301
|
Byeon SK, Madugundu AK, Jain AP, Bhat FA, Jung JH, Renuse S, Darrow J, Bakker A, Albert M, Moghekar A, Pandey A. Cerebrospinal fluid lipidomics for biomarkers of Alzheimer's disease. Mol Omics 2021; 17:454-463. [PMID: 34125126 PMCID: PMC8210464 DOI: 10.1039/d0mo00186d] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and is associated with serious neurologic sequelae resulting from neurodegenerative changes. Identification of markers of early-stage AD could be important for designing strategies to arrest the progression of the disease. The brain is rich in lipids because they are crucial for signal transduction and anchoring of membrane proteins. Cerebrospinal fluid (CSF) is an excellent specimen for studying the metabolism of lipids in AD because it can reflect changes occurring in the brain. We aimed to identify CSF lipidomic alterations associated with AD, using untargeted lipidomics, carried out in positive and negative ion modes. We found CSF lipids that were significantly altered in AD cases. In addition, comparison of CSF lipid profiles between persons with mild cognitive impairment (MCI) and AD showed a strong positive correlation between the lipidomes of the MCI and AD groups. The novel lipid biomarkers identified in this study are excellent candidates for validation in a larger set of patient samples and as predictive biomarkers of AD through future longitudinal studies. Once validated, the lipid biomarkers could lead to early detection, disease monitoring and the ability to measure the efficacy of potential therapeutic interventions in AD.
Collapse
Affiliation(s)
- Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55902, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
302
|
Translation and Validation of 10/66 Dementia Diagnostic Battery in Urdu in Karachi, Pakistan. Alzheimer Dis Assoc Disord 2021; 34:163-169. [PMID: 31651419 DOI: 10.1097/wad.0000000000000359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The 10/66 dementia research group (DRG) diagnostic tool was devised to diagnose dementia in people with low education in low and middle-income countries. This study aimed to validate the 10/66 DRG tool in Urdu in Pakistan. METHODOLOGY People older than or equal to 60 years were included: (1) With normal cognition: no/low education, high education, and depression; (2) People with mild and moderate dementia. The diagnostic and statistical manual IV-TR clinician diagnosis was used as the gold standard for dementia. The Clinician Dementia Rating scale was used to rate dementia severity. The geriatric mental status AGECAT stage I output was used to diagnose depression. The 10/66 battery was comprised of CSI-D (cognitive screening instrument for dementia), CERAD (consortium to establish a registry of Alzheimer disease), and animal naming test. RESULT The sensitivity and specificity of CSI-D COG score for diagnosing dementia was 86.7% and 72.1%, for CSI-D DF score was 71.1% and 96.1%, for CERAD-10 word list delayed recall was 85.9% and 62.2% at a cut point of ≥5 words, and 70.3% and 97.7% for 10/66 diagnostic algorithm. CONCLUSIONS The 10/66 DRG tool is a valid instrument for diagnosing dementia in the Urdu-speaking population, including with low education and depression, in Pakistan.
Collapse
|
303
|
Baik SH, Selvaraji S, Fann DY, Poh L, Jo DG, Herr DR, Zhang SR, Kim HA, Silva MD, Lai MK, Chen CLH, Drummond GR, Lim KL, Sobey CG, Arumugam TV. Hippocampal transcriptome profiling reveals common disease pathways in chronic hypoperfusion and aging. Aging (Albany NY) 2021; 13:14651-14674. [PMID: 34074801 PMCID: PMC8221317 DOI: 10.18632/aging.203123] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Vascular dementia (VaD) is a progressive cognitive impairment of vascular etiology. VaD is characterized by cerebral hypoperfusion, increased blood-brain barrier permeability and white matter lesions. An increased burden of VaD is expected in rapidly aging populations. The hippocampus is particularly susceptible to hypoperfusion, and the resulting memory impairment may play a crucial role in VaD. Here we have investigated the hippocampal gene expression profile of young and old mice subjected to cerebral hypoperfusion by bilateral common carotid artery stenosis (BCAS). Our data in sham-operated young and aged mice reveal an age-associated decline in cerebral blood flow and differential gene expression. In fact, BCAS and aging caused broadly similar effects. However, BCAS-induced changes in hippocampal gene expression differed between young and aged mice. Specifically, transcriptomic analysis indicated that in comparison to young sham mice, many pathways altered by BCAS in young mice resembled those already present in sham aged mice. Over 30 days, BCAS in aged mice had minimal effect on either cerebral blood flow or hippocampal gene expression. Immunoblot analyses confirmed these findings. Finally, relative to young sham mice the cell type-specific profile of genes in both young BCAS and old sham animals further revealed common cell-specific genes. Our data provide a genetic-based molecular framework for hypoperfusion-induced hippocampal damage and reveal common cellular signaling pathways likely to be important in the pathophysiology of VaD.
Collapse
Affiliation(s)
- Sang-Ha Baik
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sharmelee Selvaraji
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore
| | - David Y. Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Luting Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Deron R. Herr
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Shenpeng R. Zhang
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Hyun Ah Kim
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Mitchell K.P. Lai
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher Li-Hsian Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Grant R. Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Kah-Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Christopher G. Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Thiruma V. Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
304
|
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: 134] [Impact Index Per Article: 33.5] [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
|
305
|
Abd El-Fatah IM, Abdelrazek HMA, Ibrahim SM, Abdallah DM, El-Abhar HS. Dimethyl fumarate abridged tauo-/amyloidopathy in a D-Galactose/ovariectomy-induced Alzheimer's-like disease: Modulation of AMPK/SIRT-1, AKT/CREB/BDNF, AKT/GSK-3β, adiponectin/Adipo1R, and NF-κB/IL-1β/ROS trajectories. Neurochem Int 2021; 148:105082. [PMID: 34052296 DOI: 10.1016/j.neuint.2021.105082] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/30/2021] [Accepted: 05/23/2021] [Indexed: 10/21/2022]
Abstract
Since the role of estrogen in postmenauposal-associated dementia is still debatable, this issue urges the search for other medications. Dimethyl fumarate (DMF) is a drug used for the treatment of multiple sclerosis and has shown a neuroprotective effect against other neurodegenerative diseases. Accordingly, the present study aimed to evaluate the effect of DMF on an experimental model of Alzheimer disease (AD) using D-galactose (D-Gal) administered to ovariectomized (OVX) rats, resembling a postmenopausal dementia paradigm. Adult 18-month old female Wistar rats were allocated into sham-operated and OVX/D-Gal groups that were either left untreated or treated with DMF for 56 days starting three weeks after sham-operation or ovariectomy. DMF succeeded to ameliorate cognitive (learning/short- and long-term memory) deficits and to enhance the dampened overall activity (NOR, Barnes-/Y-maze tests). These behavioral upturns were associated with increased intact neurons (Nissl stain) and a reduction in OVX/D-Gal-mediated hippocampal CA1 neurodegeneration and astrocyte activation assessed as GFAP immunoreactivity. Mechanistically, DMF suppressed the hippocampal contents of AD-surrogate markers; viz., apolipoprotein (APO)-E1, BACE1, Aβ42, and hyperphosphorylated Tau. Additionally, DMF has augmented the neuroprotective parameters p-AKT, its downstream target CREB and BDNF. Besides, it activated AMPK, and enhanced SIRT-1, as well as antioxidant defenses (SOD, GSH). On the other hand, DMF inhibited the transcription factor NF-κB, IL-1β, adiponectin/adiponectin receptor type (AdipoR)1, GSK-3β, and MDA. Accordingly, in this postmenopausal AD model, DMF treatment by pursuing the adiponectin/AdipoR1, AMPK/SIRT-1, AKT/CREB/BDNF, AKT/GSK-3β, and APO-E1 quartet hampered the associated tauo-/amyloidopathy and NF-κB-mediated oxidative/inflammatory responses to advance insights into its anti-amnesic effect.
Collapse
Affiliation(s)
- Israa M Abd El-Fatah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str, 11562, Cairo, Egypt
| | - Heba M A Abdelrazek
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Egypt
| | - Sherehan M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str, 11562, Cairo, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str, 11562, Cairo, Egypt.
| | - Hanan S El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str, 11562, Cairo, Egypt
| |
Collapse
|
306
|
Yuan JL, Zhao RX, Ma YJ, Li XD, Zhou XM, Wang XF, Jiang XY, Li SJ. Prevalence/potential risk factors for motoric cognitive risk and its relationship to falls in elderly Chinese people: a cross-sectional study. Eur J Neurol 2021; 28:2680-2687. [PMID: 33905575 DOI: 10.1111/ene.14884] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Motoric cognitive risk syndrome (MCR) is characterized by slow walking speed and subjective memory complaints (SMCs). This study investigated the prevalence and potential risk factors of MCR and its association with falls in Chinese community-dwelling older adults. METHODS The analysis was based on data from the Rugao Longevity and Aging Study (RuLAS). MCR was defined as the presence of both SMCs and slow walking speed in participants free of major neurocognitive disorders. SMCs were determined according to a positive answer to the question 'Do you feel you have more problems with memory than most?' in the 15-item Geriatric Depression Scale. Slow walking speed was defined as one standard deviation or more below the mean value for patients' age and sex. Data on falls were derived from a standardized questionnaire. RESULTS The prevalence of SMCs, slow walking speed and MCR in the RuLAS cohort (N = 1592) was 51.9%, 15.6% and 8.3%, respectively. After adjusting for other covariates, an occupation of farming (odds ratio [OR] 2.358, 95% confidence interval [CI] 1.007-5.521, p = 0.048), history of cerebrovascular disease (OR 2.215, 95% CI 1.032-4.752, p = 0.041) and hospitalization (OR 2.008, 95% CI 1.120-3.602, p = 0.019) were risk factors for MCR. Binary logistic regression analysis indicated that the risk of falls was increased by MCR (OR 1.547, 95% CI 1.009-2.371), SMC (OR 1.308, 95% CI 1.003-1.707) and slow walking speed (OR 1.442, 95% CI 1.030-2.017). CONCLUSIONS Early identification of potential risk factors of MCR can prevent the occurrence of adverse health events such as falls in the elderly.
Collapse
Affiliation(s)
- Jing-Lin Yuan
- Department of Neurology, Beijing Daxing District People's Hospital, Beijing, China
| | - Rui-Xue Zhao
- Department of Neurology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ya-Jun Ma
- Department of Neurology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiao-Dong Li
- Department of Neurology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiao-Mei Zhou
- Department of Neurology, Beijing Daxing District People's Hospital, Beijing, China
| | - Xiao-Feng Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, Fudan University, Shanghai, China
| | - Xiao-Yan Jiang
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, China
| | - Shu-Juan Li
- Department of Neurology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
307
|
Wehbe Z, Hammoud SH, Yassine HM, Fardoun M, El-Yazbi AF, Eid AH. Molecular and Biological Mechanisms Underlying Gender Differences in COVID-19 Severity and Mortality. Front Immunol 2021; 12:659339. [PMID: 34025658 PMCID: PMC8138433 DOI: 10.3389/fimmu.2021.659339] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Globally, over two million people have perished due to the recent pandemic caused by SARS-CoV-2. The available epidemiological global data for SARS-CoV-2 portrays a higher rate of severity and mortality in males. Analyzing gender differences in the host mechanisms involved in SARS-CoV-2 infection and progression may offer insight into the more detrimental disease prognosis and clinical outcome in males. Therefore, we outline sexual dimorphisms which exist in particular host factors and elaborate on how they may contribute to the pronounced severity in male COVID-19 patients. This includes disparities detected in comorbidities, the ACE2 receptor, renin-angiotensin system (RAS), signaling molecules involved in SARS-CoV-2 replication, proteases which prime viral S protein, the immune response, and behavioral considerations. Moreover, we discuss sexual disparities associated with other viruses and a possible gender-dependent response to SARS-CoV-2 vaccines. By specifically highlighting these immune-endocrine processes as well as behavioral factors that differentially exist between the genders, we aim to offer a better understanding in the variations of SARS-CoV-2 pathogenicity.
Collapse
Affiliation(s)
- Zena Wehbe
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Safaa Hisham Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | | | - Manal Fardoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, Qatar University Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University Health, Qatar University, Doha, Qatar
| |
Collapse
|
308
|
Lim WLF, Huynh K, Chatterjee P, Martins I, Jayawardana KS, Giles C, Mellett NA, Laws SM, Bush AI, Rowe CC, Villemagne VL, Ames D, Drew BG, Masters CL, Meikle PJ, Martins RN. Relationships Between Plasma Lipids Species, Gender, Risk Factors, and Alzheimer's Disease. J Alzheimers Dis 2021; 76:303-315. [PMID: 32474467 PMCID: PMC7369125 DOI: 10.3233/jad-191304] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background: Lipid metabolism is altered in Alzheimer’s disease (AD); however, the relationship between AD risk factors (age, APOEɛ4, and gender) and lipid metabolism is not well defined. Objective: We investigated whether altered lipid metabolism associated with increased age, gender, and APOE status may contribute to the development of AD by examining these risk factors in healthy controls and also clinically diagnosed AD individuals. Methods: We performed plasma lipidomic profiling (582 lipid species) of the Australian Imaging, Biomarkers and Lifestyle flagship study of aging cohort (AIBL) using liquid chromatography-mass spectrometry. Linear regression and interaction analysis were used to explore the relationship between risk factors and plasma lipid species. Results: We observed strong associations between plasma lipid species with gender and increasing age in cognitively normal individuals. However, APOEɛ4 was relatively weakly associated with plasma lipid species. Interaction analysis identified differential associations of sphingolipids and polyunsaturated fatty acid esterified lipid species with AD based on age and gender, respectively. These data indicate that the risk associated with age, gender, and APOEɛ4 may, in part, be mediated by changes in lipid metabolism. Conclusion: This study extends our existing knowledge of the relationship between the lipidome and AD and highlights the complexity of the relationships between lipid metabolism and AD at different ages and between men and women. This has important implications for how we assess AD risk and also for potential therapeutic strategies involving modulation of lipid metabolic pathways.
Collapse
Affiliation(s)
- Wei Ling Florence Lim
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, WA, Australia.,Cooperative Research Centre (CRC) for Mental Health, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, VIC, Australia.,Monash University, Melbourne, Victoria, VIC, Australia
| | - Pratishtha Chatterjee
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, WA, Australia.,Department of Biomedical Sciences, Macquarie University, North Ryde, New South Wales, NSW, Australia.,KaRa Institute of Neurological Disease, Sydney, Macquarie Park, New South Wales, NSW, Australia
| | - Ian Martins
- Cooperative Research Centre (CRC) for Mental Health, Australia
| | | | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Victoria, VIC, Australia
| | - Natalie A Mellett
- Baker Heart and Diabetes Institute, Melbourne, Victoria, VIC, Australia
| | - Simon M Laws
- Cooperative Research Centre (CRC) for Mental Health, Australia.,Collaborative Genomics Group, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Western Australia, WA, Australia
| | - Ashley I Bush
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria, VIC, Australia
| | - Christopher C Rowe
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria, VIC, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, VIC, Australia
| | - Victor L Villemagne
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria, VIC, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, VIC, Australia.,Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, VIC, Australia
| | - David Ames
- National Ageing Research Institute, Parkville, Victoria, VIC, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, Victoria, VIC, Australia.,Monash University, Melbourne, Victoria, VIC, Australia
| | - Colin L Masters
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria, VIC, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, VIC, Australia.,Monash University, Melbourne, Victoria, VIC, Australia
| | - Ralph N Martins
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, WA, Australia.,Cooperative Research Centre (CRC) for Mental Health, Australia.,Department of Biomedical Sciences, Macquarie University, North Ryde, New South Wales, NSW, Australia.,KaRa Institute of Neurological Disease, Sydney, Macquarie Park, New South Wales, NSW, Australia.,School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Perth, WA, Australia.,Australian Alzheimer's Research Foundation, Nedlands, Western Australia, WA, Australia
| | | |
Collapse
|
309
|
Swinnen N, Vandenbulcke M, de Bruin ED, Akkerman R, Stubbs B, Firth J, Vancampfort D. The efficacy of exergaming in people with major neurocognitive disorder residing in long-term care facilities: a pilot randomized controlled trial. Alzheimers Res Ther 2021; 13:70. [PMID: 33785077 PMCID: PMC8008333 DOI: 10.1186/s13195-021-00806-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND It is currently unknown whether exergaming is efficacious in people with major neurocognitive disorder (MNCD) residing in long-term care facilities. This pilot randomized controlled trial (RCT) explored the efficacy of a stepping exergame program on gait speed, balance, mobility, reaction time, cognitive and neuropsychiatric outcomes, quality of life, and daily life functioning in people with MNCD residing in long-term care facilities. METHODS Participants were randomly assigned to 8 weeks, three times weekly, 15 min of exergaming versus watching preferred music videos. The exergame device consisted of a pressure-sensitive step training platform on which participants performed stepping movements to play the games. The device automatically adapted the training level to the participants' capabilities. The Short Physical Performance Battery (SPPB), step reaction time test (SRTT), Montréal Cognitive Assessment (MoCA), Neuropsychiatric Inventory (NPI), Cornell Scale for Depression in Dementia (CSDD), Dementia Quality of Life (DQoL), and Katz Activities of Daily Living (Katz ADL) were assessed at baseline and post-intervention. A Quade's non-parametric ANCOVA controlling for baseline values with post hoc Bonferroni correction (p < 0.00625) was used to analyze pre- and post-differences between the groups. Partial eta-squared (η2p) effect sizes were calculated. RESULTS Forty-five of 55 randomized inpatients with mild to moderate MNCD (Mini-Mental State Examination score = 17.2 ± 4.5; aged 70-91; 35 women) completed the study. The exergame group (n = 23) demonstrated improvements in gait speed (p < 0.001, η2p = 0.41), total SPPB (p < 0.001, η2p = 0.64), SRTT (p<0.001, η2p = 0.51), MoCA (p<0.001, η2p = 0.38), and reductions in CSDD (p<0.001, η2p = 0.43) compared to the control group (n = 22). There were no differences in NPI (p = 0.165, η2p = 0.05), DQoL (p = 0.012, η2p = 0.16), and ADL (p = 0.008, η2p = 0.16) post-intervention scores between the experimental and control group, albeit DQol and ADL measures showed large effect sizes in the exergame group. The mean attendance rate was 82.9% in the exergame group and 73.7% in the music control group. There were no study-related adverse events reported by the participants, nor observed by the research team. CONCLUSIONS The findings of this pilot RCT suggest that an individually adapted exergame training improves lower extremity functioning, cognitive functioning and step reaction time and symptoms of depression in inpatients with MNCD residing in long-term care facilities. TRIAL REGISTRATION ClinicalTrials.gov, NCT04436302.
Collapse
Affiliation(s)
- Nathalie Swinnen
- KU Leuven Department of Rehabilitation Sciences, Leuven, Belgium
- University Psychiatric Centre KU Leuven, Leuven, Kortenberg, Belgium
| | - Mathieu Vandenbulcke
- University Psychiatric Centre KU Leuven, Leuven, Kortenberg, Belgium
- KU Leuven Department of Neurosciences, Leuven, Belgium
| | - Eling D de Bruin
- Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zürich, Leopold-Ruzicka-Weg 4, 8093, Zürich, Switzerland.
- Division of Physiotherapy, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden.
| | | | - Brendon Stubbs
- South London and Maudsley NHS Foundation Trust, Denmark Hill, London, UK
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Joseph Firth
- Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- NICM Health Research Institute, Western Sydney University, Sydney, New South Wales, Australia
| | - Davy Vancampfort
- KU Leuven Department of Rehabilitation Sciences, Leuven, Belgium
- University Psychiatric Centre KU Leuven, Leuven, Kortenberg, Belgium
| |
Collapse
|
310
|
Eshraghi M, Adlimoghaddam A, Mahmoodzadeh A, Sharifzad F, Yasavoli-Sharahi H, Lorzadeh S, Albensi BC, Ghavami S. Alzheimer's Disease Pathogenesis: Role of Autophagy and Mitophagy Focusing in Microglia. Int J Mol Sci 2021; 22:3330. [PMID: 33805142 PMCID: PMC8036323 DOI: 10.3390/ijms22073330] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/14/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurological disorder, and currently, there is no cure for it. Several pathologic alterations have been described in the brain of AD patients, but the ultimate causative mechanisms of AD are still elusive. The classic hallmarks of AD, including amyloid plaques (Aβ) and tau tangles (tau), are the most studied features of AD. Unfortunately, all the efforts targeting these pathologies have failed to show the desired efficacy in AD patients so far. Neuroinflammation and impaired autophagy are two other main known pathologies in AD. It has been reported that these pathologies exist in AD brain long before the emergence of any clinical manifestation of AD. Microglia are the main inflammatory cells in the brain and are considered by many researchers as the next hope for finding a viable therapeutic target in AD. Interestingly, it appears that the autophagy and mitophagy are also changed in these cells in AD. Inside the cells, autophagy and inflammation interact in a bidirectional manner. In the current review, we briefly discussed an overview on autophagy and mitophagy in AD and then provided a comprehensive discussion on the role of these pathways in microglia and their involvement in AD pathogenesis.
Collapse
Affiliation(s)
- Mehdi Eshraghi
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA;
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Aida Adlimoghaddam
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada; (A.A.); (B.C.A.)
| | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Farzaneh Sharifzad
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (F.S.); (H.Y.-S.)
| | - Hamed Yasavoli-Sharahi
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (F.S.); (H.Y.-S.)
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Benedict C. Albensi
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada; (A.A.); (B.C.A.)
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine, Katowice School of Technology, 40-555 Katowice, Poland
| |
Collapse
|
311
|
Nagel AK, Loetscher T, Smith AE, Keage HA. What do the public really know about dementia and its risk factors? DEMENTIA 2021; 20:2424-2440. [PMID: 33745347 DOI: 10.1177/1471301221997301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Dementia has become a major public health concern globally. With no cure available and strong evidence for modifiable risk factors, it is imperative that the public are knowledgeable about dementia and to reduce their risk. The aim of this study was to measure the knowledge of dementia and its risk factors in the Australian public, as well as the number and type of information sources used. An online survey promoted through various social media platforms was completed by 596 Australian adults aged 18-78 years (59% aged 18-44 years; 78% female). Eighty-eight percent of respondents were able to recognise dementia from a vignette, more so from a moderate than from a mild symptom vignette. Only 19% of respondents had a good understanding of dementia, that is describing impairment in both cognition and function. Ninety-five percent of respondents recognised that genetics and old age contributed to a person getting dementia. However, respondents had poor knowledge of empirically supported modifiable risk factors, with most viewed as likely contributors to dementia at chance levels (50%) or below. Respondents reported using informal information sources more often than formal information sources to learn about dementia. The public appear to be able to recognise the symptoms of dementia but lack understanding of how dementia impacts both a person's cognitive functioning and ability to perform everyday tasks. Furthermore, the public remain largely unaware of empirically supported modifiable risk factors that contribute to the development of dementia. It is imperative that the public are educated on how to access and evaluate dementia-related information sources in order to increase their knowledge and understanding of dementia.
Collapse
Affiliation(s)
- Alana K Nagel
- Justice and Society, Psychology, Social Work and Social Policy, 153411University of South Australia, Adelaide, Australia
| | - Tobias Loetscher
- Justice and Society, Psychology, Social Work and Social Policy, University of South Australia, Adelaide, Australia
| | - Ashleigh E Smith
- School of Health Sciences, Alliance for Research in Exercise Nutrition and Activity (ARENA), Allied Health and Human Performance, University of South Australia, Adelaide, Australia
| | - Hannah Ad Keage
- Justice and Society, Psychology, Social Work and Social Policy, University of South Australia, Adelaide, Australia
| |
Collapse
|
312
|
Zingel R, Bohlken J, Kostev K. Association Between Inflammatory Bowel Disease and Dementia: A Retrospective Cohort Study. J Alzheimers Dis 2021; 80:1471-1478. [PMID: 33720902 DOI: 10.3233/jad-210103] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The critical role of inflammatory processes in the pathogenesis of dementia has recently been established. OBJECTIVE The aim of this study was to investigate the association between inflammatory bowel disease (IBD) and dementia risk in patients followed in general practices in Germany. METHODS This study included patients aged over 60 with an initial diagnosis of IBD (Crohn's Disease (CD), ulcerative colitis (UC)) who were followed in 1,159 German general practices between January 1995 and December 2014. IBD patients were matched to healthy patients using propensity scores based on age, gender, index year, insurance type and comorbidities. Kaplan-Meier curves were used to study the development of dementia in patients with or without IBD within up to 15 years of the index date. Cox proportional hazard regression models were used to estimate the relationship between IBD and dementia. RESULTS The study included 3,850 patients with and 3,850 patients without IBD and revealed a higher cumulative incidence of dementia in IBD patients than in non-IBD patients after the follow-up period. The cumulative incidence of dementia differed within IBD subtypes; it was significantly higher in UC patients than in CD patients. Cox proportional hazard models showed that IBD is associated with a 1.22-fold increase in the risk (95% CI: 1,07-1,39) of developing dementia. UC patients had a 1.25-fold higher risk of developing dementia (95% CI: 1.07-1.46). CD is not significantly associated with an increased risk of dementia (HR: 1.17, 95% CI: 0.93-1.47). CONCLUSION A positive association between IBD and dementia was found in patients followed in general practices in Germany.
Collapse
Affiliation(s)
| | - Jens Bohlken
- Institute for Social Medicine, Occupational Medicine, and Public Health (ISAP) of the Medical Faculty at the University of Leipzig, Leipzig, Germany
| | | |
Collapse
|
313
|
Jang YJ, Kang C, Myung W, Lim SW, Moon YK, Kim H, Kim DK. Additive interaction of mid- to late-life depression and cerebrovascular disease on the risk of dementia: a nationwide population-based cohort study. ALZHEIMERS RESEARCH & THERAPY 2021; 13:61. [PMID: 33726788 PMCID: PMC7968260 DOI: 10.1186/s13195-021-00800-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 03/02/2021] [Indexed: 12/26/2022]
Abstract
Background Dementia is a progressive neurocognitive disease with a substantial social burden. No apparent breakthroughs in treatment options have emerged so far; thus, disease prevention is essential for at-risk populations. Depression and cerebrovascular disease (CVD) are independent risk factors for dementia, but no studies have examined their interaction effect on dementia risk. This study aimed to identify the association of depression and CVD with the risk of dementia and evaluate whether dementia risk among patients with comorbid depression and CVD is higher than the sum of the individual risk due to each condition. Methods A population-based cohort study was conducted to analyze the Korean National Health Insurance Service-National Sample Cohort data of all individuals over 50 years of age. Individuals who had not been diagnosed with dementia at baseline were included and followed up from January 1, 2005, to December 31, 2013. A time-varying Cox proportional hazard regression model adjusted for potential confounding factors was used for the analysis. The interaction between depression and CVD was estimated based on the attributable proportion (AP), relative excess risk due to interaction (RERI), synergy index (SI), and multiplicative-scale interaction. Results A total of 242,237 participants were included in the analytical sample, of which 12,735 (5.3%) developed dementia. Compared to that for participants without depression or CVD, the adjusted hazard ratio for the incidence of dementia for those with depression alone was 2.35 (95% confidence interval [CI] 2.21–2.49), CVD alone was 3.25 (95% CI 3.11–3.39), and comorbid depression and CVD was 5.02 (95% CI 4.66–5.42). The additive interaction between depression and CVD was statistically significant (AP—0.08, 95% CI 0.01–0.16; RERI—0.42, 95% CI 0.03–0.82; SI—1.12, 95% CI 1.01–1.24). The multiplicative interaction was significant too, but the effect was negative (0.66, 95% CI 0.60–0.73). Conclusions In this population-based nationwide cohort with long-term follow-up, depression and CVD were associated with an increased risk of dementia, and their coexistence additively increased dementia risk more than the sum of the individual risks.
Collapse
Affiliation(s)
- Yoo Jin Jang
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Cinoo Kang
- Department of Public Health Science, Graduate School of Public Health, Seoul National University, Seoul, South Korea
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Shinn-Won Lim
- SAIHST, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Young Kyung Moon
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Ho Kim
- Department of Public Health Science, Graduate School of Public Health, Seoul National University, Seoul, South Korea. .,Institute of Health and Environment, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, South Korea.
| | - Doh Kwan Kim
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
| |
Collapse
|
314
|
Pellegrini C, Pirazzini C, Sala C, Sambati L, Yusipov I, Kalyakulina A, Ravaioli F, Kwiatkowska KM, Durso DF, Ivanchenko M, Monti D, Lodi R, Franceschi C, Cortelli P, Garagnani P, Bacalini MG. A Meta-Analysis of Brain DNA Methylation Across Sex, Age, and Alzheimer's Disease Points for Accelerated Epigenetic Aging in Neurodegeneration. Front Aging Neurosci 2021; 13:639428. [PMID: 33790779 PMCID: PMC8006465 DOI: 10.3389/fnagi.2021.639428] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/05/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by specific alterations of brain DNA methylation (DNAm) patterns. Age and sex, two major risk factors for AD, are also known to largely affect the epigenetic profiles in brain, but their contribution to AD-associated DNAm changes has been poorly investigated. In this study we considered publicly available DNAm datasets of four brain regions (temporal, frontal, entorhinal cortex, and cerebellum) from healthy adult subjects and AD patients, and performed a meta-analysis to identify sex-, age-, and AD-associated epigenetic profiles. In one of these datasets it was also possible to distinguish 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) profiles. We showed that DNAm differences between males and females tend to be shared between the four brain regions, while aging differently affects cortical regions compared to cerebellum. We found that the proportion of sex-dependent probes whose methylation is modified also during aging is higher than expected, but that differences between males and females tend to be maintained, with only a few probes showing age-by-sex interaction. We did not find significant overlaps between AD- and sex-associated probes, nor disease-by-sex interaction effects. On the contrary, we found that AD-related epigenetic modifications are significantly enriched in probes whose DNAm varies with age and that there is a high concordance between the direction of changes (hyper or hypo-methylation) in aging and AD, supporting accelerated epigenetic aging in the disease. In summary, our results suggest that age-associated DNAm patterns concur to the epigenetic deregulation observed in AD, providing new insights on how advanced age enables neurodegeneration.
Collapse
Affiliation(s)
- Camilla Pellegrini
- Istituto di Ricovero e Cura a Carattere Scientifico Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Chiara Pirazzini
- Istituto di Ricovero e Cura a Carattere Scientifico Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Claudia Sala
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Luisa Sambati
- Istituto di Ricovero e Cura a Carattere Scientifico Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Igor Yusipov
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhny Novgorod, Russia
| | - Alena Kalyakulina
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhny Novgorod, Russia
| | - Francesco Ravaioli
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Katarzyna M. Kwiatkowska
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Danielle F. Durso
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Mikhail Ivanchenko
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhny Novgorod, Russia
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Raffaele Lodi
- Istituto di Ricovero e Cura a Carattere Scientifico Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhny Novgorod, Russia
| | - Pietro Cortelli
- Istituto di Ricovero e Cura a Carattere Scientifico Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Department of Laboratory Medicine, Clinical Chemistry, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Applied Biomedical Research Center, Policlinico S.Orsola-Malpighi Polyclinic, Bologna, Italy
- National Research Council of Italy Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza,” Unit of Bologna, Bologna, Italy
| | - Maria Giulia Bacalini
- Istituto di Ricovero e Cura a Carattere Scientifico Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
315
|
Baksi S, Pradhan A. Thyroid hormone: sex-dependent role in nervous system regulation and disease. Biol Sex Differ 2021; 12:25. [PMID: 33685490 PMCID: PMC7971120 DOI: 10.1186/s13293-021-00367-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/11/2021] [Indexed: 12/13/2022] Open
Abstract
Thyroid hormone (TH) regulates many functions including metabolism, cell differentiation, and nervous system development. Alteration of thyroid hormone level in the body can lead to nervous system-related problems linked to cognition, visual attention, visual processing, motor skills, language, and memory skills. TH has also been associated with neuropsychiatric disorders including schizophrenia, bipolar disorder, anxiety, and depression. Males and females display sex-specific differences in neuronal signaling. Steroid hormones including testosterone and estrogen are considered to be the prime regulators for programing the neuronal signaling in a male- and female-specific manner. However, other than steroid hormones, TH could also be one of the key signaling molecules to regulate different brain signaling in a male- and female-specific manner. Thyroid-related diseases and neurological diseases show sex-specific incidence; however, the molecular mechanisms behind this are not clear. Hence, it will be very beneficial to understand how TH acts in male and female brains and what are the critical genes and signaling networks. In this review, we have highlighted the role of TH in nervous system regulation and disease outcome and given special emphasis on its sex-specific role in male and female brains. A network model is also presented that provides critical information on TH-regulated genes, signaling, and disease.
Collapse
Affiliation(s)
- Shounak Baksi
- Causality Biomodels, Kerala Technology Innovation Zone, Cochin, 683503, India
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, SE-701 82, Örebro, Sweden.
| |
Collapse
|
316
|
Gu Y, Guo J, Moshfegh AJ. Race/ethnicity and gender modify the association between diet and cognition in U.S. older adults: National Health and Nutrition Examination Survey 2011-2014. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12128. [PMID: 33614896 PMCID: PMC7882526 DOI: 10.1002/trc2.12128] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/20/2020] [Accepted: 11/25/2020] [Indexed: 11/12/2022]
Abstract
INTRODUCTION It is unclear whether the association between Mediterranean-type Diet (MeDi) and cognition is similar across different racial/ethnic groups or between women and men. METHODS The current study included 2435 participants (≥60 years of age) of the National Health and Nutrition Examination Survey (NHANES) 2011-2014. Linear regression models were applied to examine the association between diet and cognition, adjusted for multiple demographic variables. Interaction between MeDi and race/ethnicity (non-Hispanic White, non-Hispanic Black, Hispanic, other), and between MeDi and gender, were examined, followed by stratified analyses. RESULTS One score increase in MeDi was associated with 0.039 (95%confidence interval [CI] = 0.016-0.062) higher global cognitive z-score. Significant associations between MeDi and global cognition and between MeDi and immediate recall were found in the non-Hispanic Whites only (P-interaction = 0.057 and 0.059, respectively). MeDi was associated with increased score of animal fluency score in men but not in women (P-interaction = 0.082). DISCUSSION The positive association between MeDi and cognition might be dependent on race/ethnicity and gender.
Collapse
Affiliation(s)
- Yian Gu
- Taub Institute for Research in Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyVagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Department of EpidemiologyJoseph P. Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Jing Guo
- Taub Institute for Research in Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Alanna J Moshfegh
- Beltsville Human Nutrition Research CenterAgricultural Research ServiceUSDABeltsvilleMarylandUSA
| |
Collapse
|
317
|
Potential of Caffeine in Alzheimer's Disease-A Review of Experimental Studies. Nutrients 2021; 13:nu13020537. [PMID: 33562156 PMCID: PMC7915779 DOI: 10.3390/nu13020537] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia leading to progressive memory loss and cognitive impairment. Considering that pharmacological treatment options for AD are few and not satisfactory, increasing attention is being paid to dietary components that may affect the development of the disease. Such a dietary component may be caffeine contained in coffee, tea or energy drinks. Although epidemiological data suggest that caffeine intake may counteract the development of cognitive impairment, results of those studies are not conclusive. The aim of the present study is to review the existing experimental studies on the efficacy of caffeine against AD and AD-related cognitive impairment, focusing on the proposed protective mechanisms of action. In conclusion, the reports of studies on experimental AD models generally supported the notion that caffeine may exert some beneficial effects in AD. However, further studies are necessary to elucidate the role of caffeine in the effects of its sources on cognition and possibly AD risk.
Collapse
|
318
|
Chronic adolescent stress causes sustained impairment of cognitive flexibility and hippocampal synaptic strength in female rats. Neurobiol Stress 2021; 14:100303. [PMID: 33614865 PMCID: PMC7876631 DOI: 10.1016/j.ynstr.2021.100303] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/13/2021] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Females that experience chronic stress during development, particularly adolescence, are the most vulnerable group to stress-induced disease. While considerable attention has been devoted to stress-induced manifestation of anxiety, depression, and PTSD, evidence indicates that a history of chronic stress is also a risk factor for cognitive decline and dementia - with females again in a higher risk group. This interplay between sex and stress history indicates specific mechanisms drive neural dysfunction across the lifespan. The presence of sex and stress steroid receptors in the hippocampus provides a point of influence for these variables to drive changes in cognitive function. Here, we used a rodent model of chronic adolescent stress (CAS) to determine the extent to which CAS modifies glutamatergic signaling resulting in cognitive dysfunction. Male and female Wistar rats born in-house remained non-stressed (NS), unmanipulated aside from standard cage cleaning, or were exposed to either physical restraint (60 min) or social defeat (CAS) each day (6 trials each), along with social isolation, throughout the adolescent period (PND 35-47). Cognition was assessed in adult (PND 80-130) male and female rats (n = 10-12) using the Barnes Maze task and the Attention Set-Shift task. Whole hippocampi were extracted from a second cohort of male and female rats (NS and CAS; n = 9-10) and processed for RNA sequencing. Brain tissue from the first cohort (n = 6) was processed for density of glutamatergic synaptic markers (GluA1, NMDA1a, and synaptophysin) or whole-cell patch clamping (n = 4) to determine glutamatergic activity in the hippocampus. Females with a history of chronic stress had shorter latencies to locate the goal box than NS controls during acquisition learning but showed an increased latency to locate the new goal box during reversal learning. This reversal deficit persisted across domains as females with a history of stress required more trials to reach criterion during the reversal phases of the Attention Set-Shift task compared to controls. Ovariectomy resulted in greater performance variability overall during reversal learning with CAS females showing worse performance. Males showed no effects of CAS history on learning or memory performance. Bioinformatic prediction using gene ontology categorization indicated that in females, postsynaptic membrane gene clusters, specifically genes related to glutamatergic synapse remodeling, were enriched with a history of stress. Structural analysis indicated that CAS did not alter glutamate receptor density in females. However, functionally, CAS females had a decreased AMPA/NMDA-dependent current ratio compared to controls indicating a weakening in synaptic strength in the hippocampus. Males showed only a slight change in density of NMDA1a labeling in the CA3 region with a history of stress. The data observed here suggest that females are at risk for impaired cognitive flexibility following a history of adolescent stress, possibly driven by changes in glutamatergic signaling.
Collapse
|
319
|
Kakinen A, Javed I, Davis TP, Ke PC. In vitro and in vivo models for anti-amyloidosis nanomedicines. NANOSCALE HORIZONS 2021; 6:95-119. [PMID: 33438715 DOI: 10.1039/d0nh00548g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Amyloid diseases are global epidemics characterized by the accumulative deposits of cross-beta amyloid fibrils and plaques. Despite decades of intensive research, few solutions are available for the diagnosis, treatment, and prevention of these debilitating diseases. Since the early work on the interaction of human β2-microglobulin and nanoparticles by Linse et al. in 2007, the field of amyloidosis inhibition has gradually evolved into a new frontier in nanomedicine offering numerous interdisciplinary research opportunities, especially for materials, chemistry and biophysics. In this review we summarise, for the first time, the in vitro and in vivo models employed thus far in the field of anti-amyloidosis nanomedicines. Based on this systematic summary, we bring forth the notion that, due to the complex and often overlapping physiopathologies of amyloid diseases, there is a crucial need for the appropriate use of in vitro and in vivo models for validating novel anti-amyloidosis nanomedicines, and there is a crucial need for the development of new animal models that reflect the behavioural, symptomatic and cross-talk hallmarks of amyloid diseases such as Alzheimer's (AD), Parkinson's (PD) diseases and type 2 diabetes (T2DM).
Collapse
Affiliation(s)
- Aleksandr Kakinen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | |
Collapse
|
320
|
Bloomberg M, Dugravot A, Dumurgier J, Kivimaki M, Fayosse A, Steptoe A, Britton A, Singh-Manoux A, Sabia S. Sex differences and the role of education in cognitive ageing: analysis of two UK-based prospective cohort studies. Lancet Public Health 2021; 6:e106-e115. [PMID: 33516287 PMCID: PMC8141610 DOI: 10.1016/s2468-2667(20)30258-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous studies have shown an excess risk of Alzheimer's disease and related dementias among women. Education is thought to have a causal association with dementia onset. We aimed to investigate the role of education in influencing sex differences in cognitive ageing. METHODS We analysed data from two prospective cohort studies in the UK; the English Longitudinal Study of Ageing (ELSA) and the Whitehall II study, to assess sex differences in cognitive performance and cognitive decline by birth cohort (birth year 1930-38, 1939-45, or 1946-55), before and after adjustment for education, and by high and low education level. Memory was assessed using immediate recall, for which data were available from all waves of the ELSA (2002-14) and Whitehall II (1997-2015) studies. Fluency was assessed using a semantic fluency test based on an animal naming task, with data available from all waves of the Whitehall II study and waves one to five (2002-10) and wave seven (2014) of the ELSA study. Cognitive scores were standardised separately in each study based on the mean and SD of the corresponding test among participants aged 50-59 years with secondary education. FINDINGS 15 924 participants were included from the two studies. In pooled analyses, women had better memory scores than men in all birth cohorts, irrespective of adjustment for education (eg, at age 60 years, birth cohort 1930-38, mean difference between sexes [male scores minus female scores] -0·25 SDs [95% CI -0·32 to -0·19] after adjustment for education), and in both education level groups. Memory decline was faster in men than in women (at age 60 years, birth cohort 1946-55, mean difference in 13-year change -0·15 SDs [-0·20 to -0·09]; after adjustment for education -0·14 SDs [-0·20 to -0·08]). Men had better fluency scores than women in earlier birth cohorts and in the low education group (at age 60 years, birth cohort 1930-38, mean difference 0·20 SDs [95% CI 0·05 to 0·36]); but women had better fluency scores than men in later birth cohorts and in the high education group (at age 60 years, birth cohort 1946-55, mean difference -0·17 SDs [-0·24 to -0·10]). No sex differences were observed for fluency decline. INTERPRETATION Our findings suggest that decreasing disparities between sexes in education, due to secular increases in educational opportunities, could attenuate sex differences in dementia risk and cognitive decline in the future. FUNDING National Institute on Aging, National Institutes of Health; UK Medical Research Council; British Heart Foundation; and National Institute for Health Research.
Collapse
Affiliation(s)
- Mikaela Bloomberg
- Department of Epidemiology and Public Health, University College London, London, UK.
| | - Aline Dugravot
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
| | - Julien Dumurgier
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France; Cognitive Neurology Centre, Saint-Louis Lariboisière Fernand-Widal Hospital, AP-HP, Université de Paris, Paris, France
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Aurore Fayosse
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
| | - Andrew Steptoe
- Department of Behavioural Science and Health, University College London, London, UK
| | - Annie Britton
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Archana Singh-Manoux
- Department of Epidemiology and Public Health, University College London, London, UK; Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
| | - Séverine Sabia
- Department of Epidemiology and Public Health, University College London, London, UK; Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
| |
Collapse
|
321
|
Calsolaro V, Bottari M, Coppini G, Lemmi B, Monzani F. Endocrine dysfunction and cognitive impairment. Minerva Endocrinol (Torino) 2021; 46:335-349. [PMID: 33435644 DOI: 10.23736/s2724-6507.20.03295-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dementia is a highly prevalent chronic disease among the older population, affecting more than 50 million people worldwide and representing a huge healthcare, social and economic burden. Dementia, and in particular Alzheimer's disease, prevalence is expected to raise within the next few years. Unfortunately, no disease-modifying therapies are available so far, despite a plethora of clinical trials targeting the hallmarks of Alzheimer's disease. Given these premises, it appears crucial to address not only the neuropathological correlates of the disease, but also the modifiable risk factors. Among them, evidence suggest a role of the endocrine system not only in the brain development, but also in the maintenance of its health, having neurotrophic, antioxidant and metabolic functions crucial for the cognitive abilities. This review focuses on the evidence evaluating the impact of the endocrine systems, in particular thyroid function, insulin resistance, parathyroid hormone, vitamin D and sexual hormones on cognitive status. Results from epidemiological, preclinical and some clinical studies demonstrated the link between thyroid, parathyroid hormone and vitamin D and cognitive status, between diabetes, and insulin resistance in particular, and dementia, between sexual and adrenal hormones, particularly estrogen variation at menopause, and cognitive decline. The growing interest on the modifiable risks factors of cognitive decline increased the knowledge about the complex interplay of endocrine systems and cognition, highlighting the need and the usefulness of a multidisciplinary approach to the prevention of a complex and devastating disease.
Collapse
Affiliation(s)
- Valeria Calsolaro
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Marina Bottari
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Giulia Coppini
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Bianca Lemmi
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Fabio Monzani
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy -
| |
Collapse
|
322
|
Hong S, Han K, Park CY. The insulin resistance by triglyceride glucose index and risk for dementia: population-based study. ALZHEIMERS RESEARCH & THERAPY 2021; 13:9. [PMID: 33402193 PMCID: PMC7786939 DOI: 10.1186/s13195-020-00758-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 12/23/2020] [Indexed: 12/19/2022]
Abstract
Background Insulin resistance is suggested to have negative effects on cognition; however, results from large population studies are lacking. In this study, the potential relationships between the triglyceride glucose (TyG) index, a simple surrogate marker of insulin resistance, and dementia were evaluated using a large-scale population dataset. Methods This was a retrospective, observational, cohort study using data from the National Health Information Database from 2009 to 2015 and included 5,586,048 participants 40 years age or older. The TyG index was used as a measure of insulin resistance, and participants were divided into quartiles based on TyG index. The incidence of dementia was assessed using hazard ratios (HRs) estimated with Cox proportional hazard modeling. Results During a median follow-up of 7.21 years, dementia was diagnosed in 142,714 (2.55%) participants. Alzheimer’s disease (AD) and vascular dementia (VD) were diagnosed in 74.3% and 12.5% of the participants. Multivariate-adjusted HRs for patients in the TyG index 4th quartile were higher for dementia (HRs = 1.14; 95% confidence interval [CI] 1.12–1.16), AD (HRs = 1.12; 95% CI 1.09–1.14), and VD (HRs = 1.18; 95% CI 1.12–1.23) compared with the 1st quartile of TyG index; however, this had a small effect size (Cohen’s d = 0.10, 0.08, and 0.13, respectively). These effects were independent of age, sex, smoking status, physical activity, body mass index, systolic blood pressure, and total cholesterol. Conclusion In this large population study, TyG index was associated with an increased risk of dementia, including AD and VD, that was independent of traditional cardiovascular risk factors, although the effect size of the TyG index was small. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-020-00758-4.
Collapse
Affiliation(s)
- Sangmo Hong
- Department of Internal Medicine, Hanyang University, College of Medicine, 222, Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, 369, Sangdo-Ro, Dongjak-Gu, Seoul, Republic of Korea
| | - Cheol-Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29, Saemunan-ro, Jongno-gu, Seoul, 03181, Republic of Korea.
| |
Collapse
|
323
|
Pashkovskiy V, Safonova N, Semenova N. Epidemiological aspects of vascular dementia in the Northwestern Federal District in 2010—2019. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:84-90. [DOI: 10.17116/jnevro202112112184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
324
|
Leszek J, Mikhaylenko EV, Belousov DM, Koutsouraki E, Szczechowiak K, Kobusiak-Prokopowicz M, Mysiak A, Diniz BS, Somasundaram SG, Kirkland CE, Aliev G. The Links between Cardiovascular Diseases and Alzheimer's Disease. Curr Neuropharmacol 2021; 19:152-169. [PMID: 32727331 PMCID: PMC8033981 DOI: 10.2174/1570159x18666200729093724] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/02/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
The root cause of non-inherited Alzheimer's disease (AD) remains unknown despite hundreds of research studies performed to attempt to solve this problem. Since proper prophylaxis remains the best strategy, many scientists have studied the risk factors that may affect AD development. There is robust evidence supporting the hypothesis that cardiovascular diseases (CVD) may contribute to AD progression, as the diseases often coexist. Therefore, a lack of well-defined diagnostic criteria makes studying the relationship between AD and CVD complicated. Additionally, inflammation accompanies the pathogenesis of AD and CVD, and is not only a consequence but also implicated as a significant contributor to the course of the diseases. Of note, АроЕε4 is found to be one of the major risk factors affecting both the cardiovascular and nervous systems. According to genome wide association and epidemiological studies, numerous common risk factors have been associated with the development of AD-related pathology. Furthermore, the risk of developing AD and CVDs appears to be increased by a wide range of conditions and lifestyle factors: hypertension, dyslipidemia, hypercholesterolemia, hyperhomocysteinemia, gut/oral microbiota, physical activity, and diet. This review summarizes the literature and provides possible mechanistic links between CVDs and AD.
Collapse
Affiliation(s)
- Jerzy Leszek
- Address correspondence to these authors at the Department of Psychiatry, Wrocław Medical University, Ul. Pasteura 10, 50-367, Wroclaw, Poland;, E-mail: and GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA; Tel: +1-210-442-8625 or +1-440-263-7461; E-mails: ,
| | | | | | | | | | | | | | | | | | | | - Gjumrakch Aliev
- Address correspondence to these authors at the Department of Psychiatry, Wrocław Medical University, Ul. Pasteura 10, 50-367, Wroclaw, Poland;, E-mail: and GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA; Tel: +1-210-442-8625 or +1-440-263-7461; E-mails: ,
| |
Collapse
|
325
|
Huang LC, Hsieh SW, Tsai CC, Chen CH, Yang YH. The Role of Cilostazol and Inflammation in Cognitive Impairment After Ischemic Stroke. Am J Alzheimers Dis Other Demen 2021; 36:15333175211016185. [PMID: 34008421 PMCID: PMC10624089 DOI: 10.1177/15333175211016185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE The aim of this study is to examine the potential effect of cilostazol and inflammation on cognitive impairment after stroke in an Asian population. METHODS Forty-five patients with cognitive impairment after ischemic stroke using cilostazol were enrolled as the study group and 45 patients using aspirin or clopidogrel were enrolled as the control group. Neuropsychiatric assessments were administered at the start of the study and after 6 months. Multiple logistic regression analysis was used to estimate the association between the cognitive change and cilostazol use. Macrophage polarization were assessed using flow cytometry in 7 patients. RESULTS There were a significantly higher number of patients with peripheral arterial occlusive disease in the cilostazol group. No significant differences were observed in the cognitive change between the cilostazol and control groups. M1 macrophage subset increment were observed in the patient having a declined cognitive change. CONCLUSION Cilostazol did not make a significant difference in cognitive change after ischemic stroke. M1 macrophage subset increment may indicate post stroke cognitive decline. Due to limited number of subjects, these findings should be examined further in large-scale randomized clinical trials.
Collapse
Affiliation(s)
- Ling-Chun Huang
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan
| | - Sun-Wung Hsieh
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan
- Department of Neurology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Chia-Chan Tsai
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan
| | - Chun-Hung Chen
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan
- Department of Neurology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Yuan-Han Yang
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan
- Department of Master’s Program in Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City, Taiwan
| |
Collapse
|
326
|
Hao X, Wang A, Li C, Shao L, Li Y, Yang P. Genetic association of BIN1 and GAB2 in Alzheimer's disease: A meta-analysis and systematic review. Geriatr Gerontol Int 2020; 21:185-191. [PMID: 33331110 PMCID: PMC7898709 DOI: 10.1111/ggi.14109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/06/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022]
Abstract
Aim Heredity plays an important role in the pathogenesis of Alzheimer's disease (AD) especially for single‐nucleotide polymorphism (SNPs) of susceptible genes, which is one of the significant factors in the pathogenesis of AD. The SNPs of BIN1 rs744373, BIN1 rs7561528 and GAB2 rs2373115 are associated with AD in Asian and white people. Methods We included 34 studies with a total of 38 291 patients with AD and 55 538 controls of diverse races from four main databases. We used meta‐analysis to obtain I2‐values and odds ratios of five genetic models in three SNPs. We carried out analysis of sensitivity, subgroup, publication bias and linkage disequilibrium test. Results The forest plots showed the odds ratio value of the three SNPs was >1 in white individuals, but not Asian individuals, in their genetic model. The funnel plot was symmetrical, and the D'‐value was 0.986 between rs744373 and rs7561528. Conclusions BIN1 rs744373, BIN1 rs7561528 and GAB2 rs2373115 are pathogenicity sites for AD in white people, and also rs7561528 belongs to a risk site in Asian people. The rs7561528 and rs744373 SNPs have strong linkage disequilibrium in Chinese people. In addition, apolipoprotein E ε4 status promotes them to result in the pathogenesis of AD. Geriatr Gerontol Int 2021; 21: 185–191.
Collapse
Affiliation(s)
- Xiaoyan Hao
- Department of Neurology, Ningxia Medical University, Yinchuan, China
| | - Aijun Wang
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Chong Li
- Department of Neurology, Ningxia Medical University, Yinchuan, China
| | - Lufei Shao
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yi Li
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ping Yang
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
327
|
Abd-Elrahman KS, Albaker A, de Souza JM, Ribeiro FM, Schlossmacher MG, Tiberi M, Hamilton A, Ferguson SSG. Aβ oligomers induce pathophysiological mGluR5 signaling in Alzheimer's disease model mice in a sex-selective manner. Sci Signal 2020; 13:13/662/eabd2494. [PMID: 33323410 DOI: 10.1126/scisignal.abd2494] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The prevalence, presentation, and progression of Alzheimer's disease (AD) differ between men and women, although β-amyloid (Aβ) deposition is a pathological hallmark of AD in both sexes. Aβ-induced activation of the neuronal glutamate receptor mGluR5 is linked to AD progression. However, we found that mGluR5 exhibits distinct sex-dependent profiles. Specifically, mGluR5 isolated from male mouse cortical and hippocampal tissues bound with high affinity to Aβ oligomers, whereas mGluR5 from female mice exhibited no such affinity. This sex-selective Aβ-mGluR5 interaction did not appear to depend on estrogen, but rather Aβ interaction with cellular prion protein (PrPC), which was detected only in male mouse brain homogenates. The ternary complex between mGluR5, Aβ oligomers, and PrPC was essential to elicit mGluR5-dependent pathological suppression of autophagy in primary neuronal cultures. Pharmacological inhibition of mGluR5 reactivated autophagy, mitigated Aβ pathology, and reversed cognitive decline in male APPswe/PS1ΔE9 mice, but not in their female counterparts. Aβ oligomers also bound with high affinity to human mGluR5 isolated from postmortem donor male cortical brain tissue, but not that from female samples, suggesting that this mechanism may be relevant to patients. Our findings indicate that mGluR5 does not contribute to Aβ pathology in females, highlighting the complexity of mGluR5 pharmacology and Aβ signaling that supports the need for sex-specific stratification in clinical trials assessing AD therapeutics.
Collapse
Affiliation(s)
- Khaled S Abd-Elrahman
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Awatif Albaker
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 12371, Saudi Arabia
| | - Jessica M de Souza
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Biochemistry and Immunology, ICB, Universidade Federalde Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Fabiola M Ribeiro
- Department of Biochemistry and Immunology, ICB, Universidade Federalde Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Michael G Schlossmacher
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, K1H 8L6, Canada
| | - Mario Tiberi
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, K1H 8L6, Canada.,Department of Psychiatry, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Alison Hamilton
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Stephen S G Ferguson
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
328
|
Rahman SO, Hussain S, Alzahrani A, Akhtar M, Najmi AK. Effect of statins on amyloidosis in the rodent models of Alzheimer's disease: Evidence from the preclinical meta-analysis. Brain Res 2020; 1749:147115. [PMID: 32918868 DOI: 10.1016/j.brainres.2020.147115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Previous studies have shown contrasting results in determining efficacy of statins against amyloid beta accumulation. The aim of this study was to assess the impact of statin in AD. METHOD We searched PubMed and Embase for relevant preclinical studies. A meta-analysis of the statin's efficacy on amyloidosis and cognitive impairment was performed. Also, stratified analysis was performed on several covariates including the type of statin used, gender and age of rodents and duration of statin therapy, to account for the reported heterogeneity in the results obtained. The study protocol was registered in PROSPERO (CRD42018102557). RESULT 17 studies including 22 comparisons, containing a sample size of 446 rodents, participated in the meta-analysis of statin's effect on overall Aβ deposition. Although the effect of statin on overall Aβ deposition was found to be protective (p < 0.00001) but as we categorized the efficacy of statin on different Aβ species (soluble and insoluble Aβ40/42) and Aβ plaque load, we found that significance in the protection decreased. A stratified meta-analysis demonstrated a significant role in the duration of statin supplements and rodent's age on the heterogeneity of the results. Statin administered to rodents for the longest duration (>6 months) and younger rodents (<6 months of age) demonstrated significant efficacy of statin on Aβ deposition. CONCLUSION Statin showed reduction in Aβ level but stratified analysis revealed that this effect of statin was dependent on rodent's age and duration of the treatment.
Collapse
Affiliation(s)
- Syed Obaidur Rahman
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Salman Hussain
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abdulaziz Alzahrani
- Department of Pharmacology, College of Clinical Pharmacy, Al Baha University, Al Baha, Saudi Arabia
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
329
|
Biechele G, Franzmeier N, Blume T, Ewers M, Luque JM, Eckenweber F, Sacher C, Beyer L, Ruch-Rubinstein F, Lindner S, Gildehaus FJ, von Ungern-Sternberg B, Cumming P, Bartenstein P, Rominger A, Höglinger GU, Herms J, Brendel M. Glial activation is moderated by sex in response to amyloidosis but not to tau pathology in mouse models of neurodegenerative diseases. J Neuroinflammation 2020; 17:374. [PMID: 33317543 PMCID: PMC7737385 DOI: 10.1186/s12974-020-02046-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Background In vivo assessment of neuroinflammation by 18-kDa translocator protein positron-emission-tomography (TSPO-PET) ligands receives growing interest in preclinical and clinical research of neurodegenerative disorders. Higher TSPO-PET binding as a surrogate for microglial activation in females has been reported for cognitively normal humans, but such effects have not yet been evaluated in rodent models of neurodegeneration and their controls. Thus, we aimed to investigate the impact of sex on microglial activation in amyloid and tau mouse models and wild-type controls. Methods TSPO-PET (18F-GE-180) data of C57Bl/6 (wild-type), AppNL-G-F (β-amyloid model), and P301S (tau model) mice was assessed longitudinally between 2 and 12 months of age. The AppNL-G-F group also underwent longitudinal β-amyloid-PET imaging (Aβ-PET; 18F-florbetaben). PET results were confirmed and validated by immunohistochemical investigation of microglial (Iba-1, CD68), astrocytic (GFAP), and tau (AT8) markers. Findings in cerebral cortex were compared by sex using linear mixed models for PET data and analysis of variance for immunohistochemistry. Results Wild-type mice showed an increased TSPO-PET signal over time (female +23%, male +4%), with a significant sex × age interaction (T = − 4.171, p < 0.001). The Aβ model AppNL-G-F mice also showed a significant sex × age interaction (T = − 2.953, p = 0.0048), where cortical TSPO-PET values increased by 31% in female AppNL-G-F mice, versus only 6% in the male mice group from 2.5 to 10 months of age. Immunohistochemistry for the microglial markers Iba-1 and CD68 confirmed the TSPO-PET findings in male and female mice aged 10 months. Aβ-PET in the same AppNL-G-F mice indicated no significant sex × age interaction (T = 0.425, p = 0.673). The P301S tau model showed strong cortical increases of TSPO-PET from 2 to 8.5 months of age (female + 32%, male + 36%), without any significant sex × age interaction (T = − 0.671, p = 0.504), and no sex differences in Iba-1, CD68, or AT8 immunohistochemistry. Conclusion Female mice indicate sex-dependent microglia activation in aging and in response to amyloidosis but not in response to tau pathology. This calls for consideration of sex difference in TSPO-PET studies of microglial activation in mouse models of neurodegeneration and by extension in human studies.
Collapse
Affiliation(s)
- Gloria Biechele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Tanja Blume
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany.,DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.,DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Jose Medina Luque
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Christian Sacher
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Francois Ruch-Rubinstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Franz-Josef Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Barbara von Ungern-Sternberg
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland.,School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany.,Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany.,Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Günter U Höglinger
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.,Department of Neurology, Technical University Munich, Munich, Germany.,Center of Neuropathology and Prion Research, University of Munich, Munich, Germany
| | - Jochen Herms
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninstraße 15, 81377, Munich, Germany. .,Department of Neurology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
330
|
Qorri B, Tsay M, Agrawal A, Au R, Gracie J. Using machine intelligence to uncover Alzheimer’s disease progression heterogeneity. EXPLORATION OF MEDICINE 2020. [DOI: 10.37349/emed.2020.00026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aim: Research suggests that Alzheimer’s disease (AD) is heterogeneous with numerous subtypes. Through a proprietary interactive ML system, several underlying biological mechanisms associated with AD pathology were uncovered. This paper is an introduction to emerging analytic efforts that can more precisely elucidate the heterogeneity of AD.
Methods: A public AD data set (GSE84422) consisting of transcriptomic data of postmortem brain samples from healthy controls (n = 121) and AD (n = 380) subjects was analyzed. Data were processed by an artificial intelligence platform designed to discover potential drug repurposing candidates, followed by an interactive augmented intelligence program.
Results: Using perspective analytics, six perspective classes were identified: Class I is defined by TUBB1, ASB4, and PDE5A; Class II by NRG2 and ZNF3; Class III by IGF1, ASB4, and GTSE1; Class IV is defined by cDNA FLJ39269, ITGA1, and CPM; Class V is defined by PDE5A, PSEN1, and NDUFS8; and Class VI is defined by DCAF17, cDNA FLJ75819, and SLC33A1. It is hypothesized that these classes represent biological mechanisms that may act alone or in any combination to manifest an Alzheimer’s pathology.
Conclusions: Using a limited transcriptomic public database, six different classes that drive AD were uncovered, supporting the premise that AD is a heterogeneously complex disorder. The perspective classes highlighted genetic pathways associated with vasculogenesis, cellular signaling and differentiation, metabolic function, mitochondrial function, nitric oxide, and metal ion metabolism. The interplay among these genetic factors reveals a more profound underlying complexity of AD that may be responsible for the confluence of several biological factors. These results are not exhaustive; instead, they demonstrate that even within a relatively small study sample, next-generation machine intelligence can uncover multiple genetically driven subtypes. The models and the underlying hypotheses generated using novel analytic methods may translate into potential treatment pathways.
Collapse
Affiliation(s)
- Bessi Qorri
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Mike Tsay
- NetraMark Corp, Toronto, ON M4E 1G8, Canada
| | | | - Rhoda Au
- Department of Anatomy & Neurobiology, Neurology and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA 02218, USA
| | - Joseph Gracie
- NetraMark Corp, Toronto, ON M4E 1G8, Canada 5Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
331
|
Tseng TJ, Yen YT, Yang YH, Chen YH, Chan TC. Association between the occurrence of albuminuria and the risk of early dementia among older people upon health examination: a community-based cohort study in Taiwan. BMJ Open 2020; 10:e041664. [PMID: 33293399 PMCID: PMC7725074 DOI: 10.1136/bmjopen-2020-041664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES This study aimed to investigate the associations between biochemical markers, renal function, health behaviours and dementia among older people. DESIGN A retrospective cohort study. SETTING Community-based health examination database from Taipei city. PARTICIPANTS In total, 35 434 older people were included from February 2005 to December 2012. To assess changes in renal function, we selected participants who attended health examinations at least twice and responded to the AD8 questionnaire in 2012. We excluded those with dementia at baseline. PRIMARY OUTCOME MEASURES Early dementia was assessed using the AD8 questionnaire in 2012. Explanatory variables included demographic factors, health behaviours, biochemical markers and renal function. We used a Cox proportional hazard model to estimate the HR for early dementia onset. RESULTS Individuals with mild albuminuria (HR 1.228; 95% CI 1.066 to 1.414), lower eGFR (HR 1.549; 95% CI 1.319 to 1.820) and higher age (HR 1.022; 95% CI 1.015 to 1.028) were associated with a high risk of early dementia. Older people with no alcohol intake (HR 0.872; 95% CI 0.794 to 0.958), and higher education levels (HR 0.647; 95% CI 0.589 to 0.710) were at a low risk of early dementia. CONCLUSIONS Elevated mild albuminuria and low eGFR were associated with a high risk of early dementia in this community-based cohort. Routine health examinations for older people can help screen out the high-risk population, and clinical management might reduce or delay the risk of early dementia.
Collapse
Affiliation(s)
- Tzu-Jung Tseng
- Research Center for Humanities and Social Sciences, Academia Sinica, Taipei, Taiwan
| | - Yun-Ting Yen
- Research Center for Humanities and Social Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuan-Han Yang
- Department of Neurology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Hsu Chen
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
- School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Tropical Medicine and Infectious Diseases, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Ta-Chien Chan
- Research Center for Humanities and Social Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Public Health, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
332
|
Parodi-Rullán R, Sone JY, Fossati S. Endothelial Mitochondrial Dysfunction in Cerebral Amyloid Angiopathy and Alzheimer's Disease. J Alzheimers Dis 2020; 72:1019-1039. [PMID: 31306129 DOI: 10.3233/jad-190357] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Cerebrovascular dysfunction is one of the earliest events in the pathogenesis of AD, as well as in vascular and mixed dementias. Cerebral amyloid angiopathy (CAA), the deposition of amyloid around cerebral vessels, is observed in up to 90% of AD patients and in approximately 50% of elderly individuals over 80 years of age. CAA is a strong contributor to vascular dysfunction in AD. CAA-laden brain vessels are characterized by dysfunctional hemodynamics and leaky blood-brain barrier (BBB), contributing to clearance failure and further accumulation of amyloid-β (Aβ) in the cerebrovasculature and brain parenchyma. Mitochondrial dysfunction is increasingly recognized as an important early initiator of the pathogenesis of AD and CAA. The objective of this review is to discuss the effects of Aβ on cerebral microvascular cell function, focusing on its impact on endothelial mitochondria. After introducing CAA and its etiology and genetic risk factors, we describe the pathological relationship between cerebrovascular amyloidosis and brain microvascular endothelial cell dysfunction, critically analyzing its roles in disease progression, hypoperfusion, and BBB integrity. Then, we focus on discussing the effect of Aβ challenge on endothelial mitochondrial dysfunction pathways, and their contribution to the progression of neurovascular dysfunction in AD and dementia. Finally, we report potential pharmacological and non-pharmacological mitochondria-targeted therapeutic strategies which may help prevent or delay cerebrovascular failure.
Collapse
Affiliation(s)
- Rebecca Parodi-Rullán
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Je Yeong Sone
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, NY, USA
| | - Silvia Fossati
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
333
|
Mejia-Arango S, Garcia-Cifuentes E, Samper-Ternent R, Borda MG, Cano-Gutierrez CA. Socioeconomic Disparities and Gender Inequalities in Dementia: a Community-Dwelling Population Study from a Middle-Income Country. J Cross Cult Gerontol 2020; 36:105-118. [PMID: 33247379 DOI: 10.1007/s10823-020-09418-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2020] [Indexed: 11/29/2022]
Abstract
The proportion of the world's older adults and of its dementia cases is increasing in low and middle-income countries. This is particularly true in Colombia. There, the number of individuals with dementia may increase five-fold by 2050. Yet research is lacking on dementia in such settings. This work estimates the prevalence of dementia in a community-dwelling population in Colombia. It also assesses how gender-based differences in cardiovascular conditions and socioeconomic disparities affect dementia. This work analyzes data on 2000 respondents at least 60 years of age in the Health, Well-Being, And Aging (SABE) study in Bogota. Respondents with dementia are those who have cognitive impairment and at least two limitations in instrumental activities of daily living. The SABE study finds 8.4% of respondents have dementia. Age, cardiovascular risks, and socioeconomic disparities contributed to higher odds of dementia. The contributors to dementia can differ for men and women. For example, socioeconomic disparities were a larger contributor to dementia for women than men. The findings support the cognitive reserve hypothesis on dementia. This holds that pre-existing cognitive processes and compensatory mechanisms influence dementia. Women in Latin America are more likely to suffer from socioeconomic disparities that limit their cognitive reserve. This research points to several policy implications that can help offset these disparities and reduce the prevalence of dementia.
Collapse
Affiliation(s)
- Silvia Mejia-Arango
- Department of Population Studies, El Colegio de la Frontera Norte, Carretera Escénica Km 18.5 San Antonio del Mar, 22560, Tijuana, Baja California, Mexico.
| | - Elkin Garcia-Cifuentes
- Instituto de Envejecimiento, Facultad de Medicina, Pontificia Universidad Javeriana, Bogota, Colombia.,Semillero de Neurociencias y Envejecimiento, Facultad de Medicina, Pontificia Universidad Javeriana, Bogota, Colombia
| | | | - Miguel G Borda
- Instituto de Envejecimiento, Facultad de Medicina, Pontificia Universidad Javeriana, Bogota, Colombia.,Semillero de Neurociencias y Envejecimiento, Facultad de Medicina, Pontificia Universidad Javeriana, Bogota, Colombia.,Unidad de Geriatria, Hospital Universitario San Ignacio, Bogota, Colombia.,Center for Age-Related Diseases, Stavanger University Hospital, Stavanger, Norway
| | - Carlos A Cano-Gutierrez
- Instituto de Envejecimiento, Facultad de Medicina, Pontificia Universidad Javeriana, Bogota, Colombia.,Semillero de Neurociencias y Envejecimiento, Facultad de Medicina, Pontificia Universidad Javeriana, Bogota, Colombia.,Unidad de Geriatria, Hospital Universitario San Ignacio, Bogota, Colombia
| |
Collapse
|
334
|
Matsuoka T, Oya N, Yokota H, Akazawa K, Yamada K, Narumoto J. Pineal volume reduction in patients with mild cognitive impairment who converted to Alzheimer's disease. Psychiatry Clin Neurosci 2020; 74:587-593. [PMID: 32609399 DOI: 10.1111/pcn.13103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/09/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022]
Abstract
AIM Pineal parenchymal volume (PPV) reduction is one of the predisposing factors for Alzheimer's disease (AD). Therefore, PPV could be used as a predictor of developing AD in clinical settings. We investigated whether PPV in patients with mild cognitive impairment (MCI) was correlated with conversion of these patients to AD. METHODS A total of 237 patients with MCI underwent brain magnetic resonance imaging. A two-sample t-test was used to compare PPV at baseline in MCI patients who converted to AD (MCI-C) with those who did not convert (MCI-NC). Logistic regression analysis with forced entry was used to identify predictors of AD, with variables of PPV, age, sex, education, APOE-ε4 alleles, Mini Mental State Examination score, and total intracranial volume at baseline. Two-way repeated-measures analysis of variance was conducted to compare PPV at baseline and at the last examination in the MCI-C and MCI-NC groups. RESULTS PPV in the MCI-C group was significantly lower than that in the MCI-NC group. In logistic regression analysis, two independent predictors of AD were identified: Mini Mental State Examination and PPV. Two-way repeated-measures analysis of variance revealed a significant group effect, but no time effect. CONCLUSION The pineal volume is a predictor of AD conversion, and pineal volume reduction in AD starts early when patients are still in the MCI stage. Thus, pineal volume reduction might be useful as a predictor of developing AD in clinical settings.
Collapse
Affiliation(s)
- Teruyuki Matsuoka
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nozomu Oya
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hajime Yokota
- Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kentaro Akazawa
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Yamada
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jin Narumoto
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | |
Collapse
|
335
|
Nuthikattu S, Milenkovic D, Rutledge JC, Villablanca AC. Sex-Dependent Molecular Mechanisms of Lipotoxic Injury in Brain Microvasculature: Implications for Dementia. Int J Mol Sci 2020; 21:E8146. [PMID: 33142695 PMCID: PMC7663125 DOI: 10.3390/ijms21218146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/16/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular risk factors and biologic sex play a role in vascular dementia which is characterized by progressive reduction in cognitive function and memory. Yet, we lack understanding about the role sex plays in the molecular mechanisms whereby lipid stress contributes to cognitive decline. Five-week-old low-density lipoprotein deficient (LDL-R -/-) male and female mice and C57BL/6J wild types (WT) were fed a control or Western Diet for 8 weeks. Differential expression of protein coding and non-protein coding genes (DEG) were determined in laser captured hippocampal microvessels using genome-wide microarray, followed by bioinformatic analysis of gene networks, pathways, transcription factors and sex/gender-based analysis (SGBA). Cognitive function was assessed by Y-maze. Bioinformatic analysis revealed more DEGs in females (2412) compared to males (1972). Hierarchical clusters revealed distinctly different sex-specific gene expression profiles irrespective of diet and genotype. There were also fewer and different biologic responses in males compared to females, as well as different cellular pathways and gene networks (favoring greater neuroprotection in females), together with sex-specific transcription factors and non-protein coding RNAs. Hyperlipidemic stress also resulted in less severe cognitive dysfunction in females. This sex-specific pattern of differential hippocampal microvascular RNA expression might provide therapeutic targets for dementia in males and females.
Collapse
Affiliation(s)
- Saivageethi Nuthikattu
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| | - Dragan Milenkovic
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
- Université Clermont Auvergne, INRA, UNH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - John C. Rutledge
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| | - Amparo C. Villablanca
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| |
Collapse
|
336
|
Abstract
OBJECTIVES To investigate the cross-sectional association between measures of menstruation history (including menopausal status, age of menopause, age of menarche, and duration of reproductive stage) and brain volume. METHODS Women (aged 45 to 79 years) from the UK Biobank were included (n = 5,072) after excluding those who had (1) hysterectomy or bilateral oophorectomy, (2) ever used menopausal hormone therapy, (3) ever had a stroke, or (4) were perimenopausal. Multiple linear hierarchical regression models were computed to quantify the cross-sectional association between measures of menstruation history and brain volume. Sensitivity analysis based on propensity matching for age (and other demographic/health covariates) were applied to estimate differences in brain volumes between matched premenopausal and postmenopausal women. RESULTS Postmenopausal women had 1.06% (95% confidence interval [CI]; 1.05-1.06) and 2.17% (95% CI, 2.12-2.22) larger total brain volume (TBV) and hippocampal volumes (HV), respectively, than premenopausal women. Sensitivity analysis with age matched samples produced consistent results (TBV: 0.82%, 95% CI, 0.25-1.38; HV: 1.33%, 95% CI, 0.01-2.63). For every year increase in age above 45 years, postmenopausal women experienced 0.23% greater reduction in TBV than premenopausal women (95% CI, -0.60 to -0.14), which was not observed for HV. Moreover, every 1 year delayed onset of menopause after 45 was associated with 0.32% (95% CI, -0.35 to -0.28) and 0.31% (95% CI, -0.40 to -0.22) smaller TBV and HV, respectively. Every additional year in age of menarche was associated with 0.10% (95% CI, 0.04-0.16) larger TBV, which was not detected for HV. Similarly, every 1 year increase in duration of reproductive stage was associated with 0.09% smaller TBV (95% CI, -0.15 to -0.03), which was not detected for HV. CONCLUSIONS Menopause may contribute to brain volume beyond typical aging effects. Furthermore, early age of menarche, delayed age of menopause and increasing duration of reproductive stage were negatively associated with brain volume. Further research is required to determine whether the negative association between age of menopause and HV is potentially an indicator of future vulnerability for dementia.
Collapse
|
337
|
Effect of Caffeine Consumption on the Risk for Neurological and Psychiatric Disorders: Sex Differences in Human. Nutrients 2020; 12:nu12103080. [PMID: 33050315 PMCID: PMC7601837 DOI: 10.3390/nu12103080] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023] Open
Abstract
Caffeine occurs naturally in various foods, such as coffee, tea, and cocoa, and it has been used safely as a mild stimulant for a long time. However, excessive caffeine consumption (1~1.5 g/day) can cause caffeine poisoning (caffeinism), which includes symptoms such as anxiety, agitation, insomnia, and gastrointestinal disorders. Recently, there has been increasing interest in the effect of caffeine consumption as a protective factor or risk factor for neurological and psychiatric disorders. Currently, the importance of personalized medicine is being emphasized, and research on sex/gender differences needs to be conducted. Our review focuses on the effect of caffeine consumption on several neurological and psychiatric disorders with respect to sex differences to provide a better understanding of caffeine use as a risk or protective factor for those disorders. The findings may help establish new strategies for developing sex-specific caffeine therapies.
Collapse
|
338
|
Xu Z, Yang J, Lau KK, Yip PSF, Wong ICK, Zhang Q. Understanding the Association Between Antidepressants and the Risk of Being Diagnosed with Dementia in Older People: A Self-Controlled Case Series Study. J Alzheimers Dis 2020; 78:735-744. [PMID: 33016920 DOI: 10.3233/jad-200875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Given concerns about adverse outcomes for older people taking antidepressants in the literature, we investigated whether taking antidepressants elevates the risk of dementia. OBJECTIVE This study aims to investigate the putative association of antidepressants with the risk of dementia. METHODS We conducted a population-based self-controlled case series analysis of older people with dementia and taking antidepressants, using territory-wide medical records of 194,507 older patients collected by the Hospital Authority of Hong Kong, to investigate the association between antidepressant treatment and the risk of developing dementia in older people. RESULTS There was a significantly higher risk of being diagnosed with dementia during the pre-drug-exposed period (incidence rate ratio (IRR) 20.42 (95% CI: 18.66-22.34)) compared to the non-drug-exposed baseline period. The IRR remained high during the drug-exposed period (IRR 8.86 (7.80-10.06)) before returning to a baseline level after washout (IRR 1.12 (0.77-1.36)). CONCLUSION The higher risk of dementia before antidepressant treatment may be related to emerging psychiatric symptoms co-occurring with dementia, which trigger medical consultations that result in a decision to begin antidepressants. Our findings do not support a causal relationship between antidepressant treatment and the risk of dementia.
Collapse
Affiliation(s)
- Zhongzhi Xu
- School of Data Science, City University of Hong Kong, Kowloon, Hong Kong, China.,Hong Kong Jockey Club Centre for Suicide Research and Prevention, The University of Hong Kong, Hong Kong, China
| | - Jiannan Yang
- School of Data Science, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Kui Kai Lau
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Paul S F Yip
- Hong Kong Jockey Club Centre for Suicide Research and Prevention, The University of Hong Kong, Hong Kong, China
| | - Ian C K Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Centre for Medicines Optimisation Research and Education, Research Department of Policy and Practice, University College London School of Pharmacy, London, UK
| | - Qingpeng Zhang
- School of Data Science, City University of Hong Kong, Kowloon, Hong Kong, China
| |
Collapse
|
339
|
Drake LR, Brooks AF, Stauff J, Sherman PS, Arteaga J, Koeppe RA, Reed A, Montavon TJ, Skaddan MB, Scott PJ. Strategies for PET imaging of the receptor for advanced glycation endproducts (RAGE). J Pharm Anal 2020; 10:452-465. [PMID: 33133729 PMCID: PMC7591811 DOI: 10.1016/j.jpha.2020.07.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
The implication of the receptor for advanced glycation end-products (RAGE) in numerous diseases and neurodegenerative disorders makes it interesting both as a therapeutic target and as an inflammatory biomarker. In the context of investigating RAGE as a biomarker, there is interest in developing radiotracers that will enable quantification of RAGE using positron emission tomography (PET) imaging. We have synthesized potential small molecule radiotracers for both the intracellular ([18F]InRAGER) and extracellular ([18F]RAGER) domains of RAGE. Herein we report preclinical evaluation of both using in vitro (lead panel screens) and in vivo (rodent and nonhuman primate PET imaging) methods. Both radiotracers have high affinity for RAGE and show good brain uptake, but suffer from off-target binding. The source of the off-target PET signal is not attributable to binding to melatonin receptors, but remains unexplained. We have also investigated use of lipopolysaccharide (LPS)-treated mice as a possible animal model with upregulated RAGE for evaluation of new imaging agents. Immunoreactivity of the mouse brain sections revealed increases in RAGE in the male cohorts, but no difference in the female groups. However, it proves challenging to quantify the changes in RAGE due to off-target binding of the radiotracers. Nevertheless, they are appropriate lead scaffolds for future development of 2nd generation RAGE PET radiotracers because of their high affinity for the receptor and good CNS penetration.
Collapse
Affiliation(s)
- Lindsey R. Drake
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Allen F. Brooks
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jenelle Stauff
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Phillip S. Sherman
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Janna Arteaga
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert A. Koeppe
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Aimee Reed
- AbbVie Process Chemistry, North Chicago, IL, 60064, USA
| | | | | | - Peter J.H. Scott
- Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
340
|
Novick AM, Scott AT, Neill Epperson C, Schneck CD. Neuropsychiatric effects of tamoxifen: Challenges and opportunities. Front Neuroendocrinol 2020; 59:100869. [PMID: 32822707 PMCID: PMC7669724 DOI: 10.1016/j.yfrne.2020.100869] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/12/2020] [Accepted: 08/16/2020] [Indexed: 02/08/2023]
Abstract
Epidemiological, clinical, and basic research over the past thirty years have described the benefits of estrogen on cognition, mood, and brain health. Less is known about tamoxifen, a selective estrogen receptor modifier (SERM) commonly used in breast cancer which is able to cross the blood-brain barrier. In this article, we review the basic pharmacology of tamoxifenas well as its effects on cognition and mood. The literature reveals an overall impairing effect of tamoxifen on cognition in breast cancer patients, hinting at central antiestrogen activity. On the other hand, tamoxifen demonstrates promising effects in psychiatric disorders, like bipolar disorder, where its therapeutic action may be independent of interaction with estrogen receptors. Understanding the neuropsychiatric properties of SERMs like tamoxifen can guide future research to ameliorate unwanted side-effects and provide novel options for difficult to treat disorders.
Collapse
Affiliation(s)
- Andrew M Novick
- Department of Psychiatry, University of Colorado School of Medicine, 13001 E 17th Place, Campus Box F546, Aurora, CO 80045, United States.
| | - Anthony T Scott
- Department of Psychiatry, University of Colorado School of Medicine, 13001 E 17th Place, Campus Box F546, Aurora, CO 80045, United States
| | - C Neill Epperson
- Department of Psychiatry, University of Colorado School of Medicine, 13001 E 17th Place, Campus Box F546, Aurora, CO 80045, United States
| | - Christopher D Schneck
- Department of Psychiatry, University of Colorado School of Medicine, 13001 E 17th Place, Campus Box F546, Aurora, CO 80045, United States
| |
Collapse
|
341
|
Jiang J, Young K, Pike CJ. Second to fourth digit ratio (2D:4D) is associated with dementia in women. Early Hum Dev 2020; 149:105152. [PMID: 32781308 PMCID: PMC7484057 DOI: 10.1016/j.earlhumdev.2020.105152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease is characterized by sex differences that may involve sex hormone exposure during development. Finger length ratios, an indirect measure of prenatal androgen exposure, were found to significantly differ in women with and without dementia. This finding links a relatively feminine in utero development with vulnerability to dementia in women.
Collapse
Affiliation(s)
| | | | - Christian J. Pike
- Corresponding Author: Christian J. Pike, Ph.D., Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191 USA, Tel: 213-740-4205,
| |
Collapse
|
342
|
Mander BA. Local Sleep and Alzheimer's Disease Pathophysiology. Front Neurosci 2020; 14:525970. [PMID: 33071726 PMCID: PMC7538792 DOI: 10.3389/fnins.2020.525970] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 09/01/2020] [Indexed: 12/11/2022] Open
Abstract
Even prior to the onset of the prodromal stages of Alzheimer's disease (AD), a constellation of sleep disturbances are apparent. A series of epidemiological studies indicate that multiple forms of these sleep disturbances are associated with increased risk for developing mild cognitive impairment (MCI) and AD, even triggering disease onset at an earlier age. Through the combination of causal manipulation studies in humans and rodents, as well as targeted examination of sleep disturbance with respect to AD biomarkers, mechanisms linking sleep disturbance to AD are beginning to emerge. In this review, we explore recent evidence linking local deficits in brain oscillatory function during sleep with local AD pathological burden and circuit-level dysfunction and degeneration. In short, three deficits in the local expression of sleep oscillations have been identified in relation to AD pathophysiology: (1) frequency-specific frontal deficits in slow wave expression during non-rapid eye movement (NREM) sleep, (2) deficits in parietal sleep spindle expression, and (3) deficits in the quality of electroencephalographic (EEG) desynchrony characteristic of REM sleep. These deficits are noteworthy since they differ from that seen in normal aging, indicating the potential presence of an abnormal aging process. How each of these are associated with β-amyloid (Aβ) and tau pathology, as well as neurodegeneration of circuits sensitive to AD pathophysiology, are examined in the present review, with a focus on the role of dysfunction within fronto-hippocampal and subcortical sleep-wake circuits. It is hypothesized that each of these local sleep deficits arise from distinct network-specific dysfunctions driven by regionally-specific accumulation of AD pathologies, as well as their associated neurodegeneration. Overall, the evolution of these local sleep deficits offer unique windows into the circuit-specific progression of distinct AD pathophysiological processes prior to AD onset, as well as their impact on brain function. This includes the potential erosion of sleep-dependent memory mechanisms, which may contribute to memory decline in AD. This review closes with a discussion of the remaining critical knowledge gaps and implications of this work for future mechanistic studies and studies implementing sleep-based treatment interventions.
Collapse
Affiliation(s)
- Bryce A. Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
343
|
Swinnen N, Vandenbulcke M, de Bruin ED, Akkerman R, Stubbs B, Vancampfort D. Exergaming for people with major neurocognitive disorder: a qualitative study. Disabil Rehabil 2020; 44:2044-2052. [PMID: 32962436 DOI: 10.1080/09638288.2020.1822934] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE This study investigated the experiences of participation in a standing balance exergame program amongst people with major neurocognitive disorder (MNCD) within residential care settings. MATERIALS AND METHODS A qualitative descriptive study was conducted in participants with MNCD recruited from two residential settings. Participants exergamed for 15 min, three times per week for 8 weeks. Semi-structured interviews were conducted in all participants of the trial after 4 and 8 weeks. Audio files were transcribed and subsequently a thematic content analysis was performed using NVivo 12. Activity logs including adherence and attrition rates were kept. RESULTS Thirty-one participants with MNCD participated (median age = 85 (67-93) years; 77.4% women; Mini-Mental State Examination score = 19 (10-25)). Four broad themes emerged: (1) cognitive effects; (2) physical effects; (3) psychosocial effects and (4) motivators. The tailored exergame program was perceived as enjoyable. It stimulated participants' attention, concentration, reaction time, and memory. Participants reported improvements in balance, flexibility, and gait. Exergaming made participants energetic and calm. The attrition rate was 0% and the mean attendance rate was 79.3%. CONCLUSIONS The results indicate that standing balance exergaming is feasible, beneficial, and engaging in people with MNCD.Implications for rehabilitationExergames present a personalised intervention for engaging people with major neurocognitive disorder in physical activity.An exergame intervention is feasible and highly appreciated in this population.Exergames should be adapted to the individuals' needs and possibilities in order to enhance motivation and learning.
Collapse
Affiliation(s)
- Nathalie Swinnen
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium.,University Psychiatric Centre, KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- University Psychiatric Centre, KU Leuven, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Eling D de Bruin
- Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zürich, Zürich, Switzerland.,Department of Neurobiology, Care Sciences and Society, Division of Physiotherapy, Karolinska Institute, Stockholm, Sweden
| | | | - Brendon Stubbs
- South London and Maudsley NHS Foundation Trust, London, UK.,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Davy Vancampfort
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium.,University Psychiatric Centre, KU Leuven, Leuven, Belgium
| |
Collapse
|
344
|
Aerobic exercise increases sprouting angiogenesis in the male rat motor cortex. Brain Struct Funct 2020; 225:2301-2314. [PMID: 32918614 DOI: 10.1007/s00429-020-02100-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Exercise is beneficial to brain health, and historically, the advantageous effects of exercise on the brain have been attributed to neuronal plasticity. However, it has also become clear that the brain vascular system also exhibits plasticity in response to exercise. This plasticity occurs in areas involved in movement, such as the motor cortex. This experiment aimed to further characterize the effects of exercise on structural vascular plasticity in the male rat motor cortex, by specifically identifying whether features of angiogenesis, the growth of new capillaries, or changes in vessel diameter were present. Male rats in the exercise group engaged in a 5-week bout of voluntary wheel running, while a second group of rats remained sedentary. After the exercise regimen, vascular corrosion casts, resin replicas of the brain vasculature, were made for all animals and imaged using a scanning electron microscope. Results indicate sprouting angiogenesis was the primary form of structural vascular plasticity detected in the motor cortex under these aerobic exercise parameters. Additionally, exercised rats displayed a slight increase in capillary diameter and expanded endothelial cell nuclei diameters in this region.
Collapse
|
345
|
White L, Fishman P, Basu A, Crane PK, Larson EB, Coe NB. Dementia Is Associated With Earlier Mortality for Men and Women in the United States. Gerontol Geriatr Med 2020; 6:2333721420945922. [PMID: 32913883 PMCID: PMC7444147 DOI: 10.1177/2333721420945922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/30/2020] [Accepted: 07/06/2020] [Indexed: 11/17/2022] Open
Abstract
Objectives: Sociodemographic trends in the United States may influence future dementia-associated mortality, yet there is little evidence about their potential impact. Our study objective was to estimate the effect of dementia on survival in adults stratified by sex, education, and marital status. Methods: Using survey data from the Health and Retirement Study (HRS) linked to Medicare claims from 1991 to 2012, we identified a retrospective cohort of adults with at least one International Classification of Diseases—ninth revision—Clinical Modification (ICD-9-CM) dementia diagnosis code (n = 3,714). For each case, we randomly selected up to five comparators, matching on sex, birth year, education, and HRS entry year (n = 9,531), and assigned comparators the diagnosis date of their matched case. Participants were followed for up to 60 months following diagnosis. We estimated a survival function for the entire study population and then within successive strata defined by sex, education, and marital status. Results: On average, dementia cases were 80.5 years old at diagnosis. Most were female, had less than college-level education, and approximately 40% were married at diagnosis. In multivariate analyses, dementia diagnosis was associated with earlier mortality for women (predicted median survival of 54.5 months vs. 62.5 months; dementia coefficient = −0.13; 95% confidence interval [CI] = [−0.22, −0.04]; p = .003), but even more so among men (predicted median survival of 35.5 months vs. 54.5 months; dementia coefficient = −0.42; 95% CI = [−0.52, −0.31]; p < .001). We found substantial heterogeneity in the relationship between dementia and survival, associated with both education and marital status. Conclusion: Both sex and level of education moderate the relationship between dementia diagnosis and length of survival.
Collapse
Affiliation(s)
| | | | - Anirban Basu
- University of Washington, Seattle, WA, USA.,National Bureau of Economic Research, Cambridge, MA, USA
| | | | | | - Norma B Coe
- National Bureau of Economic Research, Cambridge, MA, USA.,University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
346
|
Argentati C, Tortorella I, Bazzucchi M, Emiliani C, Morena F, Martino S. The Other Side of Alzheimer's Disease: Influence of Metabolic Disorder Features for Novel Diagnostic Biomarkers. J Pers Med 2020; 10:E115. [PMID: 32899957 PMCID: PMC7563360 DOI: 10.3390/jpm10030115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 02/08/2023] Open
Abstract
Nowadays, the amyloid cascade hypothesis is the dominant model to explain Alzheimer's disease (AD) pathogenesis. By this hypothesis, the inherited genetic form of AD is discriminated from the sporadic form of AD (SAD) that accounts for 85-90% of total patients. The cause of SAD is still unclear, but several studies have shed light on the involvement of environmental factors and multiple susceptibility genes, such as Apolipoprotein E and other genetic risk factors, which are key mediators in different metabolic pathways (e.g., glucose metabolism, lipid metabolism, energetic metabolism, and inflammation). Furthermore, growing clinical evidence in AD patients highlighted the presence of affected systemic organs and blood similarly to the brain. Collectively, these findings revise the canonical understating of AD pathogenesis and suggest that AD has metabolic disorder features. This review will focus on AD as a metabolic disorder and highlight the contribution of this novel understanding on the identification of new biomarkers for improving an early AD diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (C.A.); (I.T.); (M.B.); (C.E.); (F.M.)
| |
Collapse
|
347
|
Forde NJ, Jeyachandra J, Joseph M, Jacobs GR, Dickie E, Satterthwaite TD, Shinohara RT, Ameis SH, Voineskos AN. Sex Differences in Variability of Brain Structure Across the Lifespan. Cereb Cortex 2020; 30:5420-5430. [PMID: 32483605 PMCID: PMC7566684 DOI: 10.1093/cercor/bhaa123] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/16/2020] [Accepted: 04/19/2020] [Indexed: 12/13/2022] Open
Abstract
Several brain disorders exhibit sex differences in onset, presentation, and prevalence. Increased understanding of the neurobiology of sex-based differences in variability across the lifespan can provide insight into both disease vulnerability and resilience. In n = 3069 participants, from 8 to 95 years of age, we found widespread greater variability in males compared with females in cortical surface area and global and subcortical volumes for discrete brain regions. In contrast, variance in cortical thickness was similar for males and females. These findings were supported by multivariate analysis accounting for structural covariance, and present and stable across the lifespan. Additionally, we examined variability among brain regions by sex. We found significant age-by-sex interactions across neuroimaging metrics, whereby in very early life males had reduced among-region variability compared with females, while in very late life this was reversed. Overall, our findings of greater regional variability, but less among-region variability in males in early life may aid our understanding of sex-based risk for neurodevelopmental disorders. In contrast, our findings in late life may provide a potential sex-based risk mechanism for dementia.
Collapse
Affiliation(s)
- Natalie J Forde
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, M5T 1R8, Toronto, Canada
| | - Jerrold Jeyachandra
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, M5T 1R8, Toronto, Canada
| | - Michael Joseph
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, M5T 1R8, Toronto, Canada
| | - Grace R Jacobs
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, M5T 1R8, Toronto, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, M5S 1A8, Toronto, Canada
| | - Erin Dickie
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, M5T 1R8, Toronto, Canada
| | - Theodore D Satterthwaite
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn-CHOP Lifespan Brain Institute, Philadelphia, PA 19104, USA
| | - Russell T Shinohara
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Biomedical Image Computing and Analytics, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19103, USA
| | - Stephanie H Ameis
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, M5T 1R8, Toronto, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, M5T 1R8, Toronto, Canada
| | - Aristotle N Voineskos
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, M5T 1R8, Toronto, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, M5T 1R8, Toronto, Canada
| |
Collapse
|
348
|
Kuring JK, Mathias JL, Ward L. Risk of Dementia in persons who have previously experienced clinically-significant Depression, Anxiety, or PTSD: A Systematic Review and Meta-Analysis. J Affect Disord 2020; 274:247-261. [PMID: 32469813 DOI: 10.1016/j.jad.2020.05.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 04/16/2020] [Accepted: 05/10/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Depression, anxiety and PTSD appear to be linked to dementia, but it is unclear whether they are risk factors (causal or prodromal) for, comorbid with, or sequelae to (secondary effect of) dementia. Existing meta-analyses have examined depression or anxiety in all-cause dementia, Alzheimer's disease (AD) and vascular dementia (VaD), but have not considered post-traumatic stress disorder (PTSD), dementia with Lewy bodies (DLB), or frontotemporal dementia (FTD). The current meta-analysis examined the risk of developing dementia (AD, VaD, DLB, FTD, all-cause) in people with and without a history of clinically-significant depression, anxiety or PTSD in order to better understand the link between mental illness and dementia (PROSPERO number: CRD42018099872). METHODS PubMed, EMBASE, PsycINFO and CINAHL searches identified 36 eligible studies. RESULTS There is a higher risk of developing all-cause dementia and AD in people with previous depression, and a higher risk of all-cause dementia in people with prior anxiety, than in persons without this history. Prior PTSD was not associated with a higher risk of later being diagnosed with dementia. LIMITATIONS The data for anxiety, PTSD, DLB and FTD were limited. CONCLUSIONS Depression and anxiety appear to be risk factors for dementia, but longitudinal studies across adulthood (young adult/mid-life/older adult) are needed to evaluate the likely causal or prodromal nature of this risk. The link between PTSD and dementia remains unclear. Regular screening for new onset mental illness and for cognitive changes in older adults with a history of mental illness may assist with earlier identification of dementia.
Collapse
Affiliation(s)
- J K Kuring
- School of Psychology, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, Australia
| | - J L Mathias
- School of Psychology, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, Australia.
| | - L Ward
- School of Psychology, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
349
|
Combining Cognitive Markers to Identify Individuals at Increased Dementia Risk: Influence of Modifying Factors and Time to Diagnosis. J Int Neuropsychol Soc 2020; 26:785-797. [PMID: 32207675 DOI: 10.1017/s1355617720000272] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE We investigated the extent to which combining cognitive markers increases the predictive value for future dementia, when compared to individual markers. Furthermore, we examined whether predictivity of markers differed depending on a range of modifying factors and time to diagnosis. METHOD Neuropsychological assessment was performed for 2357 participants (60+ years) without dementia from the population-based Swedish National Study on Aging and Care in Kungsholmen. In the main sample analyses, the outcome was dementia at 6 years. In the time-to-diagnosis analyses, a subsample of 407 participants underwent cognitive testing 12, 6, and 3 years before diagnosis, with dementia diagnosis at the 12-year follow-up. RESULTS Category fluency was the strongest individual predictor of dementia 6 years before diagnosis [area under the curve (AUC) = .903]. The final model included tests of verbal fluency, episodic memory, and perceptual speed (AUC = .913); these three domains were found to be the most predictive across a range of different subgroups. Twelve years before diagnosis, pattern comparison (perceptual speed) was the strongest individual predictor (AUC = .686). However, models 12 years before diagnosis did not show significantly increased predictivity above that of the covariates. CONCLUSIONS This study shows that combining markers from different cognitive domains leads to increased accuracy in predicting future dementia 6 years later. Markers from the verbal fluency, episodic memory, and perceptual speed domains consistently showed high predictivity across subgroups stratified by age, sex, education, apolipoprotein E ϵ4 status, and dementia type. Predictivity increased closer to diagnosis and showed highest accuracy up to 6 years before a dementia diagnosis. (JINS, 2020, 00, 1-13).
Collapse
|
350
|
Association of Sex with Serum Potassium, Sodium, and Calcium Disorders after Hypertensive Intracerebral Hemorrhage. World Neurosurg 2020; 141:e367-e373. [DOI: 10.1016/j.wneu.2020.05.137] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/31/2022]
|