1
|
Ye M, Ji Q, Liu Q, Kang X, Zhan Y. Longitudinal associations of lipid profiles with sleep disorders in patients with Parkinson's disease. Lipids 2024. [PMID: 39702743 DOI: 10.1002/lipd.12428] [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] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
To examine the associations of apolipoprotein E (APOE) carrier status and lipid profiles with sleep disorders, including excessive daytime sleepiness (EDS) and probable rapid eye movement sleep behavior disorder (pRBD), among patients with early Parkinson's disease (PD) over 5-year follow-up periods. The Parkinson's Progression Markers Initiative is a multicenter cohort study based on an ongoing and open-ended registry. Data from baseline and 5-year follow-up visits from participants of de novo PD were analyzed. Longitudinal associations of APOE carrier status and lipid profiles with sleep disorders were estimated via linear mixed-effects models. A total of 657 participants with complete APOE genotypes were enrolled at baseline. Among them, 153 (25.3%) had available lipid profiles at baseline. In the linear mixed-effects models, baseline APOE ε2/ε3/ε4 carrier status did not exhibit significant associations with EDS and pRBD (all p > 0.05) in all models. However, reduced high-density lipoprotein (HDL) and elevated triglycerides (TG) were associated with developing EDS (β = -0.04, 95% CI: -0.07, -0.00) and pRBD (β = 0.01, 95% CI: 0.00, 0.02) in PD patients, respectively. In the APOE ε4+ subgroup, decreased HDL and increased TG displayed substantial associations with developing EDS and sleep disorders (all p < 0.05) in all models, respectively, whereas no significant differences were noted in the APOE ε4- subgroup (all p > 0.05). Our study did not demonstrate a clear association between APOE ε2/ε3/ε4 and sleep disorders in PD patients. However, the presence of APOE ε4 was associated with changes in lipid profiles, notably affecting TG and HDL levels.
Collapse
Affiliation(s)
- Meijie Ye
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Qianqian Ji
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Qi Liu
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Xiaoying Kang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yiqiang Zhan
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Deng Q, Wu C, Parker E, Zhu J, Liu TCY, Duan R, Yang L. Mystery of gamma wave stimulation in brain disorders. Mol Neurodegener 2024; 19:96. [PMID: 39695746 DOI: 10.1186/s13024-024-00785-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Neuronal oscillations refer to rhythmic and periodic fluctuations of electrical activity in the central nervous system that arise from the cellular properties of diverse neuronal populations and their interactions. Specifically, gamma oscillations play a crucial role in governing the connectivity between distinct brain regions, which are essential in perception, motor control, memory, and emotions. In this context, we recapitulate various current stimulation methods to induce gamma entrainment. These methods include sensory stimulation, optogenetic modulation, photobiomodulation, and transcranial electrical or magnetic stimulation. Simultaneously, we explore the association between abnormal gamma oscillations and central nervous system disorders such as Alzheimer's disease, Parkinson's disease, stroke, schizophrenia, and autism spectrum disorders. Evidence suggests that gamma entrainment-inducing stimulation methods offer notable neuroprotection, although somewhat controversial. This review comprehensively discusses the functional role of gamma oscillations in higher-order brain activities from both physiological and pathological perspectives, emphasizing gamma entrainment as a potential therapeutic approach for neuropsychiatric disorders. Additionally, we discuss future opportunities and challenges in implementing such strategies.
Collapse
Affiliation(s)
- Qianting Deng
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Chongyun Wu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Emily Parker
- Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Jing Zhu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Timon Cheng-Yi Liu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Rui Duan
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Luodan Yang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
3
|
Cankar N, Beschorner N, Tsopanidou A, Qvist FL, Colaço AR, Andersen M, Kjaerby C, Delle C, Lambert M, Mundt F, Weikop P, Jucker M, Mann M, Skotte NH, Nedergaard M. Sleep deprivation leads to non-adaptive alterations in sleep microarchitecture and amyloid-β accumulation in a murine Alzheimer model. Cell Rep 2024; 43:114977. [PMID: 39541211 DOI: 10.1016/j.celrep.2024.114977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Impaired sleep is a common aspect of aging and often precedes the onset of Alzheimer's disease. Here, we compare the effects of sleep deprivation in young wild-type mice and their APP/PS1 littermates, a murine model of Alzheimer's disease. After 7 h of sleep deprivation, both genotypes exhibit an increase in EEG slow-wave activity. However, only the wild-type mice demonstrate an increase in the power of infraslow norepinephrine oscillations, which are characteristic of healthy non-rapid eye movement sleep. Notably, the APP/PS1 mice fail to enhance norepinephrine oscillations 24 h after sleep deprivation, coinciding with an accumulation of cerebral amyloid-β protein. Proteome analysis of cerebrospinal fluid and extracellular fluid further supports these findings by showing altered protein clearance in APP/PS1 mice. We propose that the suppression of infraslow norepinephrine oscillations following sleep deprivation contributes to increased vulnerability to sleep loss and heightens the risk of developing amyloid pathology in early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Neža Cankar
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Natalie Beschorner
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anastasia Tsopanidou
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Filippa L Qvist
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ana R Colaço
- Proteomics Research Infrastructure, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mie Andersen
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Celia Kjaerby
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Christine Delle
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Marius Lambert
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Filip Mundt
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Matthias Mann
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department for Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Niels Henning Skotte
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA.
| |
Collapse
|
4
|
Tsai CY, Su CL, Huang HT, Lin HW, Lin JW, Hei NC, Cheng WH, Chen YL, Majumdar A, Kang JH, Lee KY, Chen Z, Lin YC, Wu CJ, Kuan YC, Lin YT, Hsu CR, Lee HC, Liu WT. Mediating role of obstructive sleep apnea in altering slow-wave activity and elevating Alzheimer's disease risk: Pilot study from a northern Taiwan cohort. Sleep Health 2024:S2352-7218(24)00188-8. [PMID: 39419711 DOI: 10.1016/j.sleh.2024.08.012] [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: 05/24/2024] [Revised: 07/31/2024] [Accepted: 08/31/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES Obstructive sleep apnea is associated with alterations in slow-wave activity during sleep, potentially increasing the risk of Alzheimer's disease. This study investigated the associations between obstructive sleep apnea manifestations such as respiratory events, hypoxia, arousal, slow-wave patterns, and neurochemical biomarker levels. METHODS Individuals with suspected obstructive sleep apnea underwent polysomnography. Sleep disorder indices, oxygen metrics, and slow-wave activity data were obtained from the polysomnography, and blood samples were taken the following morning to determine the plasma levels of total tau (T-Tau) and amyloid beta-peptide 42 (Aβ42) by using an ultrasensitive immunomagnetic reduction assay. Subsequently, the participants were categorized into groups with low and high Alzheimer's disease risk on the basis of their computed product Aβ42 × T-Tau. Intergroup differences and the associations and mediation effects between sleep-related parameters and neurochemical biomarkers were analyzed. RESULTS Forty-two participants were enrolled, with 21 assigned to each of the low- and high-risk groups. High-risk individuals had a higher apnea-hypopnea index, oxygen desaturation index (≥3%, ODI-3%), fraction of total sleep time with oxygen desaturation (SpO2-90% TST), and arousal index and greater peak-to-peak amplitude and slope in slow-wave activity, with a correspondingly shorter duration, than did low-risk individuals. Furthermore, indices such as the apnea-hypopnea index, ODI-3% and SpO2-90% TST were found to indirectly affect slow-wave activity, thereby raising the Aβ42 × T-Tau level. CONCLUSIONS Obstructive sleep apnea manifestations, such as respiratory events and hypoxia, may influence slow-wave sleep activity (functioning as intermediaries) and may be linked to elevated neurochemical biomarker levels. However, a longitudinal study is necessary to determine causal relationships among these factors. STATEMENT OF SIGNIFICANCE This research aims to bridge gaps in understanding how obstructive sleep apnea is associated with an elevated risk of Alzheimer's disease, providing valuable knowledge for sleep and cognitive health.
Collapse
Affiliation(s)
- Cheng-Yu Tsai
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan; Sleep Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Chien-Ling Su
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Research Center of Biomedical Devices, Taipei Medical University, Taipei, Taiwan
| | - Huei-Tyng Huang
- Department of Medical Physics and Bioengineering, University College London, United Kingdom
| | - Hsin-Wei Lin
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jia-Wei Lin
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ng Cheuk Hei
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wun-Hao Cheng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Ling Chen
- Institute of Biomedical Informatics of National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Arnab Majumdar
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Jiunn-Horng Kang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Zhihe Chen
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Yi-Chih Lin
- Sleep Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Otolaryngology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Cheng-Jung Wu
- Department of Otolaryngology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yi-Chun Kuan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yin-Tzu Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chia-Rung Hsu
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Hsin-Chien Lee
- Institute of Medical Humanities, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, Taipei Medical University Hospital, Taipei, Taiwan.
| | - Wen-Te Liu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan; Sleep Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.
| |
Collapse
|
5
|
Yan YJ, Huang CQ. Cognitive impairment induced by circadian rhythm disorders involves hippocampal brain-derived neurotrophic factor reduction and amyloid-β deposition. Chronobiol Int 2024; 41:1299-1306. [PMID: 39311588 DOI: 10.1080/07420528.2024.2406545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/14/2024] [Accepted: 09/13/2024] [Indexed: 10/30/2024]
Abstract
Circadian rhythm disruptions have been implicated in numerous health issues, including cognitive decline and the exacerbation of neurodegenerative diseases, like Alzheimer disease (AD). Brain-derived neurotrophic factor (BDNF), vital for neuronal plasticity and cognitive function, is regulated by the circadian clock and exerts protective effects against AD. Thus, we investigated the impact of circadian rhythm disorders (CRDs) on cognitive impairment and explored the underlying neurobiological mechanisms by assessing BDNF and amyloid-β (Aβ) levels. We divided male C57BL/6 mice into three groups (n = 30): a control group (normal 12/12 hour light-dark cycle) and two CRD model groups (3/3 and 22/22 hour cycles, respectively). After 12 weeks, we assessed cognitive functions using the Morris water maze. Following behavioral tests, hippocampal levels of BDNF and Aβ were quantified using enzyme-linked immunosorbent assays. CRDs significantly impaired learning and memory, as evidenced by longer times to reach and find the platform in the CRD groups (p < 0.01). Furthermore, BDNF levels were notably decreased and Aβ levels increased in the CRD groups compared with the control group (p < 0.01). Thus, CRDs elicit cognitive impairment by reducing BDNF levels and increasing Aβ deposition in the hippocampus.
Collapse
Affiliation(s)
- Yue-Jia Yan
- Department of Geriatrics, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
| | - Chang-Quan Huang
- Department of Geriatrics, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, China
- Department of General Medicine, Chengdu Second People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Han X, Liu G, Lee SS, Yang X, Wu MN, Lu H, Wei Z. Metabolic and vascular imaging markers for investigating Alzheimer's disease complicated by sleep fragmentation in mice. Front Physiol 2024; 15:1456690. [PMID: 39371598 PMCID: PMC11449888 DOI: 10.3389/fphys.2024.1456690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 09/11/2024] [Indexed: 10/08/2024] Open
Abstract
Background Sleep problem is a common complication of Alzheimer's disease (AD). Extensive preclinical studies have been performed to investigate the AD pathology. However, the pathophysiological consequence of AD complicated by sleep problem remains to be further determined. Purpose To investigate brain metabolism and perfusion in an AD mouse model complicated by sleep problem, and subsequently identify potential imaging markers to better understand the associated pathophysiology. Methods We examined the oxygen extraction fraction (OEF), cerebral metabolic rate of oxygen (CMRO2), and cerebral blood flow (CBF) using state-of-the-art MRI techniques in a cohort of 5xFAD model mice. Additionally, neuroinflammation, indicated by activated microglia, was assessed using histology techniques. Sleep fragmentation (SF) was utilized as a representative for sleep problems. Results SF was associated with significant increases in OEF (P = 0.023) and CMRO2 (P = 0.029), indicating a state of hypermetabolism. CBF showed a significant genotype-by-sleep interaction effect (P = 0.026), particularly in the deep brain regions such as the hippocampus and thalamus. Neuroinflammation was primarily driven by genotype rather than SF, especially in regions with significant interaction effect in CBF measurements. Conclusion These results suggest that brain metabolism and perfusion measurements are promising markers for studying the co-pathogenesis of AD and SF.
Collapse
Affiliation(s)
- Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Guanshu Liu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, United States
| | - Sang Soo Lee
- Department of Neurology, Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiuli Yang
- Department of Neurology, Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark N. Wu
- Department of Neurology, Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hanzhang Lu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zhiliang Wei
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, United States
| |
Collapse
|
7
|
Rowe RK, Schulz P, He P, Mannino GS, Opp MR, Sierks MR. Acute sleep deprivation in mice generates protein pathology consistent with neurodegenerative diseases. Front Neurosci 2024; 18:1436966. [PMID: 39114483 PMCID: PMC11303328 DOI: 10.3389/fnins.2024.1436966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Insufficient or disturbed sleep is strongly associated with adverse health conditions, including various neurodegenerative disorders. While the relationship between sleep and neurodegenerative disease is likely bidirectional, sleep disturbances often predate the onset of other hallmark clinical symptoms. Neuronal waste clearance is significantly more efficient during sleep; thus, disturbed sleep may lead to the accumulation of neuronal proteins that underlie neurodegenerative diseases. Key pathological features of neurodegenerative diseases include an accumulation of misfolded or misprocessed variants of amyloid beta (Aβ), tau, alpha synuclein (α-syn), and TarDNA binding protein 43 (TDP-43). While the presence of fibrillar protein aggregates of these neuronal proteins are characteristic of neurodegenerative diseases, the presence of small soluble toxic oligomeric variants of these different proteins likely precedes the formation of the hallmark aggregates. Methods We hypothesized that sleep deprivation would lead to accumulation of toxic oligomeric variants of Aβ, tau, α-syn, and TDP-43 in brain tissue of wild-type mice. Adult mice were subjected to 6 h of sleep deprivation (zeitgeber 0-6) for 5 consecutive days or were left undisturbed as controls. Following sleep deprivation, brains were collected, and protein pathology was assessed in multiple brain regions using an immunostain panel of reagents selectively targeting neurodegenerative disease-related variants of Aβ, tau, α-syn, and TDP-43. Results Overall, sleep deprivation elevated levels of all protein variants in at least one of the brain regions of interest. The reagent PDTDP, targeting a TDP-43 variant present in Parkinson's disease, was elevated throughout the brain. The cortex, caudoputamen, and corpus callosum brain regions showed the highest accumulation of pathology following sleep deprivation. Discussion These data provide a direct mechanistic link between sleep deprivation, and the hallmark protein pathologies of neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Rachel K. Rowe
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Philip Schulz
- Chemical Engineering, The School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States
| | - Ping He
- Chemical Engineering, The School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States
| | - Grant S. Mannino
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Mark R. Opp
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Michael R. Sierks
- Chemical Engineering, The School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
8
|
Bedward A, Kaur J, Seedat S, Donohue H, Kow CS, Rasheed MK, Javed A, Hasan SS. Pharmacological interventions to improve sleep in people with Alzheimer's disease: a meta-analysis of randomized controlled trials. Expert Rev Neurother 2024; 24:527-539. [PMID: 38597219 DOI: 10.1080/14737175.2024.2341004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
INTRODUCTION This systematic review and meta-analysis evaluates the evidence from randomized controlled trials (RCTs) involving pharmacological interventions for improving sleep in people with Alzheimer's disease (AD). METHODS A systematic literature search in eight databases from January 2000 to July 2023 focusing on RCTs that compared a pharmacological intervention with a placebo for enhancing sleep in people with AD. The authors registered the study protocol at Prospero, followed the PRISMA guidelines, and produced the pooled estimates using random-effect or IVhet models. RESULTS Eight different interventions and 29 different sleep outcomes were examined in 14 RCTs included in this review. Eszopiclone positively affected sleep efficiency, as did orexin antagonists. However, there was no difference when melatonin was used. The interventions demonstrated low discontinuation rates and a few adverse drug reactions. CONCLUSION Although melatonin was the most investigated intervention, the evidence for its efficacy is inconclusive. On the other hand, trazodone and orexin receptor antagonists showed promising results; however, more RCTs are needed for definite answers.
Collapse
Affiliation(s)
- Amy Bedward
- School of Applied Sciences, University of Huddersfield, Huddersfield, West Yorkshire, UK
| | - Jasmine Kaur
- School of Applied Sciences, University of Huddersfield, Huddersfield, West Yorkshire, UK
| | - Sadiyah Seedat
- School of Applied Sciences, University of Huddersfield, Huddersfield, West Yorkshire, UK
| | - Holly Donohue
- School of Applied Sciences, University of Huddersfield, Huddersfield, West Yorkshire, UK
| | - Chia Siang Kow
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Muhammad Kamran Rasheed
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Amaan Javed
- University College of Medical Sciences, University of Delhi, New Delhi, India
| | - Syed Shahzad Hasan
- School of Applied Sciences, University of Huddersfield, Huddersfield, West Yorkshire, UK
| |
Collapse
|
9
|
Zhang M, Chen L, Ren Z, Wang Z, Luo W. Applications of TMS in individuals with methamphetamine use disorder: A review. Heliyon 2024; 10:e25565. [PMID: 38420394 PMCID: PMC10900420 DOI: 10.1016/j.heliyon.2024.e25565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/25/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Methamphetamine abuse results in a host of social and medical issues. Methamphetamine use disorder (MUD) can hinder the brain and impair cognitive functions and mental health. Transcranial magnetic stimulation (TMS) is a non-invasive approach in the treatment of MUD. Recent studies have demonstrated encouraging and positive effects of TMS on the craving, affective symptoms, sleep quality, and cognitive functions in individuals with MUD. The regulation of specific brain activities through TMS has also been found to be a contributing factor to these positive outcomes. It is essential to employ more techniques, participants, and stimulation parameters and targets in the future.
Collapse
Affiliation(s)
- Mingming Zhang
- Research Center of Brain and Cognitive Neuroscience, Liaoning Normal University, Dalian, 116029, China
- Key Laboratory of Brain and Cognitive Neuroscience, Liaoning Province, Dalian, 116029, China
| | - Lei Chen
- Research Center of Brain and Cognitive Neuroscience, Liaoning Normal University, Dalian, 116029, China
- Key Laboratory of Brain and Cognitive Neuroscience, Liaoning Province, Dalian, 116029, China
| | - Ziwei Ren
- Research Center of Brain and Cognitive Neuroscience, Liaoning Normal University, Dalian, 116029, China
- Key Laboratory of Brain and Cognitive Neuroscience, Liaoning Province, Dalian, 116029, China
| | - Zhiyan Wang
- Research Center of Brain and Cognitive Neuroscience, Liaoning Normal University, Dalian, 116029, China
- Key Laboratory of Brain and Cognitive Neuroscience, Liaoning Province, Dalian, 116029, China
| | - Wenbo Luo
- Research Center of Brain and Cognitive Neuroscience, Liaoning Normal University, Dalian, 116029, China
- Key Laboratory of Brain and Cognitive Neuroscience, Liaoning Province, Dalian, 116029, China
| |
Collapse
|
10
|
Kron JOZJ, Keenan RJ, Hoyer D, Jacobson LH. Orexin Receptor Antagonism: Normalizing Sleep Architecture in Old Age and Disease. Annu Rev Pharmacol Toxicol 2024; 64:359-386. [PMID: 37708433 DOI: 10.1146/annurev-pharmtox-040323-031929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Sleep is essential for human well-being, yet the quality and quantity of sleep reduce as age advances. Older persons (>65 years old) are more at risk of disorders accompanied and/or exacerbated by poor sleep. Furthermore, evidence supports a bidirectional relationship between disrupted sleep and Alzheimer's disease (AD) or related dementias. Orexin/hypocretin neuropeptides stabilize wakefulness, and several orexin receptor antagonists (ORAs) are approved for the treatment of insomnia in adults. Dysregulation of the orexin system occurs in aging and AD, positioning ORAs as advantageous for these populations. Indeed, several clinical studies indicate that ORAs are efficacious hypnotics in older persons and dementia patients and, as in adults, are generally well tolerated. ORAs are likely to be more effective when administered early in sleep/wake dysregulation to reestablish good sleep/wake-related behaviors and reduce the accumulation of dementia-associated proteinopathic substrates. Improving sleep in aging and dementia represents a tremendous opportunity to benefit patients, caregivers, and health systems.
Collapse
Affiliation(s)
- Jarrah O-Z J Kron
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
| | - Ryan J Keenan
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Daniel Hoyer
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia;
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Laura H Jacobson
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia;
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia;
| |
Collapse
|
11
|
Gao J, Cao J, Chen J, Wu D, Luo K, Shen G, Fang Y, Zhang W, Huang G, Su X, Zhao L. Brain morphology and functional connectivity alterations in patients with severe obstructive sleep apnea. Sleep Med 2023; 111:62-69. [PMID: 37722341 DOI: 10.1016/j.sleep.2023.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/09/2023] [Accepted: 08/29/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND It has been demonstrated that widespread structural and functional brain alterations influence the development of cognitive impairment in patients with obstructive sleep apnea (OSA). However, the literature has limited evidence regarding the neuropathophysiological mechanisms behind these impairments. This research aimed to investigate brain morphologic and functional connectivity (FC) abnormalities related to neurocognitive function in OSA. METHODS Fifty treatment-naïve males, newly diagnosed patients with severe OSA, and 50 well-matched healthy controls (HCs) were enrolled prospectively. All subjects underwent an MRI scan, cognitive psychological and sleep scale assessment. The differences of brain morphological and seed-based FC between the two groups were compared. The correlation analysis and receiver operating characteristic curve were performed for further analysis. RESULTS Compared with HCs, the right brainstem, left dorsal-lateral superior frontal gyrus (SFGdor), and superior temporal gyrus (STG) exhibited atrophy in the OSA group. In addition, FC between the left SFGdor and the right postcentral gyrus (PoCG) was increased, which was positively correlated with disease duration (r = 0.312, FDR-corrected P = 0.027). The Jacobian values of the brainstem were negatively correlated with MoCA and recall scores (r = -0.449, FDR-corrected P = 0.0025; r = -0.416, FDR-corrected P = 0.005). Furthermore, the Jacobian values of the left SFGdor demonstrated a relatively high diagnostic performance (sensitivity: 86%, specificity: 56%, AUC: 0.740, 95% CI: 0.643-0.836, P < 0.0001). CONCLUSIONS Structural atrophy in brainstem and frontotemporal lobe and altered FC may be the neurobiological hallmark of brain impairment in OSA. Notably, brainstem atrophy has been associated with cognitive impairment, which may provide new insights into understanding the neuropathophysiological mechanisms of cognitive impairment in OSA patients.
Collapse
Affiliation(s)
- Jing Gao
- The First Clinical Medical College of Gansu University of Chinese Medicine(Gansu Provincial Hospital), Lanzhou, 730000, China
| | - Jiancang Cao
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Jieyu Chen
- The First Clinical Medical College of Gansu University of Chinese Medicine(Gansu Provincial Hospital), Lanzhou, 730000, China
| | - Dan Wu
- The First Clinical Medical College of Gansu University of Chinese Medicine(Gansu Provincial Hospital), Lanzhou, 730000, China
| | - Ke Luo
- The First Clinical Medical College of Gansu University of Chinese Medicine(Gansu Provincial Hospital), Lanzhou, 730000, China
| | - Guo Shen
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Yanyan Fang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Wenwen Zhang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Gang Huang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Xiaoyan Su
- Sleep Medicine Center, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Lianping Zhao
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, China.
| |
Collapse
|
12
|
Gędek A, Koziorowski D, Szlufik S. Assessment of factors influencing glymphatic activity and implications for clinical medicine. Front Neurol 2023; 14:1232304. [PMID: 37767530 PMCID: PMC10520725 DOI: 10.3389/fneur.2023.1232304] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
The glymphatic system is a highly specialized fluid transport system in the central nervous system. It enables the exchange of the intercellular fluid of the brain, regulation of the movement of this fluid, clearance of unnecessary metabolic products, and, potentially, brain immunity. In this review, based on the latest scientific reports, we present the mechanism of action and function of the glymphatic system and look at the role of factors influencing its activity. Sleep habits, eating patterns, coexisting stress or hypertension, and physical activity can significantly affect glymphatic activity. Modifying them can help to change lives for the better. In the next section of the review, we discuss the connection between the glymphatic system and neurological disorders. Its association with many disease entities suggests that it plays a major role in the physiology of the whole brain, linking many pathophysiological pathways of individual diseases.
Collapse
Affiliation(s)
- Adam Gędek
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- Praski Hospital, Warsaw, Poland
| | - Dariusz Koziorowski
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Stanisław Szlufik
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
13
|
Harper L, McAnelly S, Walshe I, Ooms A, Tuffrey-Wijne IM. Behavioural sleep problems in children and adults with intellectual disabilities: An integrative literature review. JOURNAL OF APPLIED RESEARCH IN INTELLECTUAL DISABILITIES 2023; 36:916-928. [PMID: 37177858 DOI: 10.1111/jar.13116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND People with intellectual disabilities are more likely to experience sleep problems, which can affect quality of life, physical health, mental health and well-being. METHODS An integrative literature review was conducted to investigate what is known about behavioural sleep disturbances in people with an intellectual disability. The search used the following databases: Scopus, PsycInfo and Cinahl, to find papers published since 2015. RESULTS Within intellectual disability research, sleep appears as a common issue due to its high prevalence, negative relationships with an individual's physical and mental health, their quality of life, and impact of sleep problems on family or carers. The growing evidence base appears to support the use of behavioural, lifestyle and pharmacological interventions to improve sleep in people with an intellectual disability. CONCLUSION A wide array of literature provides evidence that people with intellectual disabilities are affected by and need support with their sleep.
Collapse
Affiliation(s)
- Lynette Harper
- Health and Life Sciences, Northumbria University, London, UK
| | - Su McAnelly
- Health and Life Sciences, Northumbria University, London, UK
| | - Ian Walshe
- Health and Life Sciences, Northumbria University, London, UK
| | - Ann Ooms
- Faculty of Health, Science, Social Care and Education, Kingston University, London, UK
| | - Irene M Tuffrey-Wijne
- Faculty of Health, Science, Social Care and Education, Kingston University, London, UK
| |
Collapse
|
14
|
Lyons LC, Vanrobaeys Y, Abel T. Sleep and memory: The impact of sleep deprivation on transcription, translational control, and protein synthesis in the brain. J Neurochem 2023; 166:24-46. [PMID: 36802068 PMCID: PMC10919414 DOI: 10.1111/jnc.15787] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/20/2023]
Abstract
In countries around the world, sleep deprivation represents a widespread problem affecting school-age children, teenagers, and adults. Acute sleep deprivation and more chronic sleep restriction adversely affect individual health, impairing memory and cognitive performance as well as increasing the risk and progression of numerous diseases. In mammals, the hippocampus and hippocampus-dependent memory are vulnerable to the effects of acute sleep deprivation. Sleep deprivation induces changes in molecular signaling, gene expression and may cause changes in dendritic structure in neurons. Genome wide studies have shown that acute sleep deprivation alters gene transcription, although the pool of genes affected varies between brain regions. More recently, advances in research have drawn attention to differences in gene regulation between the level of the transcriptome compared with the pool of mRNA associated with ribosomes for protein translation following sleep deprivation. Thus, in addition to transcriptional changes, sleep deprivation also affects downstream processes to alter protein translation. In this review, we focus on the multiple levels through which acute sleep deprivation impacts gene regulation, highlighting potential post-transcriptional and translational processes that may be affected by sleep deprivation. Understanding the multiple levels of gene regulation impacted by sleep deprivation is essential for future development of therapeutics that may mitigate the effects of sleep loss.
Collapse
Affiliation(s)
- Lisa C Lyons
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Yann Vanrobaeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Iowa City, Iowa, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Iowa City, Iowa, USA
| |
Collapse
|
15
|
Manippa V, Palmisano A, Filardi M, Vilella D, Nitsche MA, Rivolta D, Logroscino G. An update on the use of gamma (multi)sensory stimulation for Alzheimer's disease treatment. Front Aging Neurosci 2022; 14:1095081. [PMID: 36589536 PMCID: PMC9797689 DOI: 10.3389/fnagi.2022.1095081] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by reduced fast brain oscillations in the gamma band (γ, > 30 Hz). Several animal studies show that inducing gamma oscillations through (multi)sensory stimulation at 40 Hz has the potential to impact AD-related cognitive decline and neuropathological processes, including amyloid plaques deposition, neurofibrillary tangles formation, and neuronal and synaptic loss. Therefore Gamma Entrainment Using Sensory stimulation (GENUS) is among the most promising approaches for AD patients' treatment. This review summarizes the evidence on GENUS effectiveness, from animal models to AD patients. Despite the application on human is in its infancy, the available findings suggest its feasibility for the treatment of AD. We discuss such results in light of parameter improvement and possible underlying mechanisms. We finally emphasize the need for further research for its development as a disease-modifying non-pharmacological intervention.
Collapse
Affiliation(s)
- Valerio Manippa
- Department of Education, Psychology and Communication, University of Bari Aldo Moro, Bari, Italy
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Annalisa Palmisano
- Department of Education, Psychology and Communication, University of Bari Aldo Moro, Bari, Italy
| | - Marco Filardi
- Department of Basic Medicine, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione “Card. G. Panico”, Tricase, Italy
| | - Davide Vilella
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione “Card. G. Panico”, Tricase, Italy
| | - Michael A. Nitsche
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- Bielefeld University, University Hospital OWL, Protestant Hospital of Bethel Foundation, University Clinic of Psychiatry and Psychotherapy and University Clinic of Child and Adolescent Psychiatry and Psychotherapy, Bielefeld, Germany
| | - Davide Rivolta
- Department of Education, Psychology and Communication, University of Bari Aldo Moro, Bari, Italy
| | - Giancarlo Logroscino
- Department of Basic Medicine, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro at Pia Fondazione “Card. G. Panico”, Tricase, Italy
| |
Collapse
|
16
|
Tsai CY, Wu SM, Kuan YC, Lin YT, Hsu CR, Hsu WH, Liu YS, Majumdar A, Stettler M, Yang CM, Lee KY, Wu D, Lee HC, Wu CJ, Kang JH, Liu WT. Associations between risk of Alzheimer's disease and obstructive sleep apnea, intermittent hypoxia, and arousal responses: A pilot study. Front Neurol 2022; 13:1038735. [PMID: 36530623 PMCID: PMC9747943 DOI: 10.3389/fneur.2022.1038735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/04/2022] [Indexed: 01/30/2024] Open
Abstract
OBJECTIVES Obstructive sleep apnea (OSA) may increase the risk of Alzheimer's disease (AD). However, potential associations among sleep-disordered breathing, hypoxia, and OSA-induced arousal responses should be investigated. This study determined differences in sleep parameters and investigated the relationship between such parameters and the risk of AD. METHODS Patients with suspected OSA were recruited and underwent in-lab polysomnography (PSG). Subsequently, blood samples were collected from participants. Patients' plasma levels of total tau (T-Tau) and amyloid beta-peptide 42 (Aβ42) were measured using an ultrasensitive immunomagnetic reduction assay. Next, the participants were categorized into low- and high-risk groups on the basis of the computed product (Aβ42 × T-Tau, the cutoff for AD risk). PSG parameters were analyzed and compared. RESULTS We included 36 patients in this study, of whom 18 and 18 were assigned to the low- and high-risk groups, respectively. The average apnea-hypopnea index (AHI), apnea, hypopnea index [during rapid eye movement (REM) and non-REM (NREM) sleep], and oxygen desaturation index (≥3%, ODI-3%) values of the high-risk group were significantly higher than those of the low-risk group. Similarly, the mean arousal index and respiratory arousal index (R-ArI) of the high-risk group were significantly higher than those of the low-risk group. Sleep-disordered breathing indices, oxygen desaturation, and arousal responses were significantly associated with an increased risk of AD. Positive associations were observed among the AHI, ODI-3%, R-ArI, and computed product. CONCLUSIONS Recurrent sleep-disordered breathing, intermittent hypoxia, and arousal responses, including those occurring during the NREM stage, were associated with AD risk. However, a longitudinal study should be conducted to investigate the causal relationships among these factors.
Collapse
Affiliation(s)
- Cheng-Yu Tsai
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Chun Kuan
- Sleep Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
- Dementia Center, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yin-Tzu Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chia-Rung Hsu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Wen-Hua Hsu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Shin Liu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Arnab Majumdar
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Marc Stettler
- Department of Civil and Environmental Engineering, Imperial College London, London, United Kingdom
| | - Chien-Ming Yang
- Department of Psychology, National Chengchi University, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Dean Wu
- Sleep Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
- Dementia Center, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Chien Lee
- Department of Psychiatry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Cheng-Jung Wu
- Department of Otolaryngology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Jiunn-Horng Kang
- Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taipei, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Wen-Te Liu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Sleep Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Research Center of Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
17
|
Shah D, Gsell W, Wahis J, Luckett ES, Jamoulle T, Vermaercke B, Preman P, Moechars D, Hendrickx V, Jaspers T, Craessaerts K, Horré K, Wolfs L, Fiers M, Holt M, Thal DR, Callaerts-Vegh Z, D'Hooge R, Vandenberghe R, Himmelreich U, Bonin V, De Strooper B. Astrocyte calcium dysfunction causes early network hyperactivity in Alzheimer's disease. Cell Rep 2022; 40:111280. [PMID: 36001964 PMCID: PMC9433881 DOI: 10.1016/j.celrep.2022.111280] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/30/2022] [Accepted: 08/05/2022] [Indexed: 12/15/2022] Open
Abstract
Dysfunctions of network activity and functional connectivity (FC) represent early events in Alzheimer’s disease (AD), but the underlying mechanisms remain unclear. Astrocytes regulate local neuronal activity in the healthy brain, but their involvement in early network hyperactivity in AD is unknown. We show increased FC in the human cingulate cortex several years before amyloid deposition. We find the same early cingulate FC disruption and neuronal hyperactivity in AppNL-F mice. Crucially, these network disruptions are accompanied by decreased astrocyte calcium signaling. Recovery of astrocytic calcium activity normalizes neuronal hyperactivity and FC, as well as seizure susceptibility and day/night behavioral disruptions. In conclusion, we show that astrocytes mediate initial features of AD and drive clinically relevant phenotypes. The cingulate cortex of humans and mice shows early functional deficits in AD Astrocyte calcium signaling is decreased before the presence of amyloid plaques Recovery of astrocyte calcium signals mitigates neuronal hyperactivity Recovery of astrocytes normalizes cingulate connectivity and behavior disruptions
Collapse
Affiliation(s)
- Disha Shah
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium.
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Jérôme Wahis
- Laboratory of Glia Biology, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Emma S Luckett
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Tarik Jamoulle
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Ben Vermaercke
- Neuro-electronics Research Flanders, 3000 Leuven, Belgium
| | - Pranav Preman
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Daan Moechars
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Véronique Hendrickx
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Tom Jaspers
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Katleen Craessaerts
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Katrien Horré
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Leen Wolfs
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Mark Fiers
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Matthew Holt
- Laboratory of Glia Biology, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, LBI, KU Leuven, 3000 Leuven, Belgium
| | | | - Rudi D'Hooge
- Laboratory of Biological Psychology, KU-Leuven, 3000 Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, 3000 Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Vincent Bonin
- Neuro-electronics Research Flanders, 3000 Leuven, Belgium
| | - Bart De Strooper
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, KU Leuven, 3000 Leuven, Belgium; UK Dementia Research Institute at University College London, WC1E 6BT London, UK.
| |
Collapse
|
18
|
Rowe RK, Green TRF, Giordano KR, Ortiz JB, Murphy SM, Opp MR. Microglia Are Necessary to Regulate Sleep after an Immune Challenge. BIOLOGY 2022; 11:1241. [PMID: 36009868 PMCID: PMC9405260 DOI: 10.3390/biology11081241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/29/2022] [Accepted: 08/17/2022] [Indexed: 12/31/2022]
Abstract
Microglia play a critical role in the neuroimmune response, but little is known about the role of microglia in sleep following an inflammatory trigger. Nevertheless, decades of research have been predicated on the assumption that an inflammatory trigger increases sleep through microglial activation. We hypothesized that mice (n = 30) with depleted microglia using PLX5622 (PLX) would sleep less following the administration of lipopolysaccharide (LPS) to induce inflammation. Brains were collected and microglial morphology was assessed using quantitative skeletal analyses and physiological parameters were recorded using non-invasive piezoelectric cages. Mice fed PLX diet had a transient increase in sleep that dissipated by week 2. Subsequently, following a first LPS injection (0.4 mg/kg), mice with depleted microglia slept more than mice on the control diet. All mice were returned to normal rodent chow to repopulate microglia in the PLX group (10 days). Nominal differences in sleep existed during the microglia repopulation period. However, following a second LPS injection, mice with repopulated microglia slept similarly to control mice during the dark period but with longer bouts during the light period. Comparing sleep after the first LPS injection to sleep after the second LPS injection, controls exhibited temporal changes in sleep patterns but no change in cumulative minutes slept, whereas cumulative sleep in mice with repopulated microglia decreased during the dark period across all days. Repopulated microglia had a reactive morphology. We conclude that microglia are necessary to regulate sleep after an immune challenge.
Collapse
Affiliation(s)
- Rachel K. Rowe
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80301, USA
- Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Tabitha R. F. Green
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Katherine R. Giordano
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - J. Bryce Ortiz
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - Sean M. Murphy
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| | - Mark R. Opp
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80301, USA
| |
Collapse
|
19
|
Needham H, Torpey G, Flores CC, Davis CJ, Vanderheyden WM, Gerstner JR. A Dichotomous Role for FABP7 in Sleep and Alzheimer's Disease Pathogenesis: A Hypothesis. Front Neurosci 2022; 16:798994. [PMID: 35844236 PMCID: PMC9280343 DOI: 10.3389/fnins.2022.798994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/10/2022] [Indexed: 11/15/2022] Open
Abstract
Fatty acid binding proteins (FABPs) are a family of intracellular lipid chaperone proteins known to play critical roles in the regulation of fatty acid uptake and transport as well as gene expression. Brain-type fatty acid binding protein (FABP7) is enriched in astrocytes and has been implicated in sleep/wake regulation and neurodegenerative diseases; however, the precise mechanisms underlying the role of FABP7 in these biological processes remain unclear. FABP7 binds to both arachidonic acid (AA) and docosahexaenoic acid (DHA), resulting in discrete physiological responses. Here, we propose a dichotomous role for FABP7 in which ligand type determines the subcellular translocation of fatty acids, either promoting wakefulness aligned with Alzheimer's pathogenesis or promoting sleep with concomitant activation of anti-inflammatory pathways and neuroprotection. We hypothesize that FABP7-mediated translocation of AA to the endoplasmic reticulum of astrocytes increases astrogliosis, impedes glutamatergic uptake, and enhances wakefulness and inflammatory pathways via COX-2 dependent generation of pro-inflammatory prostaglandins. Conversely, we propose that FABP7-mediated translocation of DHA to the nucleus stabilizes astrocyte-neuron lactate shuttle dynamics, preserves glutamatergic uptake, and promotes sleep by activating anti-inflammatory pathways through the peroxisome proliferator-activated receptor-γ transcriptional cascade. Importantly, this model generates several testable hypotheses applicable to other neurodegenerative diseases, including amyotrophic lateral sclerosis and Parkinson's disease.
Collapse
Affiliation(s)
- Hope Needham
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Grace Torpey
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - William M. Vanderheyden
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
20
|
Xiao SY, Liu YJ, Lu W, Sha ZW, Xu C, Yu ZH, Lee SD. Possible Neuropathology of Sleep Disturbance Linking to Alzheimer's Disease: Astrocytic and Microglial Roles. Front Cell Neurosci 2022; 16:875138. [PMID: 35755779 PMCID: PMC9218054 DOI: 10.3389/fncel.2022.875138] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022] Open
Abstract
Sleep disturbances not only deteriorate Alzheimer’s disease (AD) progress by affecting cognitive states but also accelerate the neuropathological changes of AD. Astrocytes and microglia are the principal players in the regulation of both sleep and AD. We proposed that possible astrocyte-mediated and microglia-mediated neuropathological changes of sleep disturbances linked to AD, such as astrocytic adenosinergic A1, A2, and A3 regulation; astrocytic dopamine and serotonin; astrocyte-mediated proinflammatory status (TNFα); sleep disturbance-attenuated microglial CX3CR1 and P2Y12; microglial Iba-1 and astrocytic glial fibrillary acidic protein (GFAP); and microglia-mediated proinflammatory status (IL-1b, IL-6, IL-10, and TNFα). Furthermore, astrocytic and microglial amyloid beta (Aβ) and tau in AD were reviewed, such as astrocytic Aβ interaction in AD; astrocyte-mediated proinflammation in AD; astrocytic interaction with Aβ in the central nervous system (CNS); astrocytic apolipoprotein E (ApoE)-induced Aβ clearance in AD, as well as microglial Aβ clearance and aggregation in AD; proinflammation-induced microglial Aβ aggregation in AD; microglial-accumulated tau in AD; and microglial ApoE and TREM2 in AD. We reviewed astrocytic and microglial roles in AD and sleep, such as astrocyte/microglial-mediated proinflammation in AD and sleep; astrocytic ApoE in sleep and AD; and accumulated Aβ-triggered synaptic abnormalities in sleep disturbance. This review will provide a possible astrocytic and microglial mechanism of sleep disturbance linked to AD.
Collapse
Affiliation(s)
- Shu-Yun Xiao
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi-Jie Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Lu
- Department of Traditional Treatment, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhong-Wei Sha
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Che Xu
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhi-Hua Yu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shin-Da Lee
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan.,Department of Physical Therapy, Asia University, Taichung, Taiwan
| |
Collapse
|
21
|
Sunkaria A, Bhardwaj S. Sleep Disturbance and Alzheimer's Disease: The Glial Connection. Neurochem Res 2022; 47:1799-1815. [PMID: 35303225 DOI: 10.1007/s11064-022-03578-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 12/28/2022]
Abstract
Poor quality and quantity of sleep are very common in elderly people throughout the world. Growing evidence has suggested that sleep disturbances could accelerate the process of neurodegeneration. Recent reports have shown a positive correlation between sleep deprivation and amyloid-β (Aβ)/tau aggregation in the brain of Alzheimer's patients. Glial cells have long been implicated in the progression of Alzheimer's disease (AD) and recent findings have also suggested their role in regulating sleep homeostasis. However, how glial cells control the sleep-wake balance and exactly how disturbed sleep may act as a trigger for Alzheimer's or other neurological disorders have recently gotten attention. In an attempt to connect the dots, the present review has highlighted the role of glia-derived sleep regulatory molecules in AD pathogenesis. Role of glia in sleep disturbance and Alzheimer's progression.
Collapse
Affiliation(s)
- Aditya Sunkaria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| | - Supriya Bhardwaj
- Department of Dermatology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| |
Collapse
|
22
|
Wirianto M, Wang CY, Kim E, Koike N, Gomez-Gutierrez R, Nohara K, Escobedo G, Choi JM, Han C, Yagita K, Jung SY, Soto C, Lee HK, Morales R, Yoo SH, Chen Z. The clock modulator Nobiletin mitigates astrogliosis-associated neuroinflammation and disease hallmarks in an Alzheimer's disease model. FASEB J 2022; 36:e22186. [PMID: 35120261 PMCID: PMC8887996 DOI: 10.1096/fj.202101633r] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/30/2021] [Accepted: 01/18/2022] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder, and there is a pressing need to identify disease‐modifying factors and devise interventional strategies. The circadian clock, our intrinsic biological timer, orchestrates various cellular and physiological processes including gene expression, sleep, and neuroinflammation; conversely, circadian dysfunctions are closely associated with and/or contribute to AD hallmarks. We previously reported that the natural compound Nobiletin (NOB) is a clock‐enhancing modulator that promotes physiological health and healthy aging. In the current study, we treated the double transgenic AD model mice, APP/PS1, with NOB‐containing diets. NOB significantly alleviated β‐amyloid burden in both the hippocampus and the cortex, and exhibited a trend to improve cognitive function in these mice. While several systemic parameters for circadian wheel‐running activity, sleep, and metabolism were unchanged, NOB treatment showed a marked effect on the expression of clock and clock‐controlled AD gene expression in the cortex. In accordance, cortical proteomic profiling demonstrated circadian time‐dependent restoration of the protein landscape in APP/PS1 mice treated with NOB. More importantly, we found a potent efficacy of NOB to inhibit proinflammatory cytokine gene expression and inflammasome formation in the cortex, and immunostaining further revealed a specific effect to diminish astrogliosis, but not microgliosis, by NOB in APP/PS1 mice. Together, these results underscore beneficial effects of a clock modulator to mitigate pathological and cognitive hallmarks of AD, and suggest a possible mechanism via suppressing astrogliosis‐associated neuroinflammation.
Collapse
Affiliation(s)
- Marvin Wirianto
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Chih-Yen Wang
- Department of Pediatrics, Baylor College of Medicine, Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Nobuya Koike
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ruben Gomez-Gutierrez
- Department of Neurology, The University of Texas Health Science Center (UTHealth), Houston, Texas, USA.,Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain
| | - Kazunari Nohara
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Gabriel Escobedo
- Department of Neurology, The University of Texas Health Science Center (UTHealth), Houston, Texas, USA
| | - Jong Min Choi
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Chorong Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sung Yun Jung
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Claudio Soto
- Department of Neurology, The University of Texas Health Science Center (UTHealth), Houston, Texas, USA
| | - Hyun Kyoung Lee
- Department of Pediatrics, Baylor College of Medicine, Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center (UTHealth), Houston, Texas, USA.,Centro Integrativo de Biologia Y Quimica Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston (UTHealth), Houston, Texas, USA
| |
Collapse
|
23
|
Orexin A peptidergic system: comparative sleep behavior, morphology and population in brains between wild type and Alzheimer’s disease mice. Brain Struct Funct 2022; 227:1051-1065. [PMID: 35066609 PMCID: PMC8930968 DOI: 10.1007/s00429-021-02447-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 12/16/2021] [Indexed: 11/13/2022]
Abstract
Sleep disturbance is common in patients with Alzheimer’s disease (AD), and orexin A is a pivotal neurotransmitter for bidirectionally regulating the amyloid-β (Aβ) deposition of AD brain and poor sleep. In the present study, we examined the characteristic of sleep–wake architecture in APPswe/PSldE9 (APP/PS1) and Aβ-treated mice using electroencephalogram (EEG) and electromyographic (EMG) analysis. We compared the expression of orexin A, distribution, and morphology of the corresponding orexin A-positive neurons using innovative methods including three-dimensional reconstruction and brain tissue clearing between wild type (WT) and APP/PS1 mice. Results from our study demonstrated that increased wakefulness and reduced NREM sleep were seen in APP/PS1 and Aβ treated mice, while the expression of orexin A was significantly upregulated. Higher density and distribution of orexin A-positive neurons were seen in APP/PS1 mice, with a location of 1.06 mm–2.30 mm away from the anterior fontanelle compared to 1.34 mm–2.18 mm away from the anterior fontanelle in WT mice. These results suggested that the population and distribution of orexin A may play an important role in the progression of AD.
Collapse
|
24
|
Kim SE, Kim S, Kim HJ, Kim REY, Kim SA, Shin C, Lee HW. CLOCK genetic variations are associated with age-related changes in sleep duration and brain volume. J Gerontol A Biol Sci Med Sci 2021; 77:1907-1914. [PMID: 34908110 DOI: 10.1093/gerona/glab365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Although a connection between sleep disruption and brain aging has been documented, biological mechanisms need to be further clarified. Intriguingly, aging is associated with circadian rhythm and/or sleep dysfunction in a key gene regulating circadian rhythm, CLOCK, have been linked to both aging-related sleep disturbances and neurodegenerative diseases. This study aims to investigate how CLOCK genetic variation associates with sleep duration changes and/or volumetric brain alteration. METHODS This population-based cross-sectional study used data from the Korean Genome Epidemiology Study (KoGES), and analyzed sleep characteristics and genetic and brain imaging data in 2,221 subjects (mean 58.8±6.8 years, 50.2% male). Eleven single-nucleotide polymorphisms (SNPs) in CLOCK were analyzed using PLINK software v1.09 to test for their association with sleep duration and brain volume. Haplotype analysis was performed by using pair-wise linkage disequilibrium (LD) of CLOCK polymorphisms, and multivariate analysis of covariance was for statistical analysis. RESULTS Decreased sleep duration was associated with several SNPs in CLOCK intronic regions, with the highest significance for rs10002541 (P=1.58x10 -5). Five SNPs with the highest significance (rs10002541-rs6850524-rs4580704- rs3805151-rs3749474) revealed that CGTCT was the most prevalent. In the major CGTCT haplotype, decreased sleep duration over time was associated with lower cortical volumes predominantly in frontal and parietal regions. Less common haplotypes (GCCTC/CGTTC) had shorter sleep duration and more decreases in sleep duration over 8 years, which revealed smaller total and gray matter volumes, especially in frontal and temporal regions of the left hemisphere. CONCLUSION CLOCK genetic variations could be involved in age-related sleep and brain volume changes.
Collapse
Affiliation(s)
- Song E Kim
- Department of Neurology and Medical Science, Ewha Womans University School of Medicine and Ewha Medical Research Institute, Seoul, South Korea
| | - Soriul Kim
- Institute for Human Genomic Study, College of Medicine, Korea University, Ansan, South Korea
| | - Hyeon Jin Kim
- Department of Neurology and Medical Science, Ewha Womans University School of Medicine and Ewha Medical Research Institute, Seoul, South Korea.,Department of Neurology, Korea University Ansan Hospital, Ansan, South Korea
| | - Regina E Y Kim
- Institute for Human Genomic Study, College of Medicine, Korea University, Ansan, South Korea.,Department of Psychiatry, University of Iowa, Iowa City 52242, IA, USA
| | - Sol Ah Kim
- Department of Neurology and Medical Science, Ewha Womans University School of Medicine and Ewha Medical Research Institute, Seoul, South Korea.,Graduate Program in System Health Science & Engineering, Ewha Womans University, Seoul, South Korea
| | - Chol Shin
- Institute for Human Genomic Study, College of Medicine, Korea University, Ansan, South Korea.,Division of Pulmonary Sleep and Critical Care Medicine, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, South Korea
| | - Hyang Woon Lee
- Department of Neurology and Medical Science, Ewha Womans University School of Medicine and Ewha Medical Research Institute, Seoul, South Korea.,Graduate Program in System Health Science & Engineering, Ewha Womans University, Seoul, South Korea.,Computational Medicine, Graduate Program in System Health Science & Engineering, Ewha Womans University, Seoul 07985, South Korea
| |
Collapse
|
25
|
Traikapi A, Konstantinou N. Gamma Oscillations in Alzheimer’s Disease and Their Potential Therapeutic Role. Front Syst Neurosci 2021; 15:782399. [PMID: 34966263 PMCID: PMC8710538 DOI: 10.3389/fnsys.2021.782399] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/23/2021] [Indexed: 12/27/2022] Open
Abstract
Despite decades of research, Alzheimer’s Disease (AD) remains a lethal neurodegenerative disorder for which there are no effective treatments. This review examines the latest evidence of a novel and newly introduced perspective, which focuses on the restoration of gamma oscillations and investigates their potential role in the treatment of AD. Gamma brain activity (∼25–100 Hz) has been well-known for its role in cognitive function, including memory, and it is fundamental for healthy brain activity and intra-brain communication. Aberrant gamma oscillations have been observed in both mice AD models and human AD patients. A recent line of work demonstrated that gamma entrainment, through auditory and visual sensory stimulation, can effectively attenuate AD pathology and improve cognitive function in mice models of the disease. The first evidence from AD patients indicate that gamma entrainment therapy can reduce loss of functional connectivity and brain atrophy, improve cognitive function, and ameliorate several pathological markers of the disease. Even though research is still in its infancy, evidence suggests that gamma-based therapy may have a disease-modifying effect and has signified a new and promising era in AD research.
Collapse
|
26
|
Shively CA, Lacreuse A, Frye BM, Rothwell ES, Moro M. Nonhuman primates at the intersection of aging biology, chronic disease, and health: An introduction to the American Journal of Primatology Special Issue on aging, cognitive decline, and neuropathology in nonhuman primates. Am J Primatol 2021; 83:e23309. [PMID: 34403529 PMCID: PMC8935964 DOI: 10.1002/ajp.23309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/05/2021] [Indexed: 11/06/2022]
Abstract
Aging across the Primate Order is poorly understood because ages of individuals are often unknown, there is a dearth of aged animals available for study, and because aging is best characterized by longitudinal studies which are difficult to carry out in long-lived species. The human population is aging rapidly, and advanced age is a primary risk factor for several chronic diseases and conditions that impact healthspan. As lifespan has increased, diseases and disorders of the central nervous system (CNS) have become more prevalent, and Alzheimer's disease and related dementias have become epidemic. Nonhuman primate (NHP) models are key to understanding the aging primate CNS. This Special Issue presents a review of current knowledge about NHP CNS aging across the Primate Order. Similarities and differences to human aging, and their implications for the validity of NHP models of aging are considered. Topics include aging-related brain structure and function, neuropathologies, cognitive performance, social behavior and social network characteristics, and physical, sensory, and motor function. Challenges to primate CNS aging research are discussed. Together, this collection of articles demonstrates the value of studying aging in a breadth of NHP models to advance our understanding of human and nonhuman primate aging and healthspan.
Collapse
Affiliation(s)
- Carol A. Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
- Alzheimer’s Disease Research Center, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Agnès Lacreuse
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Brett M. Frye
- Department of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Emily S. Rothwell
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Manuel Moro
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Maryland, USA
| |
Collapse
|
27
|
Kaldun JC, Lone SR, Humbert Camps AM, Fritsch C, Widmer YF, Stein JV, Tomchik SM, Sprecher SG. Dopamine, sleep, and neuronal excitability modulate amyloid-β-mediated forgetting in Drosophila. PLoS Biol 2021; 19:e3001412. [PMID: 34613972 PMCID: PMC8523056 DOI: 10.1371/journal.pbio.3001412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 10/18/2021] [Accepted: 09/14/2021] [Indexed: 11/18/2022] Open
Abstract
Alzheimer disease (AD) is one of the main causes of age-related dementia and neurodegeneration. However, the onset of the disease and the mechanisms causing cognitive defects are not well understood. Aggregation of amyloidogenic peptides is a pathological hallmark of AD and is assumed to be a central component of the molecular disease pathways. Pan-neuronal expression of Aβ42Arctic peptides in Drosophila melanogaster results in learning and memory defects. Surprisingly, targeted expression to the mushroom bodies, a center for olfactory memories in the fly brain, does not interfere with learning but accelerates forgetting. We show here that reducing neuronal excitability either by feeding Levetiracetam or silencing of neurons in the involved circuitry ameliorates the phenotype. Furthermore, inhibition of the Rac-regulated forgetting pathway could rescue the Aβ42Arctic-mediated accelerated forgetting phenotype. Similar effects are achieved by increasing sleep, a critical regulator of neuronal homeostasis. Our results provide a functional framework connecting forgetting signaling and sleep, which are critical for regulating neuronal excitability and homeostasis and are therefore a promising mechanism to modulate forgetting caused by toxic Aβ peptides.
Collapse
Affiliation(s)
- Jenifer C. Kaldun
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Shahnaz R. Lone
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Department of Animal Sciences, Central University of Punjab, Bathinda, India
| | | | - Cornelia Fritsch
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Yves F. Widmer
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Jens V. Stein
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Seth M. Tomchik
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Simon G. Sprecher
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- * E-mail:
| |
Collapse
|
28
|
Yaqinuddin A, Ikram MF, Ambia AR, Alaujan R, Kashir J. 3D Models as an Adjunct for Models in Studying Alzheimer’s Disease. JOURNAL OF HEALTH AND ALLIED SCIENCES NU 2021. [DOI: 10.1055/s-0041-1731864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
AbstractAlzheimer’s disease (AD) is one of the most common causes of dementia. Disease progression is marked by cognitive decline and memory impairment due to neurodegenerative processes in the brain stemming from amyloid-β (Aβ) deposition and formation of neurofibrillary tangles. Pathogenesis in AD is dependent on two main neurological processes: formation of intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein and deposition of extracellular senile Aβ peptides. Given the nature of the disease, the pathology and progression of AD in vivo in humans have been difficult to study in vivo. To this degree, models can help to study the disease pathogenesis, biochemistry, immunological functions, genetics, and potential pharmacotherapy. While animal and two-dimensional (2D) cell culture models have facilitated significant progress in studying the disease, more recent application of novel three-dimensional (3D) culture models has exhibited several advantages. Herein, we describe a brief background of AD, and how current animal, 2D, and 3D models facilitate the study of this disease and associated therapeutics.
Collapse
Affiliation(s)
- Ahmed Yaqinuddin
- Department of Anatomy and Genetic, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Muhammad Faisal Ikram
- Department of Anatomy and Genetic, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Ayesha Rahman Ambia
- Department of Anatomy and Genetic, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Raghad Alaujan
- Department of Anatomy and Genetic, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Junaid Kashir
- Department of Anatomy and Genetic, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
29
|
Harper L, Ooms A, Tuffrey Wijne I. The impact of nutrition on sleep in people with an intellectual disability: An integrative literature review. JOURNAL OF APPLIED RESEARCH IN INTELLECTUAL DISABILITIES 2021; 34:1393-1407. [PMID: 34212459 DOI: 10.1111/jar.12911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 04/25/2021] [Accepted: 05/10/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND People with intellectual disabilities are more likely to experience sleep problems. Sleep can impact on health and well-being; therefore, evidence-based interventions are required to improve sleep in this population. METHOD An integrative literature review was conducted on the impact of nutrition on sleep in people with intellectual disabilities. Following screening of papers (n = 289), 14 papers met the inclusion criteria. RESULTS Themes related to nutrition and improved overall well-being, use of nutritional supplements, specific foods, links to health comorbidities and food fussiness. CONCLUSION This is the first comprehensive review completed on nutritional interventions to improve sleep in people with intellectual disabilities. Dietary patterns may be an important factor to improving the quality and quantity of sleep. However, the current literature regarding the benefit of improved nutrition on sleep in people with an intellectual disability needs to be interpreted with caution.
Collapse
Affiliation(s)
- Lynette Harper
- Department of Nursing, Midwifery and Health, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Ann Ooms
- Kingston University and St Georges University, London, UK
| | | |
Collapse
|
30
|
York A, Everhart A, Vitek MP, Gottschalk KW, Colton CA. Metabolism-Based Gene Differences in Neurons Expressing Hyperphosphorylated AT8- Positive (AT8+) Tau in Alzheimer's Disease. ASN Neuro 2021; 13:17590914211019443. [PMID: 34121475 PMCID: PMC8207264 DOI: 10.1177/17590914211019443] [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] [Indexed: 01/06/2023] Open
Abstract
Metabolic adaptations in the brain are critical to the establishment and maintenance of normal cellular functions and to the pathological responses to disease processes. Here, we have focused on specific metabolic pathways that are involved in immune-mediated neuronal processes in brain using isolated neurons derived from human autopsy brain sections of normal individuals and individuals diagnosed as Alzheimer's disease (AD). Laser capture microscopy was used to select specific cell types in immune-stained thin brain sections followed by NanoString technology to identify and quantify differences in mRNA levels between age-matched control and AD neuronal samples. Comparisons were also made between neurons isolated from AD brain sections expressing pathogenic hyperphosphorylated AT8- positive (AT8+) tau and non-AT8+ AD neurons using double labeling techniques. The mRNA expression data showed unique patterns of metabolic pathway expression between the subtypes of captured neurons that involved membrane based solute transporters, redox factors, and arginine and methionine metabolic pathways. We also identified the expression levels of a novel metabolic gene, Radical-S-Adenosyl Domain1 (RSAD1) and its corresponding protein, Rsad1, that impact methionine usage and radical based reactions. Immunohistochemistry was used to identify specific protein expression levels and their cellular location in NeuN+ and AT8+ neurons. APOE4 vs APOE3 genotype-specific and sex-specific gene expression differences in these metabolic pathways were also observed when comparing neurons from individuals with AD to age-matched individuals.
Collapse
Affiliation(s)
- Audra York
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| | - Angela Everhart
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| | - Michael P Vitek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| | - Kirby W Gottschalk
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| | - Carol A Colton
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States
| |
Collapse
|
31
|
Zhu D, Montagne A, Zhao Z. Alzheimer's pathogenic mechanisms and underlying sex difference. Cell Mol Life Sci 2021; 78:4907-4920. [PMID: 33844047 PMCID: PMC8720296 DOI: 10.1007/s00018-021-03830-w] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/09/2021] [Accepted: 03/29/2021] [Indexed: 01/01/2023]
Abstract
AD is a neurodegenerative disease, and its frequency is often reported to be higher for women than men: almost two-thirds of patients with AD are women. One prevailing view is that women live longer than men on average of 4.5 years, plus there are more women aged 85 years or older than men in most global subpopulations; and older age is the greatest risk factor for AD. However, the differences in the actual risk of developing AD for men and women of the same age is difficult to assess, and the findings have been mixed. An increasing body of evidence from preclinical and clinical studies as well as the complications in estimating incidence support the sex-specific biological mechanisms in diverging AD risk as an important adjunct explanation to the epidemiologic perspective. Although some of the sex differences in AD prevalence are due to differences in longevity, other distinct biological mechanisms increase the risk and progression of AD in women. These risk factors include (1) deviations in brain structure and biomarkers, (2) psychosocial stress responses, (3) pregnancy, menopause, and sex hormones, (4) genetic background (i.e., APOE), (5) inflammation, gliosis, and immune module (i.e., TREM2), and (6) vascular disorders. More studies focusing on the underlying biological mechanisms for this phenomenon are needed to better understand AD. This review presents the most recent data in sex differences in AD-the gateway to precision medicine, therefore, shaping expert perspectives, inspiring researchers to go in new directions, and driving development of future diagnostic tools and treatments for AD in a more customized way.
Collapse
Affiliation(s)
- Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
- Neuroscience Graduate Program, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Axel Montagne
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Zhen Zhao
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
32
|
Basta M, Koutentaki E, Vgontzas A, Zaganas I, Vogiatzi E, Gouna G, Bourbouli M, Panagiotakis S, Kapetanaki S, Fernandez-Mendoza J, Simos P. Objective Daytime Napping is Associated with Disease Severity and Inflammation in Patients with Mild to Moderate Dementia1. J Alzheimers Dis 2021; 74:803-815. [PMID: 32116246 DOI: 10.3233/jad-190483] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with dementia report excessive daytime sleep/sleepiness, which is associated with worse cognitive performance. Inflammatory markers may be elevated in patients with dementia and have been proposed as mediators of sleep/sleepiness. OBJECTIVE To examine the association of objective daytime napping with cognitive performance and peripheral markers of inflammation in patients with dementia as compared to not cognitively impaired (NCI) controls. METHODS A sub-sample of 46 patients with mild-to-moderate dementia and 85 NCI controls, were recruited from a large, population-based cohort of 3,140 elders (≥60 years) in Crete, Greece. All participants underwent medical history/physical examination, extensive neuropsychiatric and neuropsychological evaluation, 3-day 24 h actigraphy and a single morning measure of IL-6 and TNFα plasma levels. Comparisons of sleep parameters and inflammation markers between diagnostic groups, and between nappers and non-nappers within each diagnostic group, were conducted using ANCOVA controlling for demographics/related clinical factors. Associations between inflammatory markers, sleep variables, and neuropsychological performance were assessed within each group using partial correlation analysis controlling for confounders. RESULTS Patients with dementia slept 15 minutes longer during the day than NCI. Within dementia patients, nappers had significantly worse performance on autobiographic memory (p = 0.002), working memory (p = 0.007), episodic memory (p = 0.010), and assessment of daily function (p = 0.012) than non-nappers. Finally, IL-6 levels were significantly associated with nap duration within dementia patients who napped (r = 0.500, p = 0.01). CONCLUSIONS Daytime napping in patients with dementia is associated with worse cognitive performance and increased IL-6 levels. In dementia, objective daytime napping, may be a marker of the severity of the disease.
Collapse
Affiliation(s)
- Maria Basta
- Department of Psychiatry, University Hospital of Heraklion, Heraklion, Crete, Greece.,Sleep Research and Treatment Center, Department of Psychiatry, Penn State University, Hershey, PA, USA
| | - Eirini Koutentaki
- Department of Psychiatry, University Hospital of Heraklion, Heraklion, Crete, Greece
| | - Alexandros Vgontzas
- Department of Psychiatry, University Hospital of Heraklion, Heraklion, Crete, Greece.,Sleep Research and Treatment Center, Department of Psychiatry, Penn State University, Hershey, PA, USA
| | - Ioannis Zaganas
- Department of Neurology, University Hospital of Heraklion, Heraklion, Crete, Greece
| | - Emmanouela Vogiatzi
- Department of Neurology, University Hospital of Heraklion, Heraklion, Crete, Greece
| | - Garyfalia Gouna
- Department of Neurology, University Hospital of Heraklion, Heraklion, Crete, Greece
| | - Mara Bourbouli
- Department of Neurology, University Hospital of Heraklion, Heraklion, Crete, Greece
| | - Symeon Panagiotakis
- Department of Internal Medicine, University Hospital of Heraklion, Heraklion, Crete, Greece
| | - Stefania Kapetanaki
- Department of Neurology, University Hospital of Heraklion, Heraklion, Crete, Greece
| | - Julio Fernandez-Mendoza
- Sleep Research and Treatment Center, Department of Psychiatry, Penn State University, Hershey, PA, USA
| | - Panagiotis Simos
- Department of Psychiatry, University Hospital of Heraklion, Heraklion, Crete, Greece
| |
Collapse
|
33
|
Basta M, Zaganas I, Simos P, Koutentaki E, Dimovasili C, Mathioudakis L, Bourbouli M, Panagiotakis S, Kapetanaki S, Vgontzas A. Apolipoprotein E ɛ4 (APOE ɛ4) Allele is Associated with Long Sleep Duration Among Elderly with Cognitive Impairment. J Alzheimers Dis 2021; 79:763-771. [DOI: 10.3233/jad-200958] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Apolipoprotein E gene (APOE) ɛ4 allele increases the risk for Alzheimer’s disease (AD). Furthermore, among patients with cognitive impairment, longer sleep duration is associated with worse cognitive performance. To date, literature examining the associations between APOE ɛ4 allele and objective sleep duration is limited. Objective: Our aim was to assess the association between APOE ɛ4 and objective sleep duration, among patients with mild cognitive impairment (MCI) and AD. A sub-sample of 89 patients with AD (n = 49) and MCI (n = 40) were recruited from a large, population-based cohort of 3,140 elders (>60 years) residing on Crete, Greece. Methods: All participants underwent medical history/physical examination, extensive neuropsychiatric and neuropsychological evaluation, 3-day 24 h actigraphy and APOE ɛ4 allele genotyping. Comparisons of sleep duration variables between APOE ɛ4 allele carriers and non-carriers were assessed using ANCOVA, controlling for confounders. Results: The sample included 18 APOE ɛ4 carriers and 71 non-carriers, aged 78.6±6.6 and 78.2±6.5 years, respectively. Comparisons between the APOE ɛ4 carriers and non-carriers revealed no significant differences in terms of demographic and clinical variables. In terms of objective sleep duration across the two groups, APOE ɛ4 carriers compared to non-carriers had significantly longer nighttime Total Sleep Time (nTST) (7.7±1.4 versus 7.2±1.3 h, respectively, p = 0.011), as well as 24 h TST (8.5±1.6 versus 7.8±1.5 h, respectively, p = 0.012). Conclusion: Among patients with MCI and AD, APOE ɛ4 carriers have longer objective nighttime and 24 h sleep duration compared to non-carriers. These findings further support that objective long sleep duration is a genetically-driven pre-clinical marker associated with worse prognosis in elderly with cognitive impairment.
Collapse
Affiliation(s)
- Maria Basta
- Department of Psychiatry, University Hospital of Heraklion, Crete, Greece
- Sleep Research and Treatment Center, Department of Psychiatry, Penn State University, Hershey, PA, USA
| | - Ioannis Zaganas
- Department of Neurology, University Hospital of Heraklion, Crete, Greece
| | - Panagiotis Simos
- Department of Psychiatry, University Hospital of Heraklion, Crete, Greece
- Computational Biomedicine Lab, Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Eirini Koutentaki
- Department of Psychiatry, University Hospital of Heraklion, Crete, Greece
| | | | | | - Mara Bourbouli
- Department of Neurology, University Hospital of Heraklion, Crete, Greece
| | | | | | - Alexandros Vgontzas
- Department of Psychiatry, University Hospital of Heraklion, Crete, Greece
- Sleep Research and Treatment Center, Department of Psychiatry, Penn State University, Hershey, PA, USA
| |
Collapse
|
34
|
Rainero I, Bruni AC, Marra C, Cagnin A, Bonanni L, Cupidi C, Laganà V, Rubino E, Vacca A, Di Lorenzo R, Provero P, Isella V, Vanacore N, Agosta F, Appollonio I, Caffarra P, Bussè C, Sambati R, Quaranta D, Guglielmi V, Logroscino G, Filippi M, Tedeschi G, Ferrarese C. The Impact of COVID-19 Quarantine on Patients With Dementia and Family Caregivers: A Nation-Wide Survey. Front Aging Neurosci 2021; 12:625781. [PMID: 33536898 PMCID: PMC7849158 DOI: 10.3389/fnagi.2020.625781] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/21/2020] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Previous studies showed that quarantine for pandemic diseases is associated with several psychological and medical effects. The consequences of quarantine for COVID-19 pandemic in patients with dementia are unknown. We investigated the clinical changes in patients with Alzheimer's disease and other dementias, and evaluated caregivers' distress during COVID-19 quarantine. METHODS The study involved 87 Italian Dementia Centers. Patients with Alzheimer's Disease (AD), Dementia with Lewy Bodies (DLB), Frontotemporal Dementia (FTD), and Vascular Dementia (VD) were eligible for the study. Family caregivers of patients with dementia were interviewed by phone in April 2020, 45 days after quarantine declaration. Main outcomes were patients' changes in cognitive, behavioral, and motor symptoms. Secondary outcomes were effects on caregivers' psychological features. RESULTS 4913 patients (2934 females, 1979 males) fulfilled the inclusion criteria. Caregivers reported a worsening in cognitive functions in 55.1% of patients, mainly in subjects with DLB and AD. Aggravation of behavioral symptoms was observed in 51.9% of patients. In logistic regression analysis, previous physical independence was associated with both cognitive and behavioral worsening (odds ratio 1.85 [95% CI 1.42-2.39], 1.84 [95% CI 1.43-2.38], respectively). On the contrary, pandemic awareness was a protective factor for the worsening of cognitive and behavioral symptoms (odds ratio 0.74 [95% CI 0.65-0.85]; and 0.72 [95% CI 0.63-0.82], respectively). Approximately 25.9% of patients showed the onset of new behavioral symptoms. A worsening in motor function was reported by 36.7% of patients. Finally, caregivers reported a high increase in anxiety, depression, and distress. CONCLUSION Our study shows that quarantine for COVID-19 is associated with an acute worsening of clinical symptoms in patients with dementia as well as increase of caregivers' burden. Our findings emphasize the importance to implement new strategies to mitigate the effects of quarantine in patients with dementia.
Collapse
Affiliation(s)
- Innocenzo Rainero
- Aging Brain and Memory Clinic, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Turin, Italy
- Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Amalia C. Bruni
- Regional Neurogenetic Centre, Department of Primary Care, ASP-CZ, Catanzaro, Italy
| | - Camillo Marra
- Memory Clinic, Fondazione Policlinico Agostino Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Laura Bonanni
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti–Pescara, Chieti, Italy
| | - Chiara Cupidi
- CDCD Ospedale del Delta, AUSL Ferrara, Ferrara, Italy
| | - Valentina Laganà
- Regional Neurogenetic Centre, Department of Primary Care, ASP-CZ, Catanzaro, Italy
| | - Elisa Rubino
- Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Alessandro Vacca
- Aging Brain and Memory Clinic, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Turin, Italy
| | - Raffaele Di Lorenzo
- Regional Neurogenetic Centre, Department of Primary Care, ASP-CZ, Catanzaro, Italy
| | - Paolo Provero
- Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Turin, Italy
- Center for Omics Sciences, IRCCS S. Raffaele Scientific Institute, Milan, Italy
| | - Valeria Isella
- Department of Medicine and Surgery and Milan Center for Neuroscience (NeuroMi), University of Milano–Bicocca, Monza, Italy
| | | | - Federica Agosta
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, Neurorehabilitation Unit, and Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Ildebrando Appollonio
- Department of Medicine and Surgery and Milan Center for Neuroscience (NeuroMi), University of Milano–Bicocca, Monza, Italy
| | | | - Cinzia Bussè
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Renato Sambati
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari, Bari, Italy
- Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Davide Quaranta
- Memory Clinic, Fondazione Policlinico Agostino Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valeria Guglielmi
- Memory Clinic, Fondazione Policlinico Agostino Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giancarlo Logroscino
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari, Bari, Italy
- Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, Neurorehabilitation Unit, and Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Gioacchino Tedeschi
- Department of Medical and Surgical Sciences, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Carlo Ferrarese
- Department of Medicine and Surgery and Milan Center for Neuroscience (NeuroMi), University of Milano–Bicocca, Monza, Italy
| |
Collapse
|
35
|
Yuede CM, Wallace CE, Davis TA, Gardiner WD, Hettinger JC, Edwards HM, Hendrix RD, Doherty BM, Yuede KM, Burstein ES, Cirrito JR. Pimavanserin, a 5HT 2A receptor inverse agonist, rapidly suppresses Aβ production and related pathology in a mouse model of Alzheimer's disease. J Neurochem 2021; 156:658-673. [PMID: 33278025 DOI: 10.1111/jnc.15260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/12/2020] [Accepted: 11/25/2020] [Indexed: 12/30/2022]
Abstract
Amyloid-β (Aβ) peptide aggregation into soluble oligomers and insoluble plaques is a precipitating event in the pathogenesis of Alzheimer's disease (AD). Given that synaptic activity can regulate Aβ generation, we postulated that 5HT2A -Rs may regulate Aβ as well. We treated APP/PS1 transgenic mice with the selective 5HT2A inverse agonists M100907 or Pimavanserin systemically and measured brain interstitial fluid (ISF) Aβ levels in real-time using in vivo microdialysis. Both compounds reduced ISF Aβ levels by almost 50% within hours, but had no effect on Aβ levels in 5HT2A -R knock-out mice. The Aβ-lowering effects of Pimavanserin were blocked by extracellular-regulated kinase (ERK) and NMDA receptor inhibitors. Chronic administration of Pimavanserin by subcutaneous osmotic pump to aged APP/PS1 mice significantly reduced CSF Aβ levels and Aβ pathology and improved cognitive function in these mice. Pimavanserin is FDA-approved to treat Parkinson's disease psychosis, and also has been shown to reduce psychosis in a variety of other dementia subtypes including Alzheimer's disease. These data demonstrate that Pimavanserin may have disease-modifying benefits in addition to its efficacy against neuropsychiatric symptoms of Alzheimer's disease. Read the Editorial Highlight for this article on page 560.
Collapse
Affiliation(s)
- Carla M Yuede
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Clare E Wallace
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Todd A Davis
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Woodrow D Gardiner
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Jane C Hettinger
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah M Edwards
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel D Hendrix
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Brookelyn M Doherty
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Kayla M Yuede
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | | | - John R Cirrito
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
36
|
Huntley JD, Fleming SM, Mograbi DC, Bor D, Naci L, Owen AM, Howard R. Understanding Alzheimer's disease as a disorder of consciousness. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12203. [PMID: 34877398 PMCID: PMC8630359 DOI: 10.1002/trc2.12203] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/28/2021] [Accepted: 07/28/2021] [Indexed: 11/07/2022]
Abstract
People with Alzheimer's disease (AD) demonstrate a range of alterations in consciousness. Changes in awareness of cognitive deficit, self-awareness, and introspection are seen early in AD, and dysfunction of awareness and arousal progresses with increasing disease severity. However, heterogeneity of deficits between individuals and a lack of empirical studies in people with severe dementia highlight the importance of identifying and applying biomarkers of awareness in AD. Impairments of awareness in AD are associated with neuropathology in regions that overlap with proposed neural correlates of consciousness. Recent developments in consciousness science provide theoretical frameworks and experimental approaches to help further understand the conscious experience of people with AD. Recognition of AD as a disorder of consciousness is overdue, and important to both understand the lived experience of people with AD and to improve care.
Collapse
Affiliation(s)
- Jonathan D. Huntley
- Division of PsychiatryUniversity College LondonLondonUK
- Wellcome Centre for Human NeuroimagingUniversity College LondonLondonUK
| | - Stephen M. Fleming
- Wellcome Centre for Human NeuroimagingUniversity College LondonLondonUK
- Department of Experimental PsychologyUniversity College LondonLondonUK
- Max Planck‐UCL Centre for Computational Psychiatry and Ageing ResearchUniversity College LondonLondonUK
| | - Daniel C. Mograbi
- Department of PsychologyPontifical Catholic University of Rio de JaneiroRio de JaneiroBrazil
- Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Daniel Bor
- Department of PsychologyUniversity of CambridgeCambridgeUK
| | - Lorina Naci
- School of PsychologyGlobal Brain Health InstituteTrinity College DublinDublinIreland
| | - Adrian M. Owen
- Brain and Mind InstituteDepartment of Physiology and Pharmacology and Department of PsychologyUniversity of Western OntarioLondonOntarioCanada
| | - Robert Howard
- Division of PsychiatryUniversity College LondonLondonUK
| |
Collapse
|
37
|
Hablitz LM, Plá V, Giannetto M, Vinitsky HS, Stæger FF, Metcalfe T, Nguyen R, Benrais A, Nedergaard M. Circadian control of brain glymphatic and lymphatic fluid flow. Nat Commun 2020; 11:4411. [PMID: 32879313 PMCID: PMC7468152 DOI: 10.1038/s41467-020-18115-2] [Citation(s) in RCA: 319] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 07/24/2020] [Indexed: 12/20/2022] Open
Abstract
The glymphatic system is a network of perivascular spaces that promotes movement of cerebrospinal fluid (CSF) into the brain and clearance of metabolic waste. This fluid transport system is supported by the water channel aquaporin-4 (AQP4) localized to vascular endfeet of astrocytes. The glymphatic system is more effective during sleep, but whether sleep timing promotes glymphatic function remains unknown. We here show glymphatic influx and clearance exhibit endogenous, circadian rhythms peaking during the mid-rest phase of mice. Drainage of CSF from the cisterna magna to the lymph nodes exhibits daily variation opposite to glymphatic influx, suggesting distribution of CSF throughout the animal depends on time-of-day. The perivascular polarization of AQP4 is highest during the rest phase and loss of AQP4 eliminates the day-night difference in both glymphatic influx and drainage to the lymph nodes. We conclude that CSF distribution is under circadian control and that AQP4 supports this rhythm.
Collapse
Affiliation(s)
- Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Virginia Plá
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Michael Giannetto
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Hanna S Vinitsky
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Frederik Filip Stæger
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Tanner Metcalfe
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Rebecca Nguyen
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Abdellatif Benrais
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
38
|
Golini E, Rigamonti M, Iannello F, De Rosa C, Scavizzi F, Raspa M, Mandillo S. A Non-invasive Digital Biomarker for the Detection of Rest Disturbances in the SOD1G93A Mouse Model of ALS. Front Neurosci 2020; 14:896. [PMID: 32982678 PMCID: PMC7490341 DOI: 10.3389/fnins.2020.00896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disease that affects both central and peripheral nervous system, leading to the degeneration of motor neurons, which eventually results in muscle atrophy, paralysis, and death. Sleep disturbances are common in patients with ALS, leading to even further deteriorated quality of life. Investigating methods to potentially assess sleep and rest disturbances in animal models of ALS is thus of crucial interest. We used an automated home cage monitoring system (DVC®) to capture irregular activity patterns that can potentially be associated with sleep and rest disturbances and thus to the progression of ALS in the SOD1G93A mouse model. DVC® enables non-intrusive 24/7 long term animal activity monitoring, which we assessed together with body weight decline and neuromuscular function deterioration measured by grid hanging and grip strength tests in male and female mice from 7 until 24 weeks of age. We show that as the ALS progresses over time in SOD1G93A mice, activity patterns start becoming irregular, especially during day time, with frequent activity bouts that are neither observed in control mice nor in SOD1G93A at a younger age. The increasing irregularities of activity pattern are quantitatively captured by designing a novel digital biomarker, referred to as Regularity Disruption Index (RDI). We show that RDI is a robust measure capable of detecting home cage activity patterns that could be related to rest/sleep-related disturbances during the disease progression. Moreover, the RDI rise during the early symptomatic stage parallels grid hanging and body weight decline. The non-intrusive long-term continuous monitoring of animal activity enabled by DVC® has been instrumental in discovering novel activity patterns potentially correlated, once validated, with sleep and rest disturbances in the SOD1G93A mouse model of the ALS disease.
Collapse
Affiliation(s)
- Elisabetta Golini
- Institute of Biochemistry and Cell Biology-National Research Council (IBBC-CNR), CNR-Campus International Development (EMMA-INFRAFRONTIER-IMPC), Monterotondo, Italy
| | | | | | - Carla De Rosa
- Institute of Biochemistry and Cell Biology-National Research Council (IBBC-CNR), CNR-Campus International Development (EMMA-INFRAFRONTIER-IMPC), Monterotondo, Italy
| | - Ferdinando Scavizzi
- Institute of Biochemistry and Cell Biology-National Research Council (IBBC-CNR), CNR-Campus International Development (EMMA-INFRAFRONTIER-IMPC), Monterotondo, Italy
| | - Marcello Raspa
- Institute of Biochemistry and Cell Biology-National Research Council (IBBC-CNR), CNR-Campus International Development (EMMA-INFRAFRONTIER-IMPC), Monterotondo, Italy
| | - Silvia Mandillo
- Institute of Biochemistry and Cell Biology-National Research Council (IBBC-CNR), CNR-Campus International Development (EMMA-INFRAFRONTIER-IMPC), Monterotondo, Italy
| |
Collapse
|
39
|
Yao Y, Ying Y, Deng Q, Zhang W, Zhu H, Lin Z, Zhang S, Ma J, Zhao Y. Non-invasive 40-Hz Light Flicker Ameliorates Alzheimer's-Associated Rhythm Disorder via Regulating Central Circadian Clock in Mice. Front Physiol 2020; 11:294. [PMID: 32390857 PMCID: PMC7193101 DOI: 10.3389/fphys.2020.00294] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) patients often exhibit perturbed circadian rhythm with fragmented sleep before disease onset. This study was designed to evaluate the effect of a 40-Hz light flicker on circadian rhythm in an AD mouse model (APP/PS1). Locomotor rhythms recordings were conducted to examine the circadian clock rhythm in APP/PS1 mice. Molecular biology analyses, including western blot and real-time qPCR assays, were conducted to assess the changes in circadian locomotor output cycles kaput (CLOCK), brain and muscle arnt-like protein-1 (BMAL1), and period 2 (PER2). In addition to determining the direct effect of a 40-Hz light flicker on hypothalamic central clock, whole-cell voltage-clamp electrophysiology was employed to record individual neurons of suprachiasmatic nucleus (SCN) sections. The results reported herein demonstrate that a 40-Hz light flicker relieves circadian rhythm disorders in APP/PS1 mice and returns the expression levels of key players in the central circadian clock, including Clock, Bmal1, and Per2, to baseline. Moreover, the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) in SCN neurons is significantly lower in APP/PS1 mice than in the control, and the amplitude of sIPSCs is decreased. Exposure to a 40-Hz light flicker significantly increases the sIPSC frequency in SCN neurons of APP/PS1 mice, with little effect on the amplitude. However, the frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) are both unaffected by a 40-Hz light flicker. The data suggest that a 40-Hz light flicker can ameliorate AD-associated circadian rhythm disorders, presenting a new type of therapeutic treatment for rhythm disorders caused by AD.
Collapse
Affiliation(s)
- Youli Yao
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China.,College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| | - Ying Ying
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Qiyu Deng
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Wenjiang Zhang
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| | - Huazhang Zhu
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Zhenglong Lin
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| | - Shengli Zhang
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| | - Junxian Ma
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| | - Yingying Zhao
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
40
|
Brachem C, Winkler A, Tebrügge S, Weimar C, Erbel R, Jöckel KH, Stang A, Dragano N, Moebus S, Kowall B, Jokisch M. Associations between self-reported sleep characteristics and incident mild cognitive impairment: The Heinz Nixdorf Recall Cohort Study. Sci Rep 2020; 10:6542. [PMID: 32300149 PMCID: PMC7162850 DOI: 10.1038/s41598-020-63511-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/27/2020] [Indexed: 01/13/2023] Open
Abstract
Associations of sleep characteristics with mild cognitive impairment (MCI) have been examined in cross-sectional, but rarely in longitudinal studies. Incident MCI and sleep characteristics were assessed in 1,890 participants of the first and second follow-up of the Heinz Nixdorf Recall study, a population-based cohort study in Germany (age at first follow-up 50-80 years, mean follow-up 5.2 years). MCI was assessed with extensive cognitive tests. Sleep questionnaires including PSQI (Pittsburgh Sleep Quality Index) were used to assess sleep quality, sleep disturbances, time asleep, and time in bed. Relative risks (RR) of developing MCI when exposed to sleep characteristics were assessed in regression models adjusted for sociodemographic and cardiovascular risk factors. Poor sleep quality (PSQI > 5) (RR = 1.43, 95% CI: 1.12-1.82, fully adjusted, reference: PSQI ≤ 5) and difficulties initiating sleep (almost nightly versus never) (RR = 1.40, 0.94-2.08) were associated with incident MCI. For time in bed, the risk of MCI was increased for ≤ 5 hours (RR = 2.86, 1.24─6.60, reference:7 to <8 hours). In this longitudinal study with older participants, MCI risk was increased in persons with poor sleep quality, difficulties initiating sleep, and short time in bed.
Collapse
Affiliation(s)
- Christian Brachem
- Nutritional Epidemiology, Department of Nutrition and Food Science, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, Germany
| | - Angela Winkler
- Department of Neurology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Sarah Tebrügge
- Department of Neurology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Christian Weimar
- Department of Neurology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Raimund Erbel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Karl-Heinz Jöckel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Andreas Stang
- Center of Clinical Epidemiology, Institute for Medical Informatics, Biometry and Epidemiology, Medical Faculty, University Duisburg-Essen, Essen, Germany
- School of Public Health, Department of Epidemiology Boston University, 715 Albany Street, Talbot Building, Boston, MA, 02118, USA
| | - Nico Dragano
- Institute of Medical Sociology, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
| | - Susanne Moebus
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Bernd Kowall
- Center of Clinical Epidemiology, Institute for Medical Informatics, Biometry and Epidemiology, Medical Faculty, University Duisburg-Essen, Essen, Germany.
| | - Martha Jokisch
- Department of Neurology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
41
|
Stanhope BA, Jaggard JB, Gratton M, Brown EB, Keene AC. Sleep Regulates Glial Plasticity and Expression of the Engulfment Receptor Draper Following Neural Injury. Curr Biol 2020; 30:1092-1101.e3. [PMID: 32142708 DOI: 10.1016/j.cub.2020.02.057] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/09/2019] [Accepted: 02/20/2020] [Indexed: 12/27/2022]
Abstract
Chronic sleep disturbance is associated with numerous health consequences, including neurodegenerative disease and cognitive decline [1]. Neurite damage due to apoptosis, trauma, or genetic factors is a common feature of aging, and clearance of damaged neurons is essential for maintenance of brain function. In the central nervous system, damaged neurites are cleared by Wallerian degeneration, in which activated microglia and macrophages engulf damaged neurons [2]. The fruit fly Drosophila melanogaster provides a powerful model for investigating the relationship between sleep and Wallerian degeneration [3]. Several lines of evidence suggest that glia influence sleep duration, sleep-mediated neuronal homeostasis, and clearance of toxic substances during sleep, raising the possibility that glial engulfment of damaged axons is regulated by sleep [4]. To explore this possibility, we axotomized olfactory receptor neurons and measured the effects of sleep loss or gain on the clearance of damaged neurites. Mechanical and genetic sleep deprivation impaired the clearance of damaged neurites. Conversely, treatment with the sleep-promoting drug gaboxadol accelerated clearance, while genetic induction of sleep promotes Draper expression. In sleep-deprived animals, multiple markers of glial activation were delayed, including activation of the JAK-STAT pathway, upregulation of the cell corpse engulfment receptor Draper, and innervation of the antennal lobe by glial membranes. These markers were all enhanced following genetic and pharmacological sleep induction. Taken together, these findings reveal a critical association between sleep and glial activation following neural injury, providing a platform for further investigations of the molecular mechanisms underlying sleep-dependent modulation of glial function and neurite clearance.
Collapse
Affiliation(s)
- Bethany A Stanhope
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458
| | - James B Jaggard
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458
| | - Melanie Gratton
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458
| | - Elizabeth B Brown
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458
| | - Alex C Keene
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458.
| |
Collapse
|
42
|
Wang M, Zong HF, Chang KW, Han H, Yasir Rizvi M, Iffat Neha S, Li ZY, Yang WN, Qian YH. 5-HT 1AR alleviates Aβ-induced cognitive decline and neuroinflammation through crosstalk with NF-κB pathway in mice. Int Immunopharmacol 2020; 82:106354. [PMID: 32143008 DOI: 10.1016/j.intimp.2020.106354] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/02/2020] [Accepted: 02/24/2020] [Indexed: 12/20/2022]
Abstract
The 5-hydroxytryptamine (5-HT) receptor is significant for the regulation of mood and memory. However, the role of 5-HT1AR in β-Amyloid protein (Aβ)-induced cognitive decline, neuroinflammation and the possible mechanism remains elusive. Thus, we aimed to evaluate the effects of 5-HT1AR on Aβ-induced learning and memory decline and neuroinflammation in mice. Novel object recognition and Morris water maze tests were performed to observe learning and memory behavior in mice. Protein levels of Iba1, GFAP, MAP2, TNF-α, Tβ4, C-fos, IKK-β, IKB-α, NF-κBp65, phospho-NF-κBp65 in the hippocampus were examined by immunostaining or western blotting. Aβ1-42-treatment inducing learning and memory decline was shown in novel object recognition and Morris water maze tests; neuroinflammation shown in immunostaining. Our study found out that 5-HT1AR inhibitor WAY100635 showed significant improvement in Aβ-induced learning and memory decline. Moreover, WAY100635 decreases levels of Iba1, GFAP, and TNF-α in the hippocampus, which were related to neuroinflammation. While treatment with 5-HT1AR agonist 8-OH-DPAT or ERK inhibitor U0126 exerted no effects or even aggravated Aβ-induced learning and memory decline. In addition, WAY100635 could downregulate phospho-NF-κB in the hippocampus of Aβ1-42-injected mice. These results provide new insight into the mechanism, for 5-HT1AR in Aβ-induced cognitive impairments through crosstalk with the NF-κB signaling pathway. Our data indicated that WAY100635 was involved in the protective effects against neuroinflammation and improvement of learning and memory in Alzheimer's disease.
Collapse
Affiliation(s)
- Meng Wang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West, China
| | - Hang-Fan Zong
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West, China
| | - Ke-Wei Chang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West, China
| | - Hua Han
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West, China; Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an 710061, China
| | - Mohammad Yasir Rizvi
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West, China
| | - Saema Iffat Neha
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West, China
| | - Zhi-Yi Li
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West, China
| | - Wei-Na Yang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West, China; Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an 710061, China
| | - Yi-Hua Qian
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West, China; Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an 710061, China.
| |
Collapse
|
43
|
Kery R, Chen APF, Kirschen GW. Genetic targeting of astrocytes to combat neurodegenerative disease. Neural Regen Res 2020; 15:199-211. [PMID: 31552885 PMCID: PMC6905329 DOI: 10.4103/1673-5374.265541] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Astrocytes, glial cells that interact extensively with neurons and other support cells throughout the central nervous system, have recently come under the spotlight for their potential contribution to, or potential regenerative role in a host of neurodegenerative disorders. It is becoming increasingly clear that astrocytes, in concert with microglial cells, activate intrinsic immunological pathways in the setting of neurodegenerative injury, although the direct and indirect consequences of such activation are still largely unknown. We review the current literature on the astrocyte’s role in several neurodegenerative diseases, as well as highlighting recent advances in genetic manipulation of astrocytes that may prove critical to modulating their response to neurological injury, potentially combatting neurodegenerative damage.
Collapse
Affiliation(s)
- Rachel Kery
- Medical Scientist Training Program (MSTP), Stony Brook Medicine; Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Allen P F Chen
- Medical Scientist Training Program (MSTP), Stony Brook Medicine; Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Gregory W Kirschen
- Medical Scientist Training Program (MSTP), Stony Brook Medicine, Stony Brook, NY, USA
| |
Collapse
|
44
|
You JC, Jones E, Cross DE, Lyon AC, Kang H, Newberg AB, Lippa CF. Association of β-Amyloid Burden With Sleep Dysfunction and Cognitive Impairment in Elderly Individuals With Cognitive Disorders. JAMA Netw Open 2019; 2:e1913383. [PMID: 31617927 PMCID: PMC6806437 DOI: 10.1001/jamanetworkopen.2019.13383] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPORTANCE Evidence shows that sleep dysfunction and β-amyloid (Aβ) deposition work synergistically to impair brain function in individuals with normal cognition, increasing the risk of developing dementia later in life. However, whether Aβ continues to play an integral role in sleep dysfunction after the onset of cognitive decline in individuals with dementia is unclear. OBJECTIVE To determine whether Aβ deposition in the brain is associated with subjective measures of sleep quality and cognition in elderly individuals with cognitive disorders. DESIGN, SETTING, AND PARTICIPANTS A nested survey study was conducted at the Cognitive Disorders and Comprehensive Alzheimer Disease Center of Thomas Jefferson University Hospital in Philadelphia, Pennsylvania. Participants included patients aged 65 years and older with cognitive disorders verified by neuropsychological testing. Eligible participants were identified from a referral center-based sample of patients who underwent fluorine 18-labeled florbetaben positron emission tomography imaging at Thomas Jefferson University Hospital as part of the multicenter Imaging Dementia-Evidence for Amyloid Scanning study. Data collection and analysis occurred between November 2018 and March 2019. MAIN OUTCOMES AND MEASURES Sleep quality was measured via responses to sleep questionnaires, Aβ deposition was measured via fluorine 18-labeled florbetaben positron emission tomography, and cognition was measured via Mini-Mental State Examination (MMSE) performance. RESULTS Of the 67 eligible participants, 52 (77.6%) gave informed consent to participate in the study. Of the 52 enrolled participants (mean [SD] age, 76.6 [7.4] years), 27 (51.9%) were women. Daytime sleepiness was associated with Aβ deposition in the brainstem (B = 0.0063; 95% CI, 0.001 to 0.012; P = .02), but not MMSE performance (B = -0.01; 95% CI, -0.39 to 0.37; P = .96). The number of nocturnal awakenings was associated with Aβ deposition in the precuneus (B = 0.11; 95% CI, 0.06 to 0.17; P < .001) and poor MMSE performance (B = -2.13; 95% CI, -3.13 to -1.13; P < .001). Mediation analysis demonstrated an indirect association between Aβ deposition and poor MMSE performance that relied on nocturnal awakenings as an intermediary (B = -3.99; 95% CI, -7.88 to -0.83; P = .01). CONCLUSIONS AND RELEVANCE Nighttime sleep disruption may mediate the association between Aβ and cognitive impairment, suggesting that there is an underlying sleep-dependent mechanism that links Aβ burden in the brain to cognitive decline. Further elucidation of this mechanism may improve understanding of disease processes associated with Aβ accumulation.
Collapse
Affiliation(s)
- Jason C. You
- Cognitive Disorders and Comprehensive Alzheimer’s Disease Center, Department of Neurology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Internal Medicine, Lankenau Medical Center, Wynnewood, Pennsylvania
| | - Erica Jones
- Cognitive Disorders and Comprehensive Alzheimer’s Disease Center, Department of Neurology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Devon E. Cross
- Cognitive Disorders and Comprehensive Alzheimer’s Disease Center, Department of Neurology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Abigail C. Lyon
- Cognitive Disorders and Comprehensive Alzheimer’s Disease Center, Department of Neurology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Hyunseung Kang
- Department of Statistics, University of Wisconsin-Madison, Madison
| | - Andrew B. Newberg
- Marcus Institute for Integrative Health, Department of Integrative Medicine, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
- Department of Radiology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Carol F. Lippa
- Cognitive Disorders and Comprehensive Alzheimer’s Disease Center, Department of Neurology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| |
Collapse
|
45
|
Kaufman MJ, Kanayama G, Hudson JI, Pope HG. Supraphysiologic-dose anabolic-androgenic steroid use: A risk factor for dementia? Neurosci Biobehav Rev 2019; 100:180-207. [PMID: 30817935 PMCID: PMC6451684 DOI: 10.1016/j.neubiorev.2019.02.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/13/2019] [Accepted: 02/17/2019] [Indexed: 02/06/2023]
Abstract
Supraphysiologic-dose anabolic-androgenic steroid (AAS) use is associated with physiologic, cognitive, and brain abnormalities similar to those found in people at risk for developing Alzheimer's Disease and its related dementias (AD/ADRD), which are associated with high brain β-amyloid (Aβ) and hyperphosphorylated tau (tau-P) protein levels. Supraphysiologic-dose AAS induces androgen abnormalities and excess oxidative stress, which have been linked to increased and decreased expression or activity of proteins that synthesize and eliminate, respectively, Aβ and tau-P. Aβ and tau-P accumulation may begin soon after initiating supraphysiologic-dose AAS use, which typically occurs in the early 20s, and their accumulation may be accelerated by other psychoactive substance use, which is common among non-medical AAS users. Accordingly, the widespread use of supraphysiologic-dose AAS may increase the numbers of people who develop dementia. Early diagnosis and correction of sex-steroid level abnormalities and excess oxidative stress could attenuate risk for developing AD/ADRD in supraphysiologic-dose AAS users, in people with other substance use disorders, and in people with low sex-steroid levels or excess oxidative stress associated with aging.
Collapse
Affiliation(s)
- Marc J Kaufman
- McLean Imaging Center, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA.
| | - Gen Kanayama
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - James I Hudson
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Harrison G Pope
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
46
|
Spiers JG, Breda C, Robinson S, Giorgini F, Steinert JR. Drosophila Nrf2/Keap1 Mediated Redox Signaling Supports Synaptic Function and Longevity and Impacts on Circadian Activity. Front Mol Neurosci 2019; 12:86. [PMID: 31040766 PMCID: PMC6476960 DOI: 10.3389/fnmol.2019.00086] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/20/2019] [Indexed: 12/30/2022] Open
Abstract
Many neurodegenerative conditions and age-related neuropathologies are associated with increased levels of reactive oxygen species (ROS). The cap "n" collar (CncC) family of transcription factors is one of the major cellular system that fights oxidative insults, becoming activated in response to oxidative stress. This transcription factor signaling is conserved from metazoans to human and has a major developmental and disease-associated relevance. An important mammalian member of the CncC family is nuclear factor erythroid 2-related factor 2 (Nrf2) which has been studied in numerous cellular systems and represents an important target for drug discovery in different diseases. CncC is negatively regulated by Kelch-like ECH associated protein 1 (Keap1) and this interaction provides the basis for a homeostatic control of cellular antioxidant defense. We have utilized the Drosophila model system to investigate the roles of CncC signaling on longevity, neuronal function and circadian rhythm. Furthermore, we assessed the effects of CncC function on larvae and adult flies following exposure to stress. Our data reveal that constitutive overexpression of CncC modifies synaptic mechanisms that positively impact on neuronal function, and suppression of CncC inhibitor, Keap1, shows beneficial phenotypes on synaptic function and longevity. Moreover, supplementation of antioxidants mimics the effects of augmenting CncC signaling. Under stress conditions, lack of CncC signaling worsens survival rates and neuronal function whilst silencing Keap1 protects against stress-induced neuronal decline. Interestingly, overexpression and RNAi-mediated downregulation of CncC have differential effects on sleep patterns possibly via interactions with redox-sensitive circadian cycles. Thus, our data illustrate the important regulatory potential of CncC signaling in neuronal function and synaptic release affecting multiple aspects within the nervous system.
Collapse
Affiliation(s)
- Jereme G Spiers
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Carlo Breda
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Sue Robinson
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Joern R Steinert
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
47
|
Jones SE, van Hees VT, Mazzotti DR, Marques-Vidal P, Sabia S, van der Spek A, Dashti HS, Engmann J, Kocevska D, Tyrrell J, Beaumont RN, Hillsdon M, Ruth KS, Tuke MA, Yaghootkar H, Sharp SA, Ji Y, Harrison JW, Freathy RM, Murray A, Luik AI, Amin N, Lane JM, Saxena R, Rutter MK, Tiemeier H, Kutalik Z, Kumari M, Frayling TM, Weedon MN, Gehrman PR, Wood AR. Genetic studies of accelerometer-based sleep measures yield new insights into human sleep behaviour. Nat Commun 2019; 10:1585. [PMID: 30952852 PMCID: PMC6451011 DOI: 10.1038/s41467-019-09576-1] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 03/14/2019] [Indexed: 01/16/2023] Open
Abstract
Sleep is an essential human function but its regulation is poorly understood. Using accelerometer data from 85,670 UK Biobank participants, we perform a genome-wide association study of 8 derived sleep traits representing sleep quality, quantity and timing, and validate our findings in 5,819 individuals. We identify 47 genetic associations at P < 5 × 10-8, of which 20 reach a stricter threshold of P < 8 × 10-10. These include 26 novel associations with measures of sleep quality and 10 with nocturnal sleep duration. The majority of identified variants associate with a single sleep trait, except for variants previously associated with restless legs syndrome. For sleep duration we identify a missense variant (p.Tyr727Cys) in PDE11A as the likely causal variant. As a group, sleep quality loci are enriched for serotonin processing genes. Although accelerometer-derived measures of sleep are imperfect and may be affected by restless legs syndrome, these findings provide new biological insights into sleep compared to previous efforts based on self-report sleep measures.
Collapse
Affiliation(s)
- Samuel E Jones
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | | | - Diego R Mazzotti
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, 1011, Switzerland
| | - Séverine Sabia
- Research Department of Epidemiology and Public Health, University College London, London, WC1E 6BT, UK
- INSERM, U1153, Epidemiology of Ageing and Neurodegenerative diseases, Université de Paris, Paris, 75010, France
| | - Ashley van der Spek
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Jorgen Engmann
- UCL Institute of Cardiovascular Science, Research department of Population Science and Experimental Medicine, Centre for Translational Genomics, 222 Euston Road, London, NW1 2DA, UK
| | - Desana Kocevska
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Jessica Tyrrell
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Robin N Beaumont
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Melvyn Hillsdon
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX1 2LU, UK
| | - Katherine S Ruth
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Marcus A Tuke
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Seth A Sharp
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Yingjie Ji
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Jamie W Harrison
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Rachel M Freathy
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Anna Murray
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Jacqueline M Lane
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02111, USA
- Departments of Medicine, Brigham and Women's Hospital and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Martin K Rutter
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, M13 9PL, UK
- Manchester Diabetes Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Oxford Road, 193 Hathersage Road, Manchester, M13 0JE, UK
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
- Department of Social and Behavioral Science, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne, 1010, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Meena Kumari
- ISER, University of Essex, Colchester, Essex, CO4 3SQ, UK
| | - Timothy M Frayling
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Michael N Weedon
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK.
| | - Philip R Gehrman
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Andrew R Wood
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK.
| |
Collapse
|
48
|
Pál B. Involvement of extrasynaptic glutamate in physiological and pathophysiological changes of neuronal excitability. Cell Mol Life Sci 2018; 75:2917-2949. [PMID: 29766217 PMCID: PMC11105518 DOI: 10.1007/s00018-018-2837-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Glutamate is the most abundant neurotransmitter of the central nervous system, as the majority of neurons use glutamate as neurotransmitter. It is also well known that this neurotransmitter is not restricted to synaptic clefts, but found in the extrasynaptic regions as ambient glutamate. Extrasynaptic glutamate originates from spillover of synaptic release, as well as from astrocytes and microglia. Its concentration is magnitudes lower than in the synaptic cleft, but receptors responding to it have higher affinity for it. Extrasynaptic glutamate receptors can be found in neuronal somatodendritic location, on astroglia, oligodendrocytes or microglia. Activation of them leads to changes of neuronal excitability with different amplitude and kinetics. Extrasynaptic glutamate is taken up by neurons and astrocytes mostly via EAAT transporters, and astrocytes, in turn metabolize it to glutamine. Extrasynaptic glutamate is involved in several physiological phenomena of the central nervous system. It regulates neuronal excitability and synaptic strength by involving astroglia; contributing to learning and memory formation, neurosecretory and neuromodulatory mechanisms, as well as sleep homeostasis.The extrasynaptic glutamatergic system is affected in several brain pathologies related to excitotoxicity, neurodegeneration or neuroinflammation. Being present in dementias, neurodegenerative and neuropsychiatric diseases or tumor invasion in a seemingly uniform way, the system possibly provides a common component of their pathogenesis. Although parts of the system are extensively discussed by several recent reviews, in this review I attempt to summarize physiological actions of the extrasynaptic glutamate on neuronal excitability and provide a brief insight to its pathology for basic understanding of the topic.
Collapse
Affiliation(s)
- Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Nagyerdei krt 98, Debrecen, 4012, Hungary.
| |
Collapse
|