1
|
Wang Z, Wu D, Hu X, Hu X, Zhu Q, Lai B, Zeng C, Long Q. WuYou decoction effectively reduces neuronal damage, synaptic dysfunction, and Aβ production in rats exposed to chronic sleep deprivation by modulating the Aβ-related enzymes and SIRT1/Nrf2/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118939. [PMID: 39413939 DOI: 10.1016/j.jep.2024.118939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic sleep deprivation (CSD) can result in neuronal damage, synaptic dysfunction, Aβ production, neuroinflammation, and ultimately cognitive deterioration. WuYou Decoction (WYD), a contemporary prescription, has shown promise in enhancing sleep quality and cognitive performance in individuals with insomnia. However, the specific molecular mechanisms responsible for the neuroprotective effects of WYD on CSD remain incompletely understood. AIM OF THE STUDY This study aimed to investigate the neuroprotective effects of WYD on the CSD model and its molecular mechanism. MATERIALS AND METHODS UHPLC-MS/MS analysis was utilized to analyze the active ingredients of WYD extract. The study employed the multi-platform water environment method to establish the CSD model in rats. Subsequent to treatment with varying doses of WYD in CSD rats, cognitive function and pathological alterations in hippocampus and cortex, including neuronal damage, synaptic dysfunction, Aβ production, and neuroinflammation, were evaluated through a combination of Morris Water Maze test, HE staining, Nissl staining, Golgi-Cox staining, Transmission electron microscope, ELISA, Immunohistochemistry staining, Immunofluorescence staining and Western blot. RESULTS UHPLC-MS/MS analysis revealed a total of 99 active ingredients were identified from the WYD extract. The administration of WYD exhibited a mitigation of cognitive decline in the model of CSD, as evidenced by increased neuron count in the hippocampus and cortex, and improved density and length of dendritic spines in these brain regions. Furthermore, WYD was found to suppress the Aβ production, and inhibit the expression of BACE1, PS1, GFAP, IBA1, IL-1β, IL-6, TNF-α, phosphorylated IκBα (Ser32) and phosphorylated NF-κB p65 (Ser536) in the hippocampus and cortex, while also increasing the levels of PSD95, SYN1, ADAM10, IDE, SIRT1 and Nrf2. CONCLUSIONS WYD exhibits neuroprotective properties in CSD, potentially through modulation of the Aβ-related enzymes and SIRT1/Nrf2/NF-κB pathway.
Collapse
Affiliation(s)
- Zhengyu Wang
- Health Medical Center, Hubei Minzu University, Enshi, 445000, PR China
| | - Dan Wu
- Health Medical Center, Hubei Minzu University, Enshi, 445000, PR China
| | - Xinyi Hu
- Health Medical Center, Hubei Minzu University, Enshi, 445000, PR China
| | - Xuan Hu
- Health Medical Center, Hubei Minzu University, Enshi, 445000, PR China
| | - Qihang Zhu
- Health Medical Center, Hubei Minzu University, Enshi, 445000, PR China
| | - Bixuan Lai
- Health Medical Center, Hubei Minzu University, Enshi, 445000, PR China
| | - Chuhua Zeng
- Health Medical Center, Hubei Minzu University, Enshi, 445000, PR China; School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, 650500, PR China.
| | - Qinghua Long
- Health Medical Center, Hubei Minzu University, Enshi, 445000, PR China; Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Hubei Minzu University, Enshi, 445000, PR China.
| |
Collapse
|
2
|
Sun YR, Lv QK, Liu JY, Wang F, Liu CF. New perspectives on the glymphatic system and the relationship between glymphatic system and neurodegenerative diseases. Neurobiol Dis 2025; 205:106791. [PMID: 39778750 DOI: 10.1016/j.nbd.2025.106791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Neurodegenerative diseases (ND) are characterized by the accumulation of aggregated proteins. The glymphatic system, through its rapid exchange mechanisms between cerebrospinal fluid (CSF) and interstitial fluid (ISF), facilitates the movement of metabolic substances within the brain, serving functions akin to those of the peripheral lymphatic system. This emerging waste clearance mechanism offers a novel perspective on the removal of pathological substances in ND. This article elucidates recent discoveries regarding the glymphatic system and updates relevant concepts within its model. It discusses the potential roles of the glymphatic system in ND, including Alzheimer's disease (AD), Parkinson's disease (PD), and multiple system atrophy (MSA), and proposes the glymphatic system as a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Yan-Rui Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Qian-Kun Lv
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Jun-Yi Liu
- Department of Neurology, Dushu Lake hospital affilicated to Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China.
| |
Collapse
|
3
|
Gallo F, Myachykov A, Abutalebi J, DeLuca V, Ellis J, Rothman J, Wheeldon LR. Bilingualism, sleep, and cognition: An integrative view and open research questions. BRAIN AND LANGUAGE 2025; 260:105507. [PMID: 39644806 DOI: 10.1016/j.bandl.2024.105507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Sleep and language are fundamental to human existence and have both been shown to substantially affect cognitive functioning including memory, attentional performance, and cognitive control. Surprisingly, there is little-to-no research that examines the shared impact of bilingualism and sleep on cognitive functions. In this paper, we provide a general overview of existing research on the interplay between bilingualism and sleep with a specific focus on executive functioning. First, we highlight their interconnections and the resulting implications for cognitive performance. Second, we emphasize the need to explore how bilingualism and sleep intersect at cognitive and neural levels, offering insights into potential ways of studying the interplay between sleep, language learning, and bilingual language use. Finally, we suggest that understanding these relationships could enhance our knowledge of reserve and its role in mitigating age-related cognitive decline.
Collapse
Affiliation(s)
- F Gallo
- Uit The Arctic University of Norway, Tromsø, Norway; Higher School of Economics, Moscow, Russian Federation.
| | - A Myachykov
- Higher School of Economics, Moscow, Russian Federation; University of Macau, Taipa, Macau SAR, China
| | - J Abutalebi
- Uit The Arctic University of Norway, Tromsø, Norway; Higher School of Economics, Moscow, Russian Federation; University Vita Salute San Raffaele, Milan, Italy
| | - V DeLuca
- Uit The Arctic University of Norway, Tromsø, Norway
| | - J Ellis
- Northumbria University, Newcastle-upon-Tyne, UK
| | - J Rothman
- Uit The Arctic University of Norway, Tromsø, Norway; Lancaster University, Lancaster, UK; Nebrija Research Center in Cognition, Madrid, Spain
| | | |
Collapse
|
4
|
Mian M, Tahiri J, Habbal S, Aftan F, Reddy PH. The impact of sleep and exercise on brain atrophy in mild cognitive impairment. Mech Ageing Dev 2024; 223:112023. [PMID: 39732176 DOI: 10.1016/j.mad.2024.112023] [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: 07/11/2024] [Revised: 11/15/2024] [Accepted: 12/26/2024] [Indexed: 12/30/2024]
Abstract
Chronic sleep deprivation and lack of physical exercise may have detrimental effects on overall health, particularly in terms of brain health, with significant implications for cognitive function and well-being. This review explores the impact of chronic sleep deprivation and physical exercise on brain atrophy in mild cognitive impairment (MCI) and Alzheimer's disease (AD). Drawing insights from 40 selected studies, the review synthesizes evidence on these lifestyle factors' correlations with neurodegenerative changes. Chronic sleep deprivation disrupts circadian rhythms and neurochemical pathways, potentially accelerating brain atrophy, while physical exercise preserves brain structure by enhancing vascular health, reducing inflammation, and supporting synaptic plasticity, particularly in regions like the hippocampus. Results highlight distinct patterns of brain atrophy in AD and MCI, underscoring the potential for targeted interventions to mitigate cognitive decline. Understanding the relationship between sleep disruption and brain health provides insights into strategies for possibly delaying neurodegenerative diseases like MCI, which represents a milder form of Alzheimer's, and AD. The findings underscore the potential utility of integrating sleep therapy and physical exercise interventions in clinical practice for early detection of mild cognitive impairment and potentially delaying disease progression. This integrated approach has been found to promote healthy aging, reduce atrophy rates, and enhance cognitive resilience across aging populations.
Collapse
Affiliation(s)
- Maamoon Mian
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Jihane Tahiri
- School of Biology, Texas Tech University, Lubbock, TX 79430, USA.
| | - Saadeddine Habbal
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Fatima Aftan
- School of Biology, University of North Texas, Denton, TX 76201, USA.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
5
|
Shang Y, Yu L, Xing H, Chang Y, Dong K, Xiao Y, Liu Y, Feng M, Qin Y, Dai H. Diffusion Tensor Imaging Analysis Along the Perivascular Space (DTI-ALPS) Demonstrates That Sleep Disorders Exacerbate Glymphatic Circulatory Impairment and Cognitive Impairment in Patients with Alzheimer's Disease. Nat Sci Sleep 2024; 16:2205-2215. [PMID: 39735385 PMCID: PMC11675307 DOI: 10.2147/nss.s496607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/15/2024] [Indexed: 12/31/2024] Open
Abstract
Objective Sleep disorders are common in Alzheimer's disease (AD) patients and can impair the glymphatic system, leading to cognitive decline. This study aimed to investigate whether AD patients with sleep disorders exhibit worse glymphatic function and more severe cognitive impairment compared to those without sleep disorders and to explore the underlying molecular imaging mechanisms. Methods This study included 40 AD patients with sleep disorders (ADSD), 39 cognitively matched AD patients without sleep disorders (ADNSD), and 25 healthy middle-aged and elderly controls (NC). Participants underwent functional magnetic resonance imaging (fMRI), and cognitive and sleep assessments. The ALPS (Along the Perivascular Space) index was calculated, followed by intergroup comparisons, correlation analyses, and mediation analyses. The diagnostic utility of the ALPS index was assessed using a receiver operating characteristic (ROC) curve. Results The ALPS index was lower in the ADNSD and ADSD groups compared to the NC group. In the ADSD group, PSQI scores were negatively correlated with MMSE scores. The ALPS index was positively correlated with MMSE scores and negatively with PSQI scores. Mediation analyses indicated that the ALPS index partially mediated the effect of sleep disturbances on cognitive impairment (indirect effect = -0.134; mediation effect = 30.505%). The area under the ROC curve (AUROC) for distinguishing ADSD from ADNSD was 0.86, with a cutoff ALPS index value 1.309. Conclusion Sleep disorders worsen glymphatic function and cognitive impairment in AD patients. The ALPS index partially mediates the impact of sleep disorders on cognitive function and shows moderate accuracy in distinguishing between patients with ADSD and ADNSD.
Collapse
Affiliation(s)
- Yi Shang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Lefan Yu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Hanqi Xing
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Yue Chang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Ke Dong
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Yao Xiao
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Yuanqing Liu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Mengmeng Feng
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Yiren Qin
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Hui Dai
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
- Institute of Medical Imaging, Soochow University, Suzhou, 215006, People’s Republic of China
| |
Collapse
|
6
|
Zhou X, Du K, Mao T, Wang N, Zhang L, Tian Y, Liu T, Wang L, Wang X. BMAL1 upregulates STX17 levels to promote autophagosome-lysosome fusion in hippocampal neurons to ameliorate Alzheimer's disease. iScience 2024; 27:111413. [PMID: 39687016 PMCID: PMC11647228 DOI: 10.1016/j.isci.2024.111413] [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: 05/06/2024] [Revised: 08/02/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
We aim to investigate muscle ARNT-like protein 1 (BMAL1) regulation of syntaxin17 (STX17) in mouse hippocampal neurons, focusing on autophagy and amyloid-β (Aβ) deposition. Autophagosome-lysosome fusion in APP/PS1 hippocampal tissues was observed using transmission electron microscopy, while mRNA levels of LC3II and P62 were measured via reverse-transcription PCR (RT-PCR) after Amyloid precursor protein (APP) overexpression. STX17, linked to autophagy and differentially expressed in Alzheimer's disease (AD) brains, was knocked down or overexpressed to assess its effects. The results showed that reduced STX17 impairs autophagosome-lysosome fusion, leading to abnormal Aβ deposition. Coimmunoprecipitation (Co-IP) and immunofluorescence confirmed STX17 interaction with SNAP29 and VAMP8 to form SNARE complexes. Furthermore, BMAL1 binding to STX17 was examined using luciferase assays. Circadian rhythm disturbances and decreased BMAL1 expression in APP/PS1 mice were noted, while BMAL1 overexpression upregulated STX17 expression and promoted autophagy to reduce Aβ deposition. Thus, the BMAL1 protein can promote STX17 transcription to induce STX17-SNAP29-VAMP8 complex formation to clear intracellular Aβ through autophagy.
Collapse
Affiliation(s)
- Xiuya Zhou
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Kaili Du
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Tian Mao
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Ning Wang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Lifei Zhang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Yuan Tian
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
| | - Ting Liu
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Li Wang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Xiaohui Wang
- Basic Medical Sciences Center, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
7
|
Hauglund NL, Andersen M, Tokarska K, Radovanovic T, Kjaerby C, Sørensen FL, Bojarowska Z, Untiet V, Ballestero SB, Kolmos MG, Weikop P, Hirase H, Nedergaard M. Norepinephrine-mediated slow vasomotion drives glymphatic clearance during sleep. Cell 2024:S0092-8674(24)01343-6. [PMID: 39788123 DOI: 10.1016/j.cell.2024.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/29/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025]
Abstract
As the brain transitions from wakefulness to sleep, processing of external information diminishes while restorative processes, such as glymphatic removal of waste products, are activated. Yet, it is not known what drives brain clearance during sleep. We here employed an array of technologies and identified tightly synchronized oscillations in norepinephrine, cerebral blood volume, and cerebrospinal fluid (CSF) as the strongest predictors of glymphatic clearance during NREM sleep. Optogenetic stimulation of the locus coeruleus induced anti-correlated changes in vasomotion and CSF signal. Furthermore, stimulation of arterial oscillations enhanced CSF inflow, demonstrating that vasomotion acts as a pump driving CSF into the brain. On the contrary, the sleep aid zolpidem suppressed norepinephrine oscillations and glymphatic flow, highlighting the critical role of norepinephrine-driven vascular dynamics in brain clearance. Thus, the micro-architectural organization of NREM sleep, driven by norepinephrine fluctuations and vascular dynamics, is a key determinant for glymphatic clearance.
Collapse
Affiliation(s)
- Natalie L Hauglund
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; Danish Center for Sleep Medicine, Department of Clinical Neurophysiology, Rigshospitalet, 2600 Glostrup, Denmark
| | - Mie Andersen
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Klaudia Tokarska
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Tessa Radovanovic
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Celia Kjaerby
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Frederikke L Sørensen
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Zuzanna Bojarowska
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Verena Untiet
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Sheyla B Ballestero
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Mie G Kolmos
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Hajime Hirase
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Copenhagen, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester, Rochester, NY 14627, USA.
| |
Collapse
|
8
|
Inami S, Koh K. Sleep induced by mechanosensory stimulation provides cognitive and health benefits in Drosophila. Sleep 2024; 47:zsae226. [PMID: 39331490 DOI: 10.1093/sleep/zsae226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/08/2024] [Indexed: 09/29/2024] Open
Abstract
STUDY OBJECTIVES Sleep is a complex phenomenon regulated by various factors, including sensory input. Anecdotal observations have suggested that gentle rocking helps babies fall asleep, and experimental studies have verified that rocking promotes sleep in both humans and mice. Recent studies have expanded this understanding, demonstrating that gentle vibration also induces sleep in Drosophila. Natural sleep serves multiple functions, including learning and memory, synaptic downscaling, and reduction of harmful substances associated with neurodegenerative diseases. Here, we investigated whether vibration-induced sleep (VIS) provides similar cognitive and health benefits in Drosophila. METHODS We administered gentle vibration to flies that slept very little due to a forced activation of wake-promoting neurons and investigated how the vibration influenced learning and memory in the courtship conditioning paradigm. Additionally, we examined the effects of VIS on synaptic downscaling by counting synaptic varicosities of select neurons. Finally, we determined whether vibration could induce sleep in Drosophila models of Alzheimer's disease (AD) and suppress the accumulation of Amyloid β (Aβ) and Tubulin Associated Unit (TAU). RESULTS VIS enhanced performance in a courtship conditioning paradigm and reduced the number of synaptic varicosities in select neurons. Moreover, vibration improved sleep in Drosophila models of AD, reducing Aβ and TAU levels. CONCLUSIONS Mechanosensory stimulation offers a promising noninvasive avenue for enhancing sleep, potentially providing associated cognitive and health benefits.
Collapse
Affiliation(s)
- Sho Inami
- Department of Neuroscience and the Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, USA
| | - Kyunghee Koh
- Department of Neuroscience and the Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
9
|
Seo CR, Lee BK, Jee HJ, Yoo JR, Lee CK, Park JW, Jung YS. Ameliorating Effect of Fermented Perilla frutescens on Sleep Deprivation-Induced Cognitive Impairment Through Antioxidant and BDNF Signaling in Mice. Nutrients 2024; 16:4224. [PMID: 39683616 DOI: 10.3390/nu16234224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Adequate sleep is essential for maintaining cognitive function, as evidenced by literature. Perilla frutescens var. acuta Kudo (PF) is a traditional medicinal herb reported to improve vascular cognitive impairment and induce sedation. However, the effects of PF on cognitive impairment caused by sleep deprivation (SD) have not yet been evaluated. This study aims to evaluate the effects of fermented PF (FPF) and its underlying mechanisms in a model of SD-induced cognitive impairment. Methods: Mice were subjected to SD to establish cognitive impairment, and FPF was administered once daily for 3 days. Cognitive performance was assessed using Y-maze and passive avoidance tests, followed by molecular mechanisms analyses. Results: FPF treatment improved SD-induced cognitive impairment, as evidenced by increased spontaneous alternation and extended latency time. Histological analysis revealed that SD impaired the hippocampus, and this impairment was alleviated by FPF treatment. FPF demonstrated antioxidant activity by increasing glutathione levels and decreasing malondialdehyde levels. Furthermore, the decreased levels of brain-derived neurotrophic factor (BDNF) observed in sleep-deprived mice were restored with FPF treatment. FPF also enhanced the phosphorylation of tropomyosin receptor kinase B, extracellular signal-regulated kinase, and cAMP response element-binding protein. Conclusions: These results indicate that FPF may have beneficial effects on SD-induced cognitive impairment by protecting against oxidative stress and increasing BDNF expression.
Collapse
Affiliation(s)
- Chae-Ryeong Seo
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Bo Kyung Lee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Hye Jin Jee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Jae Ryeong Yoo
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Chul-Kyu Lee
- Headquarters of New Drug Development Support, Corestemchemon Inc., 15 F, Gyeonggi Bio Center, Suwon 16229, Republic of Korea
| | - Jin Wook Park
- Headquarters of New Drug Development Support, Corestemchemon Inc., 15 F, Gyeonggi Bio Center, Suwon 16229, Republic of Korea
| | - Yi-Sook Jung
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
10
|
Wong R, Grullon JR. Sleep disturbances and racial-ethnic disparities in 10-year dementia risk among a national sample of older adults in the USA. BJPsych Open 2024; 10:e219. [PMID: 39629606 PMCID: PMC11698159 DOI: 10.1192/bjo.2024.814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 10/07/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Race/ethnicity and sleep disturbances are associated with dementia risk. AIMS To explore racial-ethnic disparities in sleep disturbances, and whether race/ethnicity moderates the relationship between sleep disturbances and dementia risk among older adults. METHOD We analysed ten annual waves (2011-2020) of prospective cohort data from the National Health and Aging Trends Study, a nationally representative USA sample of 6284 non-Hispanic White (n = 4394), non-Hispanic Black (n = 1311), Hispanic (n = 342) and non-Hispanic Asian (n = 108) community-dwelling older adults. Sleep disturbances were converted into three longitudinal measures: (a) sleep-initiation difficulty (trouble falling asleep within 30 min), (b) sleep-maintenance difficulty (trouble falling asleep after waking up early) and (c) sleep medication usage (taking medication to sleep). Cox proportional hazards models analysed time to dementia, after applying sampling weights and adjusting for sociodemographic characteristics and health. RESULTS Black, Hispanic and Asian respondents exhibited higher frequencies of sleep-initiation and sleep-maintenance difficulties, but had less sleep medication usage, compared with White older adults. Among Hispanic respondents, sleep-initiation difficulty was associated with significantly decreased dementia risk (adjusted hazard ratio (aHR) = 0.34, 95% CI 0.15-0.76), but sleep-maintenance difficulty was associated with increased dementia risk (aHR = 2.68, 95% CI 1.17-6.13), compared with White respondents. Asian respondents using sleep medications had a significantly higher dementia risk (aHR = 3.85, 95% CI 1.64-9.04). There were no significant interactions for Black respondents. CONCLUSIONS Sleep disturbances are more frequent among older Black, Hispanic and Asian adults, and should be considered when addressing dementia disparities. Research is needed to explore how certain sleep disturbances may elevate dementia risk across different racial and ethnic subgroups.
Collapse
Affiliation(s)
- Roger Wong
- Department of Public Health and Preventive Medicine, Norton College of Medicine, SUNY Upstate Medical University, New York, USA; and Department of Geriatrics, SUNY Upstate Medical University, New York; USA
| | | |
Collapse
|
11
|
Zeller CJ, Wunderlin M, Wicki K, Teunissen CE, Nissen C, Züst MA, Klöppel S. Multi-night acoustic stimulation is associated with better sleep, amyloid dynamics, and memory in older adults with cognitive impairment. GeroScience 2024; 46:6157-6172. [PMID: 38744792 PMCID: PMC11493878 DOI: 10.1007/s11357-024-01195-z] [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: 02/19/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Sleep is a potential early, modifiable risk factor for cognitive decline and dementia. Impaired slow wave sleep (SWS) is pronounced in individuals with cognitive impairment (CI). Cognitive decline and impairments of SWS are bi-directionally linked in a vicious cycle. SWS can be enhanced non-invasively using phase-locked acoustic stimulation (PLAS), potentially breaking this vicious cycle. Eighteen healthy older adults (HC, agemean±sd, 68.3 ± 5.1) and 16 older adults (agemean±sd, 71.9 ± 3.9) with CI (Montreal Cognitive Assessment ≤ 25) underwent one baseline (sham-PLAS) night and three consecutive stimulation nights (real-PLAS). EEG responses and blood-plasma amyloid beta Aβ42/Aβ40 ratio were measured pre- and post-intervention, as was episodic memory. The latter was again evaluated 1 week and 3 months after the intervention. In both groups, PLAS induced a significant electrophysiological response in both voltage- and time-frequency analyses, and memory performance improved in association with the magnitude of this response. In the CI group, both electrophysiological and associated memory effects were delayed compared to the healthy group. After 3 intervention nights, electrophysiological response to PLAS was no longer different between CI and HC groups. Only in the CI sample, stronger electrophysiological responses were significantly associated with improving post-intervention Aβ42/Aβ40 ratios. PLAS seems to improve SWS electrophysiology, memory, and amyloid dynamics in older adults with CI. However, effects on memory require more time to unfold compared to healthy older adults. This indicates that PLAS may become a potential tool to ameliorate cognitive decline, but longer interventions are necessary to compensate for declining brain integrity. This study was pre-registered (clinicaltrials.gov: NCT04277104).
Collapse
Affiliation(s)
- Céline J Zeller
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland
- Graduate School for Health Sciences, University of Bern, 3012, Bern, Switzerland
| | - Marina Wunderlin
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland
| | - Korian Wicki
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland
- Graduate School for Health Sciences, University of Bern, 3012, Bern, Switzerland
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, Netherlands
| | - Christoph Nissen
- Division of Psychiatric Specialties, Department of Psychiatry, Geneva University Hospitals (HUG), 1201, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, 1201, Geneva, Switzerland
| | - Marc A Züst
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland.
| | - Stefan Klöppel
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, 3000, Bern 60, Switzerland
| |
Collapse
|
12
|
Pappas JA, Miner B. Sleep Deficiency in the Elderly. Sleep Med Clin 2024; 19:593-606. [PMID: 39455180 DOI: 10.1016/j.jsmc.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
With aging, there are normative changes to sleep physiology and circadian rhythmicity that may predispose older adults to sleep deficiency, whereas many health-related and psychosocial/behavioral factors may precipitate sleep deficiency. In this article, we describe age-related changes to sleep and describe how the health-related and psychosocial/behavioral factors typical of aging may converge in older adults to increase the risk for sleep deficiency. Next, we review the consequences of sleep deficiency in older adults, focusing specifically on important age-related outcomes, including mortality, cognition, depression, and physical function. Finally, we review treatments for sleep deficiency, highlighting safe and effective nonpharmacologic interventions.
Collapse
Affiliation(s)
- Jane Alexandra Pappas
- San Juan Bautista School of Medicine, Salida 21 Carr. 172 Urb. Turabo Gardens, Caguas 00726, Puerto Rico
| | - Brienne Miner
- Section of Geriatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
13
|
Brendstrup‐Brix K, Ulv Larsen SM, Lee H, Knudsen GM. Perivascular space diffusivity and brain microstructural measures are associated with circadian time and sleep quality. J Sleep Res 2024; 33:e14226. [PMID: 38676409 PMCID: PMC11512690 DOI: 10.1111/jsr.14226] [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: 10/18/2023] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
The glymphatic system is centred around brain cerebrospinal fluid flow and is enhanced during sleep, and the synaptic homeostasis hypothesis proposes that sleep acts on brain microstructure by selective synaptic downscaling. While so far primarily studied in animals, we here examine in humans if brain diffusivity and microstructure is related to time of day, sleep quality and cognitive performance. We use diffusion weighted images from 916 young healthy individuals, aged between 22 and 37 years, collected as part of the Human Connectome Project to assess diffusion tensor image analysis along the perivascular space index, white matter fractional anisotropy, intra-neurite volume fraction and extra-neurite mean diffusivity. Next, we examine if these measures are associated with circadian time of acquisition, the Pittsburgh Sleep Quality Index (high scores correspond to low sleep quality) and age-adjusted cognitive function total composite score. Consistent with expectations, we find that diffusion tensor image analysis along the perivascular space index and orbitofrontal grey matter extra-neurite mean diffusivity are negatively and white matter fractional anisotropy positively correlated with circadian time. Further, we find that grey matter intra-neurite volume fraction correlates positively with Pittsburgh Sleep Quality Index, and that this correlation is driven by sleep duration. Finally, we find positive correlations between grey matter intra-neurite volume fraction and cognitive function total composite score, as well as negative interaction effects between cognitive function total composite score and Pittsburgh Sleep Quality Index on grey matter intra-neurite volume fraction. Our findings propose that perivascular flow is under circadian control and that sleep downregulates the intra-neurite volume in healthy adults with positive impact on cognitive function.
Collapse
Affiliation(s)
- Kristoffer Brendstrup‐Brix
- Neurobiology Research UnitCopenhagen University Hospital RigshospitaletCopenhagenDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Sara Marie Ulv Larsen
- Neurobiology Research UnitCopenhagen University Hospital RigshospitaletCopenhagenDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Hong‐Hsi Lee
- Athinoula A. Martinos Center for Biomedical Imaging, Department of RadiologyMassachusetts General HospitalCharlestownMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Gitte Moos Knudsen
- Neurobiology Research UnitCopenhagen University Hospital RigshospitaletCopenhagenDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
14
|
Cankar N, Beschorner N, Tsopanidou A, Qvist FL, Colaço AR, Andersen M, Kjaerby C, Delle C, Lambert M, Mundt F, Weikop P, Jucker M, Mann M, Skotte NH, Nedergaard M. Sleep deprivation leads to non-adaptive alterations in sleep microarchitecture and amyloid-β accumulation in a murine Alzheimer model. Cell Rep 2024; 43:114977. [PMID: 39541211 DOI: 10.1016/j.celrep.2024.114977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Impaired sleep is a common aspect of aging and often precedes the onset of Alzheimer's disease. Here, we compare the effects of sleep deprivation in young wild-type mice and their APP/PS1 littermates, a murine model of Alzheimer's disease. After 7 h of sleep deprivation, both genotypes exhibit an increase in EEG slow-wave activity. However, only the wild-type mice demonstrate an increase in the power of infraslow norepinephrine oscillations, which are characteristic of healthy non-rapid eye movement sleep. Notably, the APP/PS1 mice fail to enhance norepinephrine oscillations 24 h after sleep deprivation, coinciding with an accumulation of cerebral amyloid-β protein. Proteome analysis of cerebrospinal fluid and extracellular fluid further supports these findings by showing altered protein clearance in APP/PS1 mice. We propose that the suppression of infraslow norepinephrine oscillations following sleep deprivation contributes to increased vulnerability to sleep loss and heightens the risk of developing amyloid pathology in early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Neža Cankar
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Natalie Beschorner
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anastasia Tsopanidou
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Filippa L Qvist
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ana R Colaço
- Proteomics Research Infrastructure, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mie Andersen
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Celia Kjaerby
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Christine Delle
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Marius Lambert
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Filip Mundt
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Matthias Mann
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department for Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Niels Henning Skotte
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA.
| |
Collapse
|
15
|
Sighencea MG, Popescu RȘ, Trifu SC. From Fundamentals to Innovation in Alzheimer's Disease: Molecular Findings and Revolutionary Therapies. Int J Mol Sci 2024; 25:12311. [PMID: 39596378 PMCID: PMC11594972 DOI: 10.3390/ijms252212311] [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: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut-brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy.
Collapse
Affiliation(s)
| | - Ramona Ștefania Popescu
- Department of Infectious Diseases, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| | - Simona Corina Trifu
- Department of Psychiatry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
| |
Collapse
|
16
|
Yang Z, Williams SD, Beldzik E, Anakwe S, Schimmelpfennig E, Lewis LD. Attentional failures after sleep deprivation represent moments of cerebrospinal fluid flow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623271. [PMID: 39605725 PMCID: PMC11601381 DOI: 10.1101/2024.11.15.623271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Sleep deprivation rapidly disrupts cognitive function, and in the long term contributes to neurological disease. Why sleep deprivation has such profound effects on cognition is not well understood. Here, we use simultaneous fast fMRI-EEG to test how sleep deprivation modulates cognitive, neural, and fluid dynamics in the human brain. We demonstrate that after sleep deprivation, sleep-like pulsatile cerebrospinal fluid (CSF) flow events intrude into the awake state. CSF flow is coupled to attentional function, with high flow during attentional impairment. Furthermore, CSF flow is tightly orchestrated in a series of brain-body changes including broadband neuronal shifts, pupil constriction, and altered systemic physiology, pointing to a coupled system of fluid dynamics and neuromodulatory state. The timing of these dynamics is consistent with a vascular mechanism regulated by neuromodulatory state, in which CSF begins to flow outward when attention fails, and flow reverses when attention recovers. The attentional costs of sleep deprivation may thus reflect an irrepressible need for neuronal rest periods and widespread pulsatile fluid flow.
Collapse
Affiliation(s)
- Zinong Yang
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
| | - Stephanie D. Williams
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Psychological & Brain Sciences., Boston University, Boston, MA, USA
| | - Ewa Beldzik
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston MA, USA
| | - Stephanie Anakwe
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emilia Schimmelpfennig
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Laura D. Lewis
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
17
|
Gao YZ, Liu K, Wu XM, Shi CN, He QL, Wu HP, Yang JJ, Yao H, Ji MH. Oxidative Stress-mediated Loss of Hippocampal Parvalbumin Interneurons Contributes to Memory Precision Decline After Acute Sleep Deprivation. Mol Neurobiol 2024:10.1007/s12035-024-04628-0. [PMID: 39546120 DOI: 10.1007/s12035-024-04628-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
Sleep is pivotal to memory consolidation, and sleep deprivation (SD) after learning can impede this process, leading to memory disorders. In the present study, we aimed to explore the effects of acute sleep deprivation (ASD) on memory disorders and the underlying mechanisms. ASD model was induced by subjecting the mice to 6 h of SD following fear conditioning training. Different cohorts were used for behavioral, biochemical, and electrophysiological tests. Here, we showed that memory precision decline was induced by ASD, concomitant with a notable elevation in oxidative stress within PV interneurons, loss of PV, and disturbed neuronal oscillation in the CA1 region. Notably, chemogenetic activation of PV interneurons effectively ameliorated abnormal gamma oscillation and memory precision decline observed in ASD mice. Meanwhile, chemogenetic inhibition of PV interneurons successfully mimicked the abnormal brain oscillations and memory precision decline observed in ASD mice. Additionally, prior administration of the antioxidant medication N-acetylcysteine effectively reversed memory precision decline and mitigated PV loss and abnormal oscillation triggered by ASD. Collectively, our findings indicated that ASD increased oxidative stress in PV interneurons, thereby disrupting neural oscillation in the CA1 and ultimately leading to memory precision decline.
Collapse
Affiliation(s)
- Yu-Zhu Gao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kai Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Miao Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cui-Na Shi
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiu-Li He
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai-Peng Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Hao Yao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Mu-Huo Ji
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
18
|
Muccio M, Sun Z, Chu D, Damadian BE, Minkoff L, Bonanni L, Ge Y. The impact of body position on neurofluid dynamics: present insights and advancements in imaging. Front Aging Neurosci 2024; 16:1454282. [PMID: 39582951 PMCID: PMC11582045 DOI: 10.3389/fnagi.2024.1454282] [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: 06/24/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024] Open
Abstract
The intricate neurofluid dynamics and balance is essential in preserving the structural and functional integrity of the brain. Key among these forces are: hemodynamics, such as heartbeat-driven arterial and venous blood flow, and hydrodynamics, such as cerebrospinal fluid (CSF) circulation. The delicate interplay between these dynamics is crucial for maintaining optimal homeostasis within the brain. Currently, the widely accepted framework for understanding brain functions is the Monro-Kellie's doctrine, which posits a constant sum of intracranial CSF, blood flow and brain tissue volumes. However, in recent decades, there has been a growing interest in exploring the dynamic interplay between these elements and the impact of external factors, such as daily changes in body position. CSF circulation in particular plays a crucial role in the context of neurodegeneration and dementia, since its dysfunction has been associated with impaired clearance mechanisms and accumulation of toxic substances. Despite the implementation of various invasive and non-invasive imaging techniques to investigate the intracranial hemodynamic or hydrodynamic properties, a comprehensive understanding of how all these elements interact and are influenced by body position remains wanted. Establishing a comprehensive overview of this topic is therefore crucial and could pave the way for alternative care approaches. In this review, we aim to summarize the existing understanding of intracranial hemodynamic and hydrodynamic properties, fundamental for brain homeostasis, along with factors known to influence their equilibrium. Special attention will be devoted to elucidating the effects of body position shifts, given their significance and remaining ambiguities. Furthermore, we will explore recent advancements in imaging techniques utilized for real time and non-invasive measurements of dynamic body fluid properties in-vivo.
Collapse
Affiliation(s)
- Marco Muccio
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, United States
| | - Zhe Sun
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, United States
| | - David Chu
- FONAR Corporation, Melville, NY, United States
| | - Brianna E. Damadian
- Department of Radiology, Northwell Health-Lenox Hill Hospital, New York, NY, United States
| | | | | | - Yulin Ge
- Department of Radiology, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
19
|
Ortiz-Vega N, Lobato AG, Canic T, Zhu Y, Lazopulo S, Syed S, Zhai RG. Regulation of proteostasis by sleep through autophagy in Drosophila models of Alzheimer's disease. Life Sci Alliance 2024; 7:e202402681. [PMID: 39237365 PMCID: PMC11377308 DOI: 10.26508/lsa.202402681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
Sleep and circadian rhythm dysfunctions are common clinical features of Alzheimer's disease (AD). Increasing evidence suggests that in addition to being a symptom, sleep disturbances can also drive the progression of neurodegeneration. Protein aggregation is a pathological hallmark of AD; however, the molecular pathways behind how sleep affects protein homeostasis remain elusive. Here we demonstrate that sleep modulation influences proteostasis and the progression of neurodegeneration in Drosophila models of tauopathy. We show that sleep deprivation enhanced Tau aggregational toxicity resulting in exacerbated synaptic degeneration. In contrast, sleep induction using gaboxadol led to reduced toxic Tau accumulation in neurons as a result of modulated autophagic flux and enhanced clearance of ubiquitinated Tau, suggesting altered protein processing and clearance that resulted in improved synaptic integrity and function. These findings highlight the complex relationship between sleep and regulation of protein homeostasis and the neuroprotective potential of sleep-enhancing therapeutics to slow the progression or delay the onset of neurodegeneration.
Collapse
Affiliation(s)
- Natalie Ortiz-Vega
- Department of Neurology, University of Chicago, Chicago, IL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Graduate Program in Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Amanda G Lobato
- Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Tijana Canic
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Physics, University of Miami, Coral Gables, FL, USA
| | - Yi Zhu
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL, USA
| | - R Grace Zhai
- Department of Neurology, University of Chicago, Chicago, IL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
20
|
Lacerda RAV, Desio JAF, Kammers CM, Henkes S, Freitas de Sá M, de Souza EF, da Silva DM, Teixeira Pinheiro Gusmão C, Santos JCCD. Sleep disorders and risk of alzheimer's disease: A two-way road. Ageing Res Rev 2024; 101:102514. [PMID: 39317268 DOI: 10.1016/j.arr.2024.102514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Substantial sleep impairment in patients with Alzheimer's disease (AD) is one of the emerging points for continued efforts to better understand the disease. Individuals without cognitive decline, an important marker of the clinical phase of AD, may show early alterations in the sleep-wake cycle. The objective of this critical narrative review is to explore the bidirectional pathophysiological correlation between sleep disturbances and Alzheimer's Disease. Specifically, it examines how the disruption of sleep homeostasis in individuals without dementia could contribute to the pathogenesis of AD, and conversely, how neurodegeneration in individuals with Alzheimer's Disease might lead to dysregulation of the sleep-wake cycle. Recent scientific results indicate that sleep disturbances, particularly those related to impaired glymphatic clearance, may act as an important mechanism associated with the genesis of Alzheimer's Disease. Additionally, amyloid deposition and tau protein hyperphosphorylation, along with astrocytic hyperactivation, appear to trigger changes in neurotransmission dynamics in areas related to sleep, which may explain the onset of sleep disturbances in individuals with AD. Disruption of sleep homeostasis appears to be a modifiable risk factor in Alzheimer's disease. Whenever possible, the use of non-pharmacological strategies becomes important in this context. From a different perspective, additional research is needed to understand and treat the dysfunction of the sleep-wake cycle in individuals already affected by AD. Early recognition and correction of sleep disturbances in this population could potentially mitigate the progression of dementia and improve the quality of life for those with AD.
Collapse
Affiliation(s)
| | | | | | - Silvana Henkes
- Lutheran University of Brazil - ULBRA, Carazinho, RS, Brazil
| | | | | | | | | | - Júlio César Claudino Dos Santos
- Medical School of the Christus University Center - UNICHRISTUS, Fortaleza, CE, Brazil; Post-Graduate Program of Morphofunctional Sciences, Federal University of Ceara, Fortaleza, CE, Brazil; Unifacvest University Center - UNIFACVEST, Lages, SC, Brazil.
| |
Collapse
|
21
|
Espay AJ. From Europe to the World: EMA's Leadership in Alzheimer Disease Treatment. Am J Ther 2024; 31:e686-e688. [PMID: 39792495 DOI: 10.1097/mjt.0000000000001840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Affiliation(s)
- Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
22
|
Borrelli S, Leclercq S, Pasi M, Maggi P. Cerebral small vessel disease and glymphatic system dysfunction in multiple sclerosis: A narrative review. Mult Scler Relat Disord 2024; 91:105878. [PMID: 39276600 DOI: 10.1016/j.msard.2024.105878] [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: 06/24/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
As the multiple sclerosis (MS) population ages, the prevalence of vascular comorbidities increases, potentially accelerating disease progression and brain atrophy. Recent studies highlight the prevalence of cerebral small vessel disease (CSVD) in MS, suggesting a potential link between vascular comorbidities and accelerated disability. CSVD affects the brain's small vessels, often leading to identifiable markers on MRI such as enlarged perivascular spaces (EPVS). EPVS are increasingly recognized also in MS and have been associated with vascular comorbidities, lower percentage of MS-specific perivenular lesions, brain atrophy and aging. The exact sequence of event leading to MRI visible EPVS is yet to be determined, but an impaired perivascular brain fluid drainage appears a possible physiopathological explanation for EPVS in both CSVD and MS. In this context, a dysfunction of the brain fluid clearance system - also known as "glymphatic system" - appears associated in MS to aging, neuroinflammation, and vascular dysfunction. Advanced imaging techniques show an impaired glymphatic function in both MS and CSVD. Additionally, lifestyle factors such as physical exercise, diet, and sleep quality appear to influence glymphatic function, potentially revealing novel therapeutic strategies to mitigate microangiopathy and neuroinflammation in MS. This review underscores the potential role of glymphatic dysfunction in the complex and not-yet elucidated interplay between neuroinflammation and CSVD in MS.
Collapse
Affiliation(s)
- Serena Borrelli
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium; Department of Neurology, Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Brussels, Brussels, Belgium.
| | - Sophie Leclercq
- Laboratory of Nutritional Psychiatry, Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium
| | - Marco Pasi
- Stroke Unit, Department of Neurology, CIC-IT 1415, CHRU de Tours, INSERM 1253 iBrain, Tours, France
| | - Pietro Maggi
- Neuroinflammation Imaging Lab (NIL), Institute of NeuroScience, Université catholique de Louvain, Brussels, Belgium; Department of Neurology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Av. Hippocrate 10, Brussels 1200, Belgium.
| |
Collapse
|
23
|
Wang X, Chen H, Tang T, Zhan X, Qin S, Hang T, Song M. Chronic Sleep Deprivation Altered the Expression of Memory-Related Genes and Caused Cognitive Memory Dysfunction in Mice. Int J Mol Sci 2024; 25:11634. [PMID: 39519186 PMCID: PMC11546330 DOI: 10.3390/ijms252111634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Lack of sleep, whether acute or chronic, is quite common and negatively affects an individual's memory and cognitive function. The question of whether chronic sleep deprivation (CSD) causes cognitive impairment to arise and progress is not well studied. To investigate the effects of CSD on memory and cognition, this study began by establishing a CSD mouse model. Behavioral experiments on animals revealed that CSD induced cognitive behavioral abnormalities reminiscent of Alzheimer's disease. Western blot experiments further demonstrated a considerable increase in amyloid-β (Aβ) expression in the mouse brain following CSD. Meanwhile, the hub gene Prkcg was searched for in the cerebellum using RNA-seq and bioinformatics analysis. PKCγ (Prkcg) expression was significantly reduced, as demonstrated by RT-qPCR and Western blot validations. Additionally, CSD was associated with downregulated CREB expression, decreased expression of the endothelin-converting enzyme (ECE1), and increased phosphorylation of ERK1/2 downstream of PKCγ. These findings suggested that CSD down-regulated PKCγ expression, decreased ECE1 expression, impaired Aβ degradation, and affected the PKCγ/ERK/CREB pathway and the synthesis of memory-related proteins. Overall, this study highlighted how CSD modulated PKCγ-related metabolism, impacting Aβ clearance and the production of memory-related proteins. Such insights are crucial for understanding and preventing sporadic Alzheimer's disease (sAD) associated with CSD.
Collapse
Affiliation(s)
| | | | | | | | | | - Taijun Hang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 211100, China; (X.W.); (H.C.); (T.T.); (X.Z.); (S.Q.)
| | - Min Song
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 211100, China; (X.W.); (H.C.); (T.T.); (X.Z.); (S.Q.)
| |
Collapse
|
24
|
Ma J, Chen M, Liu GH, Gao M, Chen NH, Toh CH, Hsu JL, Wu KY, Huang CM, Lin CM, Fang JT, Lee SH, Lee TMC. Effects of sleep on the glymphatic functioning and multimodal human brain network affecting memory in older adults. Mol Psychiatry 2024:10.1038/s41380-024-02778-0. [PMID: 39397082 DOI: 10.1038/s41380-024-02778-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
Understanding how sleep affects the glymphatic system and human brain networks is crucial for elucidating the neurophysiological mechanism underpinning aging-related memory declines. We analyzed a multimodal dataset collected through magnetic resonance imaging (MRI) and polysomnographic recording from 72 older adults. A proxy of the glymphatic functioning was obtained from the Diffusion Tensor Image Analysis along the Perivascular Space (DTI-ALPS) index. Structural and functional brain networks were constructed based on MRI data, and coupling between the two networks (SC-FC coupling) was also calculated. Correlation analyses revealed that DTI-ALPS was negatively correlated with sleep quality measures [e.g., Pittsburgh Sleep Quality Index (PSQI) and apnea-hypopnea index]. Regarding human brain networks, DTI-ALPS was associated with the strength of both functional connectivity (FC) and structural connectivity (SC) involving regions such as the middle temporal gyrus and parahippocampal gyrus, as well as with the SC-FC coupling of rich-club connections. Furthermore, we found that DTI-ALPS positively mediated the association between sleep quality and rich-club SC-FC coupling. The rich-club SC-FC coupling further mediated the association between DTI-ALPS and memory function in good sleepers but not in poor sleepers. The results suggest a disrupted glymphatic-brain relationship in poor sleepers, which underlies memory decline. Our findings add important evidence that sleep quality affects cognitive health through the underlying neural relationships and the interplay between the glymphatic system and multimodal brain networks.
Collapse
Affiliation(s)
- Junji Ma
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China
| | - Menglu Chen
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China
| | - Geng-Hao Liu
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Acupuncture and Moxibustion, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Sleep Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Mengxia Gao
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China
| | - Ning-Hung Chen
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Sleep Center, Respiratory Therapy, Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Cheng Hong Toh
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan
| | - Jung-Lung Hsu
- Department of Neurology, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
- Department of Neurology, at Linkou, Chang Gung Memorial Hospital and College of Medicine, Neuroscience Research Center, Chang-Gung University, Taoyuan, Taiwan
- Graduate Institute of Mind, Brain, & Consciousness, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yi Wu
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chih-Mao Huang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chih-Ming Lin
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan
- Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Ji-Tseng Fang
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan.
- Department of Nephrology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Shwu-Hua Lee
- College of Medicine, Chang Gung University, Taoyuan County, Taiwan.
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Tatia M C Lee
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China.
- Laboratory of Neuropsychology & Human Neuroscience, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
25
|
Zhao Q, Yokomizo S, Perle SJ, Lee YF, Zhou H, Miller MR, Li H, Gerashchenko D, Gomperts SN, Bacskai BJ, Kastanenka KV. Optogenetic targeting of cortical astrocytes selectively improves NREM sleep in an Alzheimer's disease mouse model. Sci Rep 2024; 14:23044. [PMID: 39362954 PMCID: PMC11450172 DOI: 10.1038/s41598-024-73082-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 10/05/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition marked by memory impairments and distinct histopathological features such as amyloid-beta (Aβ) accumulations. Alzheimer's patients experience sleep disturbances at early stages of the disease. APPswe/PS1dE9 (APP) mice exhibit sleep disruptions, including reductions in non-rapid eye movement (NREM) sleep, that contribute to their disease progression. In addition, astrocytic calcium transients associated with a sleep-dependent brain rhythm, slow oscillations prevalent during NREM sleep, are disrupted in APP mice. However, at present it is unclear whether restoration of circuit function by targeting astrocytic activity could improve sleep in APP mice. To that end, APP mice expressing channelrhodopsin-2 (ChR2) targeted to astrocytes underwent optogenetic stimulation at the slow oscillation frequency. Optogenetic stimulation of astrocytes significantly increased NREM sleep duration but not duration of rapid eye movement (REM) sleep. Optogenetic treatment increased delta power and reduced sleep fragmentation in APP mice. Thus, optogenetic activation of astrocytes increased sleep quantity and improved sleep quality in an AD mouse model. Astrocytic activity provides a novel therapeutic avenue to pursue for enhancing sleep and slowing AD progression.
Collapse
Affiliation(s)
- Qiuchen Zhao
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Shinya Yokomizo
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Stephen J Perle
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Yee Fun Lee
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Heng Zhou
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Morgan R Miller
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Hanyan Li
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Dmitry Gerashchenko
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, West Roxbury, MA, 02132, USA
| | - Stephen N Gomperts
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Brian J Bacskai
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ksenia V Kastanenka
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| |
Collapse
|
26
|
Nakata S, Iwasaki K, Funato H, Yanagisawa M, Ozaki H. Neuronal subtype-specific transcriptomic changes in the cerebral neocortex associated with sleep pressure. Neurosci Res 2024; 207:13-25. [PMID: 38537682 DOI: 10.1016/j.neures.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024]
Abstract
Sleep is homeostatically regulated by sleep pressure, which increases during wakefulness and dissipates during sleep. Recent studies have suggested that the cerebral neocortex, a six-layered structure composed of various layer- and projection-specific neuronal subtypes, is involved in the representation of sleep pressure governed by transcriptional regulation. Here, we examined the transcriptomic changes in neuronal subtypes in the neocortex upon increased sleep pressure using single-nucleus RNA sequencing datasets and predicted the putative intracellular and intercellular molecules involved in transcriptome alterations. We revealed that sleep deprivation (SD) had the greatest effect on the transcriptome of layer 2 and 3 intratelencephalic (L2/3 IT) neurons among the neocortical glutamatergic neuronal subtypes. The expression of mutant SIK3 (SLP), which is known to increase sleep pressure, also induced profound changes in the transcriptome of L2/3 IT neurons. We identified Junb as a candidate transcription factor involved in the alteration of the L2/3 IT neuronal transcriptome by SD and SIK3 (SLP) expression. Finally, we inferred putative intercellular ligands, including BDNF, LSAMP, and PRNP, which may be involved in SD-induced alteration of the transcriptome of L2/3 IT neurons. We suggest that the transcriptome of L2/3 IT neurons is most impacted by increased sleep pressure among neocortical glutamatergic neuronal subtypes and identify putative molecules involved in such transcriptional alterations.
Collapse
Affiliation(s)
- Shinya Nakata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kanako Iwasaki
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Haruka Ozaki
- Bioinformatics Laboratory, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; Center for Artificial Intelligence Research, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
27
|
Li A, Jaakkola MK, Saaresranta T, Klén R, Li XG. Analysis of sleep apnea research with a special focus on the use of positron emission tomography as a study tool. Sleep Med Rev 2024; 77:101967. [PMID: 38936220 DOI: 10.1016/j.smrv.2024.101967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/20/2024] [Accepted: 06/02/2024] [Indexed: 06/29/2024]
Abstract
The quality of sleep plays a significant role in determining human well-being, and studying sleep and sleep disorders using various methods can aid in the prevention and treatment of diseases. Positron emission tomography (PET) is a noninvasive and highly sensitive medical imaging technique that has been widely adopted in the clinic. This review article provides data on research activity related to sleep and sleep apnea and discusses the use of PET in investigating sleep apnea and other sleep disorders. We conducted a statistical analysis of the number of original research articles published on sleep and sleep apnea between 1965 and 2021 and found that there has been a dramatic increase in publications since 1990. The distribution of contributing countries and regions has also undergone significant changes. Although there is an extensive body of literature on sleep research (256,399 original research articles during 1965-2021), PET has only been used in 54 of these published studies, indicating a largely untapped area of research. Nonetheless, PET is a useful tool for identifying connections between sleep disorders and pathological changes in various diseases, including neurological, metabolic, and cardiovascular disorders, as well as cancer. To facilitate the broader use of PET in sleep apnea research, further studies are needed in both clinical and preclinical settings.
Collapse
Affiliation(s)
- Anting Li
- Turku PET Centre, University of Turku, Turku, Finland; Faculty of Medicine, University of Turku, Turku, Finland
| | - Maria K Jaakkola
- Turku PET Centre, University of Turku, Turku, Finland; Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Tarja Saaresranta
- Division of Medicine, Department of Pulmonary Diseases, Turku University Hospital, Turku, Finland; Sleep Research Centre, Department of Pulmonary Diseases and Clinical Allergology, University of Turku, Turku, Finland
| | - Riku Klén
- Turku PET Centre, University of Turku, Turku, Finland; Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, Turku, Finland; Turku PET Centre, Turku University Hospital, Turku, Finland; InFLAMES Research Flagship, University of Turku, Turku, Finland; Department of Chemistry, University of Turku, Turku, Finland.
| |
Collapse
|
28
|
Ozdinler PH. Sleep Apnea and Amyotrophic Lateral Sclerosis: Cause, Correlation, Any Relation? Brain Sci 2024; 14:978. [PMID: 39451992 PMCID: PMC11505663 DOI: 10.3390/brainsci14100978] [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: 08/30/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a motor neuron disease with progressive neurodegeneration, affecting both the cortical and the spinal component of the motor neuron circuitry in patients. The cellular and molecular basis of selective neuronal vulnerability is beginning to emerge. Yet, there are no effective cures for ALS, which affects more than 200,000 people worldwide each year. Recent studies highlight the importance of the glymphatic system and its proper function for the clearance of the cerebral spinal fluid, which is achieved mostly during the sleep period. Therefore, a potential link between problems with sleep and neurodegenerative diseases has been postulated. This paper discusses the present understanding of this potential correlation.
Collapse
Affiliation(s)
- P Hande Ozdinler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
29
|
Kroesbergen E, Riesselmann LV, Gomolka RS, Plá V, Esmail T, Stenmo VH, Kovács ER, Nielsen ES, Goldman SA, Nedergaard M, Weikop P, Mori Y. Glymphatic clearance is enhanced during sleep. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.24.609514. [PMID: 39314459 PMCID: PMC11418927 DOI: 10.1101/2024.08.24.609514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
We here revisited the concept that glymphatic clearance is enhanced by sleep and anesthesia. Utilizing dynamic magnetic resonance imaging (MRI), single photon emission computed tomography (SPECT), and fluorescent fiber photometry, we report brain glymphatic clearance is enhanced by both sleep and anesthesia, and sharply suppressed by wakefulness. Another key finding was that less tracer enters the brains of awake animals and that brain clearance across different brain states can only be compared after adjusting for the injected tracer dose.
Collapse
|
30
|
Mueller C, Nenert R, Catiul C, Pilkington J, Szaflarski JP, Amara AW. Relationship between sleep, physical fitness, brain microstructure, and cognition in healthy older adults: A pilot study. Brain Res 2024; 1839:149016. [PMID: 38768934 DOI: 10.1016/j.brainres.2024.149016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/01/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND There is a critical need for neuroimaging markers of brain integrity to monitor effects of modifiable lifestyle factors on brain health. This observational, cross-sectional study assessed relationships between brain microstructure and sleep, physical fitness, and cognition in healthy older adults. METHODS Twenty-three adults aged 60 and older underwent whole-brain multi-shell diffusion imaging, comprehensive cognitive testing, polysomnography, and exercise testing. Neurite Orientation Dispersion and Density Imaging (NODDI) was used to quantify neurite density (NDI) and orientation dispersion (ODI). Diffusion tensor imaging (DTI) was used to quantify axial diffusivity (AxD), fractional anisotropy (FA), mean diffusivity (MD), and radial diffusivity (RD). Relationships between sleep efficiency (SE), time and percent in N3 sleep, cognitive function, physical fitness (VO2 peak) and the diffusion metrics in regions of interest and the whole brain were evaluated. RESULTS Higher NDI in bilateral white and gray matter was associated with better executive functioning. NDI in the right anterior cingulate and adjacent white matter was positively associated with language skills. Higher NDI in the left posterior corona radiata was associated with faster processing speed. Physical fitness was positively associated with NDI in the left precentral gyrus and corticospinal tract. N3 % was positively associated with NDI in the left caudate and right pre- and postcentral gyri. Higher ODI in the left putamen and adjacent white matter was associated with better executive function. CONCLUSION NDI and ODI derived from NODDI are potential neuroimaging markers for associations between brain microstructure and modifiable risk factors in aging. If these associations are observable in clinical samples, NODDI could be incorporated into clinical trials assessing the effects of modifiable risk factors on brain integrity in aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Christina Mueller
- University of Alabama at Birmingham, Department of Neurology, 1719 6(th) Ave S, Birmingham, AL 35233, United States.
| | - Rodolphe Nenert
- University of Alabama at Birmingham, Department of Neurology, 1719 6(th) Ave S, Birmingham, AL 35233, United States
| | - Corina Catiul
- University of Alabama at Birmingham, Department of Neurology, 1719 6(th) Ave S, Birmingham, AL 35233, United States
| | - Jennifer Pilkington
- University of Alabama at Birmingham, Department of Neurology, 1719 6(th) Ave S, Birmingham, AL 35233, United States
| | - Jerzy P Szaflarski
- University of Alabama at Birmingham, Department of Neurology, 1719 6(th) Ave S, Birmingham, AL 35233, United States
| | - Amy W Amara
- University of Alabama at Birmingham, Department of Neurology, 1719 6(th) Ave S, Birmingham, AL 35233, United States; University of Colorado Anschutz Medical Campus, 1635 Aurora Ct, Aurora, CO 80045, United States
| |
Collapse
|
31
|
Song Y, Kim S, Joo Y, Ha E, Shim Y, Lee H, Jeong H, Lyoo I, Yoon S, Lee S. Impact of sleep disturbance in shift workers on hippocampal volume and psychomotor speed. Sleep 2024; 47:zsae100. [PMID: 38666299 DOI: 10.1093/sleep/zsae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 04/10/2024] [Indexed: 09/10/2024] Open
Abstract
STUDY OBJECTIVES Shift work interferes with circadian rhythms, affecting sleep quality and cognitive function. Poor sleep quality in shift worker (SW)s can impair psychomotor performance due to fatigue and sleepiness, increasing the risk of errors, accidents, and reduced productivity. Given the potential for atrophic changes in the hippocampus due to sleep disturbances, our study investigates how poor sleep quality correlates with hippocampal structural alterations and impacts psychomotor performance among SWs. METHODS We recruited 100 SWs, classifying them based on sleep quality into two groups: good sleep-SW group (n = 59) and poor sleep-SW group (n = 41). Sleep quality was assessed using both 7-day actigraphy for sleep efficiency and the Pittsburgh Sleep Quality Index. A control group of 106 non-SWs without sleep problems (non-SW group) was also included for comparison. The outcome measures were psychomotor speed and hippocampal volumes, both total and by subfield. RESULTS The poor sleep-SW group showed significantly smaller hippocampal volumes than both the good sleep-SW group (p < .001) and the non-SW group (p = .003). Longer shift work years correlated with greater reductions in hippocampal volume in this group (r = -0.42, p = .009), unlike in the good sleep-SW group (r = 0.08, p = .541). Furthermore, they demonstrated declines in psychomotor speed relative to the non-SW group (p = .006), which correlated with smaller hippocampal volumes (r = 0.37, p = .020). CONCLUSIONS SWs with poor sleep quality exhibit significant hippocampal volume reductions and psychomotor speed decline, underscoring the importance of early intervention and support for sleep issues in this population.
Collapse
Affiliation(s)
- Yumi Song
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Shinhye Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Yoonji Joo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
| | - Eunji Ha
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
| | - Youngeun Shim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Hyeonji Lee
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Hyeonseok Jeong
- Department of Radiology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - InKyoon Lyoo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Sujung Yoon
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Suji Lee
- College of Pharmacy, Dongduk Women's University, Seoul, South Korea
| |
Collapse
|
32
|
Zhang R, Li J, Li X, Zhang S. Therapeutic approaches to CNS diseases via the meningeal lymphatic and glymphatic system: prospects and challenges. Front Cell Dev Biol 2024; 12:1467085. [PMID: 39310229 PMCID: PMC11413538 DOI: 10.3389/fcell.2024.1467085] [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: 07/19/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
The brain has traditionally been considered an "immune-privileged" organ lacking a lymphatic system. However, recent studies have challenged this view by identifying the presence of the glymphatic system and meningeal lymphatic vessels (MLVs). These discoveries offer new opportunities for waste clearance and treatment of central nervous system (CNS) diseases. Various strategies have been developed based on these pathways, including modulation of glymphatic system function, enhancement of meningeal lymphatic drainage, and utilization of these routes for drug delivery. Consequently, this review explores the developmental features and physiological roles of the cerebral lymphatic system as well as its significance in various CNS disorders. Notably, strategies for ameliorating CNS diseases have been discussed with a focus on enhancing glymphatic system and MLVs functionality through modulation of physiological factors along with implementing pharmacological and physical treatments. Additionally, emphasis is placed on the potential use of the CNS lymphatic system in drug delivery while envisioning future directions in terms of mechanisms, applications, and translational research.
Collapse
Affiliation(s)
| | | | | | - Si Zhang
- Department of Neurosurgery, Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Carrero L, Antequera D, Municio C, Carro E. Circadian rhythm disruption and retinal dysfunction: a bidirectional link in Alzheimer's disease? Neural Regen Res 2024; 19:1967-1972. [PMID: 38227523 DOI: 10.4103/1673-5374.390962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/07/2023] [Indexed: 01/17/2024] Open
Abstract
Dysfunction in circadian rhythms is a common occurrence in patients with Alzheimer's disease. A predominant function of the retina is circadian synchronization, carrying information to the brain through the retinohypothalamic tract, which projects to the suprachiasmatic nucleus. Notably, Alzheimer's disease hallmarks, including amyloid-β, are present in the retinas of Alzheimer's disease patients, followed/associated by structural and functional disturbances. However, the mechanistic link between circadian dysfunction and the pathological changes affecting the retina in Alzheimer's disease is not fully understood, although some studies point to the possibility that retinal dysfunction could be considered an early pathological process that directly modulates the circadian rhythm.
Collapse
Affiliation(s)
- Laura Carrero
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
| | - Desireé Antequera
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Cristina Municio
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Eva Carro
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| |
Collapse
|
34
|
Chang J, Liu Y, Zhao Y, Gao P, Tang Y. Association of sleep duration with excess risk of dementia among shift workers in the UK biobank: a population-based cohort study. J Neurol 2024; 271:6056-6067. [PMID: 39033263 DOI: 10.1007/s00415-024-12580-1] [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: 05/08/2024] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Shift work was associated with elevated dementia risk. Definitive guidelines for sleep duration among shift workers have not been proposed. We aimed to identify sleep durations associated with elimination of excess dementia risk in shift workers. METHODS 285,213 dementia-free UK Biobank participants at baseline, aged 38-71 years, were enrolled between 2006 and 2010 and followed up through 2022 in this cohort study. Cox proportional hazards models were used to examine the associations between shift work, sleep duration, and risk of dementia. RESULTS The 285,213 participants included 49,079 shift workers and 236,134 non-shift workers. Over a median follow-up of 13.8 years, 1887 dementia cases were documented. Current shift workers had significantly higher dementia risk than non-shift workers (hazard ratio [HR] 1.26; 95% CI 1.11-1.42). However, this excess risk was eliminated in shift workers with 8 h of sleep (HR 1.02; 95% CI 0.80-1.29). Analysis of shift work frequency indicated that "sometimes" and "usually/always" shift work were associated with increased dementia risk compared to that of non-shift workers, but excess dementia risk was eliminated in members of either frequency group receiving 8 h of sleep ("sometimes", HR 1.05; 95% CI 0.75-1.48; "usually/always", HR 0.98; 95% CI 0.70-1.35). Both "non-night shift" and "night shift" workers showed increased dementia risk compared to non-shift workers. Workers with 8 h of sleep mitigated the excess risk (HR 1.13; 95% CI 0.84-1.53 and HR 0.86; 95% CI 0.59-1.26, respectively). CONCLUSION 8-h sleep may eliminate excess dementia risk among shift workers, suggesting a potentially effective dementia prevention guideline for shift workers.
Collapse
Affiliation(s)
- Jie Chang
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yufei Liu
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Yiwei Zhao
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Peiyang Gao
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China
| | - Yi Tang
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, 100053, China.
- Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing, 100053, China.
| |
Collapse
|
35
|
Li Y, Yan Z, Shao N, Tang S, Zhang X, Liu XM, Tang J. Dual orexin receptor antagonist ameliorates sleep deprivation-induced learning and memory impairment in APP/PS1 mice. Sleep Med 2024; 121:303-314. [PMID: 39047304 DOI: 10.1016/j.sleep.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Sleep is considered closely related to cognitive function, and cognitive impairment is the main clinical manifestation of Alzheimer's disease (AD). Sleep disturbance in AD patients is more severe than that in healthy elderly individuals. Additionally, sleep deprivation reportedly increases the activity of the hypothalamic orexin system and the risk of AD. To investigate whether intervention with the orexin system can improve sleep disturbance in AD and its impact on AD pathology. In this study, six-month-old amyloid precursor protein/presenilin 1 mice were subjected to six weeks of chronic sleep deprivation and injected intraperitoneally with almorexant, a dual orexin receptor antagonist (DORA), to investigate the effects and mechanisms of sleep deprivation and almorexant intervention on learning and memory in mice with AD. We found that sleep deprivation aggravated learning and memory impairment and increased brain β-amyloid (Aβ) deposition in mice with AD. The application of almorexant can increase the total sleep time of sleep-deprived mice and reduce cognitive impairment and Aβ deposition, which is related to the improvement in Aquaporin-4 polarity. Thus, DORA may be an effective strategy for delaying the progression of AD patients by improving the sleep disturbances.
Collapse
Affiliation(s)
- Yaran Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Zian Yan
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Na Shao
- Department of Neurology, Shandong Provincial Qian Foshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shi Tang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, China.
| | - Xiao Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Xiao Min Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Jiyou Tang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, China
| |
Collapse
|
36
|
Deng S, Hu Y, Chen S, Xue Y, Yao D, Sun Q, Nedergaard M, Wang W, Ding F. Chronic sleep fragmentation impairs brain interstitial clearance in young wildtype mice. J Cereb Blood Flow Metab 2024; 44:1515-1531. [PMID: 38639025 PMCID: PMC11418708 DOI: 10.1177/0271678x241230188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/06/2023] [Accepted: 12/27/2023] [Indexed: 04/20/2024]
Abstract
Accumulating evidence shows that most chronic neurological diseases have a link with sleep disturbances, and that patients with chronically poor sleep undergo an accelerated cognitive decline. Indeed, a single-night of sleep deprivation may increase metabolic waste levels in cerebrospinal fluid. However, it remains unknown how chronic sleep disturbances in isolation from an underlying neurological disease may affect the glymphatic system. Clearance of brain interstitial waste by the glymphatic system occurs primarily during sleep, driven by multiple oscillators including arterial pulsatility, and vasomotion. Herein, we induced sleep fragmentation in young wildtype mice and assessed the effects on glymphatic activity and cognitive functions. Chronic sleep fragmentation reduced glymphatic function and impaired cognitive functions in healthy mice. A mechanistic analysis showed that the chronic sleep fragmentation suppressed slow vasomotion, without altering cardiac-driven pulsations. Taken together, results of this study document that chronic sleep fragmentation suppresses brain metabolite clearance and impairs cognition, even in the absence of disease.
Collapse
Affiliation(s)
- Saiyue Deng
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yusi Hu
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Simiao Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, 310003, China
| | - Yang Xue
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Di Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Sun
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurology, University of Rochester Medical Center, Rochester, NY, 14642, United States
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengfei Ding
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
37
|
Wood KH, Nenert R, Miften AM, Kent GW, Sleyster M, Memon RA, Joop A, Pilkington J, Memon AA, Wilson RN, Catiul C, Szaflarski J, Amara AW. Diffusion Tensor Imaging-Along the Perivascular-Space Index Is Associated with Disease Progression in Parkinson's Disease. Mov Disord 2024; 39:1504-1513. [PMID: 38988232 PMCID: PMC11524528 DOI: 10.1002/mds.29908] [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: 02/09/2024] [Revised: 06/08/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND The glymphatic clearance pathway is a waste clearance system that allows for removal of soluble proteins such as amyloid β (Aβ) from the brain. Higher Aβ levels are associated with cognitive dysfunction in Parkinson's disease (PD). Diffusion tensor imaging-along the perivascular space (DTI-ALPS) is an imaging measure proposed to indirectly measure glymphatic function. OBJECTIVES Evaluate differences in DTI-ALPS-index between PD and healthy controls (HC) and characterize relationships between this proposed measure of glymphatic clearance, cognition, and disease severity in PD. METHODS PD (n = 32) and HC (n = 23) participants underwent brain imaging to assess DTI-ALPS. PD participants were classified as PD-normal cognition (PD-NC; n = 20) or PD-mild cognitive impairment (PD-MCI; n = 12) based on a Level II comprehensive cognitive assessment. A subgroup of PD participants (n = 21) returned for annual assessments for up to 4 years after baseline. Longitudinal outcomes included changes in performance on the comprehensive cognitive assessment and changes in the Movement Disorders Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS). RESULTS PD participants had lower DTI-ALPS-index compared to HC. PD participants classified as PD-MCI had significantly lower DTI-ALPS-index compared to PD-NC. Lower DTI-ALPS-index at baseline was associated with longitudinal cognitive decline and worse longitudinal disease severity. CONCLUSIONS Glymphatic clearance, as measured with DTI-ALPS, has potential to serve as a marker of longitudinal disease progression. Interventions targeting glymphatic function should be explored for potential to slow cognitive decline in PD. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Kimberly H. Wood
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
- Department of Psychology, Samford University, Birmingham, AL
| | - Rodolphe Nenert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Aya M. Miften
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - George W. Kent
- Department of Psychology, Samford University, Birmingham, AL
| | - Madison Sleyster
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | | | - Allen Joop
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Jennifer Pilkington
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Adeel A. Memon
- Department of Neurology, West Virginia University, Morgantown, WV
| | - Riis N. Wilson
- Department of Psychology, Samford University, Birmingham, AL
| | - Corina Catiul
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Jerzy Szaflarski
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
| | - Amy W. Amara
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
38
|
Shin S, Seok JW, Kim K, Kim J, Nam HY, Pak K. Poor sleep quality is associated with decreased regional brain glucose metabolism in healthy middle-aged adults. Neuroimage 2024; 298:120814. [PMID: 39187219 DOI: 10.1016/j.neuroimage.2024.120814] [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: 06/12/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024] Open
Abstract
Sleep disturbance is associated with the development of neurodegenerative disease. We aimed to address the effects of sleep quality on brain glucose metabolism measured by 18F-Fl uorodeoxyglucose (18F-FDG) positron emission tomography (PET) in healthy middle-aged adults. A total of 378 healthy men (mean age: 42.8±3.6 years) were included in this study. Participants underwent brain 18F-FDG PET and completed the Korean version of the Pittsburgh Sleep Quality Index (PSQI-K). Additionally, anthropometric measurements were obtained. PETs were spatially normalized to MNI space using PET templates from SPM5 with PMOD. The Automated Anatomical Labeling 2 atlas was used to define regions of interest (ROIs). The mean uptake of each ROI was scaled to the mean of the global cortical uptake of each individual and defined as the standardized uptake value ratio (SUVR). After the logarithmic transformation of the regional SUVR, the effects of the PSQI-K on the regional SUVR were investigated using Bayesian hierarchical modeling. Brain glucose metabolism of the posterior cingulate, precuneus, and thalamus showed a negative association with total PSQI-K scores in the Bayesian model ROI-based analysis. Voxel-based analysis using statistical parametric mapping revealed a negative association between the total PSQI-K scores and brain glucose metabolism of the precuneus, postcentral gyrus, posterior cingulate, and thalamus. Poor sleep quality is negatively associated with brain glucose metabolism in the precuneus, posterior cingulate, and thalamus. Therefore, the importance of sleep should not be overlooked, even in healthy middle-aged adults.
Collapse
Affiliation(s)
- Seunghyeon Shin
- Department of Nuclear Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea.
| | - Ju Won Seok
- Department of Nuclear Medicine, Chung-Ang University College of Medicine, Seoul, Republic of Korea.
| | - Keunyoung Kim
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea; School of Medicine, Pusan National University, Busan, Republic of Korea.
| | - Jihyun Kim
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.
| | - Hyun-Yeol Nam
- Department of Nuclear Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea.
| | - Kyoungjune Pak
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea; School of Medicine, Pusan National University, Busan, Republic of Korea.
| |
Collapse
|
39
|
Uchida K, Meno K, Korenaga T, Liu S, Suzuki H, Baba Y, Tagata C, Araki Y, Tsunemi S, Aso K, Inagaki S, Nakagawa S, Kobayashi M, Kakuma T, Asada T, Ota M, Takihara T, Arai T. Effect of matcha green tea on cognitive functions and sleep quality in older adults with cognitive decline: A randomized controlled study over 12 months. PLoS One 2024; 19:e0309287. [PMID: 39213264 PMCID: PMC11364242 DOI: 10.1371/journal.pone.0309287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE Lifestyle habits after middle age significantly impact the maintenance of cognitive function in older adults. Nutritional intake is closely related to lifestyle habits; therefore, nutrition is a pivotal factor in the prevention of dementia in the preclinical stages. Matcha green tea powder (matcha), which contains epigallocatechin gallate, theanine, and caffeine, has beneficial effects on cognitive function and mood. We conducted a randomized, double-blind, placebo-controlled clinical study over 12 months to examine the effect of matcha on cognitive function and sleep quality. METHODS Ninety-nine participants, including 64 with subjective cognitive decline and 35 with mild cognitive impairment were randomized, with 49 receiving 2 g of matcha and 50 receiving a placebo daily. Participants were stratified based on two factors: age at baseline and APOE genotype. Changes in cognitive function and sleep quality were analyzed using a mixed-effects model. RESULTS Matcha consumption led to significant improvements in social acuity score (difference; -1.39, 95% confidence interval; -2.78, 0.002) (P = 0.028) as evaluated by the perception of facial emotions in cognitive function. The primary outcomes, that is, Montreal Cognitive Assessment and Alzheimer's Disease Cooperative Study Activity of Daily Living scores, showed no significant changes with matcha intervention. Meanwhile, Pittsburgh Sleep Quality Index scores indicated a trend toward improvement with a difference of 0.86 (95% confidence interval; -0.002, 1.71) (P = 0.088) between the groups in changes from baseline to 12 months. CONCLUSIONS The present study suggests regular consumption of matcha could improve emotional perception and sleep quality in older adults with mild cognitive decline. Given the widespread availability and cultural acceptance of matcha green tea, incorporating it into the daily routine may offer a simple yet effective strategy for cognitive enhancement and dementia prevention.
Collapse
Affiliation(s)
- Kazuhiko Uchida
- Institute of Biomedical Research, MCBI Inc., Tsukuba, Ibaraki, Japan
- Research Division, MCBI Inc., Tsukuba, Ibaraki, Japan
| | - Kohji Meno
- Research Division, MCBI Inc., Tsukuba, Ibaraki, Japan
| | | | - Shan Liu
- Research Division, MCBI Inc., Tsukuba, Ibaraki, Japan
| | | | - Yoshitake Baba
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Chika Tagata
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Yoshiharu Araki
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Shuto Tsunemi
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Kenta Aso
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Shun Inagaki
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Sae Nakagawa
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Makoto Kobayashi
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Tatsuyuki Kakuma
- Biostatistics Center, Kurume University Graduate School of Medicine, Kurume, Japan
| | - Takashi Asada
- Memory Clinic Toride, Toride, Ibaraki, Japan
- Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Miho Ota
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takanobu Takihara
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Tetsuaki Arai
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
40
|
Livingston G, Huntley J, Liu KY, Costafreda SG, Selbæk G, Alladi S, Ames D, Banerjee S, Burns A, Brayne C, Fox NC, Ferri CP, Gitlin LN, Howard R, Kales HC, Kivimäki M, Larson EB, Nakasujja N, Rockwood K, Samus Q, Shirai K, Singh-Manoux A, Schneider LS, Walsh S, Yao Y, Sommerlad A, Mukadam N. Dementia prevention, intervention, and care: 2024 report of the Lancet standing Commission. Lancet 2024; 404:572-628. [PMID: 39096926 DOI: 10.1016/s0140-6736(24)01296-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/08/2024] [Accepted: 06/16/2024] [Indexed: 08/05/2024]
Affiliation(s)
- Gill Livingston
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK.
| | - Jonathan Huntley
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Kathy Y Liu
- Division of Psychiatry, University College London, London, UK
| | - Sergi G Costafreda
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Geir Selbæk
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Geriatric Department, Oslo University Hospital, Oslo, Norway
| | - Suvarna Alladi
- National Institute of Mental Health and Neurosciences, Bangalore, India
| | - David Ames
- National Ageing Research Institute, Melbourne, VIC, Australia; University of Melbourne Academic Unit for Psychiatry of Old Age, Melbourne, VIC, Australia
| | - Sube Banerjee
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | | | - Carol Brayne
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Nick C Fox
- The Dementia Research Centre, Department of Neurodegenerative Disease, University College London, London, UK
| | - Cleusa P Ferri
- Health Technology Assessment Unit, Hospital Alemão Oswaldo Cruz, São Paulo, Brazil; Department of Psychiatry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Laura N Gitlin
- College of Nursing and Health Professions, AgeWell Collaboratory, Drexel University, Philadelphia, PA, USA
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Helen C Kales
- Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, University of California, Sacramento, CA, USA
| | - Mika Kivimäki
- Division of Psychiatry, University College London, London, UK; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Eric B Larson
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Noeline Nakasujja
- Department of Psychiatry College of Health Sciences, Makerere University College of Health Sciences, Makerere University, Kampala City, Uganda
| | - Kenneth Rockwood
- Centre for the Health Care of Elderly People, Geriatric Medicine, Dalhousie University, Halifax, NS, Canada
| | - Quincy Samus
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Bayview, Johns Hopkins University, Baltimore, MD, USA
| | - Kokoro Shirai
- Graduate School of Social and Environmental Medicine, Osaka University, Osaka, Japan
| | - Archana Singh-Manoux
- Division of Psychiatry, University College London, London, UK; Université Paris Cité, Inserm U1153, Paris, France
| | - Lon S Schneider
- Department of Psychiatry and the Behavioural Sciences and Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Sebastian Walsh
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Yao Yao
- China Center for Health Development Studies, School of Public Health, Peking University, Beijing, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Andrew Sommerlad
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Naaheed Mukadam
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| |
Collapse
|
41
|
Favaro M, Mauri S, Bernardo G, Zordan MA, Mazzotta GM, Ziviani E. Usp14 down-regulation corrects sleep and circadian dysfunction of a Drosophila model of Parkinson's disease. Front Neurosci 2024; 18:1410139. [PMID: 39161651 PMCID: PMC11330830 DOI: 10.3389/fnins.2024.1410139] [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: 03/31/2024] [Accepted: 07/03/2024] [Indexed: 08/21/2024] Open
Abstract
PD is a complex, multifactorial neurodegenerative disease, which occurs sporadically in aged population, with some genetically linked cases. Patients develop a very obvious locomotor phenotype, with symptoms such as bradykinesia, resting tremor, muscular rigidity, and postural instability. At the cellular level, PD pathology is characterized by the presence of intracytoplasmic neurotoxic aggregates of misfolded proteins and dysfunctional organelles, resulting from failure in mechanisms of proteostasis. Nonmotor symptoms, such as constipation and olfactory deficits, are also very common in PD. They include alteration in the circadian clock, and defects in the sleep-wake cycle, which is controlled by the clock. These non-motor symptoms precede the onset of the motor symptoms by many years, offering a window of therapeutic intervention that could delay-or even prevent-the progression of the disease. The mechanistic link between aberrant circadian rhythms and neurodegeneration in PD is not fully understood, although proposed underlying mechanisms include alterations in protein homeostasis (proteostasis), which can impact protein levels of core components of the clock. Loss of proteostasis depends on the progressive pathological decline in the proteolytic activity of two major degradative systems, the ubiquitin-proteasome and the lysosome-autophagy systems, which is exacerbated in age-dependent neurodegenerative conditions like PD. Accordingly, it is known that promoting proteasome or autophagy activity increases lifespan, and rescues the pathological phenotype of animal models of neurodegeneration, presumably by enhancing the degradation of misfolded proteins and dysfunctional organelles, which are known to accumulate in these models, and to induce intracellular damage. We can enhance proteostasis by pharmacologically inhibiting or down-regulating Usp14, a proteasome-associated deubiquitinating enzyme (DUB). In a previous work, we showed that inhibition of Usp14 enhances the activity of the ubiquitin-proteasome system (UPS), autophagy and mitophagy, and abolishes motor symptoms of two well-established fly models of PD that accumulate dysfunctional mitochondria. In this work we extended the evidence on the protective effect of Usp14 down-regulation, and investigated the beneficial effect of down-regulating Usp14 in a Pink1 Drosophila model of PD that develop circadian and sleep dysfunction. We show that down-regulation of Usp14 ameliorates sleep disturbances and circadian defects that are associated to Pink1 KO flies.
Collapse
Affiliation(s)
| | | | | | | | | | - Elena Ziviani
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
42
|
Rhea EM, Leclerc M, Yassine HN, Capuano AW, Tong H, Petyuk VA, Macauley SL, Fioramonti X, Carmichael O, Calon F, Arvanitakis Z. State of the Science on Brain Insulin Resistance and Cognitive Decline Due to Alzheimer's Disease. Aging Dis 2024; 15:1688-1725. [PMID: 37611907 PMCID: PMC11272209 DOI: 10.14336/ad.2023.0814] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is common and increasing in prevalence worldwide, with devastating public health consequences. While peripheral insulin resistance is a key feature of most forms of T2DM and has been investigated for over a century, research on brain insulin resistance (BIR) has more recently been developed, including in the context of T2DM and non-diabetes states. Recent data support the presence of BIR in the aging brain, even in non-diabetes states, and found that BIR may be a feature in Alzheimer's disease (AD) and contributes to cognitive impairment. Further, therapies used to treat T2DM are now being investigated in the context of AD treatment and prevention, including insulin. In this review, we offer a definition of BIR, and present evidence for BIR in AD; we discuss the expression, function, and activation of the insulin receptor (INSR) in the brain; how BIR could develop; tools to study BIR; how BIR correlates with current AD hallmarks; and regional/cellular involvement of BIR. We close with a discussion on resilience to both BIR and AD, how current tools can be improved to better understand BIR, and future avenues for research. Overall, this review and position paper highlights BIR as a plausible therapeutic target for the prevention of cognitive decline and dementia due to AD.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Manon Leclerc
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
| | - Hussein N Yassine
- Departments of Neurology and Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Ana W Capuano
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Han Tong
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA.
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA.
| | - Xavier Fioramonti
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France.
| | - Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Frederic Calon
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
43
|
Tucker A, Goldberg TE, Kim H. Biomarkers of sleep-wake disturbance as predictors of cognitive decline and accelerated disease progression. Expert Rev Mol Diagn 2024; 24:649-657. [PMID: 39129222 DOI: 10.1080/14737159.2024.2389307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION In older adults, where sleep disturbances and cognitive impairment are common, mounting evidence suggests a potential connection between sleep and cognitive function, highlighting the significance of utilizing sleep as a biomarker for early detection of cognitive impairment to improve clinical outcomes in a noninvasive, cost-effective manner. AREAS COVERED This review describes the relationship between sleep and cognitive function in older adults, encompassing both subjective and objective measures of sleep quality, duration, architecture, and sleep-disordered breathing. The authors consider the directionality of the associations observed in prospective and cross-sectional studies, exploring whether sleep disturbances precede cognitive decline or vice versa. Furthermore, they discuss the potential bidirectional relationships between sleep and Alzheimer's disease (AD) risks in older adults while also examining the neurodegenerative pathways of this relationship. EXPERT OPINION Routine sleep monitoring in primary care settings has the potential to bolster early detection and treatment of sleep disturbance, and by extension, reduce the risk of dementia. Improving sleep assessment tools, such as wearables, provide scalable alternatives to traditional methods like polysomnography, potentially enabling widespread monitoring of sleep characteristics. Standardized measurement and inclusive participant recruitment are needed to enhance generalizability, while longitudinal studies are essential to understand the interaction between sleep and AD pathology.
Collapse
Affiliation(s)
- Aren Tucker
- Brain Aging and Mental Health Division, New York State Psychiatric Institute, New York, NY, USA
| | - Terry E Goldberg
- Brain Aging and Mental Health Division, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Psychology, New York, NY, USA
- Department of Anesthisiology, Columbia University Irving Medical Psychology, New York, NY, USA
| | - Hyun Kim
- Brain Aging and Mental Health Division, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Psychology, New York, NY, USA
| |
Collapse
|
44
|
Anwar MM, Pérez-Martínez L, Pedraza-Alva G. Exploring the Significance of Microglial Phenotypes and Morphological Diversity in Neuroinflammation and Neurodegenerative Diseases: From Mechanisms to Potential Therapeutic Targets. Immunol Invest 2024; 53:891-946. [PMID: 38836373 DOI: 10.1080/08820139.2024.2358446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Studying various microglial phenotypes and their functions in neurodegenerative diseases is crucial due to the intricate nature of their phenomics and their vital immunological role. Microglia undergo substantial phenomic changes, encompassing morphological, transcriptional, and functional aspects, resulting in distinct cell types with diverse structures, functions, properties, and implications. The traditional classification of microglia as ramified, M1 (proinflammatory), or M2 (anti-inflammatory) phenotypes is overly simplistic, failing to capture the wide range of recently identified microglial phenotypes in various brain regions affected by neurodegenerative diseases. Altered and activated microglial phenotypes deviating from the typical ramified structure are significant features of many neurodegenerative conditions. Understanding the precise role of each microglial phenotype is intricate and sometimes contradictory. This review specifically focuses on elucidating recent modifications in microglial phenotypes within neurodegenerative diseases. Recognizing the heterogeneity of microglial phenotypes in diseased states can unveil novel therapeutic strategies for targeting microglia in neurodegenerative diseases. Moreover, the exploration of the use of healthy isolated microglia to mitigate disease progression has provided an innovative perspective. In conclusion, this review discusses the dynamic landscape of mysterious microglial phenotypes, emphasizing the need for a nuanced understanding to pave the way for innovative therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Leonor Pérez-Martínez
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| | - Gustavo Pedraza-Alva
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| |
Collapse
|
45
|
Dong Z, Du X, Wang L, Zou X, Zuo H, Yan Y, Chen G, Cheng O, Zhang Y. Deep cervical lymph nodes in Parkinson's disease and atypical Parkinson's disease: A potential ultrasound biomarker for differential diagnosis. J Cent Nerv Syst Dis 2024; 16:11795735241259429. [PMID: 39086599 PMCID: PMC11289816 DOI: 10.1177/11795735241259429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 05/17/2024] [Indexed: 08/02/2024] Open
Abstract
Background Parkinson's disease (PD) is a common degenerative disease caused by abnormal accumulation of α-synuclein. The glymphatic pathway is essential for removing macromolecular proteins including α-synuclein from the brain, which flows into deep cervical lymph nodes (DCLNs) through meningeal lymphatics. As a terminal station for the cerebral lymphatic system drainage, DCLNs can be easily assessed clinically. Objectives Although the drainage function of the cerebral lymphatic system is impaired in PD, the correlation between DCLNs and PD remains unknown. Design Single-center retrospective cross-sectional study. Methods The size of the DCLNs were measured using ultrasound. The Movement Disorder Society Sponsored Revision Unified Parkinson's Disease Rating Scale and other scales were used to assess PD motor and non-motor symptoms. Results Compared with the healthy control (HC) and the atypical Parkinson's disease (AP) groups, the size of the second and third DCLNs in the Parkinson's disease (PD) group was significantly smaller (P < .05). The width diameter of the third DCLN (DCLN3(y)) was significantly smaller in the PD group than in the AP group (P = .014). DCLN3(y) combined with a variety of clinical features improved the sensitivity of AP identification (sensitivity = .813). Conclusion DCLNs were able to distinguish HC, PD and AP and were mainly located in Robbins ΙΙA level. PD and AP were associated with different factors that influenced the size of the DCLNs. DCLN3(y) plays an important role in differentiating PD from AP, which, combined with other clinical features, has the ability to distinguish PD from AP; in particular, the sensitivity of AP diagnosis was improved.
Collapse
Affiliation(s)
- Zhaoying Dong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyi Du
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoya Zou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongzhou Zuo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Yan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guojun Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Zhang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
46
|
Rowe RK, Schulz P, He P, Mannino GS, Opp MR, Sierks MR. Acute sleep deprivation in mice generates protein pathology consistent with neurodegenerative diseases. Front Neurosci 2024; 18:1436966. [PMID: 39114483 PMCID: PMC11303328 DOI: 10.3389/fnins.2024.1436966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Insufficient or disturbed sleep is strongly associated with adverse health conditions, including various neurodegenerative disorders. While the relationship between sleep and neurodegenerative disease is likely bidirectional, sleep disturbances often predate the onset of other hallmark clinical symptoms. Neuronal waste clearance is significantly more efficient during sleep; thus, disturbed sleep may lead to the accumulation of neuronal proteins that underlie neurodegenerative diseases. Key pathological features of neurodegenerative diseases include an accumulation of misfolded or misprocessed variants of amyloid beta (Aβ), tau, alpha synuclein (α-syn), and TarDNA binding protein 43 (TDP-43). While the presence of fibrillar protein aggregates of these neuronal proteins are characteristic of neurodegenerative diseases, the presence of small soluble toxic oligomeric variants of these different proteins likely precedes the formation of the hallmark aggregates. Methods We hypothesized that sleep deprivation would lead to accumulation of toxic oligomeric variants of Aβ, tau, α-syn, and TDP-43 in brain tissue of wild-type mice. Adult mice were subjected to 6 h of sleep deprivation (zeitgeber 0-6) for 5 consecutive days or were left undisturbed as controls. Following sleep deprivation, brains were collected, and protein pathology was assessed in multiple brain regions using an immunostain panel of reagents selectively targeting neurodegenerative disease-related variants of Aβ, tau, α-syn, and TDP-43. Results Overall, sleep deprivation elevated levels of all protein variants in at least one of the brain regions of interest. The reagent PDTDP, targeting a TDP-43 variant present in Parkinson's disease, was elevated throughout the brain. The cortex, caudoputamen, and corpus callosum brain regions showed the highest accumulation of pathology following sleep deprivation. Discussion These data provide a direct mechanistic link between sleep deprivation, and the hallmark protein pathologies of neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Rachel K. Rowe
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Philip Schulz
- Chemical Engineering, The School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States
| | - Ping He
- Chemical Engineering, The School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States
| | - Grant S. Mannino
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Mark R. Opp
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Michael R. Sierks
- Chemical Engineering, The School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
47
|
Yin C, Zhang M, Cheng L, Ding L, Lv Q, Huang Z, Zhou J, Chen J, Wang P, Zhang S, You Q. Melatonin modulates TLR4/MyD88/NF-κB signaling pathway to ameliorate cognitive impairment in sleep-deprived rats. Front Pharmacol 2024; 15:1430599. [PMID: 39101143 PMCID: PMC11294086 DOI: 10.3389/fphar.2024.1430599] [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: 05/10/2024] [Accepted: 07/05/2024] [Indexed: 08/06/2024] Open
Abstract
Sleep deprivation (SD) is commonplace in today's fast-paced society. SD is a severe public health problem globally since it may cause cognitive decline and even neurodegenerative disorders like Alzheimer's disease. Melatonin (MT) is a natural chemical secreted by the pineal gland with neuroprotective effects. The purpose of this study was to investigate the protective effect and mechanism of MT on chronic sleep deprivation-induced cognitive impairment. A 3-week modified multi-platform method was used to create the SD rat model. The Morris water maze test (MWM), Tissue staining (including Hematoxylin and Eosin (H & E) staining, Nissl staining, and immunofluorescence), Western blot, Enzyme-linked immunosorbent assay (ELISA), and Quantitative real-time polymerase chain reaction (qPCR) were used to investigate the protective effect and mechanism of MT in ameliorating cognitive impairment in SD rats. The results showed that MT (50 and 100 mg/kg) significantly improved cognitive function in rats, as evidenced by a shortening of escape latency and increased time of crossing the platform and time spent in the quadrant. Additionally, MT therapy alleviated hippocampus neurodegeneration and neuronal loss while lowering levels of pathogenic factors (LPS) and inflammatory indicators (IL-1β, IL-6, TNF-α, iNOS, and COX2). Furthermore, MT treatment reversed the high expression of Aβ42 and Iba1 as well as the low expression of ZO-1 and occludin, and inhibited the SD-induced TLR4/MyD88/NF-κB signaling pathway. In summary, MT ameliorated spatial recognition and learning memory dysfunction in SD rats by reducing neuroinflammation and increasing neuroprotection while inhibiting the TLR4/MyD88/NF-κB signaling pathway. Our study supports the use of MT as an alternate treatment for SD with cognitive impairment.
Collapse
Affiliation(s)
- Chao Yin
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, China
| | - Meiya Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, China
| | - Li Cheng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Li Ding
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, China
| | - Qing Lv
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Zixuan Huang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jiaqi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jianmei Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Ping Wang
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, China
| | - Shunbo Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Qiuyun You
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
48
|
Niesman IR. Stress and the domestic cat: have humans accidentally created an animal mimic of neurodegeneration? Front Neurol 2024; 15:1429184. [PMID: 39099784 PMCID: PMC11294998 DOI: 10.3389/fneur.2024.1429184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/01/2024] [Indexed: 08/06/2024] Open
Abstract
Many neurodegenerative diseases (NDD) appear to share commonality of origin, chronic ER stress. The endoplasmic reticulum (ER) is a dynamic organelle, functioning as a major site of protein synthesis and protein posttranslational modifications, required for proper folding. ER stress can occur because of external stimuli, such as oxidative stress or neuroinflammatory cytokines, creating the ER luminal environment permissive for the accumulation of aggregated and misfolded proteins. Unresolvable ER stress upregulates a highly conserved pathway, the unfolded protein response (UPR). Maladaptive chronic activation of UPR components leads to apoptotic neuronal death. In addition to other factors, physiological responses to stressors are emerging as a significant risk factor in the etiology and pathogenesis of NDD. Owned cats share a common environment with people, being exposed to many of the same stressors as people and additional pressures due to their "quasi" domesticated status. Feline Cognitive Dysfunction Syndrome (fCDS) presents many of the same disease hallmarks as human NDD. The prevalence of fCDS is rapidly increasing as more people welcome cats as companions. Barely recognized 20 years ago, veterinarians and scientists are in infancy stages in understanding what is a very complex disease. This review will describe how cats may represent an unexplored animal mimetic phenotype for human NDD with stressors as potential triggering mechanisms. We will consider how multiple variations of stressful events over the short-life span of a cat could affect neuronal loss or glial dysfunction and ultimately tip the balance towards dementia.
Collapse
Affiliation(s)
- Ingrid R. Niesman
- Department of Biology, SDSU Electron Microscopy Facility, San Diego State University, San Diego, CA, United States
| |
Collapse
|
49
|
Luo S, Guo L, Chen N, Guo Q, Xie Y, Wang Y, Wang E. CRY2 mediates the cognitive decline induced by sleep deprivation in 5xFAD mice. PLoS One 2024; 19:e0306930. [PMID: 39012854 PMCID: PMC11251589 DOI: 10.1371/journal.pone.0306930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Cryptochrome-2 (CRY2) is a core rhythm gene that plays a crucial role in DNA damage repair. The present study investigated the potential role of CRY2 in mediating sleep deprivation-induced cognitive decline in 5xFAD mice. METHODS To assess the effects of SD on different brain regions of the mouse brain, we used 18F FDG PET-CT. Cognitive function was evaluated using the Morris water maze test and Y-maze. Lentivirus was used for the overexpression of CRY2, and small interfering RNA (siRNA) was used for the downregulation of CRY2 to verify the effect of CRY2. We used qRT‒PCR and Western blotting to identify the downstream factors of CRY2 and evaluated the cognitive function of mice to confirm the effects of these factors. RESULTS The AD mice exhibited cognitive decline after 21 days of SD and had higher expression of CRY2 compared to AD mice with normal sleep. Overexpression of CRY2 led to decreased cognitive function in AD mice, and the downregulation of CRY2 attenuated the SD-induced cognitive decline in AD mice. CRY2 reduced the expression and function of CISH, which reduced the inhibition of STAT1 phosphorylation and led to synaptic dysfunction. CISH overexpression attenuated the impairing effect of sleep deprivation on cognitive function in AD mice. Furthermore, 18F FDG PET-CT revealed that SD significantly reduced glucose metabolism in different brain regions of AD mice. CONCLUSION Our study demonstrated that sleep deprivation upregulated CRY2 in the hippocampus of AD mice, which resulted in synaptic dysfunction by decreasing CISH-mediated STAT1 phosphorylation.
Collapse
Affiliation(s)
- Sumei Luo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Lizhe Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Na Chen
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Yongqiu Xie
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Yunjiao Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha, China
| | - E. Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha, China
| |
Collapse
|
50
|
Stankeviciute L, Chhatwal JP, Levin R, Pinilla V, Schultz AP, Redline S, Johnson KA, Sperling RA, Kozhemiako N, Purcell S, Djonlagic I. Amyloid beta-independent sleep markers associated with early regional tau burden and cortical thinning. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12616. [PMID: 39077684 PMCID: PMC11284643 DOI: 10.1002/dad2.12616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Sleep is crucial for memory consolidation and the clearance of toxic proteins associated with Alzheimer's disease (AD). We examined the association between sleep characteristics and imaging biomarkers of early amyloid beta (Aβ) and tau pathology as well as neurodegeneration in brain regions known to be affected in the incipient stages of AD. METHODS Thirty-nine cognitively unimpaired (CU) participants of the Harvard Aging Brain Study underwent at-home polysomnography as well as tau positron emission tomography (flortaucipir-PET), amyloid PET (Pittsburgh compound B [PiB]-PET), and magnetic resonance imaging-derived assessment of cortical thickness (CT). RESULTS Increased N1 sleep was associated with a higher tau PET signal (β = 0.009, p = 0.001) and lower CT in the temporal composite region of interest (β = -0.017, p = 0.007). Decreased slow-wave sleep (SWS) was associated with higher tau burden in the temporal composite (β = -0.008, p = 0.005) and lower CT (β = 0.008, p = 0.002), even after controlling for global PiB-PET. DISCUSSION In CU older adults, lower SWS and higher N1 sleep were associated with higher tau burden and lower CT in brain regions associated with early tau deposition and vulnerable to AD-related neurodegeneration through mechanisms dissociable from amyloid deposition. Highlights We report the results of an observational study, which leveraged -a well-characterized cohort of healthy aging (Harvard Aging Brain Study) by adding in-home full polysomnograms.By adding at-home polysomnograms to this unique and deeply phenotyped cohort, we examined variations in sleep architecture that are associated with Alzheimer's disease (AD) pathologic changes.Our results confirmed the association of sleep changes with early tau and cortical neurodegenerative changes that were independent of amyloid.The results will be of importance in monitoring sleep-related variations in relation to the natural history of AD pathology and in designing sleep-focused clinical trials.
Collapse
Affiliation(s)
- Laura Stankeviciute
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - Jasmeer P. Chhatwal
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Raina Levin
- Massachusetts General HospitalBostonMassachusettsUSA
| | | | - Aaron P. Schultz
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Susan Redline
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Keith A. Johnson
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Reisa A. Sperling
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Nataliia Kozhemiako
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Shaun Purcell
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Ina Djonlagic
- Department of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| |
Collapse
|