1
|
Mentink LJ, van Osch MJP, Bakker LJ, Olde Rikkert MGM, Beckmann CF, Claassen JAHR, Haak KV. Functional and vascular neuroimaging in maritime pilots with long-term sleep disruption. GeroScience 2024:10.1007/s11357-024-01417-4. [PMID: 39531187 DOI: 10.1007/s11357-024-01417-4] [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: 08/29/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
The mechanism underlying the possible causal association between long-term sleep disruption and Alzheimer's disease remains unclear Musiek et al. 2015. A hypothesised pathway through increased brain amyloid load was not confirmed in previous work in our cohort of maritime pilots with long-term work-related sleep disruption Thomas et al. Alzheimer's Res Ther 2020;12:101. Here, using functional MRI, T2-FLAIR, and arterial spin labeling MRI scans, we explored alternative neuroimaging biomarkers related to both sleep disruption and AD: resting-state network co-activation and between-network connectivity of the default mode network (DMN), salience network (SAL) and frontoparietal network (FPN), vascular damage and cerebral blood flow (CBF). We acquired data of 16 maritime pilots (56 ± 2.3 years old) with work-related long-term sleep disruption (23 ± 4.8 working years) and 16 healthy controls (59 ± 3.3 years old), with normal sleep patterns (Pittsburgh Sleep Quality Index ≤ 5). Maritime pilots did not show altered co-activation in either the DMN, FPN, or SAL and no differences in between-network connectivity. We did not detect increased markers of vascular damage in maritime pilots, and additionally, maritime pilots did not show altered CBF-patterns compared to healthy controls. In summary, maritime pilots with long-term sleep disruption did not show neuroimaging markers indicative of preclinical AD compared to healthy controls. These findings do not resemble those of short-term sleep deprivation studies. This could be due to resiliency to sleep disruption or selection bias, as participants have already been exposed to and were able to deal with sleep disruption for multiple years, or to compensatory mechanisms Mentink et al. PLoS ONE. 2021;15(12):e0237622. This suggests the relationship between sleep disruption and AD is not as strong as previously implied in studies on short-term sleep deprivation, which would be beneficial for all shift workers suffering from work-related sleep disruptions.
Collapse
Affiliation(s)
- Lara J Mentink
- Department of Geriatrics, Radboudumc Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.
- Department of Cognitive Science and Artificial Intelligence, School of Humanity and Digital Sciences, Tilburg University, Tilburg, The Netherlands.
| | | | - Leanne J Bakker
- Department of Geriatrics, Radboudumc Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel G M Olde Rikkert
- Department of Geriatrics, Radboudumc Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christian F Beckmann
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Centre for Functional MRI of the Brain (FMRIB), Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Jurgen A H R Claassen
- Department of Geriatrics, Radboudumc Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Koen V Haak
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cognitive Science and Artificial Intelligence, School of Humanity and Digital Sciences, Tilburg University, Tilburg, The Netherlands
| |
Collapse
|
2
|
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
|
3
|
Deng Q, Li Y, Sun Z, Gao X, Zhou J, Ma G, Qu WM, Li R. Sleep disturbance in rodent models and its sex-specific implications. Neurosci Biobehav Rev 2024; 164:105810. [PMID: 39009293 DOI: 10.1016/j.neubiorev.2024.105810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Sleep disturbances, encompassing altered sleep physiology or disorders like insomnia and sleep apnea, profoundly impact physiological functions and elevate disease risk. Despite extensive research, the underlying mechanisms and sex-specific differences in sleep disorders remain elusive. While polysomnography serves as a cornerstone for human sleep studies, animal models provide invaluable insights into sleep mechanisms. However, the availability of animal models of sleep disorders is limited, with each model often representing a specific sleep issue or mechanism. Therefore, selecting appropriate animal models for sleep research is critical. Given the significant sex differences in sleep patterns and disorders, incorporating both male and female subjects in studies is essential for uncovering sex-specific mechanisms with clinical relevance. This review provides a comprehensive overview of various rodent models of sleep disturbance, including sleep deprivation, sleep fragmentation, and circadian rhythm dysfunction. We evaluate the advantages and disadvantages of each model and discuss sex differences in sleep and sleep disorders, along with potential mechanisms. We aim to advance our understanding of sleep disorders and facilitate sex-specific interventions.
Collapse
Affiliation(s)
- Qi Deng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yuhong Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Xiang Gao
- Shanxi Bethune Hospital, Shanxi, China
| | | | - Guangwei Ma
- Peking University Sixth Hospital, Beijing, China
| | - Wei-Min Qu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China; Department of Pharmacology, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Rena Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Kim DY, Kim SM, Han IO. Chronic rapid eye movement sleep deprivation aggravates the pathogenesis of Alzheimer's disease by decreasing brain O-GlcNAc cycling in mice. J Neuroinflammation 2024; 21:180. [PMID: 39044290 PMCID: PMC11264383 DOI: 10.1186/s12974-024-03179-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
This study investigated the role of O-GlcNAc cycling in Alzheimer's disease-related changes in brain pathophysiology induced by chronic REM sleep deprivation (CSD) in mice. CSD increased amyloid beta (Aβ) and p-Tau accumulation and impaired learning and memory (L/M) function. CSD decreased dendritic length and spine density. CSD also increased the intensity of postsynaptic density protein-95 (PSD-95) staining. All of these Alzheimer's disease (AD) pathogenic changes were effectively reversed through glucosamine (GlcN) treatment by enhancing O-GlcNAcylation. Interestingly, the lelvel of O-GlcNAcylated-Tau (O-Tau) exhibited an opposite trend compared to p-Tau, as it was elevated by CSD and suppressed by GlcN treatment. CSD increased neuroinflammation, as indicated by elevated levels of glial fibrillary acidic protein and IBA-1-positive glial cells in the brain, which were suppressed by GlcN treatment. CSD promoted the phosphorylation of GSK3β and led to an upregulation in the expression of endoplasmic reticulum (ER) stress regulatory proteins and genes. These alterations were effectively suppressed by GlcN treatment. Minocycline not only suppressed neuroinflammation induced by CSD, but it also rescued the decrease in O-GlcNAc levels caused by CSD. Minocycline also reduced AD neuropathy without affecting CSD-induced ER stress. Notably, overexpressing O-GlcNAc transferase in the dentate gyrus region of the mouse brain rescued CSD-induced cognitive dysfunction, neuropathy, neuroinflammation, and ER stress responses. Collectively, our findings reveal that dysregulation of O-GlcNAc cycling underlies CSD-induced AD pathology and demonstrate that restoration of OGlcNAcylation protects against CSD-induced neurodegeneration.
Collapse
Affiliation(s)
- Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea
| | - Sang-Min Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea.
| |
Collapse
|
5
|
Liu L, Zhen J, Liu S, Ren L, Zhao G, Liang J, Xu A, Li C, Wu J, Cheung BMY. Association between sleep patterns and galectin-3 in a Chinese community population. BMC Public Health 2024; 24:1323. [PMID: 38755574 PMCID: PMC11097462 DOI: 10.1186/s12889-024-18811-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/09/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Irregular sleep patterns have been associated with inflammation. Galectin-3, a novel biomarker, plays an important role in inflammation. We investigated the relationship between sleep patterns and galectin-3 in a Chinese population. METHODS A total of 1,058 participants from the Shenzhen-Hong Kong United Network on Cardiovascular Disease study were included in the analysis. Age and sex-adjusted linear regression models were employed to investigate the relationship between galectin-3 level and traditional metabolic biomarkers. Logistic regression models were used to estimate the association among sleep disturbance, nighttime sleep duration, and daytime napping duration and elevated galectin-3, with elevated galectin-3 defined as galectin-3 level > 65.1 ng/ml. RESULTS Of study participants, the mean age was 45.3 years and 54.3% were women. Waist circumference, natural logarithm (ln)-transformed triglyceride, and ln-transformed high sensitivity C-reactive protein were positively associated with galectin-3 level (age and sex-adjusted standardized β [95% confidence interval (CI)], 0.12 [0.04, 0.21], 0.11 [0.05, 0.17], and 0.08 [0.02, 0.14], respectively). Sleep disturbance was associated with elevated galectin-3 (odds ratio [95% CI], 1.68 [1.05, 2.68], compared to those without sleep disturbance) after adjusting for traditional metabolic biomarkers. No interaction was observed between galectin-3 and age, sex, obesity, hypertension, and diabetes on sleep disturbance. No association was found between nighttime sleep duration or daytime napping duration and elevated galectin-3. CONCLUSIONS Our study provides evidence of a significant association between sleep disturbance and elevated galectin-3 level, independent of traditional metabolic biomarkers. Screening and interventions on galectin-3 could assist in preventing sleep disturbance-induced inflammatory disease.
Collapse
Affiliation(s)
- Lin Liu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Juanying Zhen
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shuyun Liu
- Department of Neurology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Lijie Ren
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Guoru Zhao
- CAS Key Laboratory of Human-Machine Intelligence-Synergy Systems, Research Center for Neural Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jianguo Liang
- Precision Health Research Center Company Limited, Hong Kong SAR, China
| | - Aimin Xu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chao Li
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Bernard Man Yung Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
- Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| |
Collapse
|
6
|
Xiao X, Rui Y, Jin Y, Chen M. Relationship of Sleep Disorder with Neurodegenerative and Psychiatric Diseases: An Updated Review. Neurochem Res 2024; 49:568-582. [PMID: 38108952 DOI: 10.1007/s11064-023-04086-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/19/2023]
Abstract
Sleep disorders affect many people worldwide and can accompany neurodegenerative and psychiatric diseases. Sleep may be altered before the clinical manifestations of some of these diseases appear. Moreover, some sleep disorders affect the physiological organization and function of the brain by influencing gene expression, accelerating the accumulation of abnormal proteins, interfering with the clearance of abnormal proteins, or altering the levels of related hormones and neurotransmitters, which can cause or may be associated with the development of neurodegenerative and psychiatric diseases. However, the detailed mechanisms of these effects are unclear. This review mainly focuses on the relationship between and mechanisms of action of sleep in Alzheimer's disease, depression, and anxiety, as well as the relationships between sleep and Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. This summary of current research hotspots may provide researchers with better clues and ideas to develop treatment solutions for neurodegenerative and psychiatric diseases associated with sleep disorders.
Collapse
Affiliation(s)
- Xiao Xiao
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yimin Rui
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yu Jin
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
7
|
Wang M, Zhao H, Zhang Z, Zhao Z, Wu H. Down-regulating insulin-like growth factor-1 receptor reduces amyloid-β deposition in mice cortex induced by chronic sleep restriction. Neurosci Lett 2023; 808:137189. [PMID: 36921668 DOI: 10.1016/j.neulet.2023.137189] [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: 08/09/2022] [Revised: 02/27/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023]
Abstract
OBJECTIVE Insufficient sleep affects cognitive function, but the underlying mechanism and potential protective ways are yet to be fully understood. This study aimed to explore the influence of chronic sleep restriction (CSR) on the insulin-like growth factor-1 (IGF-1) signaling pathway, and whether down-regulating IGF-1 signaling pathway would modulate amyloid-β (Aβ) peptides metabolism and its cortical deposition after CSR. Methods 8-week IGF-1R+/- mice and wild-type (WT) C57BL/6 (C57) mice were divided into four groups: IGF-1R+/- CSR (MUSR), IGF-1R+/- control (MUCO), C57 CSR (C57SR) and C57 control (C57CO). CSR model was established by application of slowly rotating drum for 2 months. Body weight and Lee's index were measured. The level of IGF-1 in plasma was measured by enzyme linked immunosorbent assay (ELISA). Aβ accumulation was detected by immunofluorescence. The expressions of amyloid precursor protein (APP), β-site amyloid precursor protein-cleaving enzyme-1 (BACE-1) and C99 were detected using western-blot (WB). Results Two-way ANOVA showed genotypic effect was significant on body weight and Lee's index. Neither treatment effect nor interaction reached significant difference on body weight and Lee's index. The level of IGF-1 in plasma was significantly decreased in C57SR compared with C57CO. Besides, compared with C57CO, Aβ was markedly accumulated in frontal cortex, in parallel with increased expressions of BACE-1 and C99, and with no difference of APP in C57SR group. Further, no significant changes of Aβ, BACE-1, C99 and APP were detected in MUSR compared with MUCO. Conclusions This study showed that CSR could induce the decrease of circulating IGF-1 in mice. By using the IGF-1R+/- mice, we found that down-regulating IGF-1R could reduce Aβ deposition in mice frontal cortex after CSR via inhibiting BACE-1 protein expression and activity, which were independent of the changes of body weight and Lee's index. These findings indicate that the blockage of IGF-1 signaling pathway might be a protection mechanism for alleviating the impact of CSR.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China; Department of hyperbaric oxygen and Neurology, Naval Medical Center, Shanghai 200052, China
| | - Hongyi Zhao
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China; Department of Neurology, NO 984 Hospital of PLA, Beijing 100094, China
| | - Zhaohuai Zhang
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Zhongxin Zhao
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Huijuan Wu
- Department of Neurology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
8
|
Yuan R, Yisen Z, Xiu W, Wei T, Wei W. Effects of enriched environment on the expression of β-amyloid and transport-related proteins LRP1 and RAGE in chronic sleep-deprived mice. Transl Neurosci 2023; 14:20220301. [PMID: 37692085 PMCID: PMC10487385 DOI: 10.1515/tnsci-2022-0301] [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: 04/09/2023] [Revised: 07/11/2023] [Accepted: 07/19/2023] [Indexed: 09/12/2023] Open
Abstract
Sleep plays an important role in the learning process and memory consolidation, and sleep deprivation (SD) leads to inadequate memory consolidation and plays an important role in brain development and plasticity. SD increases β-amyloid levels while impairing cognitive function. We explored the effect of enriched environment (EE) on β-amyloid and transporter protein LRP1 and receptor for advanced glycosylation end-products (RAGE) expression in chronic sleep deprived mice. We randomly divided mice into four groups (n = 10), the standard environment group (Ctrl group), the sleep deprivation group (SD group), the enriched environment intervention group (EE group), and the sleep deprivation plus environmental enrichment intervention group (SD + EE group). A modified multi-platform SD model was used to sleep deprive the mice for 19 h per day. Five hours of EE intervention was performed daily in the EE group and the SD + EE group, respectively. The behavioral measurements of mice were performed by Y-maze method and new object recognition; the expression levels of Aβ1-42, LRP1, and RAGE in prefrontal cortex and hippocampus of mice were measured by immunofluorescence; the expression levels of LRP1 and RAGE in prefrontal cortex and hippocampus were detected by Western blot. The results showed that EE could effectively ameliorate the effects of SD on cognitive impairment, reduce SD induced Aβ deposition, and decrease the expression of RAGE, while increase the expression of LRP1.
Collapse
Affiliation(s)
- Ren Yuan
- Department of Neurology, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Zhang Yisen
- Department of Neurology, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Wang Xiu
- Department of Clinical Laboratory, Wuhan Children’s Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Tang Wei
- Department of Neurology, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning Province, China
| | - Wang Wei
- Department of Basic Medicine, School of Medicine of Dalian University, Dalian, Liaoning Province, China
| |
Collapse
|
9
|
Imbimbo BP, Ippati S, Watling M, Imbimbo C. Role of monomeric amyloid-β in cognitive performance in Alzheimer's disease: Insights from clinical trials with secretase inhibitors and monoclonal antibodies. Pharmacol Res 2023; 187:106631. [PMID: 36586644 DOI: 10.1016/j.phrs.2022.106631] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
According to the β-amyloid (Aβ) hypothesis of Alzheimer's disease (AD), brain Aβ accumulation is the primary cascade event leading to cognitive deficit and dementia. Numerous anti-Aβ drugs either inhibiting production or aggregation of Aβ or stimulating its clearance have failed to show clinical benefit in large scale AD trials, with β- and γ-secretase inhibitors consistently worsening cognitive and clinical decline. In June 2021, the FDA approved aducanumab, an anti-Aβ monoclonal antibody for early AD based on its ability to reduce brain amyloid plaques, while two other amyloid-clearing antibodies (lecanemab and donanemab) have recently produced encouraging cognitive and clinical results. We reviewed AD trials using PubMed, meeting abstracts and ClinicalTrials.gov and evaluated the effects of such drugs on cerebrospinal fluid (CSF) Aβ levels, correlating them with cognitive effects. We found that β-secretase and γ-secretase inhibitors produce detrimental cognitive effects by significantly reducing CSF Aβ levels. We speculate that monoclonal antibodies targeting Aβ protofibrils, fibrils or plaques may improve cognitive performance in early AD by increasing soluble Aβ levels through Aβ aggregate disassembly and/or stabilization of existing Aβ monomers.These findings suggest that the real culprit in AD may be decreased levels of soluble monomeric Aβ due to sequestration into brain Aβ aggregates and plaques.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy.
| | - Stefania Ippati
- San Raffaele Scientific Institute, San Raffaele Hospital, 20132 Milan, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Ltd, Reading, UK
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
10
|
Peng X, Fan R, Xie L, Shi X, Wang F, Xu W, Dong K, Zhang S, Ma D, Yu X, Yang Y. Time-restricted feeding rescues circadian disruption-aggravated progression of Alzheimer's disease in diabetic mice. J Nutr Biochem 2022; 110:109128. [PMID: 35977665 DOI: 10.1016/j.jnutbio.2022.109128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 07/09/2022] [Accepted: 07/14/2022] [Indexed: 01/13/2023]
Abstract
Circadian rhythms, type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) are closely related and interacted with each other. We have previously showed circadian disruption aggravated progression of AD in T2DM mice. Time-restricted feeding (TRF) is shown to be a potential synchronizer. This study aims to determine whether TRF has a protective effect against the circadian disruption-aggravated progression of AD in T2DM. 6-week-old male diabetic (db/db) mice and wildtype (wt/wt) mice were kept under normal 12:12 light/dark cycles or altered 6:18 light/dark cycles (dark extended to 18 h) with or without TRF (food restricted to 8 h during the active (dark) period). After 8 weeks, three behavioral tests (open field test, novel object recognition test, barnes maze test) were performed and the circadian gene expression, body weight, lipid levels and AD-associated tau phosphorylation were evaluated. We found altered light/dark cycles contributed to disruptive circadian rhythms in the hippocampus of db/db mice, while TRF prevented this effect. TRF also ameliorated circadian disruption-aggravated increased body weight and lipid accumulation in db/db mice. Importantly, the db/db mice under circadian disruption showed impaired cognition accompanied by increased tau phosphorylation, whereas TRF reversed these changes. The altered light/dark cycles only affected circadian rhythms but not other indicators like plasma/liver lipids, cognition and tau phosphorylation in the wt/wt mice. Collectively, TRF has a protective effect against altered light/dark cycles-aggravated AD progression in diabetic mice.
Collapse
Affiliation(s)
- Xuemin Peng
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Rongping Fan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Lei Xie
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Xiaoli Shi
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Fen Wang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Weijie Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Kun Dong
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Shujun Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Delin Ma
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Xuefeng Yu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China.
| |
Collapse
|
11
|
Yang Y, Wang X, Xiao A, Han J, Wang Z, Wen M. Ketogenic diet prevents chronic sleep deprivation-induced Alzheimer’s disease by inhibiting iron dyshomeostasis and promoting repair via Sirt1/Nrf2 pathway. Front Aging Neurosci 2022; 14:998292. [PMID: 36118706 PMCID: PMC9475074 DOI: 10.3389/fnagi.2022.998292] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022] Open
Abstract
Sleep deprivation (SD) is one of the main risk factors for Alzheimer’s disease (AD), but the underlying mechanism is still unclear. Ketogenic diet (KD) has been shown widely neuroprotective effects but less known about its effect on SD-induced AD. In the present study, a continuous 21 days SD mouse model with or without KD was established. The changes of cognitive function, pathological hallmarks of AD, ferroptosis, and intracellular signal pathways in mice were detected by Morris water maze, ThS staining, diaminobenzidine (DAB)-enhanced Perls’ stain, antioxidant assay, immuno-histochemistry, and western blot. The results showed that KD can prevent the cognitive deficiency, amyloid deposition and hyperphosphorylated tau induced by chronic SD. Analysis of ferroptosis revealed that KD can inhibit iron dyshomeostasis by down-regulating the expression of TfR1 and DMT1 and up-regulating the expression of FTH1, FPN1. Meanwhile, KD alleviated oxidative stress with elevated xCT/GPX4 axis, FSP1 and reduced MDA. In addition, KD could promote neuronal repair by enhancing BDNF and DCX. Further studies demonstrated that KD activated Sirt1/Nrf2 signaling pathway in the hippocampus in SD-exposed mice. Our finding firstly suggested that KD could prevent chronic SD-induced AD by inhibiting ferroptosis and improving the neuronal repair ability via Sirt1/Nrf2 signaling pathway.
Collapse
|
12
|
Wong H, Buck JM, Borski C, Pafford JT, Keller BN, Milstead RA, Hanson JL, Stitzel JA, Hoeffer CA. RCAN1 knockout and overexpression recapitulate an ensemble of rest-activity and circadian disruptions characteristic of Down syndrome, Alzheimer's disease, and normative aging. J Neurodev Disord 2022; 14:33. [PMID: 35610565 PMCID: PMC9128232 DOI: 10.1186/s11689-022-09444-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Regulator of calcineurin 1 (RCAN1) is overexpressed in Down syndrome (DS), but RCAN1 levels are also increased in Alzheimer's disease (AD) and normal aging. AD is highly comorbid among individuals with DS and is characterized in part by progressive neurodegeneration that resembles accelerated aging. Importantly, abnormal RCAN1 levels have been demonstrated to promote memory deficits and pathophysiology that appear symptomatic of DS, AD, and aging. Anomalous diurnal rest-activity patterns and circadian rhythm disruptions are also common in DS, AD, and aging and have been implicated in facilitating age-related cognitive decline and AD progression. However, no prior studies have assessed whether RCAN1 dysregulation may also promote the age-associated alteration of rest-activity profiles and circadian rhythms, which could in turn contribute to neurodegeneration in DS, AD, and aging. METHODS The present study examined the impacts of RCAN1 deficiency and overexpression on the photic entrainment, circadian periodicity, intensity and distribution, diurnal patterning, and circadian rhythmicity of wheel running in young (3-6 months old) and aged (9-14 months old) mice of both sexes. RESULTS We found that daily RCAN1 levels in the hippocampus and suprachiasmatic nucleus (SCN) of light-entrained young mice are generally constant and that balanced RCAN1 expression is necessary for normal circadian locomotor activity rhythms. While the light-entrained diurnal period was unaltered, RCAN1-null and RCAN1-overexpressing mice displayed lengthened endogenous (free-running) circadian periods like mouse models of AD and aging. In light-entrained young mice, RCAN1 deficiency and overexpression also recapitulated the general hypoactivity, diurnal rest-wake pattern fragmentation, and attenuated amplitudes of circadian activity rhythms reported in DS, preclinical and clinical AD, healthily aging individuals, and rodent models thereof. Under constant darkness, RCAN1-null and RCAN1-overexpressing mice displayed altered locomotor behavior indicating circadian clock dysfunction. Using the Dp(16)1Yey/+ (Dp16) mouse model for DS, which expresses three copies of Rcan1, we found reduced wheel running activity and rhythmicity in both light-entrained and free-running young Dp16 mice like young RCAN1-overexpressing mice. Critically, these diurnal and circadian deficits were rescued in part or entirely by restoring Rcan1 to two copies in Dp16 mice. We also found that RCAN1 deficiency but not RCAN1 overexpression altered protein levels of the clock gene Bmal1 in the SCN. CONCLUSIONS Collectively, this study's findings suggest that both loss and aberrant gain of RCAN1 precipitate anomalous light-entrained diurnal and circadian activity patterns emblematic of DS, AD, and possibly aging.
Collapse
Affiliation(s)
- Helen Wong
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jordan M Buck
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Curtis Borski
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jessica T Pafford
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Bailey N Keller
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
| | - Ryan A Milstead
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jessica L Hanson
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Charles A Hoeffer
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA.
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA.
- Linda Crnic Institute, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
13
|
Nick H, Fenik P, Zhu Y, Veasey S. Hypocretin/orexin influences chronic sleep disruption injury in the hippocampus. Front Aging Neurosci 2022; 14:1025402. [PMID: 36275002 PMCID: PMC9582517 DOI: 10.3389/fnagi.2022.1025402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/20/2022] [Indexed: 01/13/2023] Open
Abstract
Chronic sleep disruption is a risk factor for Alzheimer's disease (AD), yet mechanisms by which sleep disturbances might promote or exacerbate AD are not understood. Short-term sleep loss acutely increases hippocampal amyloid β (Aβ) in wild type (WT) mice and long-term sleep loss increases amyloid plaque in AD transgenic mouse models. Both effects can be influenced by the wake-promoting neuropeptide, hypocretin (HCRT), but whether HCRT influences amyloid accumulation independent of sleep and wake timing modulation remains unclear. Here, we induced chronic fragmentation of sleep (CFS) in WT and HCRT-deficient mice to elicit similar arousal indices, sleep bout lengths and sleep bout numbers in both genotypes. We then examined the roles of HCRT in CFS-induced hippocampal Aβ accumulation and injury. CFS in WT mice resulted in increased Aβ42 in the hippocampus along with loss of cholinergic projections and loss of locus coeruleus neurons. Mice with HCRT deficiency conferred resistance to CFS Aβ42 accumulation and loss of cholinergic projections in the hippocampus yet evidenced similar CFS-induced loss of locus coeruleus neurons. Collectively, the findings demonstrate specific roles for orexin in sleep disruption hippocampal injury. Significance statement Chronic fragmentation of sleep (CFS) occurs in common conditions, including sleep apnea syndromes and chronic pain disorders, yet CFS can induce neural injury. Our results demonstrate that under conditions of sleep fragmentation, hypocretin/orexin is essential for the accumulation of amyloid-β and loss of cholinergic projections in the hippocampus observed in response to CFS yet does not influence locus coeruleus neuron response to CFS.
Collapse
Affiliation(s)
- Henry Nick
- Department of Medicine and the Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania Philadelphia, Philadelphia, PA, United States
| | - Polina Fenik
- Department of Medicine and the Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania Philadelphia, Philadelphia, PA, United States
| | - Yan Zhu
- Department of Medicine and the Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania Philadelphia, Philadelphia, PA, United States
| | - Sigrid Veasey
- Department of Medicine and the Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
14
|
Garbarino S, Lanteri P, Bragazzi NL, Magnavita N, Scoditti E. Role of sleep deprivation in immune-related disease risk and outcomes. Commun Biol 2021; 4:1304. [PMID: 34795404 PMCID: PMC8602722 DOI: 10.1038/s42003-021-02825-4] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022] Open
Abstract
Modern societies are experiencing an increasing trend of reduced sleep duration, with nocturnal sleeping time below the recommended ranges for health. Epidemiological and laboratory studies have demonstrated detrimental effects of sleep deprivation on health. Sleep exerts an immune-supportive function, promoting host defense against infection and inflammatory insults. Sleep deprivation has been associated with alterations of innate and adaptive immune parameters, leading to a chronic inflammatory state and an increased risk for infectious/inflammatory pathologies, including cardiometabolic, neoplastic, autoimmune and neurodegenerative diseases. Here, we review recent advancements on the immune responses to sleep deprivation as evidenced by experimental and epidemiological studies, the pathophysiology, and the role for the sleep deprivation-induced immune changes in increasing the risk for chronic diseases. Gaps in knowledge and methodological pitfalls still remain. Further understanding of the causal relationship between sleep deprivation and immune deregulation would help to identify individuals at risk for disease and to prevent adverse health outcomes.
Collapse
Affiliation(s)
- Sergio Garbarino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal/Child Sciences, University of Genoa, 16132, Genoa, Italy.
| | - Paola Lanteri
- Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Nicola Luigi Bragazzi
- Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, ON, M3J 1P3, Canada
| | - Nicola Magnavita
- Postgraduate School of Occupational Medicine, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Department of Woman/Child and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Egeria Scoditti
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100, Lecce, Italy
| |
Collapse
|
15
|
Ahnaou A, Drinkenburg WHIM. Sleep, neuronal hyperexcitability, inflammation and neurodegeneration: Does early chronic short sleep trigger and is it the key to overcoming Alzheimer's disease? Neurosci Biobehav Rev 2021; 129:157-179. [PMID: 34214513 DOI: 10.1016/j.neubiorev.2021.06.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 05/13/2021] [Accepted: 06/25/2021] [Indexed: 01/13/2023]
Abstract
Evidence links neuroinflammation to Alzheimer's disease (AD); however, its exact contribution to the onset and progression of the disease is poorly understood. Symptoms of AD can be seen as the tip of an iceberg, consisting of a neuropathological build-up in the brain of extracellular amyloid-β (Aβ) plaques and intraneuronal hyperphosphorylated aggregates of Tau (pTau), which are thought to stem from an imbalance between its production and clearance resulting in loss of synaptic health and dysfunctional cortical connectivity. The glymphatic drainage system, which is particularly active during sleep, plays a key role in the clearance of proteinopathies. Poor sleep can cause hyperexcitability and promote Aβ and tau pathology leading to systemic inflammation. The early neuronal hyperexcitability of γ-aminobutyric acid (GABA)-ergic inhibitory interneurons and impaired inhibitory control of cortical pyramidal neurons lie at the crossroads of excitatory/inhibitory imbalance and inflammation. We outline, with a prospective framework, a possible vicious spiral linking early chronic short sleep, neuronal hyperexcitability, inflammation and neurodegeneration. Understanding the early predictors of AD, through an integrative approach, may hold promise for reducing attrition in the late stages of neuroprotective drug development.
Collapse
Affiliation(s)
- A Ahnaou
- Dept. of Neuroscience Discovery, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, B-2340, Belgium.
| | - W H I M Drinkenburg
- Dept. of Neuroscience Discovery, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, B-2340, Belgium
| |
Collapse
|
16
|
Disruption of Circadian Clocks Promotes Progression of Alzheimer's Disease in Diabetic Mice. Mol Neurobiol 2021; 58:4404-4412. [PMID: 34018152 DOI: 10.1007/s12035-021-02425-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/07/2021] [Indexed: 01/18/2023]
Abstract
The circadian clock is an endogenous system designed to anticipate and adapt to daily changes in the environment. Alzheimer's disease (AD) is a progressive neurodegenerative disease, which is more prevalent in patients with type 2 diabetes mellitus (T2DM). However, the effects of circadian disruption on mental and physical health for T2DM patients are not yet fully understood, even though circadian disruption has been confirmed to promote the progression of AD in population. By housing db/db mice on a disrupted (a 6:18 light/dark cycle) circadian rhythm, we assessed the circadian gene expression, body weight, cognitive ability, and AD-related pathophysiology. Our results indicated that housing in these conditions led to disrupted diurnal circadian rhythms in the hippocampus of db/db mice and contributed to their weight gain. In the brain, the circadian-disrupted db/db mice showed a decreased cognitive ability and an increased hyperphosphorylation of tau protein, even though no difference was found in amyloid protein (Aβ) plaque deposition. We also found that the hyperphosphorylated tau protein exhibited more disruptive daily oscillations in db/db mice hippocampus under the 6:18 light/dark cycle. Circadian alterations could promote the development of AD in T2DM.
Collapse
|
17
|
Gao Y, Wei S, Gao F, Gao L, Dang L, Shang S, Chen C, Huo K, Wang J, Wang J, Qu Q. Sleep Disturbance is Associated With Higher Plasma Aβ Levels in Cognitively Normal Adults-A Population-Based Cross-Sectional Study. Front Aging Neurosci 2021; 12:615838. [PMID: 33536896 PMCID: PMC7848159 DOI: 10.3389/fnagi.2020.615838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/17/2020] [Indexed: 12/02/2022] Open
Abstract
Objective: Growing evidence suggests that sleep disturbance is a risk factor for Alzheimer's disease (AD). Amyloid-β (Aβ) deposition in the brain is a main pathophysiology of AD. Considering that peripheral Aβ level is associated with brain Aβ deposition, the present study investigated the relationship between sleep disturbance and plasma Aβ levels. Methods: This is a population-based cross-sectional study. A total of 1,459 participants from a village in the suburbs of Xi'an, China, were enrolled from January 3, 2017 to March 26, 2017. Sleep quality was assessed using the Pittsburgh Sleep Quality Index (PSQI), and a PSQI score of <5 points was considered as good sleep quality and a PSQI score of >10 points as poor sleep quality. Cognitive function was assessed with the Mini-Mental State Examination (MMSE). Fasting venous blood was taken in the morning, and the plasma Aβ levels were measured using ELISA. The relationships between plasma Aβ levels and sleep quality were analyzed using multiple linear regression. Results: Among the participants, 231 had poor sleep quality (15.83%). The log-transformed Aβ40 level had significant differences among the different sleep groups (F = 3.216, p = 0.040). The log-transformed Aβ40 level was higher in the poor sleep quality group than that in the general sleep quality group [87.17 (73.42, 107.34) vs. 89.69 (74.81, 125.79) pg/ml, p = 0.016]. In bivariate analysis, sleep quality was negatively associated with the log-transformed plasma Aβ40 level (β = −0.025, p = 0.011). Conclusion: In the community population, poorer sleep quality is associated with a higher plasma Aβ40 level. This indicated that sleep disturbance might also involve in dysfunction of peripheral Aβ clearance.
Collapse
Affiliation(s)
- Yao Gao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shan Wei
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fan Gao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ling Gao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangjun Dang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chen Chen
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Wang
- Huyi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
18
|
Harris SS, Schwerd-Kleine T, Lee BI, Busche MA. The Reciprocal Interaction Between Sleep and Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1344:169-188. [PMID: 34773232 DOI: 10.1007/978-3-030-81147-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
It is becoming increasingly recognized that patients with a variety of neurodegenerative diseases exhibit disordered sleep/wake patterns. While sleep impairments have typically been thought of as sequelae of underlying neurodegenerative processes in sleep-wake cycle regulating brain regions, including the brainstem, hypothalamus, and basal forebrain, emerging evidence now indicates that sleep deficits may also act as pathophysiological drivers of brain-wide disease progression. Specifically, recent work has indicated that impaired sleep can impact on neuronal activity, brain clearance mechanisms, pathological build-up of proteins, and inflammation. Altered sleep patterns may therefore be novel (potentially reversible) dynamic functional markers of proteinopathies and modifiable targets for early therapeutic intervention using non-invasive stimulation and behavioral techniques. Here we highlight research describing a potentially reciprocal interaction between impaired sleep and circadian patterns and the accumulation of pathological signs and features in Alzheimer's disease, the most prevalent neurodegenerative disease in the elderly.
Collapse
Affiliation(s)
| | | | - Byung Il Lee
- UK Dementia Research Institute at UCL, London, UK
| | | |
Collapse
|
19
|
Brice KN, Hagen CW, Peterman JL, Figg JW, Braden PN, Chumley MJ, Boehm GW. Chronic sleep restriction increases soluble hippocampal Aβ-42 and impairs cognitive performance. Physiol Behav 2020; 226:113128. [PMID: 32791178 DOI: 10.1016/j.physbeh.2020.113128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 11/17/2022]
Abstract
Currently, over 44 million people worldwide suffer from Alzheimer's disease (AD). A common feature of AD is disrupted sleep. Sleep is essential for many psychological and physiological functions, though 35.3% of adults report getting less than 7 hours per night. The present research examined whether chronic sleep restriction would elevate hippocampal amyloid-beta1-42 expression or alter cognitive ability in adult C57BL/6 mice. Chronic sleep restriction was associated with cognitive impairment and increased hippocampal amyloid-beta. Thus, chronic sleep loss may have a detrimental effect upon cognitive function, in part, via increasing amyloid-beta levels in the hippocampus, even in non-genetically modified mice.
Collapse
Affiliation(s)
- Kelly N Brice
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Christopher W Hagen
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Julia L Peterman
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - John W Figg
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Paige N Braden
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Michael J Chumley
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Gary W Boehm
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA.
| |
Collapse
|
20
|
Del Gallo F, Bianchi S, Bertani I, Messa M, Colombo L, Balducci C, Salmona M, Imeri L, Chiesa R. Sleep inhibition induced by amyloid-β oligomers is mediated by the cellular prion protein. J Sleep Res 2020; 30:e13187. [PMID: 32902030 DOI: 10.1111/jsr.13187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 11/27/2022]
Abstract
Sleep is severely impaired in patients with Alzheimer's disease. Amyloid-β deposition in the brain of Alzheimer's disease patients is a key event in its pathogenesis and is associated with disrupted sleep, even before the appearance of cognitive decline. Because soluble amyloid-β oligomers are the key mediators of synaptic and cognitive dysfunction in Alzheimer's disease and impair long-term memory in rodents, the first aim of this study was to test the hypothesis that amyloid-β oligomers would directly impair sleep in mice. The cellular prion protein is a cell surface glycoprotein of uncertain function. Because cellular prion protein binds oligomeric amyloid-β with high affinity and mediates some of its neurotoxic effects, the second aim of the study was to test whether amyloid-β oligomer-induced sleep alterations were mediated by cellular prion protein. To address these aims, wild-type and cellular prion protein-deficient mice were given acute intracerebroventricular injections (on different days, at lights on) of vehicle and synthetic amyloid-β oligomers. Compared to vehicle, amyloid-β oligomers significantly reduced the amount of time spent in non-rapid eye movement sleep by wild-type mice during both the light and dark phases of the light-dark cycle. The amount of time spent in rapid eye movement sleep was reduced during the dark phase. Sleep was also fragmented by amyloid-β oligomers, as the number of transitions between states increased in post-injection hours 9-24. No such effects were observed in cellular prion protein-deficient mice. These results show that amyloid-β oligomers do inhibit and fragment sleep, and that these effects are mediated by cellular prion protein.
Collapse
Affiliation(s)
| | - Susanna Bianchi
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Ilaria Bertani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Massimo Messa
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luca Imeri
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Roberto Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
21
|
Brown AMC, Gervais NJ. Role of Ovarian Hormones in the Modulation of Sleep in Females Across the Adult Lifespan. Endocrinology 2020; 161:5879359. [PMID: 32735650 PMCID: PMC7450669 DOI: 10.1210/endocr/bqaa128] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022]
Abstract
Ovarian hormones, including 17β-estradiol, are implicated in numerous physiological processes, including sleep. Beginning at puberty, girls report more sleep complaints than boys, which is maintained throughout the reproductive life stage. Sleep problems are exacerbated during the menopausal transition, evidenced by greater risk for sleep disorders. There is emerging evidence that menopause-associated hormone loss contributes to this elevated risk, but age is also an important factor. The extent to which menopause-associated sleep disturbance persists into postmenopause above and beyond the effects of age remains unknown. Untreated sleep disturbances have important implications for cognitive health, as they are emerging as risk factors for dementia. Given that sleep loss impairs memory, an important knowledge gap concerns the role played by menopause-associated hormone loss in exacerbating sleep disturbance and, ultimately, cognitive function in aging women. In this review, we take a translational approach to illustrate the contribution of ovarian hormones in maintaining the sleep-wake cycle in younger and middle-aged females, with evidence implicating 17β-estradiol in supporting the memory-promoting effects of sleep. Sleep physiology is briefly reviewed before turning to behavioral and neural evidence from young females linking 17β-estradiol to sleep-wake cycle maintenance. Implications of menopause-associated 17β-estradiol loss is also reviewed before discussing how ovarian hormones may support the memory-promoting effects of sleep, and why menopause may exacerbate pathological aging via effects on sleep. While still in its infancy, this research area offers a new sex-based perspective on aging research, with a focus on a modifiable risk factor for pathological aging.
Collapse
Affiliation(s)
- Alana M C Brown
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Nicole J Gervais
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Correspondence: Nicole J. Gervais, University of Toronto, Department of Psychology, 100 St. George Street, Toronto, ON, Canada M5S 3G3. E-mail:
| |
Collapse
|
22
|
Thomas J, Ooms SJ, Mentink LJ, Booij J, Olde Rikkert MGM, Overeem S, Kessels RPC, Claassen JAHR. Effects of long-term sleep disruption on cognitive function and brain amyloid-β burden: a case-control study. ALZHEIMERS RESEARCH & THERAPY 2020; 12:101. [PMID: 32847615 PMCID: PMC7450576 DOI: 10.1186/s13195-020-00668-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/13/2020] [Indexed: 12/20/2022]
Abstract
Background Recent evidence indicates that disrupted sleep could contribute to the development of Alzheimer’s disease by influencing the production and/or clearance of the amyloid-β protein. We set up a case-control study to investigate the association between long-term work-induced sleep disruption, cognitive function, and brain amyloid-β burden. Methods Nineteen male maritime pilots (aged 48–60 years) with chronic work-related sleep disruption and a sex-, age-, and education-matched control sample (n = 16, aged 50–60 years) with normal sleep completed the study. Primary sleep disorders were ruled out with in-lab polysomnography. Additional sleep measurements were obtained at home using actigraphy, sleep-wake logs, and a single-lead EEG device. Cognitive function was assessed with a neuropsychological test battery, sensitive to early symptomatic Alzheimer’s disease. Brain amyloid-β burden was assessed in maritime pilots using 18F-flutemetamol amyloid PET-CT. Results Maritime pilots reported significantly worse sleep quality (Pittsburgh Sleep Quality Index (PSQI) = 8.8 ± 2.9) during work weeks, compared to controls (PSQI = 3.2 ± 1.4; 95% CI 0.01 to 2.57; p = 0.049). This was confirmed with actigraphy-based sleep efficiency (86% ± 3.8 vs. 89.3% ± 4.3; 95% CI 0.43 to 6.03; p = 0.03). Home-EEG recordings showed less total sleep time (TST) and deep sleep time (DST) during work weeks compared to rest weeks (TST 318.56 (250.21–352.93) vs. TST 406.17 (340–425.98); p = 0.001; DST 36.75 (32.30–58.58) vs. DST 51.34 (48.37–69.30); p = 0.005)). There were no differences in any of the cognitive domains between the groups. For brain amyloid-β levels, mean global cortical standard uptake value ratios of 18F-flutemetamol were all in the normal range (1.009 ± 0.059; 95% CI 0.980 to 1.037), confirmed by visual reads. Conclusions Capitalizing on the particular work-rest schedule of maritime pilots, this study with a small sample size observed that long-term intermittent sleep disruption had no effects on global brain amyloid-β levels or cognitive function.
Collapse
Affiliation(s)
- Jana Thomas
- Department of Geriatric Medicine, Radboud University Medical Center, 6525, GC, Nijmegen, The Netherlands. .,Donders Institute for Brain, Cognition and Behaviour, 6525, HR, Nijmegen, The Netherlands. .,Radboud Alzheimer Centre, 6525, GA, Nijmegen, The Netherlands.
| | - Sharon J Ooms
- Donders Institute for Brain, Cognition and Behaviour, 6525, HR, Nijmegen, The Netherlands.,Radboud Alzheimer Centre, 6525, GA, Nijmegen, The Netherlands
| | - Lara J Mentink
- Department of Geriatric Medicine, Radboud University Medical Center, 6525, GC, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, 6525, HR, Nijmegen, The Netherlands.,Radboud Alzheimer Centre, 6525, GA, Nijmegen, The Netherlands
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, 6525, GC, Nijmegen, The Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Academic Medical Center, 1105, AZ, Amsterdam, The Netherlands
| | - Marcel G M Olde Rikkert
- Department of Geriatric Medicine, Radboud University Medical Center, 6525, GC, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, 6525, HR, Nijmegen, The Netherlands.,Radboud Alzheimer Centre, 6525, GA, Nijmegen, The Netherlands
| | - Sebastiaan Overeem
- Sleep Medicine Centre Kempenhaeghe, 5591, VE, Heeze, The Netherlands.,Eindhoven University of Technology, 5612, AZ, Eindhoven, The Netherlands
| | - Roy P C Kessels
- Donders Institute for Brain, Cognition and Behaviour, 6525, HR, Nijmegen, The Netherlands.,Radboud Alzheimer Centre, 6525, GA, Nijmegen, The Netherlands.,Department of Medical Psychology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatric Medicine, Radboud University Medical Center, 6525, GC, Nijmegen, The Netherlands. .,Donders Institute for Brain, Cognition and Behaviour, 6525, HR, Nijmegen, The Netherlands. .,Radboud Alzheimer Centre, 6525, GA, Nijmegen, The Netherlands.
| |
Collapse
|
23
|
Liew SC, Aung T. Sleep deprivation and its association with diseases- a review. Sleep Med 2020; 77:192-204. [PMID: 32951993 DOI: 10.1016/j.sleep.2020.07.048] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/08/2020] [Accepted: 07/27/2020] [Indexed: 01/02/2023]
Abstract
Sleep deprivation, a consequence of multiple health problems or a cause of many major health risks, is a significant public health concern in this era. In the recent years, numerous reports have been added to the literature to provide explanation and to answer previously unanswered questions on this important topic but comprehensive updates and reviews in this aspect remain scarce. The present study identified 135 papers that investigated the association between sleep deprivation and health risks, including cardiovascular, respiratory, neurological, gastrointestinal, immunology, dermatology, endocrine, and reproductive health. In this review, we aimed to provide insight into the association between sleep deprivation and the development of diseases. We reviewed the latest updates available in the literature and particular attention was paid to reports that detailed all possible causal relationships involving both extrinsic and intrinsic factors that may be relevant to this topic. Various mechanisms by which sleep deprivation may affect health were presented and discussed, and this review hopes to serve as a platform for ideas generation for future research.
Collapse
Affiliation(s)
- Siaw Cheok Liew
- Department of Clinical Competence, Perdana University-Royal College of Surgeons in Ireland, Kuala Lumpur, Malaysia.
| | - Thidar Aung
- Department of Biochemistry, Perdana University-Royal College of Surgeons in Ireland, Kuala Lumpur, Malaysia
| |
Collapse
|
24
|
Owen JE, Veasey SC. Impact of sleep disturbances on neurodegeneration: Insight from studies in animal models. Neurobiol Dis 2020; 139:104820. [PMID: 32087293 PMCID: PMC7593848 DOI: 10.1016/j.nbd.2020.104820] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/31/2020] [Accepted: 02/18/2020] [Indexed: 01/18/2023] Open
Abstract
Chronic short sleep or extended wake periods are commonly observed in most industrialized countries. Previously neurobehavioral impairment following sleep loss was considered to be a readily reversible occurrence, normalized upon recovery sleep. Recent clinical studies suggest that chronic short sleep and sleep disruption may be risk factors for neurodegeneration. Animal models have been instrumental in determining whether disturbed sleep can injure the brain. We now understand that repeated periods of extended wakefulness across the typical sleep period and/or sleep fragmentation can have lasting effects on neurogenesis and select populations of neurons and glia. Here we provide a comprehensive overview of the advancements made using animal models of sleep loss to understand the extent and mechanisms of chronic short sleep induced neural injury.
Collapse
Affiliation(s)
- Jessica E Owen
- Chronobiology and Sleep Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sigrid C Veasey
- Chronobiology and Sleep Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Grimmer T, Laub T, Hapfelmeier A, Eisele T, Fatke B, Hölzle P, Lüscher S, Parchmann AM, Rentrop M, Schwerthöffer D, Müller-Sarnowski F, Ortner M, Goldhardt O, Kurz A, Förstl H, Alexopoulos P. The overnight reduction of amyloid β 1-42 plasma levels is diminished by the extent of sleep fragmentation, sAPP-β, and APOE ε4 in psychiatrists on call. Alzheimers Dement 2020; 16:759-769. [PMID: 32270596 DOI: 10.1002/alz.12072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION In mice there might be an association between sleep deprivation and amyloid β plasma levels. Hence, we examined whether amyloid plasma levels are associated with sleep duration or fragmentation in 17 psychiatrists on-call. METHODS Amyloid β (Aβ)42, Aβ40, and soluble amyloid precursor protein β (sAPP-β) plasma concentrations were measured at the beginning and end of 90 on-call nights, and analyzed using generalized linear models. RESULTS In on-call nights, a 10.7% reduction of Aβ42 was revealed overnight. Every single short sleep interruption diminished this reduction by 5.4%, as well as every pg/mL of sAPP-β by 1.2%, each copy of APOE ε4 by 10.6%, and each year of professional experience by 3.0%. DISCUSSION The extent of sleep fragmentation diminishes the physiological overnight reduction of Aβ42 but not Aβ40 plasma levels in the same direction as the enzyme for Aβ42 production, the genetic risk factor for Alzheimer's disease (AD), and on-call experience. Might on-call duty and sleep fragmentation in general alter the risk for AD?
Collapse
Affiliation(s)
- Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Theresa Laub
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Alexander Hapfelmeier
- Institute of Medical Statistics and Epidemiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Tamara Eisele
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Bastian Fatke
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Patricia Hölzle
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Sandra Lüscher
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Anna-Mareike Parchmann
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Michael Rentrop
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Dirk Schwerthöffer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Felix Müller-Sarnowski
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Marion Ortner
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Alexander Kurz
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Hans Förstl
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Panagiotis Alexopoulos
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| |
Collapse
|
26
|
Zuo JX, Li M, Jiang L, Lan F, Tang YY, Kang X, Zou W, Wang CY, Zhang P, Tang XQ. Hydrogen Sulfide Prevents Sleep Deprivation-Induced Hippocampal Damage by Upregulation of Sirt1 in the Hippocampus. Front Neurosci 2020; 14:169. [PMID: 32218719 PMCID: PMC7078349 DOI: 10.3389/fnins.2020.00169] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/14/2020] [Indexed: 11/13/2022] Open
Abstract
Sleep deprivation (SD) induces hippocampal damage. Hydrogen sulfide (H2S) is a neuronal protective factor. Silence information regulating factor 1 (Sirt1) plays an important role in neuroprotection. Therefore, this study was aimed at exploring whether H2S meliorates SD-induced hippocampal damage and whether Sirt1 mediates this protective role of H2S. We found that sodium hydrosulfide (NaHS, a donor of H2S) alleviated SD-generated hippocampal oxidative stress, including increases in the activation of SOD and the level of GSH as well as a decrease in the level of MDA. Meanwhile, we found that NaHS reduced SD-exerted hippocampal endoplasmic reticulum (ER) Stress, including downregulations of GRP78, CHOP, and cleaved-caspase-12 expression. Moreover, NaHS reduced the apoptosis in the SD-exposed hippocampus, and this included decreases in the number of apoptotic cells and the activation of caspase-3, downregulation of Bax expression, and upregulation of Bcl-2 expression. NaHS upregulated the expression of Sirt1 in the hippocampus of SD-exposed rats. Furthermore, Sirtinol, the inhibitor of Sirt1, abrogated the protection of NaHS against SD-exerted hippocampal oxidative stress, ER stress, and apoptosis. These results suggested that H2S alleviates SD-induced hippocampal damage by upregulation of hippocampal Sirt1.
Collapse
Affiliation(s)
- Jin-Xi Zuo
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Min Li
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Li Jiang
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Fang Lan
- Department of Neurology, The First Affiliated Hospital, University of South China, Hengyang, China.,Institute of Neurology, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yi-Yun Tang
- Department of Physiology, Hengyang Medical College, University of South China, Hengyang, China
| | - Xuan Kang
- Institute of Neurology, The First Affiliated Hospital, University of South China, Hengyang, China.,Department of Physiology, Hengyang Medical College, University of South China, Hengyang, China
| | - Wei Zou
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Chun-Yan Wang
- Department of Physiology, Hengyang Medical College, University of South China, Hengyang, China
| | - Ping Zhang
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Xiao-Qing Tang
- Institute of Neurology, The First Affiliated Hospital, University of South China, Hengyang, China.,Department of Physiology, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
27
|
Fotuhi SN, Khalaj-Kondori M, Hoseinpour Feizi MA, Talebi M. Long Non-coding RNA BACE1-AS May Serve as an Alzheimer's Disease Blood-Based Biomarker. J Mol Neurosci 2019; 69:351-359. [PMID: 31264051 DOI: 10.1007/s12031-019-01364-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 06/25/2019] [Indexed: 12/15/2022]
Abstract
Circulating long noncoding RNAs (lncRNAs) might serve as biomarkers for different pathological conditions. BACE1-AS lncRNA upregulates in the brain of people with Alzheimer's disease (AD) and might be detected in the bloodstream. To reveal if lncRNA BACE1-AS may serve as a blood-based biomarker for AD, we compared its levels in plasma and plasma-derived exosomes between AD (n = 45) and healthy people (n = 36). Exosomes were purified from plasma by Invitrogen™ Total Exosome Isolation Kit and characterized by scanning electron microscopy (SEM) and dynamic light scattering (DLS). Total RNA was extracted from whole plasma, and plasma-derived exosomes using TRIzol® LS or TRIzol® Reagents respectively were then reverse transcribed to the cDNA using PrimeScript II cDNA synthesis kit. The BACE1-AS levels were quantified by real-time PCR, and their biomarker potencies were evaluated using ROC curve analysis. Results obtained verified the presence of BACE1-AS in the plasma samples of both AD and healthy controls. We did not observe any significant differences between the levels of BACE1-AS in the plasma or plasma-derived exosomes of AD and control people. However, there were significant differences between AD subgroups and control in the whole plasma samples. The BACE1-AS level was low in pre-AD subgroup but it was high in full-AD people compared to the healthy controls. Moreover, ROC curve analysis revealed that lncRNA BACE1-AS may discriminate pre-AD and healthy control (75% sensitivity and 100% specificity), full-AD and healthy control (68% sensitivity and 100% specificity), and pre-AD and full-AD subgroups (78% sensitivity and 100% specificity), highlighting its potential as a biomarker for AD development. In conclusion, plasma BACE1-AS level may serve as a potent blood-based biomarker for Alzheimer's disease.
Collapse
Affiliation(s)
- Seyedeh Nahid Fotuhi
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran.
| | | | - Mahnaz Talebi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Cordone S, Annarumma L, Rossini PM, De Gennaro L. Sleep and β-Amyloid Deposition in Alzheimer Disease: Insights on Mechanisms and Possible Innovative Treatments. Front Pharmacol 2019; 10:695. [PMID: 31281257 PMCID: PMC6595048 DOI: 10.3389/fphar.2019.00695] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/28/2019] [Indexed: 02/05/2023] Open
Abstract
The growing interest in the preclinical stage of Alzheimer's disease (AD) led investigators to identify modifiable risk and predictive factors useful to design early intervention strategies. The preclinical stage of AD is characterized by β-amyloid (Aβ) aggregation into amyloid plaques and tau phosphorylation and aggregation into neurofibrillary tangles. There is a consensus on the importance of sleep within this context: the bidirectional relationship between sleep and AD pathology is supported by growing evidence that proved that the occurrence of sleep changes starting from the preclinical stage of AD, many years before the onset of cognitive decline. Hence, we review the most recent studies on sleep disturbances related to Aβ and the effects of sleep deprivation on Aβ accumulation in animal and human models. We also discuss evidence on the role of sleep in clearing the brain of toxic metabolic by-products, with original findings of the clearance activity of the glymphatic system stimulated by sleep. Furthermore, starting from new recent advances about the relationship between slow-wave sleep (SWS) and Aβ burden, we review the results of recent electroencephalographic (EEG) studies in order to clarify the possible role of SWS component disruption as a novel mechanistic pathway through which Aβ pathology may contribute to cognitive decline and, conversely, the eventual useful role of SWS in facilitating Aβ clearance. Finally, we discuss some promising innovative, effective, low-risk, non-invasive interventions, although empirical evidence on the efficacy of sleep interventions in improving the course of AD is at the very beginning.
Collapse
Affiliation(s)
- Susanna Cordone
- Department of Psychology, University of Rome "Sapienza," Rome, Italy
| | | | - Paolo Maria Rossini
- Department of Neurological, Motor and Sensory Sciences, IRCCS San Raffaele Pisana, Rome, Italy.,Institute of Neurology, Catholic University of The Sacred Heart, Rome, Italy
| | - Luigi De Gennaro
- Department of Psychology, University of Rome "Sapienza," Rome, Italy
| |
Collapse
|
29
|
Zhao H, He Y, Li H, Zhu A, Ye Y, Liu G, Zhao C, Zhang X. The opposite role of alternatively spliced isoforms of LINC00477 in gastric cancer. Cancer Manag Res 2019; 11:4569-4576. [PMID: 31191018 PMCID: PMC6529032 DOI: 10.2147/cmar.s202430] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/09/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Aberrant transcript alternative splicing is an important regulatory process closely connected with oncogenesis. Purpose: The objective of this study was to determine the phenotype and function of a novel long noncoding RNA (lncRNA) LINC00477 in gastric cancer. Patients and methods: The gastric cancer samples of 140 from Oncomine database and 17 from our own hospital, as well as three gastric cancer cell lines MKN-45, AGS and KATO III were used in this study. The expression of the spliced isoforms of LINC00477 were tested. The tumor effects of LINC00477 on gastric cancer were investigated in vitro and in vivo. The mechanism of LINC00477 interacted with aconitase 1 (ACO1) was further examined by RIP and pull down assay. Results: The overall expression of LINC00477 was reduced in gastric cancers compared to normal gastric tissues. The isoform 1 of LINC00477 was down-regulated while the isoform 2 was up-regulated in gastric cancer cells. The opposite role of isoforms 1 and 2 in the proliferation and migration of cancer cells in vitro and in vivo was observed. Furthermore, isoform 1 of LINC00477 was determined to interact with ACO1 and suppress the conversion ability from citrate to isocitrate by ACO1. Conclusion: we presented the important roles of the spliced isoforms of long noncoding RNA, LINC00477 in gastric carcinogenesis.
Collapse
Affiliation(s)
- Hongchao Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yuanhang He
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Haohao Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Ali Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yanwei Ye
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Guanghui Liu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Chunlin Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xiefu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| |
Collapse
|
30
|
Dufort-Gervais J, Mongrain V, Brouillette J. Bidirectional relationships between sleep and amyloid-beta in the hippocampus. Neurobiol Learn Mem 2019; 160:108-117. [DOI: 10.1016/j.nlm.2018.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/18/2018] [Accepted: 06/14/2018] [Indexed: 12/17/2022]
|
31
|
Panza F, Lozupone M, Seripa D, Imbimbo BP. Amyloid-β immunotherapy for alzheimer disease: Is it now a long shot? Ann Neurol 2019; 85:303-315. [PMID: 30635926 DOI: 10.1002/ana.25410] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 01/01/2023]
Abstract
The amyloid-β (Aβ) cascade hypothesis of Alzheimer disease (AD) holds that brain accumulation of Aβ initiates the disease process. Accordingly, drug research has targeted Aβ production, clearance, and deposition as therapeutic strategies. Unfortunately, candidate drugs have failed to show clinical benefit in established, early, or prodromal disease, or in those with high AD risk. Currently, monoclonal antibodies specifically directed against the most neurotoxic Aβ forms are undergoing large-scale trials to confirm initially encouraging results. However, recent findings on the normal physiology of Aβ suggest that accumulation may be compensatory rather than the pathological initiator. If this is true, alternative strategies will be needed to defeat this devastating disease. ANN NEUROL 2019;85:303-315.
Collapse
Affiliation(s)
- Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy.,Neurodegenerative Disease Unit, Department of Clinical Research in Neurology, University of Bari Aldo Moro, Cardinal G. Panico Pious Foundation, Tricase, Italy.,Geriatric Unit, Home Relief of Suffering, Institute of Hospitalization and Scientific Care Foundation, San Giovanni Rotondo, Italy
| | - Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Davide Seripa
- Geriatric Unit, Home Relief of Suffering, Institute of Hospitalization and Scientific Care Foundation, San Giovanni Rotondo, Italy
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Pharmaceuticals, Parma, Italy
| |
Collapse
|
32
|
Zhao B, Liu P, Wei M, Li Y, Liu J, Ma L, Shang S, Jiang Y, Huo K, Wang J, Qu Q. Chronic Sleep Restriction Induces Aβ Accumulation by Disrupting the Balance of Aβ Production and Clearance in Rats. Neurochem Res 2019; 44:859-873. [PMID: 30632087 DOI: 10.1007/s11064-019-02719-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/04/2019] [Indexed: 11/27/2022]
Abstract
Amyloid-β (Aβ) plays an important role in Alzheimer's disease (AD) pathogenesis, and growing evidence has shown that poor sleep quality is one of the risk factors for AD, but the mechanisms of sleep deprivation leading to AD have still not been fully demonstrated. In the present study, we used wild-type (WT) rats to determine the effects of chronic sleep restriction (CSR) on Aβ accumulation. We found that CSR-21d rats had learning and memory functional decline in the Morris water maze (MWM) test. Meanwhile, Aβ42 deposition in the hippocampus and the prefrontal cortex was high after a 21-day sleep restriction. Moreover, compared with the control rats, CSR rats had increased expression of β-site APP-cleaving enzyme 1 (BACE1) and sAPPβ and decreased sAPPα levels in both the hippocampus and the prefrontal cortex, and the BACE1 level was positively correlated with the Aβ42 level. Additionally, in CSR-21d rats, low-density lipoprotein receptor-related protein 1 (LRP-1) levels were low, while receptor of advanced glycation end products (RAGE) levels were high in the hippocampus and the prefrontal cortex, and these transporters were significantly correlated with Aβ42 levels. In addition, CSR-21d rats had decreased plasma Aβ42 levels and soluble LRP1 (sLRP1) levels compared with the control rats. Altogether, this study demonstrated that 21 days of CSR could lead to brain Aβ accumulation in WT rats. The underlying mechanisms may be related to increased Aβ production via upregulation of the BACE1 pathway and disrupted Aβ clearance affecting brain and peripheral Aβ transport.
Collapse
Affiliation(s)
- Beiyu Zhao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
| | - Peng Liu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Meng Wei
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
| | - Yanbo Li
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
| | - Jie Liu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
| | - Louyan Ma
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
| | - Yu Jiang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, 710061, Xi'an, China.
| |
Collapse
|
33
|
Panza F, Lozupone M, Logroscino G, Imbimbo BP. A critical appraisal of amyloid-β-targeting therapies for Alzheimer disease. Nat Rev Neurol 2019; 15:73-88. [DOI: 10.1038/s41582-018-0116-6] [Citation(s) in RCA: 459] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Lipopolysaccharide Induces Subacute Cerebral Microhemorrhages with Involvement of Nitric Oxide Synthase in Rats. J Stroke Cerebrovasc Dis 2018; 27:1905-1913. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.02.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/09/2018] [Accepted: 02/17/2018] [Indexed: 11/22/2022] Open
|
35
|
Shokri-Kojori E, Wang GJ, Wiers CE, Demiral SB, Guo M, Kim SW, Lindgren E, Ramirez V, Zehra A, Freeman C, Miller G, Manza P, Srivastava T, De Santi S, Tomasi D, Benveniste H, Volkow ND. β-Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci U S A 2018; 115:4483-4488. [PMID: 29632177 PMCID: PMC5924922 DOI: 10.1073/pnas.1721694115] [Citation(s) in RCA: 510] [Impact Index Per Article: 72.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The effects of acute sleep deprivation on β-amyloid (Aβ) clearance in the human brain have not been documented. Here we used PET and 18F-florbetaben to measure brain Aβ burden (ABB) in 20 healthy controls tested after a night of rested sleep (baseline) and after a night of sleep deprivation. We show that one night of sleep deprivation, relative to baseline, resulted in a significant increase in Aβ burden in the right hippocampus and thalamus. These increases were associated with mood worsening following sleep deprivation, but were not related to the genetic risk (APOE genotype) for Alzheimer's disease. Additionally, baseline ABB in a range of subcortical regions and the precuneus was inversely associated with reported night sleep hours. APOE genotyping was also linked to subcortical ABB, suggesting that different Alzheimer's disease risk factors might independently affect ABB in nearby brain regions. In summary, our findings show adverse effects of one-night sleep deprivation on brain ABB and expand on prior findings of higher Aβ accumulation with chronic less sleep.
Collapse
Affiliation(s)
- Ehsan Shokri-Kojori
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892;
| | - Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892;
| | - Corinde E Wiers
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Sukru B Demiral
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Min Guo
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Sung Won Kim
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Elsa Lindgren
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Veronica Ramirez
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Amna Zehra
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Clara Freeman
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Gregg Miller
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Peter Manza
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Tansha Srivastava
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | | | - Dardo Tomasi
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510
| | - Nora D Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
36
|
Gong YH, Hua N, Zang X, Huang T, He L. Melatonin ameliorates Aβ1-42-induced Alzheimer's cognitive deficits in mouse model. J Pharm Pharmacol 2017; 70:70-80. [DOI: 10.1111/jphp.12830] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/16/2017] [Indexed: 01/09/2023]
Abstract
Abstract
Objectives
The objective of this study was to evaluate whether melatonin could ameliorate cognitive function in Aβ1-42-induced mouse model and its underlying mechanisms.
Methods
Series behaviour tests were performed to demonstrate the amelioration of cognitive function of the Alzheimer's disease (AD) mice induced by Aβ1-42. Additionally, enzyme-linked immunosorbent assay was applied to detect the expression of Aβ1-42, BACE1 and p-tau protein in the brain of the AD mice. JC-1 was performed to investigate the role in alleviating mitochondrial damage by melatonin in vitro. Western blot was used to detect the expression of melatonin on apoptosis-related factors caspase-3 and Bcl-2, as well as the expressions of GSK-3β and PP2A to further determine the mechanisms of melatonin on the expression of p-tau protein.
Key findings
Melatonin significantly ameliorated the cognitive function and mitochondrial damage in AD mice, reduced the expression levels of GSK-3β, caspase-3, Aβ1-42, BACE1, p-tau protein and increased the expressions of PP2A and Bcl-2.
Conclusion
From the overall results, we concluded that melatonin alleviated the mitochondrial damage effectively and decreased the expressions of the p-tau and some key proteins of apoptosis, leading to the improvement of cognitive function of the mice induced by Aβ1-42.
Collapse
Affiliation(s)
- Yu-Hang Gong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Nan Hua
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Xuan Zang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Tao Huang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
37
|
Li K, Wei Q, Liu FF, Hu F, Xie AJ, Zhu LQ, Liu D. Synaptic Dysfunction in Alzheimer's Disease: Aβ, Tau, and Epigenetic Alterations. Mol Neurobiol 2017; 55:3021-3032. [PMID: 28456942 DOI: 10.1007/s12035-017-0533-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/06/2017] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized in the early stages by loss of learning and memory. However, the mechanism underlying these symptoms remains unclear. The best correlation between cognitive decline and pathological changes is in synaptic dysfunction. Histopathological hallmarks of AD are the abnormal aggregation of Aβ and Tau. Evidence suggests that Aβ and Tau oligomers contribute to synaptic loss in AD. Recently, direct links between epigenetic alterations, such as dysfunction in non-coding RNAs (ncRNAs), and synaptic pathologies have emerged, raising interest in exploring the potential roles of ncRNAs in the synaptic deficits in AD. In this paper, we summarize the potential roles of Aβ, Tau, and epigenetic alterations (especially by ncRNAs) in the synaptic dysfunction of AD and discuss the novel findings in this area.
Collapse
Affiliation(s)
- Ke Li
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Qing Wei
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fang-Fang Liu
- Department of Pathology, Central Hospital of Wuhan, Wuhan, 430014, People's Republic of China
| | - Fan Hu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ao-Ji Xie
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Dan Liu
- Department of Medical Genetics, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|