1
|
Kim SM, Choi S, Lee G, Oh YH, Son JS, Ko A, Kim JS, Cho Y, Keum N, Park SM. Association of changes in predicted body composition with subsequent risk of dementia. Ann Clin Transl Neurol 2024; 11:1952-1963. [PMID: 39010668 PMCID: PMC11330214 DOI: 10.1002/acn3.52096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 04/17/2024] [Accepted: 04/29/2024] [Indexed: 07/17/2024] Open
Abstract
OBJECTIVE The effect of body composition change on the risk of dementia is not clear. This study analyzed the associations of changes in predicted lean body mass index (pLBMI), predicted appendicular skeletal muscle mass index (pASMI), and predicted body fat mass index (pBFMI) with the risk of dementia. METHODS In this nationwide cohort study, data were obtained from the Korean National Health Insurance Service database. The exposure was defined as changes in pLBMI, pASMI, and pBFMI derived from validated prediction equations. The outcome was dementia, defined based on the dementia diagnosis with prescription of anti-dementia medication. Cox proportional hazards regression analyses were performed to obtain the hazard ratio with a 95% confidence interval for risk of dementia according to changes in predicted body composition. RESULTS A total of 13,215,208 individuals with no prior record of dementia who underwent health screenings twice between 2009-2010 and 2011-2012 were included. A 1-kg/m2 increase in pLBMI and pASMI had an association with reduced risk of dementia (aHR: 0.85, 95% CI 0.84-0.87; aHR: 0.70, 95% CI 0.69-0.72, respectively for men, and aHR: 0.69, 95% CI 0.67-0.71; aHR: 0.59, 95% CI 0.57-0.61, respectively for women). A 1-kg/m2 increase in pBFMI had an association with a raised risk of dementia (aHR: 1.19, 95% CI 1.17-1.21 for men and aHR: 1.53, 95% CI 1.48-1.57 for women). These results remained consistent regardless of sex or weight change. INTERPRETATION Increase in pLBMI or pASMI, or reduction in pBFMI was linked to lower risk of dementia.
Collapse
Affiliation(s)
- Sung Min Kim
- Department of Transdisciplinary MedicineSeoul National University HospitalSeoulSouth Korea
- Department of Biomedical SciencesSeoul National University Graduate SchoolSeoulSouth Korea
| | - Seulggie Choi
- Department of Internal MedicineSeoul National University HospitalSeoulSouth Korea
| | - Gyeongsil Lee
- Life ClinicSeoulSouth Korea
- KS Healthlink InstituteSeoulSouth Korea
| | - Yun Hwan Oh
- Department of Family Medicine, Chung‐Ang University Gwangmyeong HospitalChung‐Ang University College of MedicineGwangmyeong‐siSouth Korea
| | - Joung Sik Son
- Department of Family MedicineKorea University Guro HospitalSeoulSouth Korea
| | - Ahryoung Ko
- Department of Family Medicine, Seoul National University HospitalSeoul National University College of MedicineSeoulSouth Korea
| | - Ji Soo Kim
- International Healthcare Center, Seoul National University HospitalSeoul National University College of MedicineSeoulSouth Korea
| | - Yoosun Cho
- Department of Family Medicine, Chung‐Ang University Gwangmyeong HospitalChung‐Ang University College of MedicineGwangmyeong‐siSouth Korea
| | - NaNa Keum
- Department of Food Science and BiotechnologyDongguk University Graduate SchoolSeoulSouth Korea
- Department of NutritionHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Sang Min Park
- Department of Biomedical SciencesSeoul National University Graduate SchoolSeoulSouth Korea
- Department of Family Medicine, Seoul National University HospitalSeoul National University College of MedicineSeoulSouth Korea
| |
Collapse
|
2
|
Wang MB, Rahmani F, Benzinger TLS, Raji CA. Edge Density Imaging Identifies White Matter Biomarkers of Late-Life Obesity and Cognition. Aging Dis 2024; 15:1899-1912. [PMID: 37196133 PMCID: PMC11272213 DOI: 10.14336/ad.2022.1210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 12/10/2022] [Indexed: 05/19/2023] Open
Abstract
Alzheimer disease (AD) and obesity are related to disruptions in the white matter (WM) connectome. We examined the link between the WM connectome and obesity and AD through edge-density imaging/index (EDI), a tractography-based method that characterizes the anatomical embedding of tractography connections. A total of 60 participants, 30 known to convert from normal cognition or mild-cognitive impairment to AD within a minimum of 24 months of follow up, were selected from the Alzheimer disease Neuroimaging Initiative (ADNI). Diffusion-weighted MR images from the baseline scans were used to extract fractional anisotropy (FA) and EDI maps that were subsequently averaged using deterministic WM tractography based on the Desikan-Killiany atlas. Multiple linear and logistic regression analysis were used to identify the weighted sum of tract-specific FA or EDI indices that maximized correlation to body-mass-index (BMI) or conversion to AD. Participants from the Open Access Series of Imaging Studies (OASIS) were used as an independent validation for the BMI findings. The edge-density rich, periventricular, commissural and projection fibers were among the most important WM tracts linking BMI to FA as well as to EDI. WM fibers that contributed significantly to the regression model related to BMI overlapped with those that predicted conversion; specifically in the frontopontine, corticostriatal, and optic radiation pathways. These results were replicated by testing the tract-specific coefficients found using ADNI in the OASIS-4 dataset. WM mapping with EDI enables identification of an abnormal connectome implicated in both obesity and conversion to AD.
Collapse
Affiliation(s)
- Maxwell Bond Wang
- Machine Learning Department, Carnegie Mellon University, Pittsburgh, PA, USA.
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA.
- Medical Scientist Training Program, University of Pittsburgh/Carnegie Mellon University, Pittsburgh, PA, USA.
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Division of Neuroradiology, Washington University in St. Louis, St. Louis, MO, USA.
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University, St. Louis, Missouri, USA.
| | - Tammie L. S Benzinger
- Mallinckrodt Institute of Radiology, Division of Neuroradiology, Washington University in St. Louis, St. Louis, MO, USA.
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University, St. Louis, Missouri, USA.
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Division of Neuroradiology, Washington University in St. Louis, St. Louis, MO, USA.
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University, St. Louis, Missouri, USA.
- Department of Neurology, Washington University in Saint Louis, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Le Thuc O, García-Cáceres C. Obesity-induced inflammation: connecting the periphery to the brain. Nat Metab 2024; 6:1237-1252. [PMID: 38997442 DOI: 10.1038/s42255-024-01079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Obesity is often associated with a chronic, low-grade inflammatory state affecting the entire body. This sustained inflammatory state disrupts the coordinated communication between the periphery and the brain, which has a crucial role in maintaining homeostasis through humoural, nutrient-mediated, immune and nervous signalling pathways. The inflammatory changes induced by obesity specifically affect communication interfaces, including the blood-brain barrier, glymphatic system and meninges. Consequently, brain areas near the third ventricle, including the hypothalamus and other cognition-relevant regions, become susceptible to impairments, resulting in energy homeostasis dysregulation and an elevated risk of cognitive impairments such as Alzheimer's disease and dementia. This Review explores the intricate communication between the brain and the periphery, highlighting the effect of obesity-induced inflammation on brain function.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
4
|
Campolim CM, Schimenes BC, Veras MM, Kim YB, Prada PO. Air pollution accelerates the development of obesity and Alzheimer's disease: the role of leptin and inflammation - a mini-review. Front Immunol 2024; 15:1401800. [PMID: 38933275 PMCID: PMC11199417 DOI: 10.3389/fimmu.2024.1401800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Air pollution is an urgent concern linked to numerous health problems in low- and middle-income countries, where 92% of air pollution-related deaths occur. Particulate matter 2.5 (PM2.5) is the most harmful component of air pollutants, increasing inflammation and changing gut microbiota, favoring obesity, type 2 diabetes, and Alzheimer's Disease (AD). PM2.5 contains lipopolysaccharides (LPS), which can activate the Toll-like receptor 4 (TLR4) signaling pathway. This pathway can lead to the release of pro-inflammatory markers, including interleukins, and suppressor of cytokine signaling-3 (SOCS3), which inhibits leptin action, a hormone that keeps the energy homeostasis. Leptin plays a role in preventing amyloid plaque deposition and hyperphosphorylation of tau-protein (p-tau), mechanisms involved in the neurodegeneration in AD. Approximately 50 million people worldwide are affected by dementia, with a significant proportion living in low-and middle-income countries. This number is expected to triple by 2050. This mini-review focuses on the potential impact of PM2.5 exposure on the TLR4 signaling pathway, its contribution to leptin resistance, and dysbiosis that exacerbates the link between obesity and AD.
Collapse
Affiliation(s)
- Clara Machado Campolim
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA, United States
| | | | - Mariana Matera Veras
- Laboratory of Environmental and Experimental Pathology LIM05, Department of Pathology, School of Medicine, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA, United States
| | - Patricia Oliveira Prada
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- Obesity and Comorbidities Research Center, Campinas, SP, Brazil
- Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
5
|
Roghani AK, Garcia RI, Roghani A, Reddy A, Khemka S, Reddy RP, Pattoor V, Jacob M, Reddy PH, Sehar U. Treating Alzheimer's disease using nanoparticle-mediated drug delivery strategies/systems. Ageing Res Rev 2024; 97:102291. [PMID: 38614367 DOI: 10.1016/j.arr.2024.102291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/15/2024]
Abstract
The administration of promising medications for the treatment of neurodegenerative disorders (NDDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) is significantly hampered by the blood-brain barrier (BBB). Nanotechnology has recently come to light as a viable strategy for overcoming this obstacle and improving drug delivery to the brain. With a focus on current developments and prospects, this review article examines the use of nanoparticles to overcome the BBB constraints to improve drug therapy for AD The potential for several nanoparticle-based approaches, such as those utilizing lipid-based, polymeric, and inorganic nanoparticles, to enhance drug transport across the BBB are highlighted. To shed insight on their involvement in aiding effective drug transport to the brain, methods of nanoparticle-mediated drug delivery, such as surface modifications, functionalization, and particular targeting ligands, are also investigated. The article also discusses the most recent findings on innovative medication formulations encapsulated within nanoparticles and the therapeutic effects they have shown in both preclinical and clinical testing. This sector has difficulties and restrictions, such as the need for increased safety, scalability, and translation to clinical applications. However, the major emphasis of this review aims to provide insight and contribute to the knowledge of how nanotechnology can potentially revolutionize the worldwide treatment of NDDs, particularly AD, to enhance clinical outcomes.
Collapse
Affiliation(s)
- Aryan Kia Roghani
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Frenship High School, Lubbock, TX 79382, USA.
| | - Ricardo Isaiah Garcia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Ali Roghani
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Aananya Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Lubbock High School, Lubbock, TX 79401, USA.
| | - Sachi Khemka
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Ruhananhad P Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Lubbock High School, Lubbock, TX 79401, USA.
| | - Vasanthkumar Pattoor
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; University of South Florida, Tampa, FL 33620, USA.
| | - Michael Jacob
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Services, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
6
|
Anderson T, Sharma S, Kelberman MA, Ware C, Guo N, Qin Z, Weinshenker D, Parent MB. Obesity during preclinical Alzheimer's disease development exacerbates brain metabolic decline. J Neurochem 2024; 168:801-821. [PMID: 37391269 DOI: 10.1111/jnc.15900] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/29/2023] [Accepted: 06/13/2023] [Indexed: 07/02/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Obesity in middle age increases AD risk and severity, which is alarming given that obesity prevalence peaks at middle age and obesity rates are accelerating worldwide. Midlife, but not late-life obesity increases AD risk, suggesting that this interaction is specific to preclinical AD. AD pathology begins in middle age, with accumulation of amyloid beta (Aβ), hyperphosphorylated tau, metabolic decline, and neuroinflammation occurring decades before cognitive symptoms appear. We used a transcriptomic discovery approach in young adult (6.5 months old) male and female TgF344-AD rats that overexpress mutant human amyloid precursor protein and presenilin-1 and wild-type (WT) controls to determine whether inducing obesity with a high-fat/high-sugar "Western" diet during preclinical AD increases brain metabolic dysfunction in dorsal hippocampus (dHC), a brain region vulnerable to the effects of obesity and early AD. Analyses of dHC gene expression data showed dysregulated mitochondrial and neurotransmission pathways, and up-regulated genes involved in cholesterol synthesis. Western diet amplified the number of genes that were different between AD and WT rats and added pathways involved in noradrenergic signaling, dysregulated inhibition of cholesterol synthesis, and decreased intracellular lipid transporters. Importantly, the Western diet impaired dHC-dependent spatial working memory in AD but not WT rats, confirming that the dietary intervention accelerated cognitive decline. To examine later consequences of early transcriptional dysregulation, we measured dHC monoamine levels in older (13 months old) AD and WT rats of both sexes after long-term chow or Western diet consumption. Norepinephrine (NE) abundance was significantly decreased in AD rats, NE turnover was increased, and the Western diet attenuated the AD-induced increases in turnover. Collectively, these findings indicate obesity during prodromal AD impairs memory, potentiates AD-induced metabolic decline likely leading to an overproduction of cholesterol, and interferes with compensatory increases in NE transmission.
Collapse
Affiliation(s)
- Thea Anderson
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Sumeet Sharma
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael A Kelberman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Christopher Ware
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Nanxi Guo
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, USA
| | - Zhaohui Qin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marise B Parent
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
- Department of Psychology, Georgia State University, Georgia, USA
| |
Collapse
|
7
|
Pradeepkiran JA, Baig J, Islam MA, Kshirsagar S, Reddy PH. Amyloid-β and Phosphorylated Tau are the Key Biomarkers and Predictors of Alzheimer's Disease. Aging Dis 2024:AD.2024.0286. [PMID: 38739937 DOI: 10.14336/ad.2024.0286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
Alzheimer's disease (AD) is a age-related neurodegenerative disease and is a major public health concern both in Texas, US and Worldwide. This neurodegenerative disease is mainly characterized by amyloid-beta (Aβ) and phosphorylated Tau (p-Tau) accumulation in the brains of patients with AD and increasing evidence suggests that these are key biomarkers in AD. Both Aβ and p-tau can be detected through various imaging techniques (such as positron emission tomography, PET) and cerebrospinal fluid (CSF) analysis. The presence of these biomarkers in individuals, who are asymptomatic or have mild cognitive impairment can indicate an increased risk of developing AD in the future. Furthermore, the combination of Aβ and p-tau biomarkers is often used for more accurate diagnosis and prediction of AD progression. Along with AD being a neurodegenerative disease, it is associated with other chronic conditions such as cardiovascular disease, obesity, depression, and diabetes because studies have shown that these comorbid conditions make people more vulnerable to AD. In the first part of this review, we discuss that biofluid-based biomarkers such as Aβ, p-Tau in cerebrospinal fluid (CSF) and Aβ & p-Tau in plasma could be used as an alternative sensitive technique to diagnose AD. In the second part, we discuss the underlying molecular mechanisms of chronic conditions linked with AD and how they affect the patients in clinical care.
Collapse
Affiliation(s)
| | - Javaria Baig
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sudhir Kshirsagar
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Neurology Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
8
|
Castro-Gomez S, Heneka MT. Innate immune activation in neurodegenerative diseases. Immunity 2024; 57:790-814. [PMID: 38599171 DOI: 10.1016/j.immuni.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Activation of the innate immune system following pattern recognition receptor binding has emerged as one of the major pathogenic mechanisms in neurodegenerative disease. Experimental, epidemiological, pathological, and genetic evidence underscores the meaning of innate immune activation during the prodromal as well as clinical phases of several neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal dementia. Importantly, innate immune activation and the subsequent release of inflammatory mediators contribute mechanistically to other hallmarks of neurodegenerative diseases such as aberrant proteostatis, pathological protein aggregation, cytoskeleton abnormalities, altered energy homeostasis, RNA and DNA defects, and synaptic and network disbalance and ultimately to the induction of neuronal cell death. In this review, we discuss common mechanisms of innate immune activation in neurodegeneration, with particular emphasis on the pattern recognition receptors (PRRs) and other receptors involved in the detection of damage-associated molecular patterns (DAMPs).
Collapse
Affiliation(s)
- Sergio Castro-Gomez
- Center for Neurology, Department of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, 53127 Bonn, Germany; Institute of Physiology II, University Hospital Bonn, 53115 Bonn, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg; Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
9
|
Hamilton LK, M'Bra PEH, Mailloux S, Galoppin M, Aumont A, Fernandes KJL. Central inhibition of stearoyl-CoA desaturase has minimal effects on the peripheral metabolic symptoms of the 3xTg Alzheimer's disease mouse model. Sci Rep 2024; 14:7742. [PMID: 38565895 PMCID: PMC10987571 DOI: 10.1038/s41598-024-58272-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
Evidence from genetic and epidemiological studies point to lipid metabolism defects in both the brain and periphery being at the core of Alzheimer's disease (AD) pathogenesis. Previously, we reported that central inhibition of the rate-limiting enzyme in monounsaturated fatty acid synthesis, stearoyl-CoA desaturase (SCD), improves brain structure and function in the 3xTg mouse model of AD (3xTg-AD). Here, we tested whether these beneficial central effects involve recovery of peripheral metabolic defects, such as fat accumulation and glucose and insulin handling. As early as 3 months of age, 3xTg-AD mice exhibited peripheral phenotypes including increased body weight and visceral and subcutaneous white adipose tissue as well as diabetic-like peripheral gluco-regulatory abnormalities. We found that intracerebral infusion of an SCD inhibitor that normalizes brain fatty acid desaturation, synapse loss and learning and memory deficits in middle-aged memory-impaired 3xTg-AD mice did not affect these peripheral phenotypes. This suggests that the beneficial effects of central SCD inhibition on cognitive function are not mediated by recovery of peripheral metabolic abnormalities. Given the widespread side-effects of systemically administered SCD inhibitors, these data suggest that selective inhibition of SCD in the brain may represent a clinically safer and more effective strategy for AD.
Collapse
Affiliation(s)
- Laura K Hamilton
- Research Center of the University of Montreal Hospital (CRCHUM), Montreal, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Paule E H M'Bra
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Sophia Mailloux
- Research Center of the University of Montreal Hospital (CRCHUM), Montreal, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Manon Galoppin
- Research Center of the University of Montreal Hospital (CRCHUM), Montreal, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Anne Aumont
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Karl J L Fernandes
- Research Center of the University of Montreal Hospital (CRCHUM), Montreal, Canada.
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada.
- Research Center on Aging, CIUSSS de l'Estrie-CHUS, Sherbrooke, Canada.
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada.
| |
Collapse
|
10
|
Qing W, Qian Y. Childhood obesity and risk of Alzheimer's disease: a Mendelian randomization study. Arch Public Health 2024; 82:39. [PMID: 38500220 PMCID: PMC10949616 DOI: 10.1186/s13690-024-01271-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/12/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Midlife obesity is a modifiable risk factor for Alzheimer's disease. However, the association between childhood obesity and Alzheimer's disease remains largely unknown. Therefore, we conducted a mendelian randomization analysis (MR) to assess the causal link between childhood obesity and Alzheimer's disease. METHODS Using summary statistics from publicly available genome-wide association studies (GWAS) database, we explored the genetic link between childhood obesity and Alzheimer's disease through a two-sample MR. The primary analysis employed the inverse-variance weighted (IVW) method. To complement our findings, we also employed MR-Egger, weighted median, simple model, and weighted model methods for MR estimates. Furthermore, we conducted Cochrane's Q-statistic test, Egger intercept test, and a leave-one-out sensitivity test to ensure the robustness and reliability of our results. RESULTS The IVW analysis yielded non-significant results, indicating no significant genetic association between childhood obesity and Alzheimer's disease (OR = 0.958, 95% CI = 0.910-1.008, p = 0.095). Consistent with this, the results from MR-Egger, the weighted median, simple model, and weighted model approaches all supported these findings. Furthermore, we did not detect any signs of heterogeneity or pleiotropy, and our leave-one-out analysis confirmed that no single nucleotide polymorphisms had a substantial impact on the reliability of our results. CONCLUSIONS The evidence from our MR analyses suggests that there is no causal effect of childhood obesity on the risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Wenxiang Qing
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yujie Qian
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
11
|
Zheng H, Gu C, Yang H. Identification of disease-specific bio-markers through network-based analysis of gene co-expression: A case study on Alzheimer's disease. Heliyon 2024; 10:e27070. [PMID: 38468964 PMCID: PMC10926071 DOI: 10.1016/j.heliyon.2024.e27070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/16/2024] [Accepted: 02/23/2024] [Indexed: 03/13/2024] Open
Abstract
Finding biomarker genes for complex diseases attracts persistent attention due to its application in clinics. In this paper, we propose a network-based method to obtain a set of biomarker genes. The key idea is to construct a gene co-expression network among sensitive genes and cluster the genes into different modules. For each module, we can identify its representative, i.e., the gene with the largest connectivity and the smallest average shortest path length to other genes within the module. We believe these representative genes could serve as a new set of potential biomarkers for diseases. As a typical example, we investigated Alzheimer's disease, obtaining a total of 16 potential representative genes, three of which belong to the non-transcriptome. A total of 11 out of these genes are found in literature from different perspectives and methods. The incipient groups were classified into two different subtypes using machine learning algorithms. We subjected the two subtypes to Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes analysis with healthy groups and moderate groups, respectively. The two sub-type groups were involved in two different biological processes, demonstrating the validity of this approach. This method is disease-specific and independent; hence, it can be extended to classify other kinds of complex diseases.
Collapse
Affiliation(s)
- Hexiang Zheng
- Department of Systems Science, Business School, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Changgui Gu
- Department of Systems Science, Business School, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Huijie Yang
- Department of Systems Science, Business School, University of Shanghai for Science and Technology, Shanghai, 200093, China
| |
Collapse
|
12
|
Ahmed FS, McMillan TM, Guenther BA, Dearborn P. Cognitive Performance following Single- or Multi-Session Exercise Intervention in Middle Age: A Systematic Review. Exp Aging Res 2024; 50:28-64. [PMID: 36384438 DOI: 10.1080/0361073x.2022.2137360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Research in modifiable behaviors, like exercise, on risk for dementia is increasing. Although many studies focus on older adults, brain pathology for Alzheimer's Disease can begin in middle age, suggesting an ideal target for intervention. METHODS We conducted a systematic review from exercise intervention studies on cognitive function among healthy, middle-aged participants (45-65). We searched multiple databases (PubMed, PsycINFO, MEDLINE, Cochrane Central Register of Controlled Trials, Google Scholar) for studies using standard, validated, neuropsychological measures following either single- or multi-session interventions in cognitively-unimpaired, middle-aged adults. RESULTS We identified 13 eligible studies. There was notable heterogeneity across studies, with varying design, measures, interventions, and results. Results from single-session studies showed improvement in response inhibition, while results for cognitive flexibility were mixed. No significant changes were found on measures of attention, working memory, or processing speed. Results from multi-session studies were more varied. Verbal memory was found to improve while performance on tests of attention and working memory, processing speed, and executive function were mixed. CONCLUSION Importantly, for both single-session and multi-session studies, there was no standard set of neuropsychological tests administered, making it more difficult to synthesize the findings into a single narrative. We end with a discussion on future directions and implementation.
Collapse
Affiliation(s)
- Fayeza S Ahmed
- Department of Psychology, University of Maine, Orono, Maine, USA
| | | | | | - Peter Dearborn
- Department of Psychology, University of Maine, Orono, Maine, USA
| |
Collapse
|
13
|
Ururahy RDR, do Val MS, Ciciliati AMM, Leite REP, Paes VR, Rodrigues RD, Grinberg LT, Pasqualucci CA, Jacob Filho W, Suemoto CK. The Association Between Neuropathological Lesions and Body Mass Index Is Independent of Cognitive Abilities. J Alzheimers Dis 2024; 101:773-785. [PMID: 39213060 DOI: 10.3233/jad-231366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background The association of moderate and severe dementia with low body mass index (BMI) is well described, but weight decline seems to also occur in individuals with preclinical neuropathologies. Considering that up to one-fifth of individuals with normal cognition meet the criteria for a dementia-related neuropathological diagnosis, autopsy studies are key to detecting preclinical neurodegenerative and cerebrovascular diseases that could be underlying weight changes. Objective We investigated the association between dementia-related brain lesions and BMI and evaluated whether the cognitive function was a mediator of this association. Methods In 1,170 participants, sociodemographic data, clinical history, and cognitive post-mortem evaluation were assessed with an informant. Neuropathological evaluation was performed in all cases. Linear regression models were used to investigate the association between neuropathological lesions (exposure variable) and BMI (outcome) adjusted for demographic, clinical, and cognitive variables in the whole sample, and in only those with normal cognition. Corrections for multiple comparisons were performed. In addition, a mediation analysis was performed to investigate the direct and indirect effects of cognitive abilities on the association between neuropathology and BMI. Results Individuals with lower BMI had a higher burden of neuropathological lesions and poorer cognitive abilities. Only neurofibrillary tangles (NFT) and neuropathological comorbidity were associated with low BMI, while other neurodegenerative and cerebrovascular lesions were not. NFT were indirectly associated with BMI through cognitive abilities, and also directly, even in participants with normal cognition. Conclusions Neurofibrillary tangles were directly associated with low BMI even in individuals with preclinical Alzheimer's disease.
Collapse
Affiliation(s)
- Raul Dos Reis Ururahy
- Division of Geriatrics - Laboratório de Investigação Médica no Envelhecimento (LIM 66), University of São Paulo Medical School, São Paulo, Brazil
| | | | - Aline Maria Macagnan Ciciliati
- Division of Geriatrics - Laboratório de Investigação Médica no Envelhecimento (LIM 66), University of São Paulo Medical School, São Paulo, Brazil
| | | | - Vitor Ribeiro Paes
- Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | | | - Lea Tenenholz Grinberg
- Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
- Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | | | - Wilson Jacob Filho
- Division of Geriatrics - Laboratório de Investigação Médica no Envelhecimento (LIM 66), University of São Paulo Medical School, São Paulo, Brazil
| | - Claudia Kimie Suemoto
- Division of Geriatrics - Laboratório de Patologia Cardiovascular (LIM 22), University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
14
|
Wang RT, Sun Z, Tan CC, Tan L, Xu W. Dynamic Features of Body Mass Index in Late Life Predict Cognitive Trajectories and Alzheimer's Disease: A Longitudinal Study. J Alzheimers Dis 2024; 100:1365-1378. [PMID: 39031359 DOI: 10.3233/jad-240292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2024]
Abstract
Background The causal relationships of late-life body mass index (BMI) with Alzheimer's disease (AD) remains debated. Objective We aimed to assess the associations of dynamic BMI features (ΔBMIs) with cognitive trajectories, AD biomarkers, and incident AD risk. Methods We analyzed an 8-year cohort of 542 non-demented individuals who were aged ≥65 years at baseline and had BMI measurements over the first 4 years. ΔBMIs were defined as changing extent (change ≤ or >5%), variability (standard deviation), and trajectories over the first 4 years measured using latent class trajectory modeling. Linear mixed-effect models were utilized to examine the influence of ΔBMIs on changing rates of AD pathology biomarkers, hippocampus volume, and cognitive functions. Cox proportional hazards models were used to test the associations with AD risk. Stratified analyzes were conducted by the baseline BMI group and age. Results Over the 4-year period, compared to those with stable BMI, individuals who experienced BMI decreases demonstrated accelerated declined memory function (p = 0.006) and amyloid-β deposition (p = 0.034) while BMI increases were associated with accelerated hippocampal atrophy (p = 0.036). Three BMI dynamic features, including stable BMI, low BMI variability, and persistently high BMI, were associated with lower risk of incident AD (p < 0.005). The associations were validated over the 8-year period after excluding incident AD over the first 4 years. No stratified effects were revealed by the BMI group and age. Conclusions High and stable BMI in late life could predict better cognitive trajectory and lower risk of AD.
Collapse
Affiliation(s)
- Ruo-Tong Wang
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zhen Sun
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
15
|
Ding H, Wang B, Hamel AP, Melkonyan M, Ang TFA, Au R, Lin H. Prediction of Progression from Mild Cognitive Impairment to Alzheimer's disease with Longitudinal and Multimodal Data. FRONTIERS IN DEMENTIA 2023; 2:1271680. [PMID: 38895707 PMCID: PMC11185839 DOI: 10.3389/frdem.2023.1271680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Introduction Accurate prediction of the progression from mild cognitive impairment (MCI) to Alzheimer's disease (AD) within a certain time frame is crucial for appropriate therapeutic interventions. However, it is challenging to capture the dynamic changes in cognitive and functional abilities over time, resulting in limited predictive performance. Our study aimed to investigate whether incorporating longitudinal multimodal data with advanced analytical methods could improve the capability to predict the risk of progressing to AD. Methods This study included participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI), a large-scale multi-center longitudinal study. Three data modalities, including demographic variables, neuropsychological tests, and neuroimaging measures were considered. A Long Short-Term Memory (LSTM) model using data collected at five-time points (baseline, 6-month, 12-month, 18-month, and 24-month) was developed to predict the risk of progression from MCI to AD within two years from the index exam (the exam at 24-month). In contrast, a random forest model was developed to predict the risk of progression just based on the data collected at the index exam. Results The study included 347 participants with MCI at 24-month (age: mean 75, SD 7 years; 39.8% women) from ADNI, of whom 77 converted to AD over a 2-year follow-up period. The longitudinal LSTM model showed superior prediction performance of MCI-to-AD progression (AUC 0.93±0.06) compared to the random forest model (AUC 0.90±0.09). A similar pattern was also observed across different age groups. Discussion Our study suggests that the incorporation of longitudinal data can provide better predictive performance for 2-year MCI-to-AD progression risk than relying solely on cross-sectional data. Therefore, repeated or multiple times routine health surveillance of MCI patients are essential in the early detection and intervention of AD.
Collapse
Affiliation(s)
- Huitong Ding
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Biqi Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alexander P Hamel
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mark Melkonyan
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ting F. A. Ang
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | | | - Rhoda Au
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- The Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Departments of Neurology and Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
16
|
Lee S, Byun MS, Yi D, Kim MJ, Jung JH, Kong N, Jung G, Ahn H, Lee JY, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Body mass index and two-year change of in vivo Alzheimer's disease pathologies in cognitively normal older adults. Alzheimers Res Ther 2023; 15:108. [PMID: 37312229 DOI: 10.1186/s13195-023-01259-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 06/01/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND Low body mass index (BMI) or underweight status in late life is associated with an increased risk of dementia or Alzheimer's disease (AD). However, the relationship between late-life BMI and prospective longitudinal changes of in-vivo AD pathology has not been investigated. METHODS This prospective longitudinal study was conducted as part of the Korean Brain Aging Study for Early Diagnosis and Prediction of Alzheimer's Disease (KBASE). A total of 194 cognitive normal older adults were included in the analysis. BMI at baseline was measured, and two-year changes in brain Aβ and tau deposition on PET imaging were used as the main outcomes. Linear mixed-effects (LME) models were used to examine the relationships between late-life BMI and longitudinal change in AD neuropathological biomarkers. RESULTS A lower BMI at baseline was significantly associated with a greater increase in tau deposition in AD-signature region over 2 years (β, -0.018; 95% CI, -0.028 to -0.004; p = .008), In contrast, BMI was not related to two-year changes in global Aβ deposition (β, 0.0002; 95% CI, -0.003 to 0.002, p = .671). An additional exploratory analysis for each sex showed lower baseline BMI was associated with greater increases in tau deposition in males (β, -0.027; 95% CI, -0.046 to -0.009; p = 0.007), but not in females. DISCUSSION The findings suggest that lower BMI in late-life may predict or contribute to the progression of tau pathology over the subsequent years in cognitively unimpaired older adults.
Collapse
Affiliation(s)
- Seunghoon Lee
- Department of Psychiatry, Myongji Hospital, Hanyang University College of Medicine, Goyang, 10475, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Min Jung Kim
- Department of Neuropsychiatry, Nowon Eulji University Hospital, Seoul, 01830, Republic of Korea
| | - Joon Hyung Jung
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Nayeong Kong
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Hyejin Ahn
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
17
|
So SW, Fleming KM, Nixon JP, Butterick TA. Early Life Obesity Increases Neuroinflammation, Amyloid Beta Deposition, and Cognitive Decline in a Mouse Model of Alzheimer's Disease. Nutrients 2023; 15:nu15112494. [PMID: 37299457 DOI: 10.3390/nu15112494] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity, a known risk factor of Alzheimer's disease (AD), increases the activation of microglia, leading to a proinflammatory phenotype. Our previous work shows that a high fat diet (HFD) can cause neuroinflammation and cognitive decline in mice. We hypothesized that proinflammatory activation of brain microglia in obesity exacerbates AD pathology and increases the accumulation of amyloid beta (Aβ) plaques. Presently, we tested cognitive function in 8-month-old male and female APP/PS1 mice fed a HFD, starting at 1.5 months of age. Locomotor activity, anxiety-like behavior, behavioral despair, and spatial memory were all assessed through behavioral tests. Microgliosis and Aβ deposition were measured in multiple brain regions through immunohistochemical analysis. Our results show that a HFD decreases locomotor activity, while increasing anxiety-like behavior and behavioral despair independent of genotype. A HFD led to increased memory deficits in both sexes, with HFD-fed APP/PS1 mice performing the worst out of all groups. Immunohistochemical analysis showed increased microgliosis in mice fed a HFD. This was accompanied by an increase in Aβ deposition in the HFD-fed APP/PS1 mice. Together, our results support that HFD-induced obesity exacerbates neuroinflammation and Aβ deposition in a young adult AD mouse model, leading to increased memory deficits and cognitive decline in both sexes.
Collapse
Affiliation(s)
- Simon W So
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Kendra M Fleming
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Joshua P Nixon
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Food Science and Nutrition, University of Minnesota Twin Cities, St. Paul, MN 55108, USA
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Tammy A Butterick
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN 55417, USA
- Department of Neuroscience, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
- Department of Food Science and Nutrition, University of Minnesota Twin Cities, St. Paul, MN 55108, USA
| |
Collapse
|
18
|
Müller L, Power Guerra N, Schildt A, Lindner T, Stenzel J, Behrangi N, Bergner C, Alberts T, Bühler D, Kurth J, Krause BJ, Janowitz D, Teipel S, Vollmar B, Kuhla A. [ 18F]GE-180-PET and Post Mortem Marker Characteristics of Long-Term High-Fat-Diet-Induced Chronic Neuroinflammation in Mice. Biomolecules 2023; 13:biom13050769. [PMID: 37238638 DOI: 10.3390/biom13050769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/14/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity is characterized by immoderate fat accumulation leading to an elevated risk of neurodegenerative disorders, along with a host of metabolic disturbances. Chronic neuroinflammation is a main factor linking obesity and the propensity for neurodegenerative disorders. To determine the cerebrometabolic effects of diet-induced obesity (DIO) in female mice fed a long-term (24 weeks) high-fat diet (HFD, 60% fat) compared to a group on a control diet (CD, 20% fat), we used in vivo PET imaging with the radiotracer [18F]FDG as a marker for brain glucose metabolism. In addition, we determined the effects of DIO on cerebral neuroinflammation using translocator protein 18 kDa (TSPO)-sensitive PET imaging with [18F]GE-180. Finally, we performed complementary post mortem histological and biochemical analyses of TSPO and further microglial (Iba1, TMEM119) and astroglial (GFAP) markers as well as cerebral expression analyses of cytokines (e.g., Interleukin (IL)-1β). We showed the development of a peripheral DIO phenotype, characterized by increased body weight, visceral fat, free triglycerides and leptin in plasma, as well as increased fasted blood glucose levels. Furthermore, we found obesity-associated hypermetabolic changes in brain glucose metabolism in the HFD group. Our main findings with respect to neuroinflammation were that neither [18F]GE-180 PET nor histological analyses of brain samples seem fit to detect the predicted cerebral inflammation response, despite clear evidence of perturbed brain metabolism along with elevated IL-1β expression. These results could be interpreted as a metabolically activated state in brain-resident immune cells due to a long-term HFD.
Collapse
Affiliation(s)
- Luisa Müller
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Department of Psychosomatic Medicine and Psychotherapy, Rostock University Medical Centre, 18147 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
| | - Nicole Power Guerra
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Institute of Anatomy, Rostock University Medical Centre, 18057 Rostock, Germany
- Smell & Taste Clinic, Department of Otorhinolaryngology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01034 Dresden, Germany
| | - Anna Schildt
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Tobias Lindner
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Jan Stenzel
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Newshan Behrangi
- Institute of Anatomy and Cell Biology, Medical University of Bonn, 53115 Bonn, Germany
| | - Carina Bergner
- Department of Clinic and Polyclinic for Nuclear Medicine, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Teresa Alberts
- Institute of Anatomy and Cell Biology, Medical University of Bonn, 53115 Bonn, Germany
| | - Daniel Bühler
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Jens Kurth
- Department of Clinic and Polyclinic for Nuclear Medicine, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Bernd Joachim Krause
- Department of Clinic and Polyclinic for Nuclear Medicine, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Deborah Janowitz
- Department of Psychiatry, University of Greifswald, 17475 Greifswald, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, Rostock University Medical Centre, 18147 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, 18147 Rostock, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
| | - Angela Kuhla
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
| |
Collapse
|
19
|
Grapsa I, Mamalaki E, Ntanasi E, Kosmidis MH, Dardiotis E, Hadjigeorgiou GM, Sakka P, Scarmeas N, Yannakoulia M. Longitudinal Examination of Body Mass Index and Cognitive Function in Older Adults: The HELIAD Study. Nutrients 2023; 15:nu15071795. [PMID: 37049637 PMCID: PMC10096583 DOI: 10.3390/nu15071795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
Given the increase in the aging population and thus in the prevalence of dementia, the identification of protective factors against cognitive decline is necessary. In a cohort of 1076 non-demented adults ≥ 65 years old (59.7% women) from the HELIAD study, we assessed whether changes in body mass index (BMI) were associated with changes in cognition over a 3-year follow-up period separately for those ≤ 75 and >75 years old. We identified six BMI trajectory groups based on participants' BMI status at baseline and at the first follow-up visit; normal to normal BMI was the reference group. Major cognitive domains were evaluated, and a composite index reflecting global cognition was calculated. In participants aged ≤75 years, weight loss-moving from obesity to overweight or normal BMI-was associated with less decline in the memory composite score over time (β = 0.141; p = 0.035), while 3-year maintenance of a BMI ≥ 25 kg/m2 was related to greater reduction in the visuospatial composite score over time (β = -0.093; p = 0.020). Regarding participants aged >75 years, 3-year maintenance of a BMI ≥ 30 kg/m2 contributed to a slower rate of decline in the memory composite score over time (β = 0.102; p = 0.042), whereas weight loss-from overweight to normal BMI-was associated with a decreased attention/processing speed composite score longitudinally (β = -0.275; p = 0.043). Our findings indicated that the association between changes in BMI and cognitive functioning was modified by age. Weight management may have the potential to delay cognitive decline in older adults.
Collapse
Affiliation(s)
- Ismini Grapsa
- Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece
| | - Eirini Mamalaki
- Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Eva Ntanasi
- Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Mary H Kosmidis
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece
| | | | - Paraskevi Sakka
- Athens Association of Alzheimer's Disease and Related Disorders, 11636 Maroussi, Greece
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, The Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University of Athens, 17671 Athens, Greece
| |
Collapse
|
20
|
Suzzi S, Croese T, Ravid A, Gold O, Clark AR, Medina S, Kitsberg D, Adam M, Vernon KA, Kohnert E, Shapira I, Malitsky S, Itkin M, Brandis A, Mehlman T, Salame TM, Colaiuta SP, Cahalon L, Slyper M, Greka A, Habib N, Schwartz M. N-acetylneuraminic acid links immune exhaustion and accelerated memory deficit in diet-induced obese Alzheimer's disease mouse model. Nat Commun 2023; 14:1293. [PMID: 36894557 PMCID: PMC9998639 DOI: 10.1038/s41467-023-36759-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 02/16/2023] [Indexed: 03/11/2023] Open
Abstract
Systemic immunity supports lifelong brain function. Obesity posits a chronic burden on systemic immunity. Independently, obesity was shown as a risk factor for Alzheimer's disease (AD). Here we show that high-fat obesogenic diet accelerated recognition-memory impairment in an AD mouse model (5xFAD). In obese 5xFAD mice, hippocampal cells displayed only minor diet-related transcriptional changes, whereas the splenic immune landscape exhibited aging-like CD4+ T-cell deregulation. Following plasma metabolite profiling, we identified free N-acetylneuraminic acid (NANA), the predominant sialic acid, as the metabolite linking recognition-memory impairment to increased splenic immune-suppressive cells in mice. Single-nucleus RNA-sequencing revealed mouse visceral adipose macrophages as a potential source of NANA. In vitro, NANA reduced CD4+ T-cell proliferation, tested in both mouse and human. In vivo, NANA administration to standard diet-fed mice recapitulated high-fat diet effects on CD4+ T cells and accelerated recognition-memory impairment in 5xFAD mice. We suggest that obesity accelerates disease manifestation in a mouse model of AD via systemic immune exhaustion.
Collapse
Grants
- R01 DK095045 NIDDK NIH HHS
- R01 DK099465 NIDDK NIH HHS
- the Vera and John Schwartz Family Center for Metabolic Biology.
- the National Institutes of Health (NIH) grants DK095045 and DK099465, the Cure Alzheimer’s Fund, the Chan Zuckerberg Foundation, and the Carlos Slim Foundation.
- the Israel Science Foundation (ISF) research grant no. 1709/19, the European Research Council grant 853409, the MOST-IL-China research grant no. 3-15687, and the Myers Foundation. N.H. holds the Goren-Khazzam chair in neuroscience.
- the Advanced European Research Council grants 232835 and 741744, the European Seventh Framework Program HEALTH-2011 (279017), the Israel Science Foundation (ISF)-research grant no. 991/16, the ISF-Legacy Heritage Bio-medical Science Partnership research grant no. 1354/15, and the Thompson Foundation and Adelis Foundation.
Collapse
Affiliation(s)
- Stefano Suzzi
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel.
| | - Tommaso Croese
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Adi Ravid
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Or Gold
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Abbe R Clark
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sedi Medina
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Daniel Kitsberg
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Miriam Adam
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Katherine A Vernon
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Eva Kohnert
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Inbar Shapira
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Sergey Malitsky
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Maxim Itkin
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Alexander Brandis
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Tevie Mehlman
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Tomer M Salame
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Sarah P Colaiuta
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Liora Cahalon
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Michal Slyper
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Naomi Habib
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel.
| | - Michal Schwartz
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel.
| |
Collapse
|
21
|
Luo D, Qi X, Xu X, Yang L, Yu C, Guan Q. Involvement of p38 MAPK in Leydig cell aging and age-related decline in testosterone. Front Endocrinol (Lausanne) 2023; 14:1088249. [PMID: 36950685 PMCID: PMC10025507 DOI: 10.3389/fendo.2023.1088249] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Introduction Age-related decline in testosterone is associated with Leydig cell aging with impaired testosterone synthesis in aging. Obesity accelerates the age-related decline in testosterone. However, the mechanisms underlying the Leydig cell aging and the effects of obesity on Leydig cell aging remain unclear. Method Natural aging mice and diet-induced obese mice were used to assess the process of testicular Leydig cell senescence with age or obesity. Bioinformatic analysis of the young and aged human testes was used to explore key genes related Leydig cell aging. Leydig cell-specific p38 MAPK knockout (p38LCKO) mice were used to further analyze the roles of p38 MAPK in Leydig cell aging. The levels of testosterone and steroidogenic enzymes, activity of p38 MAPK, aging status of Leydig cells, and oxidative stress and inflammation of testes or Leydig cells were detected by ELISA, immunoblotting, immunofluorescence, and senescence-associated β-galactosidase (SA-β-Gal) staining analysis, respectively. Result The serum testosterone level was significantly reduced in aged mice compared with young mice. In the testis of aged mice, the reduced mRNA and protein levels of LHCGR, SRB1, StAR, CYP11A1, and CYP17A1 and the elevated oxidative stress and inflammation were observed. KEGG analysis showed that MAPK pathway was changed in aged Leydig cells, and immunoblotting displayed that p38 MAPK was activated in aged Leydig cells. The intensity of SA-β-Gal staining on Leydig cells and the number of p21-postive Leydig cells in aged mice were more than those of young mice. Similar to aged mice, the testosterone-related indexes decreased, and the age-related indexes increased in the testicular Leydig cells of high fat diet (HFD) mice. Aged p38LCKO mice had higher levels of testosterone and steroidogenic enzymes than those of age-matched wild-type (WT) littermates, with reduced the intensity of SA-β-Gal staining and the expression of p21 protein. Conclusion Our study suggested that obesity was an important risk factor for Leydig cell aging. p38 MAPK was involved in Leydig cell aging induced by age and obesity. The inhibition of p38 MAPK could delay Leydig cell aging and alleviate decline in testosterone.
Collapse
Affiliation(s)
- Dandan Luo
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Xiangyu Qi
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Xiaoqin Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Leilei Yang
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Chunxiao Yu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Qingbo Guan
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| |
Collapse
|
22
|
Zary N, Adcock-Omlin M, de Bruin ED. Design Considerations for an Exergame-Based Training Intervention for Older Adults With Mild Neurocognitive Disorder: Qualitative Study Including Focus Groups With Experts and Health Care Professionals and Individual Semistructured In-depth Patient Interviews. JMIR Serious Games 2023; 11:e37616. [PMID: 36602851 PMCID: PMC9853342 DOI: 10.2196/37616] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/28/2022] [Accepted: 10/10/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Exergames have attracted growing interest in the prevention and treatment of neurocognitive disorders. The most effective exergame and training components (ie, exercise and training variables such as frequency, intensity, duration, or volume of training and type and content of specific exergame scenarios) however remain to be established for older adults with mild neurocognitive disorders (mNCDs). Regarding the design and development of novel exergame-based training concepts, it seems of crucial importance to explicitly include the intended users' perspective by adopting an interactive and participatory design that includes end users throughout different iterative cycles of development. OBJECTIVE This study aimed to determine the capabilities, treatment preferences, and motivators for the training of older adults with mNCD and the perspectives of individuals on training goals and settings and requirements for exergame and training components. METHODS A qualitative study including expert focus groups and individual semistructured in-depth patient interviews was conducted. Data were transcribed to a written format to perform qualitative content analysis using QCAmap software. RESULTS In total, 10 experts and health care professionals (80% females) and 8 older adults with mNCD (38% females; mean age 82.4, SD 6.2 years) were recruited until data saturation was observed. CONCLUSIONS The psychosocial consequences of patients' self-perceived cognitive deterioration might be more burdensome than the cognitive changes themselves. Older adults with mNCD prefer integrative forms of training (such as exergaming) and are primarily motivated by enjoyment or fun in exercising and the effectiveness of the training. Putting the synthesized perspectives of training goals, settings, and requirements for exergames and training components into context, our considerations point to opportunities for improvement in research and rehabilitation, either by adapting existing exergames to patients with mNCDs or by developing novel exergames and exergame-based training concepts specifically tailored to meet patient requirements and needs.
Collapse
Affiliation(s)
| | - Manuela Adcock-Omlin
- Motor Control and Learning Group - Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Eling D de Bruin
- Motor Control and Learning Group - Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.,Division of Physiotherapy, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Health, OST - Eastern Swiss University of Applied Sciences, St.Gallen, Switzerland
| |
Collapse
|
23
|
Morys F, Potvin O, Zeighami Y, Vogel J, Lamontagne-Caron R, Duchesne S, Dagher A. Obesity-Associated Neurodegeneration Pattern Mimics Alzheimer's Disease in an Observational Cohort Study. J Alzheimers Dis 2023; 91:1059-1071. [PMID: 36565111 PMCID: PMC9912737 DOI: 10.3233/jad-220535] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Excess weight in adulthood leads to health complications such as diabetes, hypertension, or dyslipidemia. Recently, excess weight has also been related to brain atrophy and cognitive decline. Reports show that obesity is linked with Alzheimer's disease (AD)-related changes, such as cerebrovascular damage or amyloid-β accumulation. However, to date no research has conducted a direct comparison between brain atrophy patterns in AD and obesity. OBJECTIVE Here, we compared patterns of brain atrophy and amyloid-β/tau protein accumulation in obesity and AD using a sample of over 1,300 individuals from four groups: AD patients, healthy controls, obese otherwise healthy individuals, and lean individuals. METHODS We age- and sex-matched all groups to the AD-patients group and created cortical thickness maps of AD and obesity. This was done by comparing AD patients with healthy controls, and obese individuals with lean individuals. We then compared the AD and obesity maps using correlation analyses and permutation-based tests that account for spatial autocorrelation. Similarly, we compared obesity brain maps with amyloid-β and tau protein maps from other studies. RESULTS Obesity maps were highly correlated with AD maps but were not correlated with amyloid-β/tau protein maps. This effect was not accounted for by the presence of obesity in the AD group. CONCLUSION Our research confirms that obesity-related grey matter atrophy resembles that of AD. Excess weight management could lead to improved health outcomes, slow down cognitive decline in aging, and lower the risk for AD.
Collapse
Affiliation(s)
- Filip Morys
- Montreal Neurological Institute, McGill University, Montréal, Canada
| | | | - Yashar Zeighami
- Montreal Neurological Institute, McGill University, Montréal, Canada
- Department of Psychiatry, McGill University, Québec, Canada
| | - Jacob Vogel
- Montreal Neurological Institute, McGill University, Montréal, Canada
| | | | - Simon Duchesne
- CERVO Brain Research Centre, Québec, Canada
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Laval University, Québec, Canada
| | - Alain Dagher
- Montreal Neurological Institute, McGill University, Montréal, Canada
| | | |
Collapse
|
24
|
Brook ES, D'Alonzo ZJ, Lam V, Chan DC, Dhaliwal SS, Watts GF, Mamo JCL, Takechi R. Plasma Amyloid-β Homeostasis Is Associated with Body Mass Index and Weight Loss in People with Overweight and Obesity. J Alzheimers Dis 2023; 93:653-664. [PMID: 37066906 DOI: 10.3233/jad-220529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND Obesity is linked to a higher incidence of Alzheimer's disease (AD). Studies show that plasma amyloid-β (Aβ) dyshomeostasis, particularly low 42/40 ratio indicates a heightened risk for developing AD. However, the relationship between body mass index (BMI) and circulating plasma Aβ has not been extensively studied. OBJECTIVE We hypothesized that people with a high BMI have altered plasma Aβ homeostasis compared with people with a lower BMI. We also tested whether reducing BMI by calorie-restriction could normalize plasma concentrations of Aβ. METHODS Plasma concentrations of Aβ40, Aβ42, and Aβ42/40 ratio were measured in 106 participants with BMIs classified as lean, overweight, or obese. From this cohort, twelve participants with overweight or obese BMIs entered a 12-week calorie-restriction weight loss program. We then tested whether decreasing BMI affected plasma Aβ concentrations. RESULTS Plasma Aβ42/40 ratio was 17.54% lower in participants with an obese BMI compared to lean participants (p < 0.0001), and 11.76% lower compared to participants with an overweight BMI (p < 0.0001). The weight loss regimen decreased BMI by an average of 4.02% (p = 0.0005) and was associated with a 6.5% decrease in plasma Aβ40 (p = 0.0425). However, weight loss showed negligible correlations with plasma Aβ40, Aβ42, and Aβ42/40 ratio. CONCLUSION Obesity is associated with aberrant plasma Aβ homeostasis which may be associated with an increased risk for AD. Weight loss appears to lower Aβ40, but large-scale longitudinal studies in addition to molecular studies are required to elucidate the underlying mechanisms of how obesity and weight loss influence plasma Aβ homeostasis.
Collapse
Affiliation(s)
- Emily S Brook
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Bentley, Australia
| | - Zachary J D'Alonzo
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Bentley, Australia
| | - Virginie Lam
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, Australia
| | - Dick C Chan
- Medical School, University of Western Australia, Perth, Australia
| | - Satvinder S Dhaliwal
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- Duke-NUS Medical School, National University of Singapore, Singapore
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Minden, Pulau Pinang, Malaysia
- Singapore University of Social Sciences, Singapore
| | - Geraldb F Watts
- Medical School, University of Western Australia, Perth, Australia
- Cardiometabolic Service, Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia
| | - John C L Mamo
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, Australia
- Perron Institute of Neurological and Translational Sciences, Nedlands, Australia
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia
- School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, Australia
| |
Collapse
|
25
|
Weise CM, Chen K, Chen Y, Devadas V, Su Y, Reiman EM. Differential impact of body mass index and leptin on baseline and longitudinal positron emission tomography measurements of the cerebral metabolic rate for glucose in amnestic mild cognitive impairment. Front Aging Neurosci 2022; 14:1031189. [PMID: 36570534 PMCID: PMC9782536 DOI: 10.3389/fnagi.2022.1031189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022] Open
Abstract
Introduction Several studies have suggested that greater adiposity in older adults is associated with a lower risk of Alzheimer's disease (AD) related cognitive decline, some investigators have postulated that this association may be due to the protective effects of the adipose tissue-derived hormone leptin. In this study we sought to demonstrate that higher body mass indices (BMIs) are associated with greater baseline FDG PET measurements of the regional cerebral metabolic rate for glucose (rCMRgl), a marker of local neuronal activity, slower rCMRgl declines in research participants with amnestic mild cognitive impairment (aMCI). We then sought to clarify the extent to which those relationships are attributable to cerebrospinal fluid (CSF) or plasma leptin concentrations. Materials and methods We used baseline PET images from 716 73 ± 8 years-old aMCI participants from the AD Neuroimaging Initiative (ADNI) of whom 453 had follow up images (≥6 months; mean follow up time 3.3 years). For the leptin analyses, we used baseline CSF samples from 81 of the participants and plasma samples from 212 of the participants. Results As predicted, higher baseline BMI was associated with greater baseline CMRgl measurements and slower declines within brain regions preferentially affected by AD. In contrast and independently of BMI, CSF, and plasma leptin concentrations were mainly related to less baseline CMRgl within mesocorticolimbic brain regions implicated in energy homeostasis. Discussion While higher BMIs are associated with greater baseline CMRgl and slower declines in persons with aMCI, these associations appear not to be primarily attributable to leptin concentrations.
Collapse
Affiliation(s)
- Christopher M. Weise
- Department of Neurology, Marti-Luther-University of Halle-Wittenberg, Halle, Germany,Department of Neurology, University of Leipzig, Leipzig, Germany,*Correspondence: Christopher M. Weise,
| | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, United States,School of Mathematics and Statistics, Arizona State University, Tempe, AZ, United States,Department of Neurology, College of Medicine, University of Arizona, Phoenix, AZ, United States,Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Yinghua Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
| | - Vivek Devadas
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
| | - Yi Su
- Banner Alzheimer’s Institute, Phoenix, AZ, United States,Department of Neurology, College of Medicine, University of Arizona, Phoenix, AZ, United States,Arizona Alzheimer’s Consortium, Phoenix, AZ, United States,School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, United States
| | - Eric M. Reiman
- Banner Alzheimer’s Institute, Phoenix, AZ, United States,Arizona Alzheimer’s Consortium, Phoenix, AZ, United States,Department of Psychiatry, College of Medicine, University of Arizona, Phoenix, AZ, United States,Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States,Arizona State University-Banner Health Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
26
|
Yau WYW, Shirzadi Z, Yang HS, Ikoba AP, Rabin JS, Properzi MJ, Kirn DR, Schultz AP, Rentz DM, Johnson KA, Sperling RA, Chhatwal JP. Tau Mediates Synergistic Influence of Vascular Risk and Aβ on Cognitive Decline. Ann Neurol 2022; 92:745-755. [PMID: 35880989 PMCID: PMC9650958 DOI: 10.1002/ana.26460] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Elevated vascular risk and beta-amyloid (Aβ) burden have been synergistically associated with cognitive decline in preclinical Alzheimer's disease (AD), although the underlying mechanisms remain unclear. We examined whether accelerated longitudinal tau accumulation mediates the vascular risk-Aβ interaction on cognitive decline. METHODS We included 175 cognitively unimpaired older adults (age 70.5 ± 8.0 years). Baseline vascular risk was quantified using the office-based Framingham Heart Study general cardiovascular disease risk score (FHS-CVD). Baseline Aβ burden was measured with Pittsburgh Compound-B positron emission tomography (PET). Tau burden was measured longitudinally (3.6 ± 1.5 years) with Flortaucipir PET, focusing on inferior temporal cortex (ITC). Cognition was assessed longitudinally (7.0 ± 2.0 years) using the Preclinical Alzheimer's Cognitive Composite. Linear mixed effects models examined the interactive effects of baseline vascular risk and Aβ on longitudinal ITC tau. Additionally, moderated mediation was used to determine whether tau accumulation mediated the FHS-CVD*Aβ effect on cognitive decline. RESULTS We observed a significant interaction between elevated baseline FHS-CVD and Aβ on greater ITC tau accumulation (p = 0.004), even in individuals with Aβ burden below the conventional threshold for amyloid positivity. Examining individual vascular risk factors, we found elevated systolic blood pressure and body mass index showed independent interactions with Aβ on longitudinal tau (both p < 0.0001). ITC tau accumulation mediated 33% of the interactive association of FHS-CVD and Aβ on cognitive decline. INTERPRETATION Vascular risks interact with subthreshold levels of Aβ to promote cognitive decline, partially by accelerating early neocortical tau accumulation. Our findings support vascular risk reduction, especially treating hypertension and obesity, to attenuate Aβ-related tau pathology and reduce late-life cognitive decline. ANN NEUROL 2022;92:745-755.
Collapse
Affiliation(s)
- Wai-Ying Wendy Yau
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Zahra Shirzadi
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
| | - Akpevweoghene P Ikoba
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Jennifer S Rabin
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Dylan R Kirn
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
27
|
Brinkley TE, Leng I, Register TC, Neth BJ, Zetterberg H, Blennow K, Craft S. Changes in Adiposity and Cerebrospinal Fluid Biomarkers Following a Modified Mediterranean Ketogenic Diet in Older Adults at Risk for Alzheimer’s Disease. Front Neurosci 2022; 16:906539. [PMID: 35720727 PMCID: PMC9202553 DOI: 10.3389/fnins.2022.906539] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/06/2022] [Indexed: 12/03/2022] Open
Abstract
Background Ketogenic diets have been used to treat both obesity and neurological disorders, including epilepsy and more recently Alzheimer’s disease (AD), likely due to favorable effects on both central and peripheral metabolism. Improvements in body composition have also been reported; however, it is unclear if diet-induced changes in adiposity are related to improvements in AD and related neuropathology. Purpose We examined the effects of a Modified Mediterranean Ketogenic (MMK) diet vs. an American Heart Association (AHA) diet on body weight, body composition, and body fat distribution and their association with cerebrospinal fluid (CSF) biomarkers in older adults at risk for AD. Methods Twenty adults (mean age: 64.3 ± 6.3 years, 35% Black, 75% female) were randomly assigned to a crossover trial starting with either the MMK or AHA diet for 6 weeks, followed by a 6-week washout and then the opposite diet for 6 weeks. At baseline and after each diet adiposity was assessed by dual-energy x-ray absorptiometry and CSF biomarkers were measured. Linear mixed effect models were used to examine the effect of diet on adiposity. Spearman correlations were examined to assess associations between adiposity and CSF biomarkers. Results At baseline there was a high prevalence of overweight/obesity and central adiposity, and higher visceral fat and lower peripheral fat were associated with an adverse CSF biomarker profile. The MMK and AHA diets led to similar improvements in body composition and body fat distribution. Significant correlations were found between changes in adiposity and changes in CSF biomarkers (r’s = 0.63–0.92, p’s < 0.05), with notable differences by diet. Decreases in body fat on the MMK diet were related to changes in Aβ biomarkers, whereas decreases in body fat on the AHA diet were related to changes in tau biomarkers and cholinesterase activity. Interestingly, increases in CSF Aβ on the MMK diet occurred in those with less fat loss. Conclusion An MMK diet leads to favorable changes in body composition, body fat distribution, and CSF biomarkers. Our data suggest that modest weight loss that maximizes visceral fat loss and preserves peripheral fat, may have the greatest impact on brain health. Clinical Trial Registration [www.ClinicalTrials.gov], identifier [NCT02984540].
Collapse
Affiliation(s)
- Tina E. Brinkley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Winston-Salem, NC, United States
- *Correspondence: Tina E. Brinkley,
| | - Iris Leng
- Division of Public Health Sciences, Department of Biostatistics and Data Sciences, Winston-Salem, NC, United States
| | - Thomas C. Register
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Bryan J. Neth
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
- United Kingdom Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, Hong Kong SAR, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Winston-Salem, NC, United States
| |
Collapse
|
28
|
Austad SN, Ballinger S, Buford TW, Carter CS, Smith DL, Darley-Usmar V, Zhang J. Targeting whole body metabolism and mitochondrial bioenergetics in the drug development for Alzheimer's disease. Acta Pharm Sin B 2022; 12:511-531. [PMID: 35256932 PMCID: PMC8897048 DOI: 10.1016/j.apsb.2021.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Aging is by far the most prominent risk factor for Alzheimer's disease (AD), and both aging and AD are associated with apparent metabolic alterations. As developing effective therapeutic interventions to treat AD is clearly in urgent need, the impact of modulating whole-body and intracellular metabolism in preclinical models and in human patients, on disease pathogenesis, have been explored. There is also an increasing awareness of differential risk and potential targeting strategies related to biological sex, microbiome, and circadian regulation. As a major part of intracellular metabolism, mitochondrial bioenergetics, mitochondrial quality-control mechanisms, and mitochondria-linked inflammatory responses have been considered for AD therapeutic interventions. This review summarizes and highlights these efforts.
Collapse
Key Words
- ACE2, angiotensin I converting enzyme (peptidyl-dipeptidase A) 2
- AD, Alzheimer's disease
- ADP, adenosine diphosphate
- ADRD, AD-related dementias
- Aβ, amyloid β
- CSF, cerebrospinal fluid
- Circadian regulation
- DAMPs
- DAMPs, damage-associated molecular patterns
- Diabetes
- ER, estrogen receptor
- ETC, electron transport chain
- FCCP, trifluoromethoxy carbonylcyanide phenylhydrazone
- FPR-1, formyl peptide receptor 1
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide-1
- HBP, hexoamine biosynthesis pathway
- HTRA, high temperature requirement A
- Hexokinase biosynthesis pathway
- I3A, indole-3-carboxaldehyde
- IRF-3, interferon regulatory factor 3
- LC3, microtubule associated protein light chain 3
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- MAVS, mitochondrial anti-viral signaling
- MCI, mild cognitive impairment
- MRI, magnetic resonance imaging
- MRS, magnetic resonance spectroscopy
- Mdivi-1, mitochondrial division inhibitor 1
- Microbiome
- Mitochondrial DNA
- Mitochondrial electron transport chain
- Mitochondrial quality control
- NLRP3, leucine-rich repeat (LRR)-containing protein (NLR)-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- NeuN, neuronal nuclear protein
- PET, fluorodeoxyglucose (FDG)-positron emission tomography
- PKA, protein kinase A
- POLβ, the base-excision repair enzyme DNA polymerase β
- ROS, reactive oxygen species
- Reactive species
- SAMP8, senescence-accelerated mice
- SCFAs, short-chain fatty acids
- SIRT3, NAD-dependent deacetylase sirtuin-3
- STING, stimulator of interferon genes
- STZ, streptozotocin
- SkQ1, plastoquinonyldecyltriphenylphosphonium
- T2D, type 2 diabetes
- TCA, Tricarboxylic acid
- TLR9, toll-like receptor 9
- TMAO, trimethylamine N-oxide
- TP, tricyclic pyrone
- TRF, time-restricted feeding
- cAMP, cyclic adenosine monophosphate
- cGAS, cyclic GMP/AMP synthase
- hAPP, human amyloid precursor protein
- hPREP, human presequence protease
- i.p., intraperitoneal
- mTOR, mechanistic target of rapamycin
- mtDNA, mitochondrial DNA
- αkG, alpha-ketoglutarate
Collapse
Affiliation(s)
- Steven N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christy S. Carter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
29
|
Coad BM, Ghomroudi PA, Sims R, Aggleton JP, Vann SD, Metzler-Baddeley C. Apolipoprotein ε4 modifies obesity-related atrophy in the hippocampal formation of cognitively healthy adults. Neurobiol Aging 2022; 113:39-54. [PMID: 35303671 PMCID: PMC9084919 DOI: 10.1016/j.neurobiolaging.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/18/2022] [Accepted: 02/12/2022] [Indexed: 12/02/2022]
Abstract
Age-related inverted U-shaped curve of hippocampal myelin/neurite packing. Reduced hippocampal myelin/neurite packing and size/complexity in obesity. APOE modifies the effects of obesity on hippocampal size/complexity. Age-related slowing of spatial navigation but no risk effects on cognition. CA/DG predict episodic memory and subiculum predicts spatial navigation performance.
Characterizing age- and risk-related hippocampal vulnerabilities may inform about the neural underpinnings of cognitive decline. We studied the impact of three risk-factors, Apolipoprotein (APOE)-ε4, a family history of dementia, and central obesity, on the CA1, CA2/3, dentate gyrus and subiculum of 158 cognitively healthy adults (38-71 years). Subfields were labelled with the Automatic Segmentation of Hippocampal Subfields and FreeSurfer (version 6) protocols. Volumetric and microstructural measurements from quantitative magnetization transfer and Neurite Orientation Density and Dispersion Imaging were extracted for each subfield and reduced to three principal components capturing apparent myelin/neurite packing, size/complexity, and metabolism. Aging was associated with an inverse U-shaped curve on myelin/neurite packing and affected all subfields. Obesity led to reductions in myelin/neurite packing and size/complexity regardless of APOE and family history of dementia status. However, amongst individuals with a healthy Waist-Hip-Ratio, APOE ε4 carriers showed lower size/complexity than non-carriers. Segmentation protocol type did not affect this risk pattern. These findings reveal interactive effects between APOE and central obesity on the hippocampal formation of cognitively healthy adults.
Collapse
|
30
|
Manser P, de Bruin ED. Making the Best Out of IT: Design and Development of Exergames for Older Adults With Mild Neurocognitive Disorder - A Methodological Paper. Front Aging Neurosci 2021; 13:734012. [PMID: 34955806 PMCID: PMC8698204 DOI: 10.3389/fnagi.2021.734012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/03/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Utilizing information technology (IT) systems, for example in form of computerized cognitive screening or exergame-based (also called active videogames) training, has gained growing interest for supporting healthy aging and to detect, prevent and treat neurocognitive disorders (NCD). To ameliorate the effectiveness of exergaming, the neurobiological mechanisms as well as the most effective components for exergame-based training remain to be established. At the same time, it is important to account for the end-users' capabilities, preferences, and therapeutic needs during the design and development process to foster the usability and acceptance of the resulting program in clinical practice. This will positively influence adherence to the resulting exergame-based training program, which, in turn, favors more distinct training-related neurobiological effects. Objectives and Methods: This methodological paper describes the design and development process of novel exergame-based training concepts guided by a recently proposed methodological framework: The 'Multidisciplinary Iterative Design of Exergames (MIDE): A Framework for Supporting the Design, Development, and Evaluation of Exergames for Health' (Li et al., 2020). Case Study: A step-by-step application of the MIDE-framework as a specific guidance in an ongoing project aiming to design, develop, and evaluate an exergame-based training concept with the aim to halt and/or reduce cognitive decline and improve quality of life in older adults with mild neurocognitive disorder (mNCD) is illustrated. Discussion and Conclusion: The development of novel exergame-based training concepts is greatly facilitated when it is based on a theoretical framework (e.g., the MIDE-framework). Applying this framework resulted in a structured, iterative, and evidence-based approach that led to the identification of multiple key requirements for the exergame design as well as the training components that otherwise may have been overlooked or neglected. This is expected to foster the usability and acceptance of the resulting exergame intervention in "real life" settings. Therefore, it is strongly recommended to implement a theoretical framework (e.g., the MIDE-framework) for future research projects in line with well-known checklists to improve completeness of reporting and replicability when serious games for motor-cognitive rehabilitation purposes are to be developed.
Collapse
Affiliation(s)
- Patrick Manser
- Movement Control and Learning - Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Eling D de Bruin
- Movement Control and Learning - Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland.,Division of Physiotherapy, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,OST - Eastern Switzerland University of Applied Sciences, St. Gallen, Switzerland
| |
Collapse
|
31
|
Olsthoorn L, Vreeken D, Kiliaan AJ. Gut Microbiome, Inflammation, and Cerebrovascular Function: Link Between Obesity and Cognition. Front Neurosci 2021; 15:761456. [PMID: 34938153 PMCID: PMC8685335 DOI: 10.3389/fnins.2021.761456] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/16/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity affects 13% of the adult population worldwide and this number is only expected to increase. Obesity is known to have a negative impact on cardiovascular and metabolic health, but it also impacts brain structure and function; it is associated with both gray and white matter integrity loss, as well as decreased cognitive function, including the domains of executive function, memory, inhibition, and language. Especially midlife obesity is associated with both cognitive impairment and an increased risk of developing dementia at later age. However, underlying mechanisms are not yet fully revealed. Here, we review recent literature (published between 2010 and March 2021) and discuss the effects of obesity on brain structure and cognition, with a main focus on the contributions of the gut microbiome, white adipose tissue (WAT), inflammation, and cerebrovascular function. Obesity-associated changes in gut microbiota composition may cause increased gut permeability and inflammation, therewith affecting cognitive function. Moreover, excess of WAT in obesity produces pro-inflammatory adipokines, leading to a low grade systemic peripheral inflammation, which is associated with decreased cognition. The blood-brain barrier also shows increased permeability, allowing among others, peripheral pro-inflammatory markers to access the brain, leading to neuroinflammation, especially in the hypothalamus, hippocampus and amygdala. Altogether, the interaction between the gut microbiota, WAT inflammation, and cerebrovascular integrity plays a significant role in the link between obesity and cognition. Future research should focus more on the interplay between gut microbiota, WAT, inflammation and cerebrovascular function to obtain a better understanding about the complex link between obesity and cognitive function in order to develop preventatives and personalized treatments.
Collapse
Affiliation(s)
- Lisette Olsthoorn
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands
| | - Debby Vreeken
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands.,Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, Arnhem, Netherlands
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands
| |
Collapse
|
32
|
Motegi N, Yamaoka Y, Moriichi A, Morisaki N. Causes of death in patients with Down syndrome in 2014-2016: A population study in Japan. Am J Med Genet A 2021; 188:224-236. [PMID: 34622557 PMCID: PMC9292866 DOI: 10.1002/ajmg.a.62526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/28/2021] [Accepted: 09/18/2021] [Indexed: 11/06/2022]
Abstract
Despite the higher mortality rates in patients with Down syndrome compared with the general Japanese population, the life span has dramatically increased in Japan and other countries. We aimed to clarify recent causes of death in patients with Down syndrome in Japan. We calculated proportionate mortality and standardized mortality odds ratios (SMORs) among all deaths registered with Down syndrome as the cause of death (ICD‐10 code, Q90) in the Japanese National Death Registry Database in 2014–2016. In the study period, 762 in patients with Down syndrome died. The main causes of death were pneumonia/respiratory infections (20.5%), congenital malformations of the circulatory system (11.2%), other diseases of the circulatory system (9.2%), and aspiration pneumonia (8.4%). The SMORs (95% confidence intervals) were higher for natural death, defined as death of an elderly person with no other cause of death to be mentioned (55.73 [36.92–84.12]), early‐onset Alzheimer's disease, defined as Alzheimer's disease with onset <65 years of age (29.36 [16.44–52.44]), aspiration pneumonia (18.33 [14.03–23.96]), pneumonia/respiratory infections (8.11 [6.76–9.73]), congenital malformations of the circulatory system (8.07 [5.98–10.88]), and leukemia/lymphoma (2.16 [1.55–2.99]) but lower for malignant solid tumors (0.04 [0.02–0.06]) in patients with Down syndrome. Patients with Down syndrome had the greatest relative risk of dying from natural death, early‐onset Alzheimer's disease, and respiratory illnesses, highlighting the need for appropriate medical, health, and welfare services.
Collapse
Affiliation(s)
- Narumi Motegi
- Department of Specific Pediatric Chronic Disease Information, National Center for Child Health and Development, Tokyo, Japan.,Graduate School of Advanced Integrated Studies in Human Survivability, Kyoto University, Kyoto, Japan
| | - Yui Yamaoka
- Department of Global Health Promotion, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akinori Moriichi
- Department of Specific Pediatric Chronic Disease Information, National Center for Child Health and Development, Tokyo, Japan
| | - Naho Morisaki
- Department of Social Medicine, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
33
|
Sanborn V, Preis SR, Ang A, Devine S, Mez J, DeCarli C, Au R, Alosco ML, Gunstad J. Association Between Leptin, Cognition, and Structural Brain Measures Among "Early" Middle-Aged Adults: Results from the Framingham Heart Study Third Generation Cohort. J Alzheimers Dis 2021; 77:1279-1289. [PMID: 32831199 DOI: 10.3233/jad-191247] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND There is growing interest in the pathophysiological processes of preclinical Alzheimer's disease (AD), including the potential role of leptin. Human studies have shown that both low and high levels of leptin can be associated with worse neurocognitive outcomes, suggesting this relationship may be moderated by another risk factor. OBJECTIVE We examined the association between plasma leptin levels and both neuropsychological test performance and structural neuroimaging and assessed whether body mass index (BMI) is an effect modifier of these associations. METHODS Our study sample consisted of 2,223 adults from the Framingham Heart Study Third Generation Cohort (average age = 40 years, 53% women). RESULTS Among the entire sample, there was no association between leptin and any of the neuropsychological domain measures or any of the MRI brain volume measures, after adjustment for BMI, APOE4, and other clinical factors. However, we did observe that BMI category was an effect modifier for the association between leptin and verbal memory (p for interaction = 0.03), where higher levels of leptin were associated with better performance among normal weight participants (BMI 18.5-24.9) kg/m2 (beta = 0.12, p = 0.02). No association was observed between leptin level and verbal memory test performance among participants who were overweight or obese. CONCLUSION These findings suggest that the association between leptin and cognitive function is moderated by BMI category. Prospective examination of individuals transitioning from middle age to older adulthood will help to clarify the contribution of leptin to AD and other neurodegenerative conditions.
Collapse
Affiliation(s)
- Victoria Sanborn
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
| | - Sarah R Preis
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alvin Ang
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
| | - Sherral Devine
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Jesse Mez
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Boston University Alzheimer's Disease Center and Boston University CTE Center, Boston University School of Medicine, Boston, MA, USA
| | - Charles DeCarli
- Department of Neurology, University of California at Davis Health System, Sacramento, CA, USA
| | - Rhoda Au
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Boston University Alzheimer's Disease Center and Boston University CTE Center, Boston University School of Medicine, Boston, MA, USA.,Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA.,Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Michael L Alosco
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Boston University Alzheimer's Disease Center and Boston University CTE Center, Boston University School of Medicine, Boston, MA, USA
| | - John Gunstad
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
34
|
Möllers T, Stocker H, Perna L, Nabers A, Rujescu D, Hartmann AM, Holleczek B, Schöttker B, Gerwert K, Brenner H. Aβ misfolding in blood plasma is inversely associated with body mass index even in middle adulthood. Alzheimers Res Ther 2021; 13:145. [PMID: 34461995 PMCID: PMC8406782 DOI: 10.1186/s13195-021-00889-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND To understand the potential for early intervention and prevention measures in Alzheimer's disease, the association between risk factors and early pathological change needs to be assessed. Hence, the aim of this study was to determine whether risk factors of Alzheimer's clinical syndrome (clinical AD), such as body mass index (BMI), are associated with Aβ misfolding in blood, a strong risk marker for AD among older adults. METHODS Information on risk factors and blood samples were collected at baseline in the ESTHER study, a population-based cohort study of older adults (age 50-75 years) in Germany. Aβ misfolding in blood plasma was analyzed using an immuno-infrared-sensor in a total of 872 participants in a nested case-control design among incident dementia cases and matched controls. Associations between risk factors and Aβ misfolding were assessed by multiple logistic regression. For comparison, the association between the risk factors and AD incidence during 17 years of follow-up was investigated in parallel among 5987 cohort participants. RESULTS An inverse association with Aβ misfolding was seen for BMI at age 50 based on reported weight history (aOR 0.64, 95% CI 0.43-0.96, p = 0.03). Similar but not statistically significant associations were seen for BMI at baseline (i.e., mean age 68) and at age 40. No statistically significant associations with Aβ misfolding were found for other risk factors, such as diabetes, smoking, and physical activity. On the other hand, low physical activity was associated with a significantly reduced risk of developing clinical AD compared to physical inactivity. CONCLUSIONS Our results support that AD pathology may be detectable and associated with reduced weight even in middle adulthood, many years before clinical diagnosis of AD. Physical activity might reduce the risk of onset of AD symptoms.
Collapse
Affiliation(s)
- Tobias Möllers
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, Germany.
- Network Aging Research, Heidelberg University, Bergheimer Straße 20, Heidelberg, Germany.
| | - Hannah Stocker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, Germany
- Network Aging Research, Heidelberg University, Bergheimer Straße 20, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Im Neuenheimer Feld 572, Heidelberg, Germany
| | - Laura Perna
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, Germany
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, München, Germany
| | - Andreas Nabers
- Department of Biophysics, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
- Department of Biophysics, Center for Protein Diagnostics (ProDi), Biospectroscopy, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
| | - Dan Rujescu
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Julius-Kühn-Straße 7, Halle (Saale), Germany
| | - Annette M Hartmann
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Julius-Kühn-Straße 7, Halle (Saale), Germany
| | - Bernd Holleczek
- Saarland Cancer Registry, Präsident-Baltz-Straße 5, Saarbrücken, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, Germany
- Network Aging Research, Heidelberg University, Bergheimer Straße 20, Heidelberg, Germany
| | - Klaus Gerwert
- Department of Biophysics, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
- Department of Biophysics, Center for Protein Diagnostics (ProDi), Biospectroscopy, Ruhr-University Bochum, Universitätsstraße 150, Bochum, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, Heidelberg, Germany
- Network Aging Research, Heidelberg University, Bergheimer Straße 20, Heidelberg, Germany
| |
Collapse
|
35
|
The risk of Alzheimer's disease according to dynamic changes in metabolic health and obesity: a nationwide population-based cohort study. Aging (Albany NY) 2021; 13:16974-16989. [PMID: 34237705 PMCID: PMC8312469 DOI: 10.18632/aging.203255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/18/2021] [Indexed: 12/29/2022]
Abstract
We evaluated the association of metabolic health and obesity phenotypes with the risk of Alzheimer's disease (AD). This study enrolled 136,847 elderly participants aged 60 or above from the Korean National Health Insurance System. At baseline examinations in 2009 and 2010, subjects were categorized into four groups: the metabolically healthy non-obese (MHNO), metabolically healthy obese (MHO), metabolically unhealthy non-obese (MUNO), and metabolically unhealthy obese (MUO) groups. Based on the phenotypic transition after 2 years, the subjects were further categorized into 16 subgroups. They were followed from 2009 to 2015 to monitor for AD development. The MHO phenotype protected subjects from AD, relative to the MHNO phenotype (HR, 0.73; 95% CI, 0.65-0.81). Among subjects initially classified as MHO, 41.8% remained MHO, with a significantly lower risk of AD compared with the stable MHNO group (HR, 0.62; 95% CI, 0.50-0.77). Among MUO subjects at baseline, those who changed phenotype to MUNO were at higher risk of AD (HR, 1.47; 95% CI, 1.28-1.70), and the transition to the MHO phenotype protected subjects from AD (HR, 0.62; 95% CI, 0.50-0.78). The MHO phenotype conferred a decreased risk of AD. Maintenance or recovery of metabolic health might mitigate AD risk among obese individuals.
Collapse
|
36
|
Litke R, Garcharna LC, Jiwani S, Neugroschl J. Modifiable Risk Factors in Alzheimer Disease and Related Dementias: A Review. Clin Ther 2021; 43:953-965. [PMID: 34108080 DOI: 10.1016/j.clinthera.2021.05.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Although Alzheimer disease and related dementias (ADRDs) have long been considered nonpreventable and even an inevitable consequence of aging, recent findings from longitudinal studies indicate a downtrend in age-adjusted incidence and prevalence of ADRDs in Western countries. This remarkable trend might be the result of improved management of so-called modifiable risk factors. The aim of this review is to present evidence of modifiable factors of ADRDs in a life-course approach. METHODS A PubMed database search was conducted between November and December 2020 to identify relevant studies evaluating the role of modifiable risk factors in the development of ADRDs. Key words (Alzheimer's disease and modifiable risk factors) were used and specific inclusion and exclusion criteria applied. FINDINGS This review identifies modifiable factors for ADRDs divided into early-life, middle-life, and late-life risk factors, depending on the available window of preventive action. According to life course exposure, factors can be protective or deleterious for ADRDs that participate in the underlying pathophysiologic complexity of these diseases as well as the complexity for public health measures implementations. IMPLICATIONS The available evidence derived from epidemiologic, preclinical, interventional studies suggest that modifiable risk factors for ADRDs offer opportunities for therapeutic and preventive actions.
Collapse
Affiliation(s)
- Rachel Litke
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.
| | | | - Salima Jiwani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Judith Neugroschl
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
37
|
Li X, Tian Y, Yang YX, Ma YH, Shen XN, Chen SD, Dong Q, Tan L, Yu JT. Life Course Adiposity and Alzheimer's Disease: A Mendelian Randomization Study. J Alzheimers Dis 2021; 82:503-512. [PMID: 34057091 DOI: 10.3233/jad-210345] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Several studies showed that life course adiposity was associated with Alzheimer's disease (AD). However, the underlying causality remains unclear. OBJECTIVE We aimed to examine the causal relationship between life course adiposity and AD using Mendelian randomization (MR) analysis. METHODS Instrumental variants were obtained from large genome-wide association studies (GWAS) for life course adiposity, including birth weight (BW), childhood body mass index (BMI), adult BMI, waist circumference (WC), waist-to-hip ratio (WHR), and body fat percentage (BFP). A meta-analysis of GWAS for AD including 71,880 cases and 383,378 controls was used in this study. MR analyses were performed using inverse variance weighted (IVW), weighted median, and MR-Egger regression methods. We calculated odds ratios (ORs) per genetically predicted standard deviation (1-SD) unit increase in each trait for AD. RESULTS Genetically predicted 1-SD increase in adult BMI was significantly associated with higher risk of AD (IVW: OR = 1.03, 95% confidence interval [CI] = 1.01-1.05, p = 2.7×10-3) after Bonferroni correction. The weighted median method indicated a significant association between BW and AD (OR = 0.94, 95% CI = 0.90-0.98, p = 1.8×10-3). We also found suggestive associations of AD with WC (IVW: OR = 1.03, 95% CI = 1.00-1.07, p = 0.048) and WHR (weighted median: OR = 1.04, 95% CI = 1.00-1.07, p = 0.029). No association was detected of AD with childhood BMI and BFP. CONCLUSION Our study demonstrated that lower BW and higher adult BMI had causal effects on increased AD risk.
Collapse
Affiliation(s)
- Xian Li
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
| | - Yan Tian
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu-Xiang Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China.,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Vinuesa A, Pomilio C, Gregosa A, Bentivegna M, Presa J, Bellotto M, Saravia F, Beauquis J. Inflammation and Insulin Resistance as Risk Factors and Potential Therapeutic Targets for Alzheimer's Disease. Front Neurosci 2021; 15:653651. [PMID: 33967682 PMCID: PMC8102834 DOI: 10.3389/fnins.2021.653651] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Overnutrition and modern diets containing high proportions of saturated fat are among the major factors contributing to a low-grade state of inflammation, hyperglycemia and dyslipidemia. In the last decades, the global rise of type 2 diabetes and obesity prevalence has elicited a great interest in understanding how changes in metabolic function lead to an increased risk for premature brain aging and the development of neurodegenerative disorders such as Alzheimer's disease (AD). Cognitive impairment and decreased neurogenic capacity could be a consequence of metabolic disturbances. In these scenarios, the interplay between inflammation and insulin resistance could represent a potential therapeutic target to prevent or ameliorate neurodegeneration and cognitive impairment. The present review aims to provide an update on the impact of metabolic stress pathways on AD with a focus on inflammation and insulin resistance as risk factors and therapeutic targets.
Collapse
Affiliation(s)
- Angeles Vinuesa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Pomilio
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Amal Gregosa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melisa Bentivegna
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jessica Presa
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melina Bellotto
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Saravia
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Beauquis
- Laboratorio de Neurobiología del Envejecimiento, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
39
|
de Bem AF, Krolow R, Farias HR, de Rezende VL, Gelain DP, Moreira JCF, Duarte JMDN, de Oliveira J. Animal Models of Metabolic Disorders in the Study of Neurodegenerative Diseases: An Overview. Front Neurosci 2021; 14:604150. [PMID: 33536868 PMCID: PMC7848140 DOI: 10.3389/fnins.2020.604150] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022] Open
Abstract
The incidence of metabolic disorders, as well as of neurodegenerative diseases—mainly the sporadic forms of Alzheimer’s and Parkinson’s disease—are increasing worldwide. Notably, obesity, diabetes, and hypercholesterolemia have been indicated as early risk factors for sporadic forms of Alzheimer’s and Parkinson’s disease. These conditions share a range of molecular and cellular features, including protein aggregation, oxidative stress, neuroinflammation, and blood-brain barrier dysfunction, all of which contribute to neuronal death and cognitive impairment. Rodent models of obesity, diabetes, and hypercholesterolemia exhibit all the hallmarks of these degenerative diseases, and represent an interesting approach to the study of the phenotypic features and pathogenic mechanisms of neurodegenerative disorders. We review the main pathological aspects of Alzheimer’s and Parkinson’s disease as summarized in rodent models of obesity, diabetes, and hypercholesterolemia.
Collapse
Affiliation(s)
- Andreza Fabro de Bem
- Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brazilia, Brazil
| | - Rachel Krolow
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Hémelin Resende Farias
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Victória Linden de Rezende
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - José Cláudio Fonseca Moreira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - João Miguel das Neves Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Jade de Oliveira
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
40
|
Kadey KR, Woodard JL, Moll A, Nielson KA, Smith JC, Durgerian S, Rao SM. Five-Year Change in Body Mass Index Predicts Conversion to Mild Cognitive Impairment or Dementia Only in APOE ɛ4 Allele Carriers. J Alzheimers Dis 2021; 81:189-199. [PMID: 33749649 PMCID: PMC10735532 DOI: 10.3233/jad-201360] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Body mass index (BMI) has been identified as an important modifiable lifestyle risk factor for dementia, but less is known about how BMI might interact with Apolipoprotein E ɛ4 (APOE ɛ4) carrier status to predict conversion to mild cognitive impairment (MCI) and dementia. OBJECTIVE The aim of this study was to investigate the interaction between APOE ɛ4 status and baseline (bBMI) and five-year BMI change (ΔBMI) on conversion to MCI or dementia in initially cognitively healthy older adults. METHODS The associations between bBMI, ΔBMI, APOE ɛ4 status, and conversion to MCI or dementia were investigated among 1,289 cognitively healthy elders from the National Alzheimer's Coordinating Center (NACC) database. RESULTS After five years, significantly more carriers (30.6%) converted to MCI or dementia than noncarriers (17.6%), p < 0.001, OR = 2.06. Neither bBMI (OR = 0.99, 95%CI = 0.96-1.02) nor the bBMI by APOE interaction (OR = 1.02, 95%CI = 0.96-1.08) predicted conversion. Although ΔBMI also did not significantly predict conversion (OR = 0.90, 95%CI = 0.78-1.04), the interaction between ΔBMI and carrier status was significant (OR = 0.72, 95%CI = 0.53-0.98). For carriers only, each one-unit decline in BMI over five years was associated with a 27%increase in the odds of conversion (OR = 0.73, 95%CI = 0.57-0.94). CONCLUSION A decline in BMI over five years, but not bBMI, was strongly associated with conversion to MCI or dementia only for APOE ɛ4 carriers. Interventions and behaviors aimed at maintaining body mass may be important for long term cognitive health in older adults at genetic risk for AD.
Collapse
Affiliation(s)
- Kylie R. Kadey
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - John L. Woodard
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Allison Moll
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Kristy A. Nielson
- Department of Psychology, Marquette University, Milwaukee, WI, USA
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - J. Carson Smith
- Department of Kinesiology, University of Maryland, College Park, MD, USA
| | | | - Stephen M. Rao
- Schey Center for Cognitive Neuroimaging, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
41
|
Tolea MI, Heo J, Chrisphonte S, Galvin JE. A Modified CAIDE Risk Score as a Screening Tool for Cognitive Impairment in Older Adults. J Alzheimers Dis 2021; 82:1755-1768. [PMID: 34219721 PMCID: PMC8483620 DOI: 10.3233/jad-210269] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Although an efficacious dementia-risk score system, Cardiovascular Risk Factors, Aging, and Dementia (CAIDE) was derived using midlife risk factors in a population with low educational attainment that does not reflect today's US population, and requires laboratory biomarkers, which are not always available. OBJECTIVE Develop and validate a modified CAIDE (mCAIDE) system and test its ability to predict presence, severity, and etiology of cognitive impairment in older adults. METHODS Population consisted of 449 participants in dementia research (N = 230; community sample; 67.9±10.0 years old, 29.6%male, 13.7±4.1 years education) or receiving dementia clinical services (N = 219; clinical sample; 74.3±9.8 years old, 50.2%male, 15.5±2.6 years education). The mCAIDE, which includes self-reported and performance-based rather than blood-derived measures, was developed in the community sample and tested in the independent clinical sample. Validity against Framingham, Hachinski, and CAIDE risk scores was assessed. RESULTS Higher mCAIDE quartiles were associated with lower performance on global and domain-specific cognitive tests. Each one-point increase in mCAIDE increased the odds of mild cognitive impairment (MCI) by up to 65%, those of AD by 69%, and those for non-AD dementia by > 85%, with highest scores in cases with vascular etiologies. Being in the highest mCAIDE risk group improved ability to discriminate dementia from MCI and controls and MCI from controls, with a cut-off of ≥7 points offering the highest sensitivity, specificity, and positive and negative predictive values. CONCLUSION mCAIDE is a robust indicator of cognitive impairment in community-dwelling seniors, which can discriminate well between dementia severity including MCI versus controls. The mCAIDE may be a valuable tool for case ascertainment in research studies, helping flag primary care patients for cognitive testing, and identify those in need of lifestyle interventions for symptomatic control.
Collapse
Affiliation(s)
- Magdalena I. Tolea
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine
| | - Jaeyeong Heo
- Department of Neurology, Harbor UCLA Medical Center
| | - Stephanie Chrisphonte
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine
| | - James E. Galvin
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine
| |
Collapse
|
42
|
Abstract
In India, increasing lifespan and decreasing fertility rates have resulted in a growing number of older persons. By 2050, people over 60 years of age are predicted to constitute 19.1% of the total population. This ageing of the population is expected to be accompanied by a dramatic increase in the prevalence of dementia. The aetiopathogenesis of dementia has been the subject of a number of prospective longitudinal studies in North America and Europe; however, the findings from these studies cannot simply be translated to the Indian population. The population of India is extremely diverse in terms of socio-economic, cultural, linguistic, geographical, lifestyle-related and genetic factors. Indeed, preliminary data from recently initiated longitudinal studies in India indicate that the prevalence of vascular and metabolic risk factors, as well as white matter hyperintensities, differs between urban and rural cohorts. More information on the complex role of vascular risk factors, gender and genetic influences on dementia prevalence and progression in Indian populations is urgently needed. Low-cost, culturally appropriate and scalable interventions need to be developed expeditiously and implemented through public health measures to reduce the growing burden of dementia. Here, we review the literature concerning dementia epidemiology and risk factors in the Indian population and discuss the future work that needs to be performed to put in place public health interventions to mitigate the burden of dementia.
Collapse
|
43
|
Shabir O, Moll TA, Matuszyk MM, Eyre B, Dake MD, Berwick J, Francis SE. Preclinical models of disease and multimorbidity with focus upon cardiovascular disease and dementia. Mech Ageing Dev 2020; 192:111361. [DOI: 10.1016/j.mad.2020.111361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
|
44
|
Perera G, Rijnbeek PR, Alexander M, Ansell D, Avillach P, Duarte-Salles T, Gordon MF, Lapi F, Mayer MA, Pasqua A, Pedersen L, van Der Lei J, Visser PJ, Stewart R. Vascular and metabolic risk factor differences prior to dementia diagnosis: a multidatabase case-control study using European electronic health records. BMJ Open 2020; 10:e038753. [PMID: 33191253 PMCID: PMC7668358 DOI: 10.1136/bmjopen-2020-038753] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/16/2020] [Accepted: 09/17/2020] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE The objective of the study is to compare body mass index (BMI), systolic/diastolic blood pressure (SBP/DBP) and serum total cholesterol levels between dementia cases and controls at multiple time intervals prior to dementia onset, and to test time interval as a modifying factor for these associations. DESIGN Case-control study. SETTING Six European electronic health records databases. PARTICIPANTS 291 780 cases at the date of first-recorded dementia diagnosis, compared with 29 170 549 controls randomly selected from the same databases, age matched and sex matched at this index date. EXPOSURE The following measures were extracted whenever recorded within each dataset: BMI (kg/m2), SBP and DBP (mm Hg) and serum total cholesterol (mmol/L). Levels for each of these variables were defined within six 2-year time intervals over the 12 years prior to the index date. MAIN OUTCOMES Case-control differences in exposures of interest were modelled for each time period and adjusted for demographic and clinical factors (ischaemic/unspecified stroke, type 2 diabetes mellitus, acute myocardial infarction, hypertension diagnosis, antihypertensive medication, cholesterol-lowering medication). Coefficients and interactions with time period were meta-analysed across the six databases. RESULTS Mean BMI (coefficient -1.16 kg/m2; 95% CI -1.38 to 0.93) and SBP (-2.83 mm Hg; 95% CI -4.49 to -1.16) were lower in cases at diagnosis, and case-control differences were greater in more recent time periods, as indicated by significant case-x-time interaction and case-x-time-squared interaction terms. Time variations in coefficients for cholesterol levels were less consistent between databases and those for DBP were largely not significant. CONCLUSION Routine clinical data show emerging divergence in levels of BMI and SBP prior to the diagnosis of dementia but less evidence for DBP or total cholesterol levels. These divergences should receive at least some consideration in routine dementia risk screening, although underlying mechanisms still require further investigation.
Collapse
Affiliation(s)
- Gayan Perera
- Psychological Medicine, King's College London (Institute of Psychiatry, Psychology and Neuroscience), London, UK
| | - P R Rijnbeek
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - David Ansell
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Paul Avillach
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Aarhus University, Aarhus, Denmark
| | | | - Mark Forrest Gordon
- Specialty Clinical Development, Neurology and Psychiatry, Teva Pharmaceuticals USA Inc, North Wales, Pennsylvania, USA
| | - Francesco Lapi
- Health Search, Italian College of General Practitioners and Primary Care, Florence, Italy
| | | | - Alessandro Pasqua
- Health Search, Italian College of General Practitioners and Primary Care, Florence, Italy
| | - Lars Pedersen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Johan van Der Lei
- Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- Department of Neurology, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - Robert Stewart
- Psychological Medicine, King's College London (Institute of Psychiatry, Psychology and Neuroscience), London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
45
|
Mole JP, Fasano F, Evans J, Sims R, Kidd E, Aggleton JP, Metzler-Baddeley C. APOE-ε4-related differences in left thalamic microstructure in cognitively healthy adults. Sci Rep 2020; 10:19787. [PMID: 33188215 PMCID: PMC7666117 DOI: 10.1038/s41598-020-75992-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/15/2020] [Indexed: 01/05/2023] Open
Abstract
APOE-ε4 is a main genetic risk factor for developing late onset Alzheimer's disease (LOAD) and is thought to interact adversely with other risk factors on the brain. However, evidence regarding the impact of APOE-ε4 on grey matter structure in asymptomatic individuals remains mixed. Much attention has been devoted to characterising APOE-ε4-related changes in the hippocampus, but LOAD pathology is known to spread through the whole of the Papez circuit including the limbic thalamus. Here, we tested the impact of APOE-ε4 and two other risk factors, a family history of dementia and obesity, on grey matter macro- and microstructure across the whole brain in 165 asymptomatic individuals (38-71 years). Microstructural properties of apparent neurite density and dispersion, free water, myelin and cell metabolism were assessed with Neurite Orientation Density and Dispersion (NODDI) and quantitative magnetization transfer (qMT) imaging. APOE-ε4 carriers relative to non-carriers had a lower macromolecular proton fraction (MPF) in the left thalamus. No risk effects were present for cortical thickness, subcortical volume, or NODDI indices. Reduced thalamic MPF may reflect inflammation-related tissue swelling and/or myelin loss in APOE-ε4. Future prospective studies should investigate the sensitivity and specificity of qMT-based MPF as a non-invasive biomarker for LOAD risk.
Collapse
Affiliation(s)
- Jilu P Mole
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Fabrizio Fasano
- Siemens Healthcare, Henkestrasse 127, 91052, Erlangen, Germany
| | - John Evans
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Haydn Ellis Building, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Emma Kidd
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue,, Cardiff, CF10 3NB, UK
| | - John P Aggleton
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Claudia Metzler-Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
46
|
Tsai CL, Pai MC. Circulating levels of Irisin in obese individuals at genetic risk for Alzheimer's disease: Correlations with amyloid-β, metabolic, and neurocognitive indices. Behav Brain Res 2020; 400:113013. [PMID: 33186636 DOI: 10.1016/j.bbr.2020.113013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022]
Abstract
Irisin is involved in various metabolic pathways and is suggested to be a potential agent capable of preventing onset of Alzheimer's disease (AD) and ameliorating AD neuropathology and cognitive deficits. In the present study, the serum levels of Irisin and Amyloid-β (Aβ) peptides and the neurocognitive performance among obese individuals at genetic risk for AD were investigated. The correlations between Irisin and AD-related neuropathological and neurocognitive indices were also explored. Thirty-two individuals with a family history of AD (ADFH) and obesity (ADFH-obesity group) and 32 controls (ADFH-non-obesity group) were recruited. Circulating levels of Irisin, Aβ peptides, and metabolic biomarkers, as well as neurocognitive performance [e.g., behavior and brain even-related potentials (ERP)] were measured during a visuospatial working memory task. Although the ADFH-obesity group exhibited comparable reaction times, ERP N2 latency and amplitudes, and P3 latency as compared to the ADFH-non-obesity group when performing the cognitive task, they exhibited significantly lower rates of accuracy and smaller P3 amplitudes in the higher memory-load condition, even when controlling for the blood pressure and cardiorespiratory fitness co-variables. The serum levels of leptin, insulin, and glucose, and HOMA-IR were significantly higher in the ADFH-obesity group relative to the ADFH-non-obesity group, but this was not the case for the levels of Aβ1-40 and Aβ1-42. The Irisin levels approached between-group significance. Partial correlations adjusting for cardiorespiratory fitness and blood pressure showed that Irisin levels were positively associated with neurophysiological (i.e., P3 amplitude) performance in the ADFH-obesity group. The Irisin levels were not significantly correlated with the levels of Aβ1-40 and Aβ1-42. The present findings suggest that ADFH individuals with obesity exhibited neurocognitive deficits when performing the visuospatial working memory task, and serum Irisin levels could be one of the influencing factors. However, the relationship between the circulating levels of Irisin and Aβ peptides needs more evidence to support this assumption.
Collapse
Affiliation(s)
- Chia-Liang Tsai
- Institute of Physical Education, Health and Leisure Studies, National Cheng Kung University, Taiwan.
| | - Ming-Chyi Pai
- Division of Behavioral Neurology, Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Taiwan; Alzheimer's Disease Research Center, National Cheng Kung University Hospital, Taiwan
| |
Collapse
|
47
|
Mattson MP. Applying available knowledge and resources to alleviate familial and sporadic neurodegenerative disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:91-107. [PMID: 33453944 DOI: 10.1016/bs.pmbts.2020.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Here I present the scientific rationale and implementation strategies for elimination of early-onset neurodegenerative disorders (EONDD) from future generations, and for risk reduction and treatments for the more common late-onset neurodegenerative disorders (LONDD). Young adults with a family history of an EONDD should be educated on the genetics and familial burden of EONDD. They can then be genotyped and, if positive for the mutation, counseled as to how they can ensure that none of their children will be affected by choosing either adoption or in vitro fertilization and preimplantation genetic testing. LONDD risk reduction will require education of physicians and patients on the benefits of regular intermittent bioenergetic and cognitive challenges (exercise, intermittent fasting, intellectual challenges and social engagement) for brain health, and on specific risk-reduction regimens. Regulations will be required to counteract the disease-promoting mercenary practices of the processed food and pharmaceutical industries. Clinical trials of pharmacological interventions should shift to small trials of agents that substantially mimic mechanisms of action of exercise and intermittent fasting to bolster neuronal bioenergetics and stress resistance.
Collapse
Affiliation(s)
- Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
48
|
Mole JP, Fasano F, Evans J, Sims R, Hamilton DA, Kidd E, Metzler-Baddeley C. Genetic risk of dementia modifies obesity effects on white matter myelin in cognitively healthy adults. Neurobiol Aging 2020; 94:298-310. [PMID: 32736120 DOI: 10.1016/j.neurobiolaging.2020.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 01/05/2023]
Abstract
APOE-ε4 is a major genetic risk factor for late-onset Alzheimer's disease that interacts with other risk factors, but the nature of such combined effects remains poorly understood. We quantified the impact of APOE-ε4, family history (FH) of dementia, and obesity on white matter (WM) microstructure in 165 asymptomatic adults (38-71 years old) using quantitative magnetization transfer and neurite orientation dispersion and density imaging. Microstructural properties of the fornix, parahippocampal cingulum, and uncinate fasciculus were compared with those in motor and whole-brain WM regions. Widespread interaction effects between APOE, FH, and waist-hip ratio were found in the myelin-sensitive macromolecular proton fraction from quantitative magnetization transfer. Among individuals with the highest genetic risk (FH+ and APOE-ε4), obesity was associated with reduced macromolecular proton fraction in the right parahippocampal cingulum, whereas no effects were present for those without FH. Risk effects on apparent myelin were moderated by hypertension and inflammation-related markers. These findings suggest that genetic risk modifies the impact of obesity on WM myelin consistent with neuroglia models of aging and late-onset Alzheimer's disease.
Collapse
Affiliation(s)
- Jilu P Mole
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | | | - John Evans
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neuroscience, School of Medicine, Cardiff University, Cardiff, UK
| | - Derek A Hamilton
- Department of Psychology, The University of New Mexico, Albuquerque, NM, USA
| | - Emma Kidd
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Claudia Metzler-Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK.
| |
Collapse
|
49
|
Pettigrew C, Soldan A, Wang J, Wang MC, Arthur K, Moghekar A, Gottesman RF, Albert M. Association of midlife vascular risk and AD biomarkers with subsequent cognitive decline. Neurology 2020; 95:e3093-e3103. [PMID: 32989109 DOI: 10.1212/wnl.0000000000010946] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/22/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether vascular risk and Alzheimer disease (AD) biomarkers have independent or synergistic effects on cognitive decline and whether vascular risk is associated with the accumulation of AD pathology as measured by change in biomarkers over time. METHODS At baseline, participants (n = 168) were cognitively normal and primarily middle-aged (mean 56.4 years, SD 10.9 years) and had both vascular risk factor status and proximal CSF biomarkers available. Baseline vascular risk was quantified with a composite vascular risk score reflecting the presence or absence of hypertension, hypercholesterolemia, diabetes, current smoking, and obesity. CSF biomarkers of β-amyloid (Aβ)1-42, total tau (t-tau), and phosphorylated tau (p-tau) were used to create dichotomous high and low AD biomarker groups (based on Aβ1-42 and tau). Linear mixed-effects models were used to examine change in a cognitive composite score (mean follow-up 13.9 years) and change in CSF biomarkers (mean follow-up 4.2 years). RESULTS There was no evidence of a synergistic relationship between the vascular risk score and CSF AD biomarkers and cognitive decline. Instead, the vascular risk score (estimate -0.022, 95% confidence interval [CI] -0.043 to -0.002, p = 0.03) and AD biomarkers (estimate -0.060, 95% CI -0.096 to -0.024, p = 0.001) were independently and additively associated with cognitive decline. In addition, the vascular risk score was unrelated to levels of or rate of change in CSF Aβ1-42, t-tau, or p-tau. CONCLUSIONS The results of this observational cohort study suggest that vascular risk and biomarkers of AD pathology, when measured in midlife, act along independent pathways and underscore the importance of accounting for multiple risk factors for identifying cognitively normal individuals at the greatest risk of cognitive decline.
Collapse
Affiliation(s)
- Corinne Pettigrew
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD.
| | - Anja Soldan
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jiangxia Wang
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Mei-Cheng Wang
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Karissa Arthur
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Abhay Moghekar
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Rebecca F Gottesman
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Marilyn Albert
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
50
|
Ahn S, Mathiason MA, Lindquist R, Yu F. Factors predicting episodic memory changes in older adults with subjective cognitive decline: A longitudinal observational study. Geriatr Nurs 2020; 42:268-275. [PMID: 32919799 DOI: 10.1016/j.gerinurse.2020.08.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 01/25/2023]
Abstract
Episodic memory is affected early in the neuropathological process of Alzheimer's dementia. This study was performed to identify longitudinal associations between baseline vascular/neuropsychiatric risk factors and episodic memory changes over 4.1 ± 2.4 years in 1,401 older adults with subjective cognitive decline (age 74.0 ± 8.2 years). Data were from the National Alzheimer's Coordinating Center-Uniform Data Set and linear mixed effects regression models were used. Reference was those without risk factors. Participants with hypercholesterolemia and with former cigarette smoking had higher episodic memory scores, but current smokers had fewer points than reference at their first and follow-up visits. Despite no difference at baseline, episodic memory scores decreased in those with depressive symptoms relative to reference over time. In older adults with subjective cognitive decline, interventions managing current smoking and depressive symptoms could preserve episodic memory, which may result in delaying the onset of Alzheimer's dementia. Further research is required for the role of cholesterol and smoking.
Collapse
Affiliation(s)
- Sangwoo Ahn
- University of Tennessee College of Nursing, Knoxville, TN, United States.
| | | | - Ruth Lindquist
- University of Minnesota School of Nursing, Minneapolis, MN, United States.
| | - Fang Yu
- University of Minnesota School of Nursing, Minneapolis, MN, United States.
| |
Collapse
|