Imperial College London, London W12 0BZ and University of Surrey, Guildford GU2 7XP, United Kingdom
| | - Agampodi Mendis
- Surrey Clinical Trials Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
| | - Keith Wafford
- Lilly Research Centre, Eli Lilly and Company, Windlesham GU20 6PH, United Kingdom
| | - Gary Gilmour
- Lilly Research Centre, Eli Lilly and Company, Windlesham GU20 6PH, United Kingdom
| | - Derk-Jan Dijk
- Surrey Sleep Research Centre, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
- UK Dementia Research Institute Care Research and Technology Centre, Imperial College London, London W12 0BZ and University of Surrey, Guildford GU2 7XP, United Kingdom
| | - Raphaelle Winsky-Sommerer
- Surrey Sleep Research Centre, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
| |
Collapse
|
33
|
Suresh S, Begum RF, Singh S A, V C. Anthocyanin as a therapeutic in Alzheimer's disease: A systematic review of preclinical evidences. Ageing Res Rev 2022; 76:101595. [PMID: 35217244 DOI: 10.1016/j.arr.2022.101595] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/15/2022] [Accepted: 02/19/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND The aim of this systematic review is to ponder the possible mechanism of action of anthocyanin in Alzheimer's disease (AD), to prompt the development of anthocyanin-based dietary supplementation or therapeutic intervention for AD and to explore the natural sources of anthocyanins. METHODS Electronic bibliographic databases such as PubMed, ScienceDirect, Proquest, DOAJ, Scopus, and Google Scholar were searched for preclinical studies probing the efficacy of anthocyanin on AD. The search strategy included no time limit, but was restricted to English. The review protocol is registered on PROSPERO, registration no. CRD42021272972. The systematic review followed the PICO approach for inclusion of reports. All the reports were appraised for risk of bias using the SYRCLE's RoB tool. RESULTS Bibliographic details of the article, animal strain/weight/age, induction model, anthocyanin source, type of anthocyanin, dose, route of administration, duration, and the outcome measures were extracted from 12 retrieved reports explicitly. The implication of food-based anthocyanin in acute and long-term cognition and Aβ mediated neurodegeneration appears alluring. Majority of the studies comprehended in this review had moderate methodological quality. DISCUSSION Efficacy of anthocyanin in alleviating oxidative stress, reactive astrogliosis, cholinergic dysfunction, apoptosis, synaptotoxicity, neuroinflammation, tau hyperphosphorylation, dysregulated membrane potential, neuronal extracellular calcium, dysfunctional amyloidogenic pathway, and cognitive deficits in various rodent models of AD is manifested compositely in 12 studies.
Collapse
|
34
|
Guan Y, Zhang L, Wang S, Deng Y, Zhou H, Chen D, Zhang L. The role of microglia in Alzheimer's disease and progress of treatment. IBRAIN 2022; 8:37-47. [PMID: 37786418 PMCID: PMC10529349 DOI: 10.1002/ibra.12023] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 10/04/2023]
Abstract
Microglia are permanent immune cells of the central nervous system. Microglia play an important role in the pathological process of Alzheimer's disease (AD). They are mainly involved in the uptake and clearance of amyloid-β (Aβ), as well as the formation of neuroinflammation. We found that overactivated microglia increase Aβ and Tau, and Aβ and Tau in turn act as activators of microglia. Additionally, various cytokines and proteins, high cholesterol, and telomere shortening are all associated with microglia activation. More activated microglia induce the release of inflammatory and anti-inflammatory factors to regulate inflammation, while microglia express multiple homologous receptors that bind to neuroimmunomodulators to prevent microglia overactivation. Moreover, aging of the body promotes neuroinflammation by increasing the response to IFN-γ (interferon-γ), and aging of the microglia themselves promotes AD by inducing the accumulation of large amounts of iron and reducing autophagy by regulating protein levels. Cognitive dysfunction occurs when activated microglia induce an increase in beta oligomers, promoting the production of pro-inflammatory factors that alter the shape, composition, and density of synapses. Based on their correlation, microglia-mediated AD therapy as well as the corresponding targets and drugs are discussed. In contrast to similar reviews, this article also summarizes some novel microglia-mediated AD treatment methods over the recent years.
Collapse
Affiliation(s)
- Yi‐Huan Guan
- Department of AnesthesiologyZunyi Medical UniversityZunyiGuizhouChina
| | - Ling‐Jing Zhang
- Department of AnesthesiologyZunyi Medical UniversityZunyiGuizhouChina
| | - Shi‐Ya Wang
- Department of AnesthesiologyZunyi Medical UniversityZunyiGuizhouChina
| | - Ya‐Dan Deng
- Department of AnesthesiologyZunyi Medical UniversityZunyiGuizhouChina
| | - Hong‐Su Zhou
- Department of AnesthesiaGraduate School of Zunyi Medical UniversityZunyiGuizhouChina
| | - Dong‐Qing Chen
- Department of AnesthesiaGraduate School of Zunyi Medical UniversityZunyiGuizhouChina
| | - Lan‐Chun Zhang
- Department of ZoologyKunming Medical UniversityKunmingChina
| |
Collapse
|
35
|
Ohara T, Hata J, Tanaka M, Honda T, Yamakage H, Inoue T, Hirakawa Y, Kusakabe T, Shibata M, Kitazono T, Nakao T, Satoh-Asahara N, Ninomiya T. Association of daily sleep duration with the incident dementia by serum soluble TREM2 in a community. J Am Geriatr Soc 2021; 70:1147-1156. [PMID: 34970991 DOI: 10.1111/jgs.17634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Little is known about the influence of serum level of soluble triggering receptor expressed on myeloid cells 2 (sTREM2), which is a soluble type of an innate immune receptor expressed on the microglia, on the association of the daily sleep duration with the risk of dementia. METHODS A total of 1230 Japanese community-residents aged 60 and older without dementia were followed prospectively for 10 years (2002-2012). Serum sTREM2 levels were divided into two groups using the median value (334.8 pg/ml). Self-reported daily sleep duration was grouped into three categories of <5.0, 5.0-7.9, and ≥8.0 h. A Cox proportional hazards model was used to estimate the hazard ratios (HRs) and their 95% confidence intervals (CIs) of daily sleep duration on the risk of dementia according to serum sTREM2 levels. RESULTS During the follow-up, 262 subjects developed dementia. In subjects with low serum sTREM2 levels, subjects with ≥8.0 h of daily sleep had a significantly greater risk of dementia (multivariable-adjusted HR 2.05 [95% CI 1.32-3.19]) than those with 5.0-7.9 h of daily sleep, but those with <5.0 h did not. In contrast, the risk of dementia increased significantly in subjects with both <5.0 (1.95 [1.03-3.68]) and ≥8.0 h of daily sleep (1.48 [1.06-2.07]) in the subjects with high serum sTREM2 levels. CONCLUSIONS The influence of daily sleep duration on risk of dementia differed according to serum sTREM2 levels in the older Japanese population. Short daily sleep may be associated with greater risk of dementia only in subjects with a high serum sTREM2 level.
Collapse
Affiliation(s)
- Tomoyuki Ohara
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Hata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masashi Tanaka
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takanori Honda
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hajime Yamakage
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takayuki Inoue
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Yoichiro Hirakawa
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toru Kusakabe
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Mao Shibata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Nakao
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriko Satoh-Asahara
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
36
|
Fu Y, Wang ZT, Qu Y, Wang XT, Ma YH, Bi YL, Dong Q, Tan L, Yu JT. Sleep Characteristics and Cognitive Function in Older Adults Without Dementia: The CABLE Study. J Alzheimers Dis 2021; 84:1029-1038. [PMID: 34602483 DOI: 10.3233/jad-215017] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The associations between sleep characteristics and cognition are complicated. Alzheimer's disease (AD) pathologies have been proven to be associated with sleep characteristics. OBJECTIVE We aimed to investigate the associations between sleep characteristics and cognitive function and examine the roles of AD pathologies in modulating the association of sleep duration with cognition. METHODS A total of 974 participants who had measurements of cerebrospinal fluid (CSF) amyloid-β (Aβ), phosphorylated tau (P-tau), total tau proteins (T-tau), cognitive function, and sleep characteristics were included from the Chinese Alzheimer's Biomarker and Lifestyle (CABLE) study. Linear regression analyses were utilized to explore the associations of sleep characteristics with cognition. Non-linear regression analyses were utilized to explore the associations of sleep habits with cognition. Causal mediation analyses were conducted to explore the mediation effects of AD pathologies on cognition. RESULTS The Pittsburgh Sleep Quality Index (PSQI) total score was significantly negatively correlated with Montreal Cognitive Assessment (MoCA) score (p = 0.0176). Long latency (p = 0.0054) and low efficiency (p = 0.0273) were associated with cognitive impairment. Habitual nap behavior was associated with lower MoCA scores (p = 0.0045). U-shaped associations were observed between sleep habits (bedtime and nocturnal sleep duration) and cognition. A causal mediation analysis indicated that P-tau/Aβ42 mediated the association of sleep duration with cognition. CONCLUSION These findings showed sleep characteristics were associated with cognitive functions. Sleep habits (duration, bedtime) had U-shaped associations with cognition. AD core pathologies might partially mediate the influence of sleep duration on cognitive impairments.
Collapse
Affiliation(s)
- Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zuo-Teng Wang
- College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Yi Qu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xiao-Tong Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan-Lin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.,College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Healy KL, Morris AR, Liu AC. Circadian Synchrony: Sleep, Nutrition, and Physical Activity. FRONTIERS IN NETWORK PHYSIOLOGY 2021; 1:732243. [PMID: 35156088 PMCID: PMC8830366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/20/2021] [Indexed: 11/11/2022]
Abstract
The circadian clock in mammals regulates the sleep/wake cycle and many associated behavioral and physiological processes. The cellular clock mechanism involves a transcriptional negative feedback loop that gives rise to circadian rhythms in gene expression with an approximately 24-h periodicity. To maintain system robustness, clocks throughout the body must be synchronized and their functions coordinated. In mammals, the master clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. The SCN is entrained to the light/dark cycle through photic signal transduction and subsequent induction of core clock gene expression. The SCN in turn relays the time-of-day information to clocks in peripheral tissues. While the SCN is highly responsive to photic cues, peripheral clocks are more sensitive to non-photic resetting cues such as nutrients, body temperature, and neuroendocrine hormones. For example, feeding/fasting and physical activity can entrain peripheral clocks through signaling pathways and subsequent regulation of core clock genes and proteins. As such, timing of food intake and physical activity matters. In an ideal world, the sleep/wake and feeding/fasting cycles are synchronized to the light/dark cycle. However, asynchronous environmental cues, such as those experienced by shift workers and frequent travelers, often lead to misalignment between the master and peripheral clocks. Emerging evidence suggests that the resulting circadian disruption is associated with various diseases and chronic conditions that cause further circadian desynchrony and accelerate disease progression. In this review, we discuss how sleep, nutrition, and physical activity synchronize circadian clocks and how chronomedicine may offer novel strategies for disease intervention.
Collapse
Affiliation(s)
| | | | - Andrew C. Liu
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
38
|
O'Hearn LA. The therapeutic properties of ketogenic diets, slow-wave sleep, and circadian synchrony. Curr Opin Endocrinol Diabetes Obes 2021; 28:503-508. [PMID: 34269711 DOI: 10.1097/med.0000000000000660] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW To summarize emerging connections between sleep, ketogenic diets, and health. RECENT FINDINGS Mechanisms involved in the therapeutic benefits of ketogenic diets continue to be elucidated. Concurrently, the importance of sleep quality and circadian rhythms in their effects on metabolic and cognitive health is increasingly appreciated. Advances in the understanding of the actions of adenosine, nicotinamide adenine dinucleotide, and slow-wave sleep underscore connections between these areas of research. SUMMARY Many molecular pathways activated during ketogenic diets are known to modulate sleep-wake cycles, circadian rhythms, and sleep stages. Ketogenic diets often have beneficial effects on sleep at the same time as having beneficial effects on particular medical conditions. Enhancement of slow-wave sleep and rejuvenation of circadian programming may be synergistic with or causally involved in the benefits of ketogenic diets.
Collapse
|
39
|
Healy KL, Morris AR, Liu AC. Circadian Synchrony: Sleep, Nutrition, and Physical Activity. FRONTIERS IN NETWORK PHYSIOLOGY 2021; 1:732243. [PMID: 35156088 PMCID: PMC8830366 DOI: 10.3389/fnetp.2021.732243] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/20/2021] [Indexed: 08/01/2023]
Abstract
The circadian clock in mammals regulates the sleep/wake cycle and many associated behavioral and physiological processes. The cellular clock mechanism involves a transcriptional negative feedback loop that gives rise to circadian rhythms in gene expression with an approximately 24-h periodicity. To maintain system robustness, clocks throughout the body must be synchronized and their functions coordinated. In mammals, the master clock is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. The SCN is entrained to the light/dark cycle through photic signal transduction and subsequent induction of core clock gene expression. The SCN in turn relays the time-of-day information to clocks in peripheral tissues. While the SCN is highly responsive to photic cues, peripheral clocks are more sensitive to non-photic resetting cues such as nutrients, body temperature, and neuroendocrine hormones. For example, feeding/fasting and physical activity can entrain peripheral clocks through signaling pathways and subsequent regulation of core clock genes and proteins. As such, timing of food intake and physical activity matters. In an ideal world, the sleep/wake and feeding/fasting cycles are synchronized to the light/dark cycle. However, asynchronous environmental cues, such as those experienced by shift workers and frequent travelers, often lead to misalignment between the master and peripheral clocks. Emerging evidence suggests that the resulting circadian disruption is associated with various diseases and chronic conditions that cause further circadian desynchrony and accelerate disease progression. In this review, we discuss how sleep, nutrition, and physical activity synchronize circadian clocks and how chronomedicine may offer novel strategies for disease intervention.
Collapse
|
40
|
Yong SJ, Yong MH, Teoh SL, Soga T, Parhar I, Chew J, Lim WL. The Hippocampal Vulnerability to Herpes Simplex Virus Type I Infection: Relevance to Alzheimer's Disease and Memory Impairment. Front Cell Neurosci 2021; 15:695738. [PMID: 34483839 PMCID: PMC8414573 DOI: 10.3389/fncel.2021.695738] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) as a possible infectious etiology in Alzheimer’s disease (AD) has been proposed since the 1980s. The accumulating research thus far continues to support the association and a possible causal role of HSV-1 in the development of AD. HSV-1 has been shown to induce neuropathological and behavioral changes of AD, such as amyloid-beta accumulation, tau hyperphosphorylation, as well as memory and learning impairments in experimental settings. However, a neuroanatomical standpoint of HSV-1 tropism in the brain has not been emphasized in detail. In this review, we propose that the hippocampal vulnerability to HSV-1 infection plays a part in the development of AD and amnestic mild cognitive impairment (aMCI). Henceforth, this review draws on human studies to bridge HSV-1 to hippocampal-related brain disorders, namely AD and aMCI/MCI. Next, experimental models and clinical observations supporting the neurotropism or predilection of HSV-1 to infect the hippocampus are examined. Following this, factors and mechanisms predisposing the hippocampus to HSV-1 infection are discussed. In brief, the hippocampus has high levels of viral cellular receptors, neural stem or progenitor cells (NSCs/NPCs), glucocorticoid receptors (GRs) and amyloid precursor protein (APP) that support HSV-1 infectivity, as well as inadequate antiviral immunity against HSV-1. Currently, the established diseases HSV-1 causes are mucocutaneous lesions and encephalitis; however, this review revises that HSV-1 may also induce and/or contribute to hippocampal-related brain disorders, especially AD and aMCI/MCI.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Min Hooi Yong
- Department of Psychology, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia.,Aging Health and Well-being Research Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Tomoko Soga
- Jeffrey Cheah School of Medicine and Health Sciences, Brain Research Institute Monash Sunway, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ishwar Parhar
- Jeffrey Cheah School of Medicine and Health Sciences, Brain Research Institute Monash Sunway, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jactty Chew
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia.,Aging Health and Well-being Research Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| |
Collapse
|
41
|
Choudhury ME, Miyanishi K, Takeda H, Tanaka J. Microglia and the Aging Brain: Are Geriatric Microglia Linked to Poor Sleep Quality? Int J Mol Sci 2021; 22:ijms22157824. [PMID: 34360590 PMCID: PMC8345993 DOI: 10.3390/ijms22157824] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022] Open
Abstract
Poor sleep quality and disrupted circadian behavior are a normal part of aging and include excessive daytime sleepiness, increased sleep fragmentation, and decreased total sleep time and sleep quality. Although the neuronal decline underlying the cellular mechanism of poor sleep has been extensively investigated, brain function is not fully dependent on neurons. A recent antemortem autographic study and postmortem RNA sequencing and immunohistochemical studies on aged human brain have investigated the relationship between sleep fragmentation and activation of the innate immune cells of the brain, microglia. In the process of aging, there are marked reductions in the number of brain microglial cells, and the depletion of microglial cells disrupts circadian rhythmicity of brain tissue. We also showed, in a previous study, that pharmacological suppression of microglial function induced sleep abnormalities. However, the mechanism underlying the contribution of microglial cells to sleep homeostasis is only beginning to be understood. This review revisits the impact of aging on the microglial population and activation, as well as microglial contribution to sleep maintenance and response to sleep loss. Most importantly, this review will answer questions such as whether there is any link between senescent microglia and age-related poor quality sleep and how this exacerbates neurodegenerative disease.
Collapse
Affiliation(s)
- Mohammed E. Choudhury
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Shitsukawa, Toon 791-0295, Ehime, Japan
- Correspondence: (M.E.C.); (J.T.)
| | - Kazuya Miyanishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan;
| | - Haruna Takeda
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Aoba, Sendai 980-8575, Miyagi, Japan;
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Shitsukawa, Toon 791-0295, Ehime, Japan
- Correspondence: (M.E.C.); (J.T.)
| |
Collapse
|
42
|
Bhagavan SM, Sahota PK. Sleep Fragmentation and Atherosclerosis: is There a Relationship? MISSOURI MEDICINE 2021; 118:272-276. [PMID: 34149089 PMCID: PMC8210987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Sleep fragmentation refers to the disruption of sleep architecture with poor quality of sleep despite optimal duration of sleep. Sleep fragmentation has been shown to have multiple effects on different body systems. This article reviews the effect of sleep fragmentation on the rate of atherosclerosis which has been linked to comorbidities like myocardial infarction, stroke, and coronary artery disease with an aim to educate patients regarding the importance of sleep hygiene and to incorporate a good amount and quality of sleep as life style modification along with diet and exercise.
Collapse
Affiliation(s)
- Sachin M Bhagavan
- Resident Physician in the Department of Neurology, University of Missouri-Columbia, Columbia, Missouri (UMC CM)
| | - Pradeep K Sahota
- Professor and Chairman, Department of Neurology and Director, Sleep Disorder Center, UMC CM
| |
Collapse
|
43
|
Qiu Y, Yu L, Ge X, Sun Y, Wang Y, Wu X, Xu Q, Zhou Y, Xu J. Loss of Integrity of Corpus Callosum White Matter Hyperintensity Penumbra Predicts Cognitive Decline in Patients With Subcortical Vascular Mild Cognitive Impairment. Front Aging Neurosci 2021; 13:605900. [PMID: 33679371 PMCID: PMC7930322 DOI: 10.3389/fnagi.2021.605900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 01/25/2021] [Indexed: 12/04/2022] Open
Abstract
Loss of white matter (WM) integrity contributes to subcortical vascular mild cognitive impairment (svMCI). Diffusion tensor imaging (DTI) has revealed damage beyond the area of WM hyperintensity (WMH) including in normal-appearing WM (NAWM); however, the functional significance of this observation is unclear. To answer this question, in this study we investigated the relationship between microstructural changes in the WMH penumbra (WMH-P) and cognitive function in patients with svMCI by regional tract-based analysis. A total of 111 patients with svMCI and 72 patients with subcortical ischemic vascular disease (SIVD) without cognitive impairment (controls) underwent DTI and neuropsychological assessment. WMH burden was determined before computing mean values of fractional anisotropy (FA) and mean diffusivity (MD) within WMHs and WMH-Ps. Pearson’s partial correlations were used to assess the relationship between measurements showing significant intergroup differences and composite Z-scores representing global cognitive function. Multiple linear regression analysis was carried out to determine the best model for predicting composite Z-scores. We found that WMH burden in the genu, body, and splenium of the corpus callosum (GCC, BCC, and SCC respectively); bilateral anterior, superior, and posterior corona radiata; left sagittal stratum was significantly higher in the svMCI group than in the control group (p < 0.05). The WMH burden of the GCC, BCC, SCC, and bilateral anterior corona radiata was negatively correlated with composite Z-scores. Among diffusion parameters showing significant differences across the 10 WM regions, mean FA values of WMH and WMH-P of the BCC were correlated with composite Z-scores in svMCI patients. The results of the multiple linear regression analysis showed that the FA of WMH-P of the BCC and WMH burden of the SCC and GCC were independent predictors of composite Z-score, with the FA of WMH-P of the BCC making the largest contribution. These findings indicate that disruption of the CC microstructure—especially the WMH-P of the BCC—may contribute to the cognitive deficits associated with SIVD.
Collapse
Affiliation(s)
- Yage Qiu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Yu
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Ge
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yawen Sun
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Wang
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowei Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qun Xu
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Health Manage Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhou
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianrong Xu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
44
|
Harris SS, Schwerd-Kleine T, Lee BI, Busche MA. The Reciprocal Interaction Between Sleep and Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1344:169-188. [PMID: 34773232 DOI: 10.1007/978-3-030-81147-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
It is becoming increasingly recognized that patients with a variety of neurodegenerative diseases exhibit disordered sleep/wake patterns. While sleep impairments have typically been thought of as sequelae of underlying neurodegenerative processes in sleep-wake cycle regulating brain regions, including the brainstem, hypothalamus, and basal forebrain, emerging evidence now indicates that sleep deficits may also act as pathophysiological drivers of brain-wide disease progression. Specifically, recent work has indicated that impaired sleep can impact on neuronal activity, brain clearance mechanisms, pathological build-up of proteins, and inflammation. Altered sleep patterns may therefore be novel (potentially reversible) dynamic functional markers of proteinopathies and modifiable targets for early therapeutic intervention using non-invasive stimulation and behavioral techniques. Here we highlight research describing a potentially reciprocal interaction between impaired sleep and circadian patterns and the accumulation of pathological signs and features in Alzheimer's disease, the most prevalent neurodegenerative disease in the elderly.
Collapse
Affiliation(s)
| | | | - Byung Il Lee
- UK Dementia Research Institute at UCL, London, UK
| | | |
Collapse
|
45
|
Liu Y, Han J, Ning L, Chen L, Jiang X, Ke A, Zhang Y. Cognitive function and life quality of patients with moderate-to-severe obstructive sleep apnea-hypopnea syndrome in China. Expert Rev Respir Med 2020; 15:435-440. [PMID: 33315466 DOI: 10.1080/17476348.2021.1852081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Objective: To investigate the correlation between cognitive function and quality of life in patients with moderate to severe OSAHS, and to further analyze the impact of cognitive domains on QoL in patients with OSAHS.Methods: Pearson analysis was applied to explore the correlation between MoCA score and SAQLI score. It was conducted by collecting the clinical characteristics of patients and the Epworth Sleepiness Scale and Sleep Quality Scale. T-test, a non-parametric test (Mann-Whitney) or chi-square test, and Pearson analysis were used to analyze the relationship between MoCA score and patient clinical characteristics, sleep variables, and SAQLI score.Results: The SAQLI score of the MCI group significantly decreased. Patients in the MCI group had a higher WHR, higher alcohol intake, and a history of hypertension were firmly related to the poor SAQLI (P < 0.05). The Epworth sleepiness scale (ESS) score increased in the MCI group, and the AHI and ODI elevated significantly, which was related to the poorer SAQLI as well (P < 0.05).Conclusion: Cognitive impairment in OSAHS patients is related to QoL reduction.Keywords: cognitive function; Montreal cognitive assessment; obstructive sleep apnea hypopnea syndrome; quality of life; sleep apnea quality of life index.
Collapse
Affiliation(s)
- YeHai Liu
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - JiangYin Han
- Department of Nursing, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Lin Ning
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - LiJuan Chen
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Xiao Jiang
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - AnRan Ke
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - YinHua Zhang
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
46
|
Rockwood K, Andrew MK, Aubertin‐Leheudre M, Belleville S, Bherer L, Bowles SK, Kehler DS, Lim A, Middleton L, Phillips N, Wallace LM. CCCDTD5: Reducing the risk of later-life dementia. Evidence informing the Fifth Canadian Consensus Conference on the Diagnosis and Treatment of Dementia (CCCDTD-5). ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12083. [PMID: 33204818 PMCID: PMC7656906 DOI: 10.1002/trc2.12083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 11/23/2022]
Abstract
The Fifth Canadian Consensus Conference on the Diagnosis and Treatment of Dementia (CCCDTD-5) was a year-long process to synthesize the best available evidence on several topics. Our group undertook evaluation of risk reduction, in eight domains: nutrition; physical activity; hearing; sleep; cognitive training and stimulation; social engagement and education; frailty; and medications. Here we describe the rationale for the undertaking and summarize the background evidence-this is also tabulated in the Appendix. We further comment specifically on the relationship between age and dementia, and offer some suggestions for how reducing the risk of dementia in the seventh decade and beyond might be considered if we are to improve prospects for prevention in the near term. We draw to attention that a well-specified model of success in dementia prevention need not equate to the elimination of cognitive impairment in late life.
Collapse
Affiliation(s)
- Kenneth Rockwood
- Division of Geriatric MedicineDalhousie UniversityHalifaxNova ScotiaCanada
| | - Melissa K. Andrew
- Division of Geriatric MedicineDalhousie UniversityHalifaxNova ScotiaCanada
| | | | - Sylvie Belleville
- Research CenterInstitut Universitaire de Gériatrie de MontréalMontréalQuebecCanada
- Psychology DepartmentUniversité de MontréalMontréalCanada
| | - Louis Bherer
- Département de Médecine, Faculté de médecine, Université de Montréal, Centre de recherche, Institut de cardiologie de Montréal, Centre de rechercheInstitut universitaire de gériatrie de MontréalMontréalQuébecCanada
| | - Susan K. Bowles
- Division of Geriatric MedicineDalhousie UniversityHalifaxNova ScotiaCanada
- College of PharmacyDalhousie UniversityHalifaxNova ScotiaCanada
| | - D Scott Kehler
- School of PhysiotherapyDalhousie UniversityHalifaxNova ScotiaCanada
| | - Andrew Lim
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences CentreUniversity of TorontoTorontoOntarioCanada
| | - Laura Middleton
- Department of KinesiologyUniversity of WaterlooWaterlooOntarioCanada
| | - Natalie Phillips
- Department of PsychologyConcordia UniversityMontréalQuébecCanada
| | | |
Collapse
|
47
|
Wallace NK, Pollard F, Savenkova M, Karatsoreos IN. Effect of Aging on Daily Rhythms of Lactate Metabolism in the Medial Prefrontal Cortex of Male Mice. Neuroscience 2020; 448:300-310. [PMID: 32717298 DOI: 10.1016/j.neuroscience.2020.07.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 07/11/2020] [Accepted: 07/18/2020] [Indexed: 01/24/2023]
Abstract
Aging is associated with reduced amplitude and earlier timing of circadian (daily) rhythms in sleep, brain function, and behavior. We examined whether age-related circadian dysfunction extends to the metabolic function of the brain, particularly in the prefrontal cortex (PFC). Using enzymatic amperometric biosensors, we recorded lactate concentration changes in the PFC in Young (7 mos) and Aged (19 mos) freely-behaving C57BL/6N male mice. Both Young and Aged mice displayed diurnal and circadian rhythms of lactate, with the Aged rhythm slightly phase advanced. Under constant conditions, the Aged rhythm showed a reduced amplitude not seen in the Young mice. We simultaneously observed a relationship between arousal state and PFC lactate rhythm via electroencephalography, which was modified by aging. Finally, using RT-qPCR, we found that aging affects the daily expression pattern of Glucose Transporter 1 (GLUT-1).
Collapse
Affiliation(s)
- Naomi K Wallace
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Felicity Pollard
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Marina Savenkova
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Ilia N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
48
|
Montero‐Odasso M, Pieruccini‐Faria F, Ismail Z, Li K, Lim A, Phillips N, Kamkar N, Sarquis‐Adamson Y, Speechley M, Theou O, Verghese J, Wallace L, Camicioli R. CCCDTD5 recommendations on early non cognitive markers of dementia: A Canadian consensus. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2020; 6:e12068. [PMID: 33094146 PMCID: PMC7568425 DOI: 10.1002/trc2.12068] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022]
Abstract
Introduction Cognitive impairment is the hallmark of Alzheimer's disease (AD) and related dementias. However, motor decline has been recently described as a prodromal state that can help to detect at‐risk individuals. Similarly, sensory changes, sleep and behavior disturbances, and frailty have been associated with higher risk of developing dementia. These clinical findings, together with the recognition that AD pathology precedes the diagnosis by many years, raises the possibility that non‐cognitive changes may be early and non‐invasive markers for AD or, even more provocatively, that treating non‐cognitive aspects may help to prevent or treat AD and related dementias. Methods A subcommittee of the Canadian Consensus Conference on Diagnosis and Treatment of Dementia reviewed areas of emerging evidence for non‐cognitive markers of dementia. We examined the literature for five non‐cognitive domains associated with future dementia: motor, sensory (hearing, vision, olfaction), neuro‐behavioral, frailty, and sleep. The Grading of Recommendations Assessment, Development, and Evaluation system was used to assign the strength of the evidence and quality of the recommendations. We provide recommendations to primary care clinics and to specialized memory clinics, answering the following main questions: (1) What are the non‐cognitive and functional changes associated with risk of developing dementia? and (2) What is the evidence that sensory, motor, behavioral, sleep, and frailty markers can serve as potential predictors of dementia? Results Evidence supported that gait speed, dual‐task gait speed, grip strength, frailty, neuropsychiatric symptoms, sleep measures, and hearing loss are predictors of dementia. There was insufficient evidence for recommending assessing olfactory and vision impairments as a predictor of dementia. Conclusions Non‐cognitive markers can assist in identifying people at risk for cognitive decline or dementia. These non‐cognitive markers may represent prodromal symptoms and several of them are potentially amenable to treatment that might delay the onset of cognitive decline.
Collapse
Affiliation(s)
- Manuel Montero‐Odasso
- Gait and Brain LaboratoryParkwood InstituteLawson Health Research InstituteLondonOntarioCanada
- Division of Geriatric MedicineDepartment of MedicineSchulich School of Medicine and DentistryLondonOntarioCanada
- Department of Epidemiology and BiostatisticsUniversity of Western OntarioLondonOntarioCanada
| | - Frederico Pieruccini‐Faria
- Gait and Brain LaboratoryParkwood InstituteLawson Health Research InstituteLondonOntarioCanada
- Division of Geriatric MedicineDepartment of MedicineSchulich School of Medicine and DentistryLondonOntarioCanada
| | - Zahinoor Ismail
- Departments of PsychiatryClinical Neurosciencesand Community Health SciencesHotchkiss Brain Institute and O'Brien Institute for Public HealthUniversity of CalgaryCalgaryAlbertaCanada
| | - Karen Li
- Centre for Research in Human DevelopmentConcordia UniversityMontrealQuebecCanada
- Department of PsychologyConcordia UniversityQuebecCanada
| | - Andrew Lim
- Division of NeurologyDepartment of MedicineSunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Natalie Phillips
- Centre for Research in Human DevelopmentConcordia UniversityMontrealQuebecCanada
- Department of PsychologyConcordia UniversityQuebecCanada
| | - Nellie Kamkar
- Gait and Brain LaboratoryParkwood InstituteLawson Health Research InstituteLondonOntarioCanada
| | - Yanina Sarquis‐Adamson
- Gait and Brain LaboratoryParkwood InstituteLawson Health Research InstituteLondonOntarioCanada
| | - Mark Speechley
- Department of Epidemiology and BiostatisticsUniversity of Western OntarioLondonOntarioCanada
| | - Olga Theou
- School of PhysiotherapyDalhousie UniversityHalifaxNova ScotiaCanada
- Department of MedicineDalhousie UniversityHalifaxNova ScotiaCanada
| | - Joe Verghese
- Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Lindsay Wallace
- Department of MedicineDalhousie UniversityHalifaxNova ScotiaCanada
| | - Richard Camicioli
- Division of NeurologyDepartment of MedicineUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
49
|
Li Z, Li J, Yu G, Yu F, Li K, Szanton S. The effect of resistance training on sleep in Chinese older adults: A randomized controlled trial. Geriatr Nurs 2020; 42:289-294. [PMID: 32977992 DOI: 10.1016/j.gerinurse.2020.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
Sleep disturbances affect approximately half of the older adult population and add additional risks of developing Alzheimer's disease. This study is to test the effects of a 12-week resistance training (RT) program on sleep in older adults residing in an assisted living facility in China. A total of 62 eligible participants were randomly assigned to RT (n = 31) or control group (n = 31). Participants in RT group participated in three 1-hour moderate intensity RT sessions per week for 12 weeks (at least 48 h between sessions). In the overall sample, sleep efficiency increased (P < 0.01), wake after sleep onset (P < 0.01) decreased in participants in RT group at post-intervention, compared to control group. Similar results were found in participants with mild cognitive impairment (MCI). The findings support that RT improves sleep efficiency and decreases sleep fragmentation in older adults in general and in those with MCI.
Collapse
Affiliation(s)
- Zhihui Li
- School of Nursing, Jilin University, 965 Xinjiang Street, Changchun 130021, China; Nursing department, Qingdao Women and Children's Hospital, No.6 Tongfu Road, Shibei District, Qingdao 266011, China
| | - Junxin Li
- School of Nursing, Johns Hopkins University, Baltimore, MD, United States
| | - Guiling Yu
- Nursing department, Qingdao Women and Children's Hospital, No.6 Tongfu Road, Shibei District, Qingdao 266011, China
| | - Fang Yu
- School of Nursing, University of Minnesota, 5-140 Weaver-Densford Hall 308 Harvard Street SE Minneapolis, MN 55455, United States
| | - Kun Li
- School of Nursing, Jilin University, 965 Xinjiang Street, Changchun 130021, China.
| | - Sarah Szanton
- School of Nursing, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
50
|
Ennerfelt HE, Lukens JR. The role of innate immunity in Alzheimer's disease. Immunol Rev 2020; 297:225-246. [PMID: 32588460 PMCID: PMC7783860 DOI: 10.1111/imr.12896] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/23/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
The amyloid hypothesis has dominated Alzheimer's disease (AD) research for almost 30 years. This hypothesis hinges on the predominant clinical role of the amyloid beta (Aβ) peptide in propagating neurofibrillary tangles (NFTs) and eventual cognitive impairment in AD. Recent research in the AD field has identified the brain-resident macrophages, known as microglia, and their receptors as integral regulators of both the initiation and propagation of inflammation, Aβ accumulation, neuronal loss, and memory decline in AD. Emerging studies have also begun to reveal critical roles for distinct innate immune pathways in AD pathogenesis, which has led to great interest in harnessing the innate immune response as a therapeutic strategy to treat AD. In this review, we will highlight recent advancements in our understanding of innate immunity and inflammation in AD onset and progression. Additionally, there has been mounting evidence suggesting pivotal contributions of environmental factors and lifestyle choices in AD pathogenesis. Therefore, we will also discuss recent findings, suggesting that many of these AD risk factors influence AD progression via modulation of microglia and immune responses.
Collapse
Affiliation(s)
- Hannah E. Ennerfelt
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|