1
|
Kawada T. Type 2 diabetes mellitus duration and subsequent risk of Alzheimer disease. Eur J Neurol 2024; 31:e16274. [PMID: 38450870 PMCID: PMC11235985 DOI: 10.1111/ene.16274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 02/22/2024] [Indexed: 03/08/2024]
Affiliation(s)
- Tomoyuki Kawada
- Department of Hygiene and Public HealthNippon Medical SchoolTokyoJapan
| |
Collapse
|
2
|
Canoy RJ, Sy JC, Deguit CD, Castro CB, Dimaapi LJ, Panlaqui BG, Perian W, Yu J, Velasco JM, Sevilleja JE, Gibson A. Non-coding RNAs involved in the molecular pathology of Alzheimer's disease: a systematic review. Front Neurosci 2024; 18:1421675. [PMID: 39005845 PMCID: PMC11243705 DOI: 10.3389/fnins.2024.1421675] [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: 04/22/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia globally, having a pathophysiology that is complex and multifactorial. Recent findings highlight the significant role of non-coding RNAs (ncRNAs), specifically microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and piwi-interacting RNAs (piRNAs) in the molecular mechanisms underlying AD. These ncRNAs are involved in critical biological processes such as cell proliferation, apoptosis, oxidative stress, amyloid-beta aggregation, tau phosphorylation, neuroinflammation, and autophagy, which are pivotal in AD development and progression. This systematic review aims to consolidate current scientific knowledge on the role of ncRNAs in AD, making it the first to encompass the four types of ncRNAs associated with the disease. Our comprehensive search and analysis reveal that ncRNAs not only play crucial roles in the pathogenesis of AD but also hold potential as biomarkers for its early detection and as novel therapeutic targets. Specifically, the findings underscore the significance of miRNAs in regulating genes involved in key AD pathways such as activin receptor signaling pathway, actomyosin contractile ring organization, and advanced glycation endproducts-receptor advanced glycation endproducts (AGE-RAGE) signaling pathway. This review also highlights the potential of ncRNAs in unveiling novel diagnostic and therapeutic strategies, emphasizing the need for further research to validate their clinical utility. Our systematic exploration provides a foundation for future bioinformatic analyses and the development of ncRNA-based precision medicine approaches for AD, offering new insights into the disease's molecular pathology and paving the way for innovative treatment strategies. Systematic review registration PROSPERO, https://www.crd.york.ac.uk/prospero/, CRD42022355307.
Collapse
Affiliation(s)
- Reynand Jay Canoy
- SciLore LLC, Kingsbury, TX, United States
- Instiute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Jenica Clarisse Sy
- SciLore LLC, Kingsbury, TX, United States
- Center for Research and Innovation, Ateneo de Manila University School of Medicine and Public Health, Pasig City, Philippines
| | - Christian Deo Deguit
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Caitlin Bridgette Castro
- National Institute of Molecular Biology and Biotechnology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Lyoneil James Dimaapi
- National Institute of Molecular Biology and Biotechnology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Beatrice Gabrielle Panlaqui
- National Institute of Molecular Biology and Biotechnology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Wenzel Perian
- National Institute of Molecular Biology and Biotechnology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Justine Yu
- Institute for Dementia Care Asia, Quezon City, Philippines
| | - John Mark Velasco
- Institute of Molecular Biology and Biotechnology, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | | | - Anna Gibson
- SciLore LLC, Kingsbury, TX, United States
- Center for Research and Innovation, Ateneo de Manila University School of Medicine and Public Health, Pasig City, Philippines
| |
Collapse
|
3
|
Augusto-Oliveira M, Arrifano GP, Leal-Nazaré CG, Santos-Sacramento L, Lopes-Araújo A, Royes LFF, Crespo-Lopez ME. Exercise Reshapes the Brain: Molecular, Cellular, and Structural Changes Associated with Cognitive Improvements. Mol Neurobiol 2023; 60:6950-6974. [PMID: 37518829 DOI: 10.1007/s12035-023-03492-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023]
Abstract
Physical exercise is well known as a non-pharmacological and holistic therapy believed to prevent and mitigate numerous neurological conditions and alleviate ageing-related cognitive decline. To do so, exercise affects the central nervous system (CNS) at different levels. It changes brain physiology and structure, promoting cognitive improvements, which ultimately improves quality of life. Most of these effects are mediated by neurotrophins release, enhanced adult hippocampal neurogenesis, attenuation of neuroinflammation, modulation of cerebral blood flow, and structural reorganisation, besides to promote social interaction with beneficial cognitive outcomes. In this review, we discuss, based on experimental and human research, how exercise impacts the brain structure and function and how these changes contribute to cognitive improvements. Understanding the mechanisms by which exercise affects the brain is essential to understand the brain plasticity following exercise, guiding therapeutic approaches to improve the quality of life, especially in obesity, ageing, neurodegenerative disorders, and following traumatic brain injury.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil.
| | - Gabriela P Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil
| | - Caio G Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil
| | - Letícia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil
| | - Amanda Lopes-Araújo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil
| | - Luiz Fernando Freire Royes
- Laboratório de Bioquímica Do Exercício, Centro de Educacão Física E Desportos, Universidade Federal de Santa Maria, Santa Maria, RGS, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil.
| |
Collapse
|
4
|
Erichsen J, Craft S. Targeting immunometabolic pathways for combination therapy in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12423. [PMID: 37786483 PMCID: PMC10541802 DOI: 10.1002/trc2.12423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 10/04/2023]
Abstract
The recent success of disease-modifying anti-amyloid monoclonal antibodies in slowing Alzheimer's disease (AD) symptoms has been an exciting step forward for the field. Despite successfully clearing amyloid from the brain, however, only modest symptomatic improvement has been demonstrated, and treatment-related side effects such as amyloid-related imaging abnormalities (ARIA) limit use for some. These limitations suggest that fully efficacious AD treatment may require combination therapy regimens, as are used in other complex disorders such as cancer and HIV. One reasonable strategy may be to use agents that address the biological changes that predict future amyloid accumulation, or accompany amyloid accumulation in preclinical disease states. Immunometabolic pathways, including the insulin signaling pathway, are dysregulated at the earliest stages of AD, concomitant with amyloid accumulation. It is plausible that agents that target these pathways may work synergistically with anti-amyloid therapies to halt AD progression. Insulin signaling is integrally involved in innate and adaptive immune systems, with pleiotropic effects that moderate pro- and anti-inflammatory responses. Metabolic modulators that enhance insulin sensitivity and function, such as GLP-1 receptor agonists, SGLT2 inhibitors, and insulin itself have been shown to improve immune function and reduce chronic inflammation. Additional effects of insulin and metabolic modulators demonstrated in preclinical and clinical studies of AD include increased clearance of amyloid-β, slowed tau progression, improved vascular function and lipid metabolism, reduced synaptotoxicity, and improved cognitive and functional outcomes. A large number of compounds that treat metabolic disorders have been extensively characterized with respect to mechanism of action and safety, and thus are readily available to be repurposed for combination therapy protocols. Determining the most successful combination regimens of these agents together with disease-modifying therapies, and the appropriate timing of treatment, are promising next steps in the quest to treat and prevent AD.
Collapse
Affiliation(s)
- Jennifer Erichsen
- Department of Internal MedicineDivision of Gerontology and Geriatric MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Suzanne Craft
- Department of Internal MedicineDivision of Gerontology and Geriatric MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
5
|
Cheng Y, Zamrini E, Faselis C, Redd D, Shao Y, Morgan CJ, Sheriff HM, Ahmed A, Kokkinos P, Zeng-Treitler Q. Cardiorespiratory fitness and risk of Alzheimer's disease and related dementias among American veterans. Alzheimers Dement 2023; 19:4325-4334. [PMID: 36946469 PMCID: PMC10729726 DOI: 10.1002/alz.12998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Cardiorespiratory fitness (CRF) is associated with improved health and survival. Less is known about its association with Alzheimer's disease and related dementias (ADRD). METHODS We identified 649,605 US veterans 30 to 95 years of age and free of ADRD who completed a standardized exercise tolerance test between 2000 and 2017 with no evidence of ischemia. We examined the association between five age- and sex-specific CRF categories and ADRD incidence using multivariate Cox regression models. RESULTS During up to 20 (median 8.3) years of follow-up, incident ADRD occurred in 44,105 (6.8%) participants, with an incidence rate of 7.7/1000 person-years. Compared to the least-fit, multivariable-adjusted hazard ratios (95% confidence intervals) for incident ADRD were: 0.87 (0.85-0.90), 0.80 (0.78-0.83), 0.74 (0.72-0.76), and 0.67 (0.65-0.70), for low-fit, moderate-fit, fit, and high-fit individuals, respectively. DISSCUSSION These findings demonstrate an independent, inverse, and graded association between CRF and incident ADRD. Future studies may determine the amount and duration of physical activity needed to optimize ADRD risk reduction.
Collapse
Affiliation(s)
- Yan Cheng
- Washington DC VA Medical Center, Washington, DC
- George Washington University, Washington, DC
| | - Edward Zamrini
- Washington DC VA Medical Center, Washington, DC
- George Washington University, Washington, DC
- University of Utah, Salt Lake City, Utah
- Irvine Clinical Research, Irvine, CA
| | - Charles Faselis
- Washington DC VA Medical Center, Washington, DC
- George Washington University, Washington, DC
- Uniformed Services University, Washington, DC
| | - Douglas Redd
- Washington DC VA Medical Center, Washington, DC
- George Washington University, Washington, DC
| | - Yijun Shao
- Washington DC VA Medical Center, Washington, DC
- George Washington University, Washington, DC
| | | | - Helen M. Sheriff
- Washington DC VA Medical Center, Washington, DC
- George Washington University, Washington, DC
| | - Ali Ahmed
- Washington DC VA Medical Center, Washington, DC
- George Washington University, Washington, DC
- Georgetown University, Washington, DC
| | - Peter Kokkinos
- Washington DC VA Medical Center, Washington, DC
- George Washington University, Washington, DC
- Rutgers University, New Brunswick, NJ
| | - Qing Zeng-Treitler
- Washington DC VA Medical Center, Washington, DC
- George Washington University, Washington, DC
| |
Collapse
|
6
|
Nguyen S, LaCroix AZ, Hayden KM, Di C, Palta P, Stefanick ML, Manson JE, Rapp SR, LaMonte MJ, Bellettiere J. Accelerometer-measured physical activity and sitting with incident mild cognitive impairment or probable dementia among older women. Alzheimers Dement 2023; 19:3041-3054. [PMID: 36695426 PMCID: PMC10366337 DOI: 10.1002/alz.12908] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Physical activity (PA) is prospectively inversely associated with dementia risk, but few studies examined accelerometer measures of PA and sitting with rigorously-adjudicated mild cognitive impairment (MCI) and dementia risk. METHODS We examined the associations of accelerometer measures (PA and sitting) with incident MCI/probable dementia in the Women's Health Initiative (n = 1277; mean age = 82 ± 6 years) RESULTS: Over a median follow-up of 4.2 years, 267 MCI/probable dementia cases were identified. Adjusted Cox regression HRs (95% CI) across moderate-to-vigorous PA (MVPA) min/d quartiles were 1.00 (reference), 1.28 (0.90 to 1.81), 0.79 (0.53 to 1.17), and 0.69 (0.45 to 1.06); P-trend = 0.01. Adjusted HRs (95% CI) across steps/d quartiles were 1.00 (reference), 0.73 (0.51 to 1.03), 0.64 (0.43 to 0.94), and 0.38 (0.23 to 0.61); P-trend < 0.001. The HR (95% CI) for each 1-SD increment in MVPA (31 min/d) and steps/d (1865) were 0.79 (0.67 to 0.94) and 0.67 (0.54 to 0.82), respectively. Sitting was not associated with MCI/probable dementia. DISCUSSION Findings suggest ≥ moderate intensity PA, particularly stepping, associates with lower MCI and dementia risk. HIGHLIGHTS Few studies have examined accelerometer-measured physical activity, including steps, and sitting with incident ADRD. Moderate-to-vigorous physical activity and steps, but not light physical activity or sitting, were inversely associated with lower ADRD risk. Among older women, at least moderate intensity physical activity may be needed to reduce ADRD risk.
Collapse
Affiliation(s)
- Steve Nguyen
- Division of Epidemiology, Herbert Wertheim School of Public Health, University of California San Diego, La Jolla, CA, USA
| | - Andrea Z. LaCroix
- Division of Epidemiology, Herbert Wertheim School of Public Health, University of California San Diego, La Jolla, CA, USA
| | - Kathleen M. Hayden
- Department of Social Sciences & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Chongzhi Di
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Priya Palta
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Marcia L. Stefanick
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - JoAnn E. Manson
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen R. Rapp
- Department of Psychiatry & Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Michael J. LaMonte
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo - SUNY, Buffalo, NY, USA
| | - John Bellettiere
- Division of Epidemiology, Herbert Wertheim School of Public Health, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
7
|
Haghighi AH, Barzoei M, Kakhak SAH, Budini F, Shahrabadi H. Effect of multimodal exercise training on physical fitness indices, cognitive status, and depressive symptoms in Alzheimer's disease. Dement Neuropsychol 2023; 17:e20220008. [PMID: 37261252 PMCID: PMC10229082 DOI: 10.1590/1980-5764-dn-2022-0008] [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: 02/04/2022] [Revised: 11/10/2022] [Accepted: 12/16/2022] [Indexed: 06/02/2023] Open
Abstract
In Alzheimer's disease (AD) patients, low levels of physical fitness (PF) and cognitive status are associated with high rates of depression. However, this condition can be improved through physical training. Objective The aim of the present study was to investigate the effect of multimodal exercise training (MET) on aerobic endurance, muscular strength, agility, dynamic balance, cognitive status, and depressive symptoms in men with mild-to-moderate AD. Methods A total of 25 elderly men with a diagnosis of mild-to-moderate AD were randomly categorized into an MET or a control group. The subjects in the MET group participated in a 12-week, three sessions per week MET program that included resistance, balance, and aerobic exercises. While the participants in the control group did not perform any regular exercise training during this period. Patients' cognitive status and depressive symptoms were assessed by Mini-Mental State Examination and the Geriatric Depression Scale-15 (GDS-15) questionnaires. PF indicators such as aerobic endurance, muscular strength, agility, and dynamic balance, as well as cognitive status and depressive symptoms, were taken from all the subjects before and after MET. Results The participants in the MET group improved handgrip, upper and lower body strength, agility, dynamic balance, and depressive symptoms (p<0.05). The intervention had no significant effect on aerobic endurance and cognitive status (p>0.05). Conclusions MET is an effective strategy to improve muscular strength, agility, dynamic balance, and depressive symptoms in men with mild-to-moderate AD. It is recommended for AD patients to engage in this type of exercise to reduce AD complications.
Collapse
Affiliation(s)
- Amir Hossein Haghighi
- Hakim Sabzevari University, Faculty of Sport Sciences, Department of Exercise Physiology, Sabzevar, Iran
| | - Masoud Barzoei
- Hakim Sabzevari University, Faculty of Sport Sciences, Department of Exercise Physiology, Sabzevar, Iran
| | - Seyed Alireza Hosseini Kakhak
- Ferdowsi University of Mashhad and Hakim Sabzevari University, Faculty of Sport Sciences, Department of Exercise Physiology, Mashhad and Sabzevar, Iran
| | - Francesco Budini
- University of Graz, Institute of Human Movement Science, Sport and Health, Graz, Austria
| | - Hadi Shahrabadi
- Hakim Sabzevari University, Faculty of Sport Sciences, Department of Exercise Physiology, Sabzevar, Iran
| |
Collapse
|
8
|
Dong R, Denier-Fields DN, Van Hulle CA, Kollmorgen G, Suridjan I, Wild N, Lu Q, Anderson RM, Zetterberg H, Blennow K, Carlsson CM, Johnson SC, Engelman CD. Identification of plasma metabolites associated with modifiable risk factors and endophenotypes reflecting Alzheimer's disease pathology. Eur J Epidemiol 2023; 38:559-571. [PMID: 36964431 PMCID: PMC11070200 DOI: 10.1007/s10654-023-00988-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 03/05/2023] [Indexed: 03/26/2023]
Abstract
Modifiable factors can influence the risk for Alzheimer's disease (AD) and serve as targets for intervention; however, the biological mechanisms linking these factors to AD are unknown. This study aims to identify plasma metabolites associated with modifiable factors for AD, including MIND diet, physical activity, smoking, and caffeine intake, and test their association with AD endophenotypes to identify their potential roles in pathophysiological mechanisms. The association between each of the 757 plasma metabolites and four modifiable factors was tested in the wisconsin registry for Alzheimer's prevention cohort of initially cognitively unimpaired, asymptomatic middle-aged adults. After Bonferroni correction, the significant plasma metabolites were tested for association with each of the AD endophenotypes, including twelve cerebrospinal fluid (CSF) biomarkers, reflecting key pathophysiologies for AD, and four cognitive composite scores. Finally, causal mediation analyses were conducted to evaluate possible mediation effects. Analyses were performed using linear mixed-effects regression. A total of 27, 3, 23, and 24 metabolites were associated with MIND diet, physical activity, smoking, and caffeine intake, respectively. Potential mediation effects include beta-cryptoxanthin in the association between MIND diet and preclinical Alzheimer cognitive composite score, hippurate between MIND diet and immediate learning, glutamate between physical activity and CSF neurofilament light, and beta-cryptoxanthin between smoking and immediate learning. Our study identified several plasma metabolites that are associated with modifiable factors. These metabolites can be employed as biomarkers for tracking these factors, and they provide a potential biological pathway of how modifiable factors influence the human body and AD risk.
Collapse
Affiliation(s)
- Ruocheng Dong
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53726, USA
| | - Diandra N Denier-Fields
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53726, USA
- Department of Nutrition Science, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Carol A Van Hulle
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | | | | | - Norbert Wild
- Roche Diagnostics GmbH, 82377, Penzberg, Germany
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Rozalyn M Anderson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
- Geriatric Research Education and Clinical Center, William. S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, S-43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-43180, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1H 0AL, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, S-43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-43180, Mölndal, Sweden
| | - Cynthia M Carlsson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
- Geriatric Research Education and Clinical Center, William. S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA
| | - Sterling C Johnson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
- Geriatric Research Education and Clinical Center, William. S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53719, USA
| | - Corinne D Engelman
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53726, USA.
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA.
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53719, USA.
| |
Collapse
|
9
|
Park SY, Setiawan VW, White LR, Wu AH, Cheng I, Haiman CA, Wilkens LR, Le Marchand L, Lim U. Modifying effects of race and ethnicity and APOE on the association of physical activity with risk of Alzheimer's disease and related dementias. Alzheimers Dement 2023; 19:507-517. [PMID: 35476309 PMCID: PMC9810117 DOI: 10.1002/alz.12677] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 01/07/2023]
Abstract
INTRODUCTION We investigated whether the protective association of physical activity with risk of Alzheimer's disease and related dementias (ADRD) has genetic or behavioral variations. METHODS In the Multiethnic Cohort, we analyzed moderate or vigorous physical activity (MVPA) reported at ages 45 to 75 among 88,047 participants in relation to 13,039 incident diagnoses of late-onset ADRD identified in Medicare claims (1999 to 2014), by five racial and ethnic groups, hours sitting, and in a subset (16%), apolipoprotein E (APOE) genotype. RESULTS MVPA was inversely associated with ADRD (hazard ratio for ≥14 vs <2.5 hours/week: 0.83, 95% confidence interval [CI]: 0.76 to 0.90 in men; 0.88, 5% CI: 0.81 to 0.95 in women). The association was inverse in all racial and ethnic groups except Black participants (P-heterogeneity = 0.52), but stronger in individuals with lower levels of sitting duration or those who do not carry the APOE e4 risk allele. DISCUSSION The different effects of physical activity by sitting duration and APOE genotype warrant further research.
Collapse
Affiliation(s)
- Song-Yi Park
- Cancer Epidemiology Program, University of Hawaii Cancer Center, University of Hawaii, Honolulu, Hawaii, USA
| | - Veronica Wendy Setiawan
- Department of Population and Public Health Sciences, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Lon R. White
- Pacific Health Research and Education Institute, Honolulu, Hawaii, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Anna H. Wu
- Department of Population and Public Health Sciences, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Iona Cheng
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Christopher A. Haiman
- Department of Population and Public Health Sciences, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Lynne R. Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, University of Hawaii, Honolulu, Hawaii, USA
| | - Loїc Le Marchand
- Cancer Epidemiology Program, University of Hawaii Cancer Center, University of Hawaii, Honolulu, Hawaii, USA
| | - Unhee Lim
- Cancer Epidemiology Program, University of Hawaii Cancer Center, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
10
|
Campos HC, Ribeiro DE, Hashiguchi D, Glaser T, Milanis MDS, Gimenes C, Suchecki D, Arida RM, Ulrich H, Monteiro Longo B. Neuroprotective effects of resistance physical exercise on the APP/PS1 mouse model of Alzheimer's disease. Front Neurosci 2023; 17:1132825. [PMID: 37090809 PMCID: PMC10116002 DOI: 10.3389/fnins.2023.1132825] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/16/2023] [Indexed: 04/25/2023] Open
Abstract
Introduction Physical exercise has beneficial effects by providing neuroprotective and anti-inflammatory responses to AD. Most studies, however, have been conducted with aerobic exercises, and few have investigated the effects of other modalities that also show positive effects on AD, such as resistance exercise (RE). In addition to its benefits in developing muscle strength, balance and muscular endurance favoring improvements in the quality of life of the elderly, RE reduces amyloid load and local inflammation, promotes memory and cognitive improvements, and protects the cortex and hippocampus from the degeneration that occurs in AD. Similar to AD patients, double-transgenic APPswe/PS1dE9 (APP/PS1) mice exhibit Αβ plaques in the cortex and hippocampus, hyperlocomotion, memory deficits, and exacerbated inflammatory response. Therefore, the aim of this study was to investigate the effects of 4 weeks of RE intermittent training on the prevention and recovery from these AD-related neuropathological conditions in APP/PS1 mice. Methods For this purpose, 6-7-month-old male APP/PS1 transgenic mice and their littermates, negative for the mutations (CTRL), were distributed into three groups: CTRL, APP/PS1, APP/PS1+RE. RE training lasted four weeks and, at the end of the program, the animals were tested in the open field test for locomotor activity and in the object recognition test for recognition memory evaluation. The brains were collected for immunohistochemical analysis of Aβ plaques and microglia, and blood was collected for plasma corticosterone by ELISA assay. Results APP/PS1 transgenic sedentary mice showed increased hippocampal Aβ plaques and higher plasma corticosterone levels, as well as hyperlocomotion and reduced central crossings in the open field test, compared to APP/PS1 exercised and control animals. The intermittent program of RE was able to recover the behavioral, corticosterone and Aβ alterations to the CTRL levels. In addition, the RE protocol increased the number of microglial cells in the hippocampus of APP/PS1 mice. Despite these alterations, no memory impairment was observed in APP/PS1 mice in the novel object recognition test. Discussion Altogether, the present results suggest that RE plays a role in alleviating AD symptoms, and highlight the beneficial effects of RE training as a complementary treatment for AD.
Collapse
Affiliation(s)
- Henrique Correia Campos
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deidiane Elisa Ribeiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Debora Hashiguchi
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- Instituto do Cérebro - ICe, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Milena da Silva Milanis
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Christiane Gimenes
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deborah Suchecki
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ricardo Mario Arida
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- *Correspondence: Henning Ulrich, ; Beatriz Monteiro Longo, ;
| | - Beatriz Monteiro Longo
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- *Correspondence: Henning Ulrich, ; Beatriz Monteiro Longo, ;
| |
Collapse
|
11
|
Rahman MR, Akter R, Neelotpol S, Mayesha II, Afrose A. The Neuropathological Impacts of COVID-19: Challenges and Alternative Treatment Options for Alzheimer's Like Brain Changes on Severely SARS-CoV-2 Infected Patients. Am J Alzheimers Dis Other Demen 2023; 38:15333175231214974. [PMID: 37972355 PMCID: PMC10655662 DOI: 10.1177/15333175231214974] [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: 11/19/2023]
Abstract
Recently, some researchers claimed neuropathological changes lead to Alzheimer's-like brains after severe infection of SARS-CoV-2. Several mechanisms have been postulated on how SARS-CoV-2 neurological damage leads to Alzheimer's disease (AD) development. Neurobiochemical changes during infection may significantly induce Alzheimer's disease in severely COVID-19 infected people. The immune system is also compromised while infected by this novel coronavirus. However, recent studies are insufficient to conclude the relationship between Alzheimer's disease and COVID-19. This review demonstrates the possible pathways of neuropathological changes induced by the SARS-CoV-2 virus in AD patients or leading to AD in COVID-19 patients. Therefore, this study delineates the challenges for COVID-19 infected AD patients and the mechanism of actions of natural compounds and alternative treatments to overcome those. Furthermore, animal studies and a large cohort of COVID-19 survivors who showed neuroinflammation and neurological changes may augment the research to discover the relationship between Alzheimer's disease and COVID-19.
Collapse
Affiliation(s)
- Md. Rashidur Rahman
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, Bangladesh
| | | | | | | | - Afrina Afrose
- School of Pharmacy, BRAC University, Dhaka, Bangladesh
| |
Collapse
|
12
|
Bielak AAM, Gow AJ. A Decade Later on How to "Use It" So We Don't "Lose It": An Update on the Unanswered Questions about the Influence of Activity Participation on Cognitive Performance in Older Age. Gerontology 2023; 69:336-355. [PMID: 35709704 DOI: 10.1159/000524666] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 04/19/2022] [Indexed: 01/13/2023] Open
Abstract
Activity engagement is a modifiable factor that has been widely-cited as being good for the aging brain and cognition and represents a valuable target for reducing dementia risk. However, specific issues about activity engagement (mental, social, and physical) and cognition in older adulthood remain, and Bielak [Gerontology 2010;56: 507-519] reviewed seven major methodological and theoretical questions about this relationship. We present an updated reflection on these key questions, focusing on research published in the last 10 years. For some questions, a significant amount of work has been done and conclusions have become clearer; for others, there have been few additions to the literature and our knowledge remains much the same as it was a decade ago. We review the issues identified in the 2010 paper including the directionality and temporal nature of the relationship; whether specific activity domains offer different benefits to cognition and what domain(s) of cognition are affected; variation in the relation by age, gender, or education; potential mechanisms involved; and how activity engagement is assessed. For each, we present the most up-to-date research, discuss remaining challenges and possible future directions. This formal unifying of the information in the field is intended as a guide to support continued progress by spurring on studies addressing specific questions while reminding researchers of critical issues. We conclude with recommendations that future studies investigating the link between activity engagement and cognitive performance in adulthood should consider.
Collapse
Affiliation(s)
- Allison A M Bielak
- Department of Human Development and Family Studies, Colorado State University, Fort Collins, Colorado, USA
| | - Alan J Gow
- Centre for Applied Behavioural Sciences and Department of Psychology, Heriot-Watt University, Edinburgh, UK
| |
Collapse
|
13
|
Geng R, Zhang Y, Liu M, Deng S, Ding J, Zhong H, Tu Q. Elevated serum uric acid is associated with cognitive improvement in older American adults: A large, population-based-analysis of the NHANES database. Front Aging Neurosci 2022; 14:1024415. [PMID: 36570535 PMCID: PMC9772611 DOI: 10.3389/fnagi.2022.1024415] [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: 08/21/2022] [Accepted: 11/15/2022] [Indexed: 12/14/2022] Open
Abstract
Background The many studies revealing a connection between serum uric acid (SUA) and dementia have reported conflicting results. This study sought to investigate the relations between SUA and cognitive function in older adults. Materials and methods The sample was 2,767 American adults aged ≥60 years from the National Health and Nutrition Examination Survey 2011-2014. Cognitive performance was evaluated by the Consortium to Establish a Registry for Alzheimer's Disease test, animal fluency test, digit symbol substitution test, and composite z-score. Multivariate linear regression analyses were conducted to estimate the association between SUA and cognitive function. Results SUA level and cognitive function were significantly, positively correlated. Age significantly correlated with the association between SUA and cognitive function. Conclusion These findings support a connection between SUA and cognition, showing a positive link between SUA and cognitive scores among older American adults. We contend that a slight rise in uric acid within the normal range is advantageous for enhanced cognition. To confirm the precise dose-time-response relation, more tests will be needed.
Collapse
|
14
|
Abstract
In this article, the authors discuss primarily what is known about the epidemiology of all-cause dementia. Dementia is caused by a complex interplay of genetics, comorbidities, and lifestyle factors, and drug development has been challenging. However, evidence from large, prospective, observational studies has identified a variety of factors that may prevent or delay the onset of dementia. Several of these factors are modifiable and lend themselves to well to treatments currently available. The authors discuss the state of current evidence on dementia risk factors, the most promising avenues, and future directions for dementia prevention and management.
Collapse
Affiliation(s)
- Christina S Dintica
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA.
| |
Collapse
|
15
|
Sex- and Neuropsychiatric-Dependent Circadian Alterations in Daily Voluntary Physical Activity Engagement and Patterns in Aged 3xTg-AD Mice. Int J Mol Sci 2022; 23:ijms232213671. [PMID: 36430150 PMCID: PMC9696337 DOI: 10.3390/ijms232213671] [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: 10/02/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease (AD) patients suffer from circadian rhythm alterations affecting their daily physical activity patterns with less willingness to perform a voluntary exercise. In preclinical studies, there is no clarity on whether animal models of AD can replicate these impairments. Here, we provide a proof of concept of the performance and behavioral effects of four weeks of voluntary wheel running (VWR) in a group of 14-month-old male and female 3xTg-AD mice at advanced stages of AD and the daily variance (behavioral circadian rhythmicity) of VWR associated with sex and their neuropsychiatric-like phenotype. Higher levels of horizontal exploration in the open field (OF) test were found in mice submitted to exercise. A linear mixed effect model showed significant sex-dependent differences in the VWR activity performed on the first night of follow-up, with high-NIBI males running less than high-NIBI females. Thus, an influence of NPS-like symptoms on the circadian patterns of VWR may account for such differences. In addition, males remained more active than females during diurnal periods. We hypothesize that this increment in energy expenditure during resting periods may be related to hyperactive behavior, similar to that observed in humans' exacerbated agitation or sundowning behavior. These findings support the usage of the 3xTg-AD mouse as a reliable model for studying circadian rhythm alterations in AD and, at the translational level, the importance of tailored and individualized physical activity programs in clinical settings.
Collapse
|
16
|
A Bibliometric and Visual Analysis of Exercise Intervention Publications for Alzheimer’s Disease (1998–2021). J Clin Med 2022; 11:jcm11195903. [PMID: 36233770 PMCID: PMC9571385 DOI: 10.3390/jcm11195903] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia worldwide, posing a considerable economic burden to patients and society as a whole. Exercise has been confirmed as a non-drug intervention method in the related literature on AD. However, at present, there are still few bibliometric studies on AD exercise research. In order to fill the gap, this paper aims to intuitively analyze the growth in AD exercise literature published from 1998 to 2021 using bibliometrics, providing historical insights for scientific research circles. The main source of literature retrieval is the Web of Science database. Using the Boolean operator tools “OR” and “AND” combined with keywords related to “exercise” and “Alzheimer’s disease”, we conducted a title search and obtained 247 documents. Using Microsoft Excel, Datawrapper, and Biblioshiny, this study carried out a bibliometric analysis of countries, institutions, categories, journals, documents, authors, and keyword plus terms. The study found that the number of papers published from 2016 to 2021 had the greatest increase, which may have been influenced by the Global Dementia Report 2015 and COVID-19. Interdisciplinary cooperation and the research results published in high-scoring journals actively promoted research and development in the AD exercise field. The United States and the University of Minnesota system play a central role in this field. In future, it will be necessary to explore the effectiveness and feasibility of multi-mode interventions on an active lifestyle, including exercise, in different groups and environments worldwide. This study may provide a direction and path for future research by showing the global overview, theme evolution, and future trends of research results in the AD exercise field.
Collapse
|
17
|
Huang SY, Li YZ, Zhang YR, Huang YY, Wu BS, Zhang W, Deng YT, Chen SD, He XY, Chen SF, Dong Q, Zhang C, Chen RJ, Suckling J, Rolls ET, Feng JF, Cheng W, Yu JT. Sleep, physical activity, sedentary behavior, and risk of incident dementia: a prospective cohort study of 431,924 UK Biobank participants. Mol Psychiatry 2022; 27:4343-4354. [PMID: 35701596 DOI: 10.1038/s41380-022-01655-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 02/07/2023]
Abstract
Although sleep, physical activity and sedentary behavior have been found to be associated with dementia risk, findings are inconsistent and their joint relationship remains unclear. This study aimed to investigate independent and joint associations of these three modifiable behaviors with dementia risks. A total of 431,924 participants (median follow-up 9.0 years) without dementia from UK Biobank were included. Multiple Cox regressions were used to estimate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs). Models fitted with restricted cubic spline were conducted to test for linear and nonlinear shapes of each association. Sleep duration, leisure-time physical activity (LTPA), and screen-based sedentary behavior individually associated with dementia risks in different non-linear patterns. Sleep duration associated with dementia in a U-shape with a nadir at 7 h/day. LTPA revealed a curvilinear relationship with dementia in diminishing tendency, while sedentary behavior revealed a J-shaped relationship. The dementia risk was 17% lower in the high LTPA group (HR[95%CI]: 0.83[0.76-0.91]) and 22% higher in the high sedentary behavior group (1.22[1.10-1.35]) compared to the corresponding low-level group, respectively. A combination of seven-hour/day sleep, moderate-to-high LTPA, and low-to-moderate sedentary behavior showed the lowest dementia risk (0.59[0.50-0.69]) compared to the referent group (longer or shorter sleep/low LTPA/high sedentary behavior). Notably, each behavior was non-linearly associated with brain structures in a pattern similar to its association with dementia, suggesting they may affect dementia risk by affecting brain structures. Our findings highlight the potential to change these three daily behaviors individually and simultaneously to reduce the risk of dementia.
Collapse
Affiliation(s)
- Shu-Yi Huang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Zhu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Yuan Huang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Yu He
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shu-Fen Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ren-Jie Chen
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University, Shanghai, China
| | - John Suckling
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Edmund T Rolls
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Oxford Centre for Computational Neuroscience, Oxford, UK
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China.
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer Center, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Fox FAU, Diers K, Lee H, Mayr A, Reuter M, Breteler MMB, Aziz NA. Association Between Accelerometer-Derived Physical Activity Measurements and Brain Structure: A Population-Based Cohort Study. Neurology 2022; 99:e1202-e1215. [PMID: 35918154 PMCID: PMC9536740 DOI: 10.1212/wnl.0000000000200884] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/11/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES While there is growing evidence that physical activity promotes neuronal health, studies examining the relation between physical activity and brain morphology remain inconclusive. We therefore examined whether objectively quantified physical activity is related to brain volume, cortical thickness, and gray matter density in a large cohort study. In addition, we assessed molecular pathways that may underlie the effects of physical activity on brain morphology. METHODS We used cross-sectional baseline data from 2,550 eligible participants (57.6% women; mean age: 54.7 years, range: 30-94 years) of a prospective cohort study. Physical activity dose (metabolic equivalent hours and step counts) and intensity (sedentary and light-intensity and moderate-to-vigorous intensity activities) were recorded with accelerometers. Brain volumetric, gray matter density, and cortical thickness measures were obtained from 3T MRI scans using FreeSurfer and Statistical Parametric Mapping. The relation of physical activity (independent variable) and brain structure (outcome) was examined with polynomial multivariable regression, while adjusting for age, sex, intracranial volume, education, and smoking. Using gene expression profiles from the Allen Brain Atlas, we extracted molecular signatures associated with the effects of physical activity on brain morphology. RESULTS Physical activity dose and intensity were independently associated with larger brain volumes, gray matter density, and cortical thickness of several brain regions. The effects of physical activity on brain volume were most pronounced at low physical activity quantities and differed between men and women and across age. For example, more time spent in moderate-to-vigorous intensity activities was associated with greater total gray matter volume, but the relation leveled off with more activity (standardized β [95% CIs]: 1.37 [0.35-2.39] and -0.70 [-1.25 to -0.15] for the linear and quadratic terms, respectively). The strongest effects of physical activity were observed in motor regions and cortical regions enriched for genes involved in mitochondrial respiration. DISCUSSION Our findings suggest that physical activity benefits brain health, with the strongest effects in motor regions and regions with a high oxidative demand. While young adults may particularly profit from additional high-intensity activities, older adults may already benefit from light-intensity activities. Physical activity and reduced sedentary time may be critical in the prevention of age-associated brain atrophy and neurodegenerative diseases.
Collapse
Affiliation(s)
- Fabienne A U Fox
- From the Population Health Sciences (F.A.U.F., H.L., M.M.B.B., N.A.A.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Image Analysis (K.D., M.R.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Institute for Medical Biometry (A.M., M.M.B.B.), Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany; A.A. Martinos Center for Biomedical Imaging (M.R.), Massachusetts General Hospital, Boston; Department of Radiology (M.R.), Harvard Medical School, Boston, MA; and Department of Neurology (N.A.A.), Faculty of Medicine, University of Bonn, Germany
| | - Kersten Diers
- From the Population Health Sciences (F.A.U.F., H.L., M.M.B.B., N.A.A.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Image Analysis (K.D., M.R.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Institute for Medical Biometry (A.M., M.M.B.B.), Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany; A.A. Martinos Center for Biomedical Imaging (M.R.), Massachusetts General Hospital, Boston; Department of Radiology (M.R.), Harvard Medical School, Boston, MA; and Department of Neurology (N.A.A.), Faculty of Medicine, University of Bonn, Germany
| | - Hweeling Lee
- From the Population Health Sciences (F.A.U.F., H.L., M.M.B.B., N.A.A.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Image Analysis (K.D., M.R.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Institute for Medical Biometry (A.M., M.M.B.B.), Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany; A.A. Martinos Center for Biomedical Imaging (M.R.), Massachusetts General Hospital, Boston; Department of Radiology (M.R.), Harvard Medical School, Boston, MA; and Department of Neurology (N.A.A.), Faculty of Medicine, University of Bonn, Germany
| | - Andreas Mayr
- From the Population Health Sciences (F.A.U.F., H.L., M.M.B.B., N.A.A.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Image Analysis (K.D., M.R.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Institute for Medical Biometry (A.M., M.M.B.B.), Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany; A.A. Martinos Center for Biomedical Imaging (M.R.), Massachusetts General Hospital, Boston; Department of Radiology (M.R.), Harvard Medical School, Boston, MA; and Department of Neurology (N.A.A.), Faculty of Medicine, University of Bonn, Germany
| | - Martin Reuter
- From the Population Health Sciences (F.A.U.F., H.L., M.M.B.B., N.A.A.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Image Analysis (K.D., M.R.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Institute for Medical Biometry (A.M., M.M.B.B.), Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany; A.A. Martinos Center for Biomedical Imaging (M.R.), Massachusetts General Hospital, Boston; Department of Radiology (M.R.), Harvard Medical School, Boston, MA; and Department of Neurology (N.A.A.), Faculty of Medicine, University of Bonn, Germany
| | - Monique M B Breteler
- From the Population Health Sciences (F.A.U.F., H.L., M.M.B.B., N.A.A.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Image Analysis (K.D., M.R.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Institute for Medical Biometry (A.M., M.M.B.B.), Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany; A.A. Martinos Center for Biomedical Imaging (M.R.), Massachusetts General Hospital, Boston; Department of Radiology (M.R.), Harvard Medical School, Boston, MA; and Department of Neurology (N.A.A.), Faculty of Medicine, University of Bonn, Germany
| | - N Ahmad Aziz
- From the Population Health Sciences (F.A.U.F., H.L., M.M.B.B., N.A.A.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Image Analysis (K.D., M.R.), German Center for Neurodegenerative Diseases (DZNE), Bonn; Institute for Medical Biometry (A.M., M.M.B.B.), Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany; A.A. Martinos Center for Biomedical Imaging (M.R.), Massachusetts General Hospital, Boston; Department of Radiology (M.R.), Harvard Medical School, Boston, MA; and Department of Neurology (N.A.A.), Faculty of Medicine, University of Bonn, Germany.
| |
Collapse
|
19
|
Asher S, Stephen R, Mäntylä P, Suominen AL, Solomon A. Periodontal health, cognitive decline, and dementia: A systematic review and meta-analysis of longitudinal studies. J Am Geriatr Soc 2022; 70:2695-2709. [PMID: 36073186 PMCID: PMC9826143 DOI: 10.1111/jgs.17978] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Emerging evidence indicates that poor periodontal health adversely impacts cognition. This review examined the available longitudinal evidence concerning the effect of poor periodontal health on cognitive decline and dementia. METHODS Comprehensive literature search was conducted on five electronic databases for relevant studies published until April 2022. Longitudinal studies having periodontal health as exposure and cognitive decline and/or dementia as outcomes were considered. Random effects pooled estimates and 95% confidence intervals were generated (pooled odds ratio for cognitive decline and hazards ratio for dementia) to assess whether poor periodontal health increases the risk of cognitive decline and dementia. Heterogeneity between studies was estimated by I2 and the quality of available evidence was assessed through quality assessment criteria. RESULTS Adopted search strategy produced 2132 studies for cognitive decline and 2023 for dementia, from which 47 studies (24 for cognitive decline and 23 for dementia) were included in this review. Poor periodontal health (reflected by having periodontitis, tooth loss, deep periodontal pockets, or alveolar bone loss) was associated with both cognitive decline (OR = 1.23; 1.05-1.44) and dementia (HR = 1.21; 1.07-1.38). Further analysis, based on measures of periodontal assessment, found tooth loss to independently increase the risk of both cognitive decline (OR = 1.23; 1.09-1.39) and dementia (HR = 1.13; 1.04-1.23). Stratified analysis based on the extent of tooth loss indicated partial tooth loss to be important for cognitive decline (OR = 1.50; 1.02-2.23) and complete tooth loss for dementia (HR = 1.23; 1.05-1.45). However, the overall quality of evidence was low, and associations were at least partly due to reverse causality. CONCLUSIONS Poor periodontal health and tooth loss appear to increase the risk of both cognitive decline and dementia. However, the available evidence is limited (e.g., highly heterogenous, lacking robust methodology) to draw firm conclusions. Further well-designed studies involving standardized periodontal and cognitive health assessment and addressing reverse causality are highly warranted.
Collapse
Affiliation(s)
- Sam Asher
- Institute of DentistryUniversity of Eastern FinlandKuopioFinland
| | - Ruth Stephen
- Neurology, Institute of Clinical MedicineUniversity of Eastern FinlandKuopioFinland
| | - Päivi Mäntylä
- Institute of DentistryUniversity of Eastern FinlandKuopioFinland
- Department of Oral and Maxillofacial DiseasesKuopio University HospitalKuopioFinland
| | - Anna Liisa Suominen
- Institute of DentistryUniversity of Eastern FinlandKuopioFinland
- Department of Oral and Maxillofacial DiseasesKuopio University HospitalKuopioFinland
| | - Alina Solomon
- Neurology, Institute of Clinical MedicineUniversity of Eastern FinlandKuopioFinland
- Division of Clinical Geriatrics, NVSKarolinska InstituteStockholmSweden
- Aging Epidemiology Research Unit, School of Public HealthImperial College LondonLondonUK
| |
Collapse
|
20
|
Yang JJ, Keohane LM, Pan XF, Qu R, Shu XO, Lipworth L, Braun K, Steinwandel MD, Dai Q, Shrubsole M, Zheng W, Blot WJ, Yu D. Association of Healthy Lifestyles With Risk of Alzheimer Disease and Related Dementias in Low-Income Black and White Americans. Neurology 2022; 99:e944-e953. [PMID: 35697505 PMCID: PMC9502739 DOI: 10.1212/wnl.0000000000200774] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/08/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Although the importance of healthy lifestyles for preventing Alzheimer disease and related dementias (ADRD) has been recognized, epidemiologic evidence remains limited for non-White or low-income individuals who bear disproportionate burdens of ADRD. This population-based cohort study aims to investigate associations of lifestyle factors, individually and together, with the risk of ADRD among socioeconomically disadvantaged Americans. METHODS In the Southern Community Cohort Study, comprising two-thirds self-reported Black and primarily low-income Americans, we identified incident ADRD using claims data among participants enrolled in Medicare for at least 12 consecutive months after age 65 years. Five lifestyle factors-tobacco smoking, alcohol consumption, leisure-time physical activity (LTPA), sleep hours, and diet quality-were each scored 0 (unhealthy), 1 (intermediate), or 2 (healthy) based on the health guidelines. A composite lifestyle score was created by summing all scores. Cox regression was used to estimate hazard ratios (HRs, 95% CIs) for incident ADRD, treating death as a competing risk. RESULTS We identified 1,694 patients with newly diagnosed ADRD among 17,209 participants during a median follow-up of 4.0 years in claims data; the mean age at ADRD diagnosis was 74.0 years. Healthy lifestyles were individually associated with an 11%-25% reduced risk of ADRD: multivariable-adjusted HR (95% CI) was 0.87 (0.76-0.99) for never vs current smoking, 0.81 (0.72-0.92) for low-to-moderate vs no alcohol consumption, 0.89 (0.77-1.03) for ≥150 minutes of moderate or ≥75 minutes of vigorous LTPA each week vs none, 0.75 (0.64-0.87) for 7-9 hours vs >9 hours of sleep, and 0.85 (0.75-0.96) for the highest vs lowest tertiles of the Healthy Eating Index. The composite lifestyle score showed a dose-response association with up to 36% reduced risk of ADRD: multivariable-adjusted HRs (95% CIs) across quartiles were 1 (ref), 0.88 (0.77-0.99), 0.79 (0.70-0.90), and 0.64 (0.55-0.74); p trend <0.001. The beneficial associations were observed regardless of participants' sociodemographics (e.g., race, education, and income) and health conditions (e.g., history of cardiometabolic diseases and depression). DISCUSSION Our findings support significant benefits of healthy lifestyles for ADRD prevention among socioeconomically disadvantaged Americans, suggesting that promoting healthy lifestyles and reducing barriers to lifestyle changes are crucial to tackling the growing burden and disparities posed by ADRD.
Collapse
Affiliation(s)
- Jae Jeong Yang
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Laura M Keohane
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Xiong-Fei Pan
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Ruiqi Qu
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Xiao-Ou Shu
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Loren Lipworth
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Kyle Braun
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Mark D Steinwandel
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Qi Dai
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Martha Shrubsole
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Wei Zheng
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - William J Blot
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Danxia Yu
- From the Division of Epidemiology (J.J.Y., X.-F.P., R.Q., X.-O.S., L.L., Q.D., M.S., W.Z., W.J.B., D.Y.), Department of Medicine, Vanderbilt University Medical Center; Department of Health Policy (L.M.K., K.B.), Vanderbilt University School of Medicine; and International Epidemiology Field Station (M.D.S.), Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
21
|
Samuel I, Ben-Haroush Schyr R, Arad Y, Attali T, Azulai S, Bergel M, Halfon A, Hefetz L, Hirsch T, Israeli H, Lax N, Nitzan K, Sender D, Sweetat S, Okun E, Rosenmann H, Ben-Zvi D. Sleeve Gastrectomy Reduces Glycemia but Does Not Affect Cognitive Impairment in Lean 5xFAD Mice. Front Neurosci 2022; 16:937663. [PMID: 36033613 PMCID: PMC9403181 DOI: 10.3389/fnins.2022.937663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity and hyperglycemia are risk factors for cognitive decline and for the development of Alzheimer’s Disease (AD). Bariatric surgery is an effective treatment for obesity that was shown to improve cognitive decline in obese patients. Bariatric surgery was shown to exert weight loss independent effects on metabolic diseases such as type 2 diabetes. We tested whether sleeve gastrectomy (SG), a common bariatric surgery, can affect the cognitive impairment in lean, normoglycemic female 5xFAD mice, a genetic model for AD. 5xFAD mice and wild-type (WT) littermates underwent SG or sham surgery at the age of 5 months and were tested for metabolic, behavioral, and molecular phenotypes 90 days later. SG led to a reduction in blood glucose levels and total plasma cholesterol levels in 5xFAD mice without inducing weight loss. However, the surgery did not affect the outcomes of long-term spatial memory tests in these mice. Analysis of β-Amyloid plaques corroborated the behavioral studies in showing no effect of surgery on the molecular phenotype of 5xFAD mice. In conclusion, SG leads to an improved metabolic profile in lean female 5xFAD mice without inducing weight loss but does not affect the brain pathology or behavioral phenotype. Our results suggest that the positive effects of bariatric surgery on cognitive decline in obese patients are likely attributed to weight loss and improvement in obesity sequelae, and not to weight loss independent effects of surgery.
Collapse
Affiliation(s)
- Itia Samuel
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Rachel Ben-Haroush Schyr
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Yhara Arad
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
- Department of Military Medicine and Tzameret, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tamar Attali
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Shira Azulai
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Michael Bergel
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Aviv Halfon
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
- Department of Military Medicine and Tzameret, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Liron Hefetz
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
- Department of Military Medicine and Tzameret, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tamir Hirsch
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer’s Disease Research, Bar-Ilan University, Ramat Gan, Israel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Hadar Israeli
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Neta Lax
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Keren Nitzan
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Dana Sender
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Sahar Sweetat
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer’s Disease Research, Bar-Ilan University, Ramat Gan, Israel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Hanna Rosenmann
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
- *Correspondence: Hanna Rosenmann,
| | - Danny Ben-Zvi
- Department of Developmental Biology and Cancer Research, The Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
- Danny Ben-Zvi,
| |
Collapse
|
22
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
23
|
Liang YJ, Su QW, Sheng ZR, Weng QY, Niu YF, Zhou HD, Liu CB. Effectiveness of Physical Activity Interventions on Cognition, Neuropsychiatric Symptoms, and Quality of Life of Alzheimer’s Disease: An Update of a Systematic Review and Meta-Analysis. Front Aging Neurosci 2022; 14:830824. [PMID: 35309887 PMCID: PMC8926300 DOI: 10.3389/fnagi.2022.830824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/08/2022] [Indexed: 01/05/2023] Open
Abstract
The topic of physical activity interventions for the treatment of Alzheimer’s disease (AD) has been discussed for decades, but there are still inconsistent views on the effect of its intervention in different studies. With the increase in randomized controlled trials (RCTs), it is necessary to update newly published studies and systematically evaluate the effects of physical activity interventions. Scientific citation databases (e.g., PubMed, EMBASE, etc.) and registration databases (e.g., ISRCTN, CHICTR, etc.) were checked to screen RCTs and systematic reviews of physical activity interventions in AD. Then extract and review the intervention methods and their evaluation results in the included studies. Spearman correlation method was used to test the association between the mean difference (MD) of intervention results and activity time. The Hedges’g method was used to combine continuous data to analyze the standard MD (SMD) of different intervention types or time subgroups. The overall results show that physical activity intervention can improve the cognition, neuropsychiatric symptoms and quality of life (Qol) of AD patients, but the duration of the intervention significantly affected the outcome of the assessment. Subgroup analysis results showed that an intervention duration of 2–5 months had a significant advantage: cognitive function (Minimum Mental State Examination: SMD = 0.47, 95% CI = 0.33 ∼ 0.61, P < 0.01), neuropsychiatric symptoms (Neuropsychiatric Inventory: SMD = −0.48, 95% CI = −0.85 ∼−0.11, P < 0.01), and quality of life (Qol-AD: SMD = 0.47, 95% CI = 0.23 ∼ 0.71, P < 0.01). The systematic review and analysis results of updated RCTs suggested that short-term (2–5 months) physical activity interventions were more beneficial in improving cognitive function, neuropsychiatric symptoms and Qol in patients with AD. And there was no evidence of differences in the effectiveness of different physical activity interventions.
Collapse
Affiliation(s)
- Ya-Jing Liang
- Department of Nursing, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- *Correspondence: Ya-Jing Liang,
| | - Qing-Wen Su
- Department of Health Management Center, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Zhi-Ren Sheng
- Department of Nursing, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Qiu-Yan Weng
- Department of Neurology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Yan-Fang Niu
- Department of Neurology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Hong-Di Zhou
- Department of Nursing, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Chun-Bo Liu
- Department of Nursing, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- Chun-Bo Liu,
| |
Collapse
|
24
|
Lamberti V, Palermi S, Franceschin A, Scapol G, Lamberti V, Lamberti C, Vecchiato M, Spera R, Sirico F, Della Valle E. The Effectiveness of Adapted Personalized Motor Activity (AMPA) to Improve Health in Individuals with Mental Disorders and Physical Comorbidities: A Randomized Controlled Trial. Sports (Basel) 2022; 10:sports10030030. [PMID: 35324639 PMCID: PMC8952683 DOI: 10.3390/sports10030030] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
Mental disorders are highly prevalent worldwide and have a high impact on daily functioning. Exercise therapy was found to improve health of individuals with physical and mental disorders. This study aims to investigate the effectiveness of an Adapted Personalized Motor Activity (AMPA) in improving health in individuals with physical and mental disorders. Forty-three patients affected by both mental and chronic nontransmissible conditions were randomly assigned to intervention group (AMPA intervention) and control group (no intervention). Perceived physical and mental health were assessed using the Short Form 12 (SF-12) questionaries. Moreover, subjects underwent an accurate medical screening process, complete clinical evaluation, body composition evaluation, and cardiopulmonary assessment. Repeated Measurement Analysis of the Variance (RM-ANOVA) was used to compare any changes in health and physiological parameters in-between groups. AMPA group showed a statistically significant improvement in both perceived mental and physical health. Moreover, Body Mass Index (BMI), glycolipid profile, aerobic functional capacity and cardiopulmonary parameters improved significantly among individuals from the intervention group compared with the individuals from the control group. AMPA may be considered a possible intervention to improve health in individuals suffering from multiple physical and mental disorders. Future studies should examine the effectiveness in larger and heterogeneous sample of chronically ill patients and the long-term effect of AMPA.
Collapse
Affiliation(s)
- Vito Lamberti
- Sport Medicine and Motor Activity Institute c.FMSI-CONI, Vittorio Veneto, 31029 Treviso, Italy; (V.L.); (A.F.); (G.S.); (V.L.); (C.L.)
| | - Stefano Palermi
- Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (R.S.); (F.S.); (E.D.V.)
- Correspondence:
| | - Andrea Franceschin
- Sport Medicine and Motor Activity Institute c.FMSI-CONI, Vittorio Veneto, 31029 Treviso, Italy; (V.L.); (A.F.); (G.S.); (V.L.); (C.L.)
| | - Giovanni Scapol
- Sport Medicine and Motor Activity Institute c.FMSI-CONI, Vittorio Veneto, 31029 Treviso, Italy; (V.L.); (A.F.); (G.S.); (V.L.); (C.L.)
| | - Vincenzo Lamberti
- Sport Medicine and Motor Activity Institute c.FMSI-CONI, Vittorio Veneto, 31029 Treviso, Italy; (V.L.); (A.F.); (G.S.); (V.L.); (C.L.)
| | - Chiara Lamberti
- Sport Medicine and Motor Activity Institute c.FMSI-CONI, Vittorio Veneto, 31029 Treviso, Italy; (V.L.); (A.F.); (G.S.); (V.L.); (C.L.)
| | - Marco Vecchiato
- Sports and Exercise Medicine Division, Department of Medicine, University of Padova, 35100 Padova, Italy;
| | - Rocco Spera
- Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (R.S.); (F.S.); (E.D.V.)
| | - Felice Sirico
- Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (R.S.); (F.S.); (E.D.V.)
| | - Elisabetta Della Valle
- Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (R.S.); (F.S.); (E.D.V.)
| |
Collapse
|
25
|
Schreiner TG, Popescu BO. Impact of Caffeine on Alzheimer’s Disease Pathogenesis—Protective or Risk Factor? Life (Basel) 2022; 12:life12030330. [PMID: 35330081 PMCID: PMC8952218 DOI: 10.3390/life12030330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), the most common dementia worldwide, remains without an effective treatment to this day despite intensive research conducted during the last decades. In this context, researchers have turned their attention towards the prevention of this pathology, focusing on early detection and better control of the most important risk factors, concomitantly with trying to find potentially protective factors that may delay the onset of AD. From the multitude of factors studied, coffee (especially its main component, caffeine) is a current interesting research topic, taking into consideration the contradictory results of recent years’ studies. On the one hand, much of the evidence from fundamental research suggests the potentially protective trait of caffeine in AD, while other data mainly from human studies lean toward no correlation or even suggesting that caffeine is a veritable risk factor for dementia. Given the methodological heterogeneity of the studies, this review aims to bring new evidence regarding this topic and to try to clearly establish a correlation between the two entities. Thus, in the first part, the authors make a clear distinction between the effects of coffee and the effects of caffeine in AD, presenting a rich basis of clinical trials on both animal models and the human subject. Subsequently, the main pathophysiological mechanisms that would explain the action of caffeine in the etiopathogenesis of AD are reviewed. Finally, the role of computational models is presented, having beneficial impact on both better understanding of the disease mechanism and the development of new therapeutic approaches for AD prevention.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania;
- Department of Neurology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
- Correspondence:
| | - Bogdan Ovidiu Popescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania;
- Neurology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
26
|
Falsetti L, Viticchi G, Zaccone V, Guerrieri E, Moroncini G, Luzzi S, Silvestrini M. Shared Molecular Mechanisms among Alzheimer’s Disease, Neurovascular Unit Dysfunction and Vascular Risk Factors: A Narrative Review. Biomedicines 2022; 10:biomedicines10020439. [PMID: 35203654 PMCID: PMC8962428 DOI: 10.3390/biomedicines10020439] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/27/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia, affecting 24 million individuals. Clinical and epidemiological studies have found several links between vascular risk factors (VRF), neurovascular unit dysfunction (NVUd), blood-brain barrier breakdown (BBBb) and AD onset and progression in adulthood, suggesting a pathogenetic continuum between AD and vascular dementia. Shared pathways between AD, VRF, and NVUd/BBB have also been found at the molecular level, underlining the strength of this association. The present paper reviewed the literature describing commonly shared molecular pathways between adult-onset AD, VRF, and NVUd/BBBb. Current evidence suggests that VRF and NVUd/BBBb are involved in AD neurovascular and neurodegenerative pathology and share several molecular pathways. This is strongly supportive of the hypothesis that the presence of VRF can at least facilitate AD onset and progression through several mechanisms, including NVUd/BBBb. Moreover, vascular disease and several comorbidities may have a cumulative effect on VRF and worsen the clinical manifestations of AD. Early detection and correction of VRF and vascular disease by improving NVUd/BBBd could be a potential target to reduce the overall incidence and delay cognitive impairment in AD.
Collapse
Affiliation(s)
- Lorenzo Falsetti
- Internal and Subintensive Medicine Department, Azienda Ospedaliero-Universitaria “Ospedali Riuniti” di Ancona, 60100 Ancona, Italy;
- Correspondence: ; Tel.: +39-071-596-5269
| | - Giovanna Viticchi
- Neurologic Clinic, Marche Polytechnic University, 60126 Ancona, Italy; (G.V.); (S.L.); (M.S.)
| | - Vincenzo Zaccone
- Internal and Subintensive Medicine Department, Azienda Ospedaliero-Universitaria “Ospedali Riuniti” di Ancona, 60100 Ancona, Italy;
| | - Emanuele Guerrieri
- Emergency Medicine Residency Program, Università Politecnica delle Marche, 60121 Ancona, Italy;
| | | | - Simona Luzzi
- Neurologic Clinic, Marche Polytechnic University, 60126 Ancona, Italy; (G.V.); (S.L.); (M.S.)
| | - Mauro Silvestrini
- Neurologic Clinic, Marche Polytechnic University, 60126 Ancona, Italy; (G.V.); (S.L.); (M.S.)
| |
Collapse
|
27
|
Associations of Objectively-Measured Sedentary Time and Patterns with Cognitive Function in Non-Demented Japanese Older Adults: A Cross-Sectional Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19041999. [PMID: 35206188 PMCID: PMC8871939 DOI: 10.3390/ijerph19041999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/08/2022] [Indexed: 02/01/2023]
Abstract
This study aimed to investigate the cross-sectional associations of objectively-measured sedentary time and patterns with cognitive function in Japanese older adults. A total of 1681 non-demented community-dwelling older adults (aged 73 ± 6, 62.1% women) were included. Total sedentary time, prolonged sedentary time (accumulated in ≥30 min bouts) and mean sedentary bout length were assessed using a tri-axial accelerometer. Global and domain-specific cognitive functions were measured using the Montreal Cognitive Assessment. The average of total sedentary time and prolonged sedentary time were 462 ± 125 and 186 ± 111 min/day, respectively. Greater prolonged sedentary time, but not total sedentary time, was significantly associated with poorer performance in the orientation domain even after controlling for moderate-to-vigorous physical activity (p for trend = 0.002). A significant inverse association was also observed between mean sedentary bout length and the orientation domain (p for trend = 0.009). No significant associations were observed for global cognitive function or other cognitive domains. Sedentary time accumulated in prolonged bouts, but not total sedentary time, was inversely associated with orientation ability among older adults. Our results encourage further researches to confirm the role of prolonged sedentary time in changes to cognitive domains over time among older adults.
Collapse
|
28
|
Yoon J, Isoda H, Ueda T, Okura T. Cognitive and physical benefits of a game‐like dual‐task exercise among the oldest nursing home residents in Japan. ALZHEIMER'S & DEMENTIA: TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2022; 8:e12276. [PMID: 35493411 PMCID: PMC9043709 DOI: 10.1002/trc2.12276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022]
Abstract
Introduction Dual‐task (DT) exercise can act as a substitute, which can help improve both physical and cognitive functions. Thus, this study investigated the effects of a game‐like cognitive DT exercise called “Synapsology” (SYNAP) among the oldest residents of a nursing home. Methods Participants (aged 85–97 years) were assigned to the intervention group (n = 12) and the control group (n = 12). The intervention group underwent 60‐minute sessions, twice a week for 24 weeks. Results A comparison of the Mini‐Mental State Examination scores and six physical function tests, before and after the intervention, shows that the SYNAP had a positive impact on the cognitive and physical functions among the intervention group. Discussion These findings suggest that SYNAP may help maintain or improve cognitive and physical functions among older adults compared to no interventions. Therefore, SYNAP would act as a beneficial tool amidst a “superaging” society like Japan.
Collapse
Affiliation(s)
- Jieun Yoon
- R&D Center for Tailor‐Made QOL University of Tsukuba Tsukuba Japan
| | - Hiroko Isoda
- R&D Center for Tailor‐Made QOL University of Tsukuba Tsukuba Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA) University of Tsukuba Tsukuba Japan
- Faculty of Life and Environmental Sciences University of Tsukuba Tsukuba Japan
| | | | - Tomohiro Okura
- R&D Center for Tailor‐Made QOL University of Tsukuba Tsukuba Japan
- Faculty of Health and Sport Sciences University of Tsukuba Tsukuba Japan
| |
Collapse
|
29
|
Kopf D. Diabetes mellitus und Demenz. DIABETOL STOFFWECHS 2021. [DOI: 10.1055/a-1146-7607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Daniel Kopf
- Geriatrische Klinik und Zentrum für Altersmedizin, Klinikum Ludwigsburg, Ludwigsburg, Deutschland
| |
Collapse
|
30
|
Wang S, Liu HY, Cheng YC, Su CH. Exercise Dosage in Reducing the Risk of Dementia Development: Mode, Duration, and Intensity-A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182413331. [PMID: 34948942 PMCID: PMC8703896 DOI: 10.3390/ijerph182413331] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/02/2022]
Abstract
Senile dementia, also known as dementia, is the mental deterioration which is associated with aging. It is characterized by a decrease in cognitive abilities, inability to concentrate, and especially the loss of higher cerebral cortex function, including memory, judgment, abstract thinking, and other loss of personality, even behavior changes. As a matter of fact, dementia is the deterioration of mental and intellectual functions caused by brain diseases in adults when they are mature, which affects the comprehensive performance of life and work ability. Most dementia cases are caused by Alzheimer’s disease (AD) and multiple infarct dementia (vascular dementia, multi-infarct dementia). Alzheimer’s disease is characterized by atrophy, shedding, and degenerative alterations in brain cells, and its occurrence is linked to age. The fraction of the population with dementia is smaller before the age of 65, and it increases after the age of 65. Since women live longer than men, the proportion of women with Alzheimer’s disease is higher. Multiple infarct dementia is caused by a cerebral infarction, which disrupts blood supply in multiple locations and impairs cerebral cortex function. Researchers worldwide are investigating ways to prevent Alzheimer’s disease; however, currently, there are no definitive answers for Alzheimer’s prevention. Even so, research has shown that we can take steps to reduce the risk of developing it. Prospective studies have found that even light to moderate physical activity can lower the risk of dementia and Alzheimer’s disease. Exercise has been proposed as a potential lifestyle intervention to help reduce the occurrence of dementia and Alzheimer’s disease. Various workout modes will be introduced based on various physical conditions. In general, frequent exercise for 6–8 weeks lessens the risk of dementia development.
Collapse
Affiliation(s)
- Sukai Wang
- College of Physical Education, Huaqiao University, Quanzhou 362021, China;
| | - Hong-Yu Liu
- Department of Exercise and Health Promotion, Chinese Culture University, Taipei 111369, Taiwan; (H.-Y.L.); (Y.-C.C.)
| | - Yi-Chen Cheng
- Department of Exercise and Health Promotion, Chinese Culture University, Taipei 111369, Taiwan; (H.-Y.L.); (Y.-C.C.)
| | - Chun-Hsien Su
- College of Kinesiology and Health, Chinese Culture University, Taipei 111369, Taiwan
- Correspondence: ; Tel.: +886-975159678
| |
Collapse
|
31
|
Effects of Aerobic Exercise Combined with Attentional Bias Training on Cognitive Function and Psychiatric Symptoms of Individuals with Methamphetamine Dependency: a Randomized Controlled Trial. Int J Ment Health Addict 2021. [DOI: 10.1007/s11469-021-00686-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
32
|
Potential causal effect of physical activity on reducing the risk of dementia: a 6-year cohort study from the Japan Gerontological Evaluation Study. Int J Behav Nutr Phys Act 2021; 18:140. [PMID: 34715877 PMCID: PMC8555243 DOI: 10.1186/s12966-021-01212-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/08/2021] [Indexed: 12/03/2022] Open
Abstract
Background The causal effect of physical activity on reducing dementia risk has been questioned due to the possibility of reverse causation. This study examined the potential causal effects of physical activity on reducing dementia risk using residency in a snowy area as an instrumental variable (IV) representing the physical activity of older adults. Methods We used cohort data from the Japan Gerontological Evaluation Study, a longitudinal cohort enrolling people aged 65 or older who were physically and cognitively independent in 2013; study participants were followed for an average of 5.7 years. Participants in the present study included 73,260 individuals living in 19 municipalities in Japan. Physical activity was measured by self-report questionnaires and the incidence of dementia was ascertained by linking participants to the public registries of long-term care insurance. IV estimation was obtained from a piecewise Cox proportional hazard model using a two-stage regression procedure. Results During the study period, we ascertained 8714 cases (11.9%) of dementia onset. In the IV analysis, we found that the frequency of physical activity per week was negatively associated with dementia risk, though the association weakened over time (Year 1: hazard ratio = 0.53, 95% confidence interval: 0.39–0.74; Year 4: 0.69, 0.53–0.90; Year 6: 0.85, 0.66–1.10). Conclusions Our IV analysis indicated a potential causal effect of physical activity on reducing dementia risk that persisted for at least 4 years of follow-up. Thus, we conclude that physical activity should be recommended for older adults to reduce dementia risk. Supplementary Information The online version contains supplementary material available at 10.1186/s12966-021-01212-w.
Collapse
|
33
|
Insulin and Insulin Resistance in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22189987. [PMID: 34576151 PMCID: PMC8472298 DOI: 10.3390/ijms22189987] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023] Open
Abstract
Insulin plays a range of roles as an anabolic hormone in peripheral tissues. It regulates glucose metabolism, stimulates glucose transport into cells and suppresses hepatic glucose production. Insulin influences cell growth, differentiation and protein synthesis, and inhibits catabolic processes such as glycolysis, lipolysis and proteolysis. Insulin and insulin-like growth factor-1 receptors are expressed on all cell types in the central nervous system. Widespread distribution in the brain confirms that insulin signaling plays important and diverse roles in this organ. Insulin is known to regulate glucose metabolism, support cognition, enhance the outgrowth of neurons, modulate the release and uptake of catecholamine, and regulate the expression and localization of gamma-aminobutyric acid (GABA). Insulin is also able to freely cross the blood–brain barrier from the circulation. In addition, changes in insulin signaling, caused inter alia insulin resistance, may accelerate brain aging, and affect plasticity and possibly neurodegeneration. There are two significant insulin signal transduction pathways: the PBK/AKT pathway which is responsible for metabolic effects, and the MAPK pathway which influences cell growth, survival and gene expression. The aim of this study is to describe the role played by insulin in the CNS, in both healthy people and those with pathologies such as insulin resistance and Alzheimer’s disease.
Collapse
|
34
|
Janoutová J, Kovalová M, Machaczka O, Ambroz P, Zatloukalová A, Němček K, Janout V. Risk Factors for Alzheimer's Disease: An Epidemiological Study. Curr Alzheimer Res 2021; 18:372-379. [PMID: 34420505 DOI: 10.2174/1567205018666210820124135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/04/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Dementia becomes a major public health challenge in both the Czech Republic and worldwide. The most common form of dementia is Alzheimer's disease (AD). OBJECTIVE We conducted two successive epidemiological projects in 2012-2015 and 2016-2019. Their aim was to study the effect of selected potential genetic, vascular and psychosocial risk factors on the development of AD by comparing their frequencies in AD patients and controls. METHODS Epidemiological case-control studies were conducted. In total, data from 2106 participants (1096 cases, 1010 controls) were analyzed. RESULTS Three times more females than males suffered from AD. The highest proportion of cases were those with primary education, unlike controls. There were statistically significantly more manual workers among cases than among controls. Of selected vascular risk factors, coronary heart disease was found to be statistically significantly more frequent in cases than in controls. The onset of hypertension and diabetes mellitus was earlier in controls than in cases. As for hobbies and interests, there were statistically significant differences in physical activity, reading and solving crosswords between the groups, with these activities being more common in controls. CONCLUSION The prevalence of chronic neurodegenerative diseases, in particular AD, is currently increasing. Given the aging of the population, these conditions may be expected to rise in prevalence. Potential risk of AD needs to be studied, analyzed and confirmed; a detailed knowledge of the risks of AD and early detection of the pathology may therefore be very beneficial for prevention and early treatment of this condition.
Collapse
Affiliation(s)
- Jana Janoutová
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Martina Kovalová
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Ondřej Machaczka
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Petr Ambroz
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Anna Zatloukalová
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Kateřina Němček
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Vladimír Janout
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
35
|
Effects of Functional Fitness Enhancement through Taekwondo Training on Physical Characteristics and Risk Factors of Dementia in Elderly Women with Depression. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18157961. [PMID: 34360254 PMCID: PMC8345697 DOI: 10.3390/ijerph18157961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/24/2022]
Abstract
The purpose of this research is to identify the correlations between functional fitness enhancement through a long-term Taekwondo training program and the physical characteristics and risk factors of dementia among elderly women with depression. The study has found that conducting three 60-min Taekwondo training sessions a week for the duration of 12 weeks has enhanced a number of functional fitness indexes, including hand grip strength/weight (p < 0.01), 4-m gait speed (p < 0.001), 3-m timed up and go (p < 0.05), and figure-of-eight track (p < 0.05), and significantly improved general health condition indexes as well, including percent fat (p < 0.05), appendicular skeletal muscle mass index (p < 0.01), systolic blood pressure (p < 0.01), and diastolic blood pressure (p < 0.001). Furthermore, the arteriosclerosis index and cognitive function have been found to be improved with an increase of brain-derived neurotrophic factor (BDNF; which prevents dementia) and a significant decrease of β-amyloid—a risk factor of dementia—as a result of enhancements in serum lipids and adiponectin, confirming the positive effects of functional fitness enhancement on fighting depression, promoting physical characteristics, and reducing the risk factors of dementia.
Collapse
|
36
|
Baek JY, Lee E, Kim WJ, Jang IY, Jung HW. A Cumulative Muscle Index and Its Parameters for Predicting Future Cognitive Decline: Longitudinal Outcomes of the ASPRA Cohort. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18147350. [PMID: 34299801 PMCID: PMC8303449 DOI: 10.3390/ijerph18147350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/28/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022]
Abstract
Sarcopenia and cognitive decline share the major risk factors of physical inactivity; previous studies have shown inconsistent associations. We aimed to identify the association of sarcopenia and its parameters with cognitive decline. The 3-year longitudinal outcomes of 1327 participants from the Aging Study of the Pyeongchang Rural Area (ASPRA) cohort were analyzed. Cognitive performance was evaluated using the Mini-Mental State Examination (MMSE), and sarcopenia was defined by the following: the original and revised Asian Working Group for Sarcopenia (AWGS), the original and revised European Working Group on Sarcopenia in Older People (EWGSOP), and the Cumulative Muscle Index (CMI), a novel index based on the number of impaired domains of sarcopenia. Approximately half of the participants showed meaningful cognitive decline. Sarcopenia by the original EWGSOP and the CMI were associated with cognitive decline. Only the CMI showed consistent predictability for cognitive impairment even with different criteria of the MMSE score (OR 1.23 [1.04-1.46]; OR 1.34 [1.12-1.59]; OR 1.22 [1.01-1.49], using the 1, 2, and 3 cut-off value, respectively). Of the CMI parameters, gait speed was satisfactorily predictive of 3-year cognitive impairment (OR 0.54 [0.30-0.97]). In conclusion, sarcopenia based on the CMI may be predictive of future cognitive impairment. Gait speed was the single most important indicator of cognitive decline.
Collapse
Affiliation(s)
- Ji-Yeon Baek
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.-Y.B.); (E.L.)
| | - Eunju Lee
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.-Y.B.); (E.L.)
| | - Woo Jung Kim
- Department of Psychiatry, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Korea;
| | - Il-Young Jang
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.-Y.B.); (E.L.)
- Correspondence: (I.-Y.J.); (H.-W.J.); Tel.: +82-2-3010-1658 (I.-Y.J.); +82-2-3010-1852 (H.-W.J.)
| | - Hee-Won Jung
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.-Y.B.); (E.L.)
- Correspondence: (I.-Y.J.); (H.-W.J.); Tel.: +82-2-3010-1658 (I.-Y.J.); +82-2-3010-1852 (H.-W.J.)
| |
Collapse
|
37
|
Lisko I, Kulmala J, Annetorp M, Ngandu T, Mangialasche F, Kivipelto M. How can dementia and disability be prevented in older adults: where are we today and where are we going? J Intern Med 2021; 289:807-830. [PMID: 33314384 PMCID: PMC8248434 DOI: 10.1111/joim.13227] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ageing of the population, together with population growth, has brought along an ample increase in the number of older individuals living with dementia and disabilities. Dementia is the main cause of disability in old age, and promoting healthy brain ageing is considered as a key element in diminishing the burden of age-related disabilities. The World Health Organization recently launched the first risk reduction guidelines for cognitive impairment and dementia. According to recent estimates, approximately 40% of dementia cases worldwide could be attributable to 12 modifiable risk factors: low education; midlife hypertension and obesity; diabetes, smoking, excessive alcohol use, physical inactivity, depression, low social contact, hearing loss, traumatic brain injury and air pollution indicating clear prevention potential. Dementia and physical disability are closely linked with shared risk factors and possible shared underlying mechanisms supporting the possibility of integrated preventive interventions. FINGER trial was the first large randomized controlled trial indicating that multidomain lifestyle-based intervention can prevent cognitive and functional decline amongst at-risk older adults from the general population. Within the World-Wide FINGERS network, the multidomain FINGER concept is now tested and adapted worldwide proving evidence and tools for effective and easily implementable preventive strategies. Close collaboration between researchers, policymakers and healthcare practitioners, involvement of older adults and utilization of new technologies to support self-management is needed to facilitate the implementation of the research findings. In this scoping review, we present the current scientific evidence in the field of dementia and disability prevention and discuss future directions in the field.
Collapse
Affiliation(s)
- I. Lisko
- From theDivision of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
- Faculty of Sport and Health Sciences and Gerontology Research CenterUniversity of JyväskyläJyväskyläFinland
| | - J. Kulmala
- From theDivision of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
- Public Health Promotion UnitFinnish Institute for Health and WelfareHelsinkiFinland
- School of Health Care and Social WorkSeinäjoki University of Applied SciencesSeinäjokiFinland
| | - M. Annetorp
- Karolinska University Hospital, Theme AgingStockholmSweden
| | - T. Ngandu
- From theDivision of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
- Public Health Promotion UnitFinnish Institute for Health and WelfareHelsinkiFinland
| | - F. Mangialasche
- From theDivision of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
- Aging Research CenterDepartment of Neurobiology, Care Sciences and SocietyKarolinska Institutet and Stockholm UniversityStockholmSweden
| | - M. Kivipelto
- From theDivision of Clinical GeriatricsCenter for Alzheimer ResearchDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetStockholmSweden
- Karolinska University Hospital, Theme AgingStockholmSweden
- Institute of Public Health and Clinical NutritionUniversity of Eastern FinlandHelsinkiFinland
- Ageing and Epidemiology (AGE) Research UnitSchool of Public HealthImperial College LondonLondonUK
| |
Collapse
|
38
|
Wang H, Lo MT, Rosenthal SB, Makowski C, Andreassen OA, Salem RM, McEvoy LK, Fiecas M, Chen CH. Similar Genetic Architecture of Alzheimer's Disease and Differential APOE Effect Between Sexes. Front Aging Neurosci 2021; 13:674318. [PMID: 34122051 PMCID: PMC8194397 DOI: 10.3389/fnagi.2021.674318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/27/2021] [Indexed: 01/09/2023] Open
Abstract
Sex differences have been observed in the clinical manifestations of Alzheimer’s disease (AD) and elucidating their genetic basis is an active research topic. Based on autosomal genotype data of 7,216 men and 10,680 women, including 8,136 AD cases and 9,760 controls, we explored sex-related genetic heterogeneity in AD by investigating SNP heritability, genetic correlation, as well as SNP- and gene-based genome-wide analyses. We found similar SNP heritability (men: 19.5%; women: 21.5%) and high genetic correlation (Rg = 0.96) between the sexes. The heritability of APOE ε4-related risks for AD, after accounting for effects of all SNPs excluding chromosome 19, was nominally, but not significantly, higher in women (10.6%) than men (9.7%). In age-stratified analyses, ε3/ε4 was associated with a higher risk of AD among women than men aged 65–75 years, but not in the full sample. Apart from APOE, no new significant locus was identified in sex-stratified gene-based analyses. Our result of the high genetic correlation indicates overall similar genetic architecture of AD in both sexes at the genome-wide averaged level. Our study suggests that clinically observed sex differences may arise from sex-specific variants with small effects or more complicated mechanisms involving epigenetic alterations, sex chromosomes, or gene-environment interactions.
Collapse
Affiliation(s)
- Hao Wang
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, San Diego, CA, United States
| | - Min-Tzu Lo
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, San Diego, CA, United States
| | - Sara Brin Rosenthal
- Center for Computational Biology and Bioinformatics, University of California, San Diego, San Diego, CA, United States
| | - Carolina Makowski
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, San Diego, CA, United States
| | - Ole A Andreassen
- Division of Mental Health and Addiction, NORMENT Centre, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Rany M Salem
- Department of Family Medicine and Public Health, Division of Epidemiology, University of California, San Diego, San Diego, CA, United States
| | - Linda K McEvoy
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, San Diego, CA, United States.,Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, San Diego, CA, United States
| | - Mark Fiecas
- School of Public Health, Division of Biostatistics, University of Minnesota, Minneapolis, MN, United States
| | - Chi-Hua Chen
- Department of Radiology, Center for Multimodal Imaging and Genetics, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
39
|
Xia X, Wang Y, Zheng J. COVID-19 and Alzheimer's disease: how one crisis worsens the other. Transl Neurodegener 2021; 10:15. [PMID: 33941272 PMCID: PMC8090526 DOI: 10.1186/s40035-021-00237-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/05/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) has emerged as a key comorbidity of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The morbidity and mortality of COVID-19 are elevated in AD due to multiple pathological changes in AD patients such as the excessive expression of viral receptor angiotensin converting enzyme 2 and pro-inflammatory molecules, various AD complications including diabetes, lifestyle alterations in AD, and drug-drug interactions. Meanwhile, COVID-19 has also been reported to cause various neurologic symptoms including cognitive impairment that may ultimately result in AD, probably through the invasion of SARS-CoV-2 into the central nervous system, COVID-19-induced inflammation, long-term hospitalization and delirium, and post-COVID-19 syndrome. In addition, the COVID-19 crisis also worsens behavioral symptoms in uninfected AD patients and poses new challenges for AD prevention. In this review, we first introduce the symptoms and pathogenesis of COVID-19 and AD. Next, we provide a comprehensive discussion on the aggravating effects of AD on COVID-19 and the underlying mechanisms from molecular to social levels. We also highlight the influence of COVID-19 on cognitive function, and propose possible routes of viral invasion into the brain and potential mechanisms underlying the COVID-19-induced cognitive impairment. Last, we summarize the negative impacts of COVID-19 pandemic on uninfected AD patients and dementia prevention.
Collapse
Affiliation(s)
- Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Tenth People's Hospital of Tongji University, Shanghai, 200072, China.
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200434, China.
| | - Yi Wang
- Center for Translational Neurodegeneration and Regenerative Therapy, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200434, China
| | - Jialin Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Tenth People's Hospital of Tongji University, Shanghai, 200072, China.
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200434, China.
- Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092, China.
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5930, USA.
| |
Collapse
|
40
|
Cancela-Carral JM, López-Rodríguez A, Mollinedo-Cardalda I. Effect of physical exercise on cognitive function in older adults' carriers versus noncarriers of apolipoprotein E4: systematic review and meta-analysis. J Exerc Rehabil 2021; 17:69-80. [PMID: 34012932 PMCID: PMC8103191 DOI: 10.12965/jer.2142130.065] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/13/2021] [Indexed: 12/15/2022] Open
Abstract
The presence of apolipoprotein (Apo) E4 is a genetic risk factor in cognitive impairment. Physical exercise contributes to slowing cognitive impairment in older adults, but little is known about the influence of exercise on ApoE4 carriers and noncarriers. The objective of systematic review is to study the role of physical exercise in older adults' ApoE4 carriers and noncarriers. A systematic literature search was carried out in five international databases: PubMed, Web of Science, PeDro, Scopus, and SPORTDiscus. A total of nine randomized controlled trials were included with a sample size of 2,025 subjects (901 ApoE4 carriers). The exercise reported a significant improvement on cognitive performance in older adults' ApoE4 noncarriers (standardized mean difference [SMD]=0.653; 95% confidence interval [CI], 0.29-1.00; chi2=35.36; degrees of freedom [df ]=7; P<0.0001; l 2=80%). It was also reported that a total program duration greater than 50 sessions generated different and significant effects on cognitive performance in older adults' ApoE4 noncarriers (SMD=0.878; 95% CI, 0.14-1.61; chi2=31.82; df=3; P<0.0001; l 2=91%). The results reported that high intensity generated a differential effect on cognitive performance in older adults' ApoE4 carriers versus noncarriers (SMD=0.963; 95% CI, 0.25-1.67; chi2=18.11; df=3; P<0.0004; l 2=83%). The effect of physical exercise on cognitive performance in older adults is conditioned by the presence or not of ApoE4.
Collapse
Affiliation(s)
- José María Cancela-Carral
- HealthyFit Research Group, University of Vigo, Pontevedra, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur), Sergas-UVIGO, Pontevedra, Spain
- Faculty of Education and Sports Science, University of Vigo, Pontevedra, Spain
| | - Adriana López-Rodríguez
- Galicia Sur Health Research Institute (IIS Galicia Sur), Sergas-UVIGO, Pontevedra, Spain
- Faculty of Education and Sports Science, University of Vigo, Pontevedra, Spain
| | - Irimia Mollinedo-Cardalda
- Galicia Sur Health Research Institute (IIS Galicia Sur), Sergas-UVIGO, Pontevedra, Spain
- Faculty of Physiotherapy, University of Vigo, Pontevedra, Spain
| |
Collapse
|
41
|
López-Ortiz S, Pinto-Fraga J, Valenzuela PL, Martín-Hernández J, Seisdedos MM, García-López O, Toschi N, Di Giuliano F, Garaci F, Mercuri NB, Nisticò R, Emanuele E, Lista S, Lucia A, Santos-Lozano A. Physical Exercise and Alzheimer's Disease: Effects on Pathophysiological Molecular Pathways of the Disease. Int J Mol Sci 2021; 22:ijms22062897. [PMID: 33809300 PMCID: PMC7999827 DOI: 10.3390/ijms22062897] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD), the most common form of neurodegenerative dementia in adults worldwide, is a multifactorial and heterogeneous disorder characterized by the interaction of genetic and epigenetic factors and the dysregulation of numerous intracellular signaling and cellular/molecular pathways. The introduction of the systems biology framework is revolutionizing the study of complex diseases by allowing the identification and integration of cellular/molecular pathways and networks of interaction. Here, we reviewed the relationship between physical activity and the next pathophysiological processes involved in the risk of developing AD, based on some crucial molecular pathways and biological process dysregulated in AD: (1) Immune system and inflammation; (2) Endothelial function and cerebrovascular insufficiency; (3) Apoptosis and cell death; (4) Intercellular communication; (5) Metabolism, oxidative stress and neurotoxicity; (6) DNA damage and repair; (7) Cytoskeleton and membrane proteins; (8) Synaptic plasticity. Moreover, we highlighted the increasingly relevant role played by advanced neuroimaging technologies, including structural/functional magnetic resonance imaging, diffusion tensor imaging, and arterial spin labelling, in exploring the link between AD and physical exercise. Regular physical exercise seems to have a protective effect against AD by inhibiting different pathophysiological molecular pathways implicated in AD.
Collapse
Affiliation(s)
- Susana López-Ortiz
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - Jose Pinto-Fraga
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - Pedro L. Valenzuela
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
| | - Juan Martín-Hernández
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - María M. Seisdedos
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - Oscar García-López
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (N.T.); (F.G.)
- Department of Radiology, “Athinoula A. Martinos” Center for Biomedical Imaging, Boston, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Francesca Di Giuliano
- Neuroradiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (N.T.); (F.G.)
- Casa di Cura “San Raffaele Cassino”, 03043 Cassino, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy;
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Robert Nisticò
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, 00161 Rome, Italy;
- School of Pharmacy, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | | | - Simone Lista
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
- School of Pharmacy, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
- Research Institute of the Hospital 12 de Octubre (“imas12”), 28041 Madrid, Spain
- Centro de Investigación Biomeédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), 28029 Madrid, Spain
- Correspondence:
| | - Alejandro Santos-Lozano
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
- Research Institute of the Hospital 12 de Octubre (“imas12”), 28041 Madrid, Spain
| |
Collapse
|
42
|
Abstract
A three-part mechanism is proposed for the induction of Alzheimer’s disease: (1) decreased blood lactic acid; (2) increased blood ceramide and adipokines; (3) decreased blood folic acid. The age-related nature of these mechanisms comes from age-associated decreased muscle mass, increased visceral fat and changes in diet. This mechanism also explains why many people do not develop Alzheimer’s disease. Simple changes in lifestyle and diet can prevent Alzheimer’s disease. Alzheimer’s disease is caused by a cascade of events that culminates in damage to the blood–brain barrier and damage to neurons. The blood–brain barrier keeps toxic molecules out of the brain and retains essential molecules in the brain. Lactic acid is a nutrient to the brain and is produced by exercise. Damage to endothelial cells and pericytes by inadequate lactic acid leads to blood–brain barrier damage and brain damage. Inadequate folate intake and oxidative stress induced by activation of transient receptor potential cation channels and endothelial nitric oxide synthase damage the blood–brain barrier. NAD depletion due to inadequate intake of nicotinamide and alterations in the kynurenine pathway damages neurons. Changes in microRNA levels may be the terminal events that cause neuronal death leading to Alzheimer’s disease. A new mechanism of Alzheimer’s disease induction is presented involving lactic acid, ceramide, IL-1β, tumor necrosis factor α, folate, nicotinamide, kynurenine metabolites and microRNA.
Collapse
|
43
|
Greenspace Inversely Associated with the Risk of Alzheimer's Disease in the Mid-Atlantic United States. ACTA ACUST UNITED AC 2021; 2:140-150. [PMID: 34322679 PMCID: PMC8312690 DOI: 10.3390/earth2010009] [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] [Indexed: 12/21/2022]
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disease and the most common form of dementia in older adults. Treatment of AD symptoms is very challenging and expensive. Appropriate diet as well as mental and physical activity may delay or reduce the occurrence of AD. It is unknown whether environmental factors offer potentially protective effects against the development of AD. We explored the possible beneficial effects of greenspace (trees and herbaceous cover) on the rate of AD in the mid-Atlantic US. Data for initial AD medical claims during 2011–2013 were obtained from Medicare records for 2999 ZIP codes. The percentages of land cover classes in each ZIP code were calculated based on high-resolution land cover imagery. Associations between AD and greenspace, blue space (water), and other variables were examined using zero-inflated Poisson models. The rate of AD was negatively associated with greenspace (for a greenspace increase of 10%, risk ratio (RR) = 0.91, 95% confidence interval (CI): 0.89–0.94), and blue space (for a water area increase of 10%, RR = 0.85, 95% CI: 0.81–0.89). The inverse relationships between greenspace and the risk of AD held across season, gender, and race. The rate of AD was positively associated with the concentration of fine particulate matter (PM2.5) (RR = 1.03, 95% CI: 1.02–1.05 for an increase in PM2.5 of 1 μg/m3). Our results suggest that greenspace may have protective effects for AD, although potential mechanisms are unclear and require further investigation.
Collapse
|
44
|
Leblhuber F, Ehrlich D, Steiner K, Geisler S, Fuchs D, Lanser L, Kurz K. The Immunopathogenesis of Alzheimer's Disease Is Related to the Composition of Gut Microbiota. Nutrients 2021; 13:361. [PMID: 33504065 PMCID: PMC7912578 DOI: 10.3390/nu13020361] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
The microbiota-gut-brain axis plays an important role in the development of neurodegenerative diseases. Commensal and pathogenic enteric bacteria can influence brain and immune system function by the production of lipopolysaccharides and amyloid. Dysbiosis of the intestinal microbiome induces local and consecutively systemic immune-mediated inflammation. Proinflammatory cytokines then trigger neuroinflammation and finally neurodegeneration. Immune-mediated oxidative stress can lead to a deficiency of vitamins and essential micronutrients. Furthermore, the wrong composition of gut microbiota might impair the intake and metabolization of nutrients. In patients with Alzheimer's disease (AD) significant alterations of the gut microbiota have been demonstrated. Standard Western diet, infections, decreased physical activity and chronic stress impact the composition and diversity of gut microbiota. A higher abundancy of "pro-inflammatory" gut microbiota goes along with enhanced systemic inflammation and neuroinflammatory processes. Thus, AD beginning in the gut is closely related to the imbalance of gut microbiota. Modulation of gut microbiota by Mediterranean diet, probiotics and curcumin can slow down cognitive decline and alter the gut microbiome significantly. A multi-domain intervention approach addressing underlying causes of AD (inflammation, infections, metabolic alterations like insulin resistance and nutrient deficiency, stress) appears very promising to reduce or even reverse cognitive decline by exerting positive effects on the gut microbiota.
Collapse
Affiliation(s)
- Friedrich Leblhuber
- Department of Gerontology, Neuromed Campus, Kepler University Clinic, Linz A-4020, Austria; (F.L.); (D.E.); (K.S.)
| | - Daniela Ehrlich
- Department of Gerontology, Neuromed Campus, Kepler University Clinic, Linz A-4020, Austria; (F.L.); (D.E.); (K.S.)
| | - Kostja Steiner
- Department of Gerontology, Neuromed Campus, Kepler University Clinic, Linz A-4020, Austria; (F.L.); (D.E.); (K.S.)
| | - Simon Geisler
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck A-6020, Austria; (S.G.); (D.F.)
| | - Dietmar Fuchs
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck A-6020, Austria; (S.G.); (D.F.)
| | - Lukas Lanser
- Department of Internal Medicine, Medical University of Innsbruck, Innsbruck A-6020, Austria;
| | - Katharina Kurz
- Department of Internal Medicine, Medical University of Innsbruck, Innsbruck A-6020, Austria;
| |
Collapse
|
45
|
Grazioli E, Nigro E, Cerulli C, Borriello G, Mancini A, Tranchita E, Polito R, Parisi A, Buono P, Daniele A. Case Report: Concurrent Resistance and Aerobic Training Regulate Adiponectin Expression and Disease Severity in Multiple Sclerosis: A Case Study. Front Neurosci 2021; 14:567302. [PMID: 33414700 PMCID: PMC7783198 DOI: 10.3389/fnins.2020.567302] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Abstract
Adapted exercise is an effective non-pharmacological tool to improve functional, cognitive, and psychological parameters in multiple sclerosis (MS), in association with increased quality of life (QoL) and decreased disease severity. Adipose tissue, through the production of different adipokines, is involved in regulating energy metabolism and inflammation. Adiponectin, increased in MS, circulates as oligomers of low (LMW), medium (MMW), and high molecular weight (HMW), the latter mediating the main biological effects. The aim of study was to evaluate the effects of 4 months training at moderate intensity [65% heart rate reserve (HRR)] on BMI, adiponectin, and QoL in a volunteer with secondary progressive MS. The parameters were evaluated before (T0), after 4 months training (T1), and 6 months after the end of training (T2); total serum adiponectin and its oligomeric profile were evaluated. We found a reduction in BMI (−0.9%) and FAT (−2.6%), an improvement in perceived QoL and a reduced expression of total adiponectin and HMW oligomers together with decreased MS disability level at T1 measured by EDSS. Despite the limitations of a case study, this represent a starting point to understand the influence of exercise in MS and the relationship with adiponectin expression.
Collapse
Affiliation(s)
- Elisa Grazioli
- Department of Exercise, Human and Health Sciences, Foro Italico University of Rome, Rome, Italy
| | - Ersilia Nigro
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy.,CEINGE Advanced Biotechnologies, Naples, Italy
| | - Claudia Cerulli
- Department of Exercise, Human and Health Sciences, Foro Italico University of Rome, Rome, Italy
| | - Giovanna Borriello
- Department of Neurological Sciences, Sapienza University of Rome, Rome, Italy
| | - Annamaria Mancini
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Eliana Tranchita
- Department of Exercise, Human and Health Sciences, Foro Italico University of Rome, Rome, Italy
| | - Rita Polito
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Attilio Parisi
- Department of Exercise, Human and Health Sciences, Foro Italico University of Rome, Rome, Italy
| | - Pasqualina Buono
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Aurora Daniele
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Caserta, Italy.,CEINGE Advanced Biotechnologies, Naples, Italy
| |
Collapse
|
46
|
Effects of 6-Month Multimodal Physical Exercise Program on Bone Mineral Density, Fall Risk, Balance, and Gait in Patients with Alzheimer's Disease: A Controlled Clinical Trial. Brain Sci 2021; 11:brainsci11010063. [PMID: 33419016 PMCID: PMC7825330 DOI: 10.3390/brainsci11010063] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
We aimed to determine the short- and medium-term effects of a multimodal physical exercise program (MPEP) on bone health status, fall risk, balance, and gait in patients with Alzheimer’s disease. A single-blinded, controlled clinical trial was performed where 72 subjects were allocated in a 3:1 ratio to an intervention group (IG; n = 53) and control group (CG; n = 19), where the IG’s subjects were admitted to live in a State Reference Center of Alzheimer’s disease, which offers the targeted exercise program, while the CG’s subjects resided in independent living. A multidisciplinary health team assessed all patients before allocation, and dependent outcomes were again assessed at one, three, and six months. During the study, falls were recorded, and in all evaluations, bone mineral density was measured using a calcaneal quantitative ultrasound densitometer; balance and gait were measured using the performance-oriented mobility assessment (POMA), the timed up and go test (TUG), the one-leg balance test (OLB), and the functional reach test (FR). There were no differences between groups at baseline for all outcome measures. The prevalence of falls was significantly lower in the IG (15.09%) than in the CG (42.11%) (χ2 = 5.904; p = 0.015). We also found that there was a significant time*group interaction, with a post hoc Šidák test finding significant differences of improved physical function, especially in gait, for the IG, as assessed by POMA-Total, POMA-Gait, and TUG with a large effect size (ƞ2p = 0.185–0.201). In balance, we found significant differences between groups, regardless of time, and a medium effect size as assessed by POMA-Balance and the OLB (ƞ2p = 0.091–0.104). Clinically relevant effects were observed, although without significant differences in bone health, with a slowing of bone loss. These results show that a multimodal physical exercise program reduces fall risk and produces an improvement in gait, balance, and bone mineral density in the short and medium term in institutionalized patients with Alzheimer’s disease.
Collapse
|
47
|
Abstract
Brain insulin signaling contributes to memory function and might be a viable target in the prevention and treatment of memory impairments including Alzheimer's disease. This short narrative review explores the potential of central nervous system (CNS) insulin administration via the intranasal pathway to improve memory performance in health and disease, with a focus on the most recent results. Proof-of-concept studies and (pilot) clinical trials in individuals with mild cognitive impairment or Alzheimer's disease indicate that acute and prolonged intranasal insulin administration enhances memory performance, and suggest that brain insulin resistance is a pathophysiological factor in Alzheimer's disease with or without concomitant metabolic dysfunction. Intranasally administered insulin is assumed to trigger improvements in synaptic plasticity and regional glucose uptake as well as alleviations of Alzheimer's disease neuropathology; additional contributions of changes in hypothalamus-pituitary-adrenocortical axis activity and sleep-related mechanisms are discussed. While intranasal insulin delivery has been conclusively demonstrated to be effective and safe, the recent outcomes of large-scale clinical studies underline the need for further investigations, which might also yield new insights into sex differences in the response to intranasal insulin and contribute to the optimization of delivery devices to grasp the full potential of intranasal insulin for Alzheimer's disease.
Collapse
Affiliation(s)
- Manfred Hallschmid
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Otfried-Müller-Str. 25, 72076, Tübingen, Germany.
- German Center for Diabetes Research (DZD), Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.
| |
Collapse
|
48
|
Kim YJ, Han KD, Baek MS, Cho H, Lee EJ, Lyoo CH. Association between physical activity and conversion from mild cognitive impairment to dementia. ALZHEIMERS RESEARCH & THERAPY 2020; 12:136. [PMID: 33176851 PMCID: PMC7661208 DOI: 10.1186/s13195-020-00707-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 10/15/2020] [Indexed: 11/10/2022]
Abstract
Background Physical activity has been suggested to prevent the conversion of mild cognitive impairment (MCI) to dementia in patients. We investigated the association between the continuance and regularity of physical activity and the risk of developing dementia in patients with MCI. Methods We analyzed 6-year followed up data for 247,149 individuals in the National Health Insurance Service (NHIS) cohort of Korea who were enrolled between January 1, 2009, and December 31, 2015. The patients were divided into four groups: those who did not engage in physical activity consistently (Never-PA group), those who initiated physical activity (Initiation-PA group), those who ceased physical activity (Withdrawal-PA group), and those who performed physical activity consistently (Maintenance-PA group). We also divided the patients into two groups: those who engaged in physical activity irregularly (Irregular-PA) and those who undertook physical activity regularly (Regular-PA). Results When the risk for the Never-PA group was set as the benchmark (ref = 1), the Maintenance-PA group had the lowest incidence of dementia of the Alzheimer type (DAT) compared to the other groups (HR = 0.82, 95% CI 0.79–0.86). The DAT risk of the Initiation-PA group (HR = 0.89, 95% CI 0.85–0.93) was lower than the Never-PA group. In addition, compared to the Irregular-PA group, the Regular-PA group had a 15% reduced risk for developing DAT. Conclusions Although no causal inference could be made, continued regular physical activity in patients with MCI is associated with a protective effect against developing DAT. Moreover, ceasing physical activity could halt this protective effect. Supplementary information Supplementary information accompanies this paper at 10.1186/s13195-020-00707-1.
Collapse
Affiliation(s)
- Yeo Jin Kim
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Kyung-Do Han
- Department of Biostatics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Seok Baek
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211, Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211, Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea.
| | - Eun Joo Lee
- Big Data Steering Department, National Health Insurance Service, Wonju, Republic of Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211, Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea
| |
Collapse
|
49
|
Baumeister SE, Karch A, Bahls M, Teumer A, Leitzmann MF, Baurecht H. Physical activity and risk of Alzheimer disease: A 2-sample mendelian randomization study. Neurology 2020; 95:e1897-e1905. [PMID: 32680943 PMCID: PMC7963349 DOI: 10.1212/wnl.0000000000010013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 04/10/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Evidence from observational studies for the effect of physical activity on the risk of Alzheimer disease (AD) is inconclusive. We performed a 2-sample mendelian randomization analysis to examine whether physical activity is protective for AD. METHODS Summary data of genome-wide association studies on physical activity and AD were used. The primary study population included 21,982 patients with AD and 41,944 cognitively normal controls. Eight single nucleotide polymorphisms (SNPs) known at p < 5 × 10-8 to be associated with average accelerations and 8 SNPs associated at p < 5 × 10-7 with vigorous physical activity (fraction of accelerations >425 milligravities) served as instrumental variables. RESULTS There was no association between genetically predicted average accelerations with the risk of AD (inverse variance weighted odds ratio [OR] per SD increment: 1.03, 95% confidence interval 0.97-1.10, p = 0.332). Genetic liability for fraction of accelerations >425 milligravities was unrelated to AD risk. CONCLUSION The present study does not support a causal association between physical activity and risk of AD.
Collapse
Affiliation(s)
- Sebastian E Baumeister
- From Epidemiology (S.E.B.), LMU München, UNIKA-T Augsburg; Independent Research Group, Clinical Epidemiology (S.E.B.), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich; Institute of Epidemiology and Social Medicine (A.K.), University of Münster; Department of Internal Medicine B (M.B.) and Institute for Community Medicine (A.T.), University Medicine Greifswald; DZHK (German Centre for Cardiovascular Research) (M.B., A.T.), Partner Site Greifswald; and Department of Epidemiology and Preventive Medicine (M.F.L., H.B.), University of Regensburg, Germany.
| | - André Karch
- From Epidemiology (S.E.B.), LMU München, UNIKA-T Augsburg; Independent Research Group, Clinical Epidemiology (S.E.B.), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich; Institute of Epidemiology and Social Medicine (A.K.), University of Münster; Department of Internal Medicine B (M.B.) and Institute for Community Medicine (A.T.), University Medicine Greifswald; DZHK (German Centre for Cardiovascular Research) (M.B., A.T.), Partner Site Greifswald; and Department of Epidemiology and Preventive Medicine (M.F.L., H.B.), University of Regensburg, Germany
| | - Martin Bahls
- From Epidemiology (S.E.B.), LMU München, UNIKA-T Augsburg; Independent Research Group, Clinical Epidemiology (S.E.B.), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich; Institute of Epidemiology and Social Medicine (A.K.), University of Münster; Department of Internal Medicine B (M.B.) and Institute for Community Medicine (A.T.), University Medicine Greifswald; DZHK (German Centre for Cardiovascular Research) (M.B., A.T.), Partner Site Greifswald; and Department of Epidemiology and Preventive Medicine (M.F.L., H.B.), University of Regensburg, Germany
| | - Alexander Teumer
- From Epidemiology (S.E.B.), LMU München, UNIKA-T Augsburg; Independent Research Group, Clinical Epidemiology (S.E.B.), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich; Institute of Epidemiology and Social Medicine (A.K.), University of Münster; Department of Internal Medicine B (M.B.) and Institute for Community Medicine (A.T.), University Medicine Greifswald; DZHK (German Centre for Cardiovascular Research) (M.B., A.T.), Partner Site Greifswald; and Department of Epidemiology and Preventive Medicine (M.F.L., H.B.), University of Regensburg, Germany
| | - Michael F Leitzmann
- From Epidemiology (S.E.B.), LMU München, UNIKA-T Augsburg; Independent Research Group, Clinical Epidemiology (S.E.B.), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich; Institute of Epidemiology and Social Medicine (A.K.), University of Münster; Department of Internal Medicine B (M.B.) and Institute for Community Medicine (A.T.), University Medicine Greifswald; DZHK (German Centre for Cardiovascular Research) (M.B., A.T.), Partner Site Greifswald; and Department of Epidemiology and Preventive Medicine (M.F.L., H.B.), University of Regensburg, Germany
| | - Hansjörg Baurecht
- From Epidemiology (S.E.B.), LMU München, UNIKA-T Augsburg; Independent Research Group, Clinical Epidemiology (S.E.B.), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich; Institute of Epidemiology and Social Medicine (A.K.), University of Münster; Department of Internal Medicine B (M.B.) and Institute for Community Medicine (A.T.), University Medicine Greifswald; DZHK (German Centre for Cardiovascular Research) (M.B., A.T.), Partner Site Greifswald; and Department of Epidemiology and Preventive Medicine (M.F.L., H.B.), University of Regensburg, Germany
| |
Collapse
|
50
|
Butterfield DA, Boyd-Kimball D. Mitochondrial Oxidative and Nitrosative Stress and Alzheimer Disease. Antioxidants (Basel) 2020; 9:E818. [PMID: 32887505 PMCID: PMC7554713 DOI: 10.3390/antiox9090818] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/22/2020] [Accepted: 08/29/2020] [Indexed: 12/29/2022] Open
Abstract
Oxidative and nitrosative stress are widely recognized as critical factors in the pathogenesis and progression of Alzheimer disease (AD) and its earlier stage, amnestic mild cognitive impairment (MCI). A major source of free radicals that lead to oxidative and nitrosative damage is mitochondria. This review paper discusses oxidative and nitrosative stress and markers thereof in the brain, along with redox proteomics, which are techniques that have been pioneered in the Butterfield laboratory. Selected biological alterations in-and oxidative and nitrosative modifications of-mitochondria in AD and MCI and systems of relevance thereof also are presented. The review article concludes with a section on the implications of mitochondrial oxidative and nitrosative stress in MCI and AD with respect to imaging studies in and targeted therapies toward these disorders. Taken together, this review provides support for the notion that brain mitochondrial alterations in AD and MCI are key components of oxidative and nitrosative stress observed in these two disorders, and as such, they provide potentially promising therapeutic targets to slow-and hopefully one day stop-the progression of AD, which is a devastating dementing disorder.
Collapse
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Debra Boyd-Kimball
- Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH 44601, USA;
| |
Collapse
|