1
|
Wen X, Hu J. Targeting STAT3 signaling pathway in the treatment of Alzheimer's disease with compounds from natural products. Int Immunopharmacol 2024; 141:112936. [PMID: 39163684 DOI: 10.1016/j.intimp.2024.112936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is difficult to cure and of global concern. Neuroinflammation is closely associated with the onset and progression of AD, making its treatment increasingly important. Compounds from natural products, with fewer side effects than synthetic drugs, are of high research interest. STAT3, a multifunctional transcription factor, is involved in various cellular processes including inflammation, cell growth, and apoptosis. Its activation and inhibition can have different effects under various pathological conditions. In AD, the STAT3 protein plays a crucial role in promoting neuroinflammation and contributing to disease progression. This occurs primarily through the JAK2-STAT3 signaling pathway, which impacts microglia, astrocytes, and hippocampal neurons. This paper reviews the STAT3 signaling pathway in AD and 25 compounds targeting STAT3 up to 2024. Notably, Rutin, Paeoniflorin, and Geniposide up-regulate STAT3 in hippocampal and cortex neurons, showing neuroprotective effects in various AD models. Other 23 compounds downregulate AD by suppressing neuroinflammation through inhibition of STAT3 activation in microglia and astrocytes. These findings highlight the potential of compounds from natural products in improving AD by targeting STAT3, offering insights into the prevention and management of AD.
Collapse
Affiliation(s)
- Xiyue Wen
- Department of Clinical Laboratory, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, China
| | - Jinyue Hu
- Medical Research Center, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, China.
| |
Collapse
|
2
|
Szilágyi A, Takács B, Szekeres R, Tarjányi V, Nagy D, Priksz D, Bombicz M, Kiss R, Szabó AM, Lehoczki A, Gesztelyi R, Juhász B, Szilvássy Z, Varga B. Effects of voluntary and forced physical exercise on the retinal health of aging Wistar rats. GeroScience 2024; 46:4707-4728. [PMID: 38795184 PMCID: PMC11336036 DOI: 10.1007/s11357-024-01208-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/13/2024] [Indexed: 05/27/2024] Open
Abstract
Aging is accompanied by an increased prevalence of degenerative conditions, including those affecting ocular health, which significantly impact quality of life and increase the burden on healthcare systems. Among these, retinal aging is of particular concern due to its direct link to vision impairment, a leading cause of disability in the elderly. Vision loss in the aging population is associated with heightened risks of cognitive decline, social isolation, and morbidity. This study addresses the critical gap in our understanding of modifiable lifestyle factors, such as physical exercise, that may mitigate retinal aging and its related pathologies. We investigated the effects of different exercise regimens-voluntary (recreational-type) and forced (high-intensity)-on the retinal health of aging Wistar rats (18-month-old), serving as a model for studying the translational potential of exercise interventions in humans. Male Wistar rats were divided into four groups: a young control (3-month-old) for baseline comparison, an aged sedentary control, an aged group engaging in voluntary exercise via a running wheel in their cage, and an aged group subjected to forced exercise on a treadmill for six sessions of 20 min each per week. After a 6-month experimental period, we assessed retinal function via electroretinography (ERG), measured retinal thickness histologically, and analyzed protein expression changes relevant to oxidative stress, inflammation, and anti-aging mechanisms. Our findings reveal that voluntary exercise positively impacts retinal function and morphology, reducing oxidative stress and inflammation markers while enhancing anti-aging protein expression. In contrast, forced exercise showed diminished benefits. These insights underscore the importance of exercise intensity and preference in preserving retinal health during aging. The study highlights the potential of recreational physical activity as a non-invasive strategy to counteract retinal aging, advocating for further research into exercise regimens as preventative therapies for age-related ocular degenerations.
Collapse
Affiliation(s)
- Anna Szilágyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Barbara Takács
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Réka Szekeres
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Vera Tarjányi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Dávid Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Dániel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Rita Kiss
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Adrienn Mónika Szabó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Andrea Lehoczki
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Zoltán Szilvássy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Balázs Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary.
| |
Collapse
|
3
|
Sinclair D, Canty AJ, Ziebell JM, Woodhouse A, Collins JM, Perry S, Roccati E, Kuruvilla M, Leung J, Atkinson R, Vickers JC, Cook AL, King AE. Experimental laboratory models as tools for understanding modifiable dementia risk. Alzheimers Dement 2024; 20:4260-4289. [PMID: 38687209 PMCID: PMC11180874 DOI: 10.1002/alz.13834] [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: 12/13/2023] [Revised: 02/29/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024]
Abstract
Experimental laboratory research has an important role to play in dementia prevention. Mechanisms underlying modifiable risk factors for dementia are promising targets for dementia prevention but are difficult to investigate in human populations due to technological constraints and confounds. Therefore, controlled laboratory experiments in models such as transgenic rodents, invertebrates and in vitro cultured cells are increasingly used to investigate dementia risk factors and test strategies which target them to prevent dementia. This review provides an overview of experimental research into 15 established and putative modifiable dementia risk factors: less early-life education, hearing loss, depression, social isolation, life stress, hypertension, obesity, diabetes, physical inactivity, heavy alcohol use, smoking, air pollution, anesthetic exposure, traumatic brain injury, and disordered sleep. It explores how experimental models have been, and can be, used to address questions about modifiable dementia risk and prevention that cannot readily be addressed in human studies. HIGHLIGHTS: Modifiable dementia risk factors are promising targets for dementia prevention. Interrogation of mechanisms underlying dementia risk is difficult in human populations. Studies using diverse experimental models are revealing modifiable dementia risk mechanisms. We review experimental research into 15 modifiable dementia risk factors. Laboratory science can contribute uniquely to dementia prevention.
Collapse
Affiliation(s)
- Duncan Sinclair
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Alison J. Canty
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
- Global Brain Health Institute, Trinity CollegeDublinIreland
| | - Jenna M. Ziebell
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Jessica M. Collins
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Sharn Perry
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Eddy Roccati
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Maneesh Kuruvilla
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Jacqueline Leung
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Rachel Atkinson
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - James C. Vickers
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Anthony L. Cook
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Anna E. King
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| |
Collapse
|
4
|
Rodriguez-Ayllon M, Solis-Urra P, Arroyo-Ávila C, Álvarez-Ortega M, Molina-García P, Molina-Hidalgo C, Gómez-Río M, Brown B, Erickson KI, Esteban-Cornejo I. Physical activity and amyloid beta in middle-aged and older adults: A systematic review and meta-analysis. JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 13:133-144. [PMID: 37558161 PMCID: PMC10980893 DOI: 10.1016/j.jshs.2023.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/11/2023] [Accepted: 06/30/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND One of the pathological hallmarks distinguishing Alzheimer's disease from other dementias is the accumulation of amyloid beta (Aβ). Higher physical activity is associated with decreased dementia risk, and one potential path could be through Aβ levels modulation. We aimed to explore the relationship between physical activity and Aβ in middle-aged and older adults. METHODS A systematic search of PubMed, Web of Science, PsycINFO, Cochrane Central Register of Controlled Trials, and SPORTDiscus was performed from inception to April 28, 2022. Studies were eligible if they included physical activity and Aβ data in adults aged 45 years or older. Multi-level meta-analyses of intervention and observational studies were performed to examine the role of physical activity in modulating Aβ levels. RESULTS In total, 37 articles were included (8 randomized controlled trials, 3 non-randomized controlled trials, 4 prospective longitudinal studies, and 22 cross-sectional studies). The overall effect size of physical activity interventions on changes in blood Aβ was medium (pooled standardized mean difference = -0.69, 95% confidence interval (95%CI): -1.41 to 0.03; I2 = 74.6%). However, these results were not statistically significant, and there were not enough studies to explore the effects of physical activity on cerebrospinal fluid (CSF) and brain Aβ. Data from observational studies were examined based on measurements of Aβ in the brain using positron emission tomography scans, CSF, and blood. Higher physical activity was positively associated with Aβ only in the CSF (Estimate r = 0.12; 95%CI: 0.05-0.18; I2 = 38.00%). CONCLUSION Physical activity might moderately reduce blood Aβ in middle-aged and older adults. However, results were only near statistical significance and might be interpreted with caution given the methodological limitations observed in some of the included studies. In observational studies, higher levels of physical activity were positively associated with Aβ only in CSF. Therefore, further research is needed to understand the modulating role of physical activity in the brain, CSF, and blood Aβ, as well as its implication for cognitive health.
Collapse
Affiliation(s)
- María Rodriguez-Ayllon
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, GD 3015, the Netherlands
| | - Patricio Solis-Urra
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada 18071, Spain; Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2531015, Chile; Department of Nuclear Medicine, Virgen de las Nieves University Hospital, Institute of Biosanitary Research of Granada (IBS), Granada 18014, Spain
| | - Cristina Arroyo-Ávila
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada 18071, Spain
| | - Miriam Álvarez-Ortega
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada 18071, Spain
| | - Pablo Molina-García
- Physical Medicine and Rehabilitation Service, Virgen de las Nieves University Hospital, Institute of Biosanitary Research of Granada (IBS), Granada 18014, Spain
| | | | - Manuel Gómez-Río
- Department of Nuclear Medicine, Virgen de las Nieves University Hospital, Institute of Biosanitary Research of Granada (IBS), Granada 18014, Spain
| | - Belinda Brown
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Kirk I Erickson
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada 18071, Spain; Department of Psychology, University of Pittsburgh, Pittsburgh, PA 15260, USA; Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia; Advent Health Research Institute, Neuroscience Institute Orlando, Orlando, FL 32803, USA
| | - Irene Esteban-Cornejo
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada 18071, Spain; Physiopathology of Obesity and Nutrition Research Center (CIBERobn), Institute of Health Carlos III (ISCIII), Madrid 28029, Spain.
| |
Collapse
|
5
|
Gori A, Topino E, Griffiths MD. Family Functioning Styles and Exercise Addiction: Disengaged, Enmeshed, and Rigid Family Patterns Are Associated with Exercise Addiction. Eur J Investig Health Psychol Educ 2024; 14:148-163. [PMID: 38248130 PMCID: PMC10814248 DOI: 10.3390/ejihpe14010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/20/2023] [Accepted: 12/31/2023] [Indexed: 01/23/2024] Open
Abstract
Physical exercise is a widely recommended practice for promoting health, but for some individuals, this activity can result in pathological and morbid behaviour. Therefore, the study of the factors contributing to the onset, development, and progression of exercise addiction is particularly relevant. Within this framework, the present study assessed the effect of family functioning, body image concerns, age, and gender on exercise addiction. A sample of 300 regular exercisers (Mage = 30.3 years, SD = 11.6; 69.7% females, 30.3% males) participated in the study and completed the Family Adaptability and Cohesion Evaluation Scales-IV, Body Image Concern Inventory, and Exercise Addiction Inventory. Data were analysed by implementing a series of moderated moderated-mediations. Results showed that three significant models were relevant. First, positive associations of disengaged (p < 0.05), enmeshed (p < 0.05), and rigid (p < 0.01) family functioning with exercise addiction were found. Furthermore, body image concerns mediated all these relationships, and the interaction between gender and age significantly moderated the effects of body image concerns on exercise addiction (p < 0.05). Such data may be useful for a deeper understanding of the variables associated with the development of exercise addiction, suggesting key elements on which it might be useful to focus in clinical and/or preventive activity.
Collapse
Affiliation(s)
- Alessio Gori
- Department of Health Sciences, University of Florence, Via di San Salvi 12, Pad. 26, 50135 Florence, Italy
| | - Eleonora Topino
- Department of Human Sciences, LUMSA University of Rome, Via della Traspontina 21, 00193 Rome, Italy;
| | - Mark D. Griffiths
- Psychology Department, Nottingham Trent University, 50 Shakespeare Street, Nottingham NG1 4FQ, UK;
| |
Collapse
|
6
|
Widjaya MA, Lee SD, Cheng WC, Wu BT. Effects of Exercise Training on Immune-Related Genes and Pathways in the Cortex of Animal Models of Alzheimer's Disease: A Systematic Review. J Alzheimers Dis 2024; 98:1219-1234. [PMID: 38578886 DOI: 10.3233/jad-230803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
Background Alzheimer's disease (AD) is a chronic neurodegenerative disease that affects the immune system due to the accumulation of amyloid-β (Aβ) and tau associated molecular pathology and other pathogenic processes. To address AD pathogenesis, various approaches had been conducted from drug development to lifestyle modification to reduce the prevalence of AD. Exercise is considered a prominent lifestyle modification to combat AD. Objective This observation prompted us to review the literature on exercise related to immune genes in the cortex of animal models of AD. We focused on animal model studies due to their prevalence in this domain. Methods The systematic review was conducted according to PRISMA standards using Web of Science (WoS) and PubMed databases. Any kind of genes, proteins, and molecular molecules were included in this systematic review. The list of these immune-related molecules was analyzed in the STRING database for functional enrichment analysis. Results We found that 17 research studies discussed immune-related molecules and 30 immune proteins. These studies showed that exercise had the ability to ameliorate dysfunction in AD-related pathways, which led to decreasing the expression of microglia-related pathways and Th17-related immune pathways. As a result of decreasing the expression of immune-related pathways, the expression of apoptosis-related pathways was also decreasing, and neuronal survival was increased by exercise activity. Conclusions Based on functional enrichment analysis, exercise not only could reduce apoptotic factors and immune components but also could increase cell survival and Aβ clearance in cortex samples. PROSPERO ID: CRD42022326093.
Collapse
Affiliation(s)
- Michael Anekson Widjaya
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Shin-Da Lee
- Department of Physical Therapy, PhD program in Healthcare Science, China Medical University, Taichung, Taiwan
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Bor-Tsang Wu
- Department of Senior Citizen Service Management, National Taichung University of Science and Technology, Taichung, Taiwan
| |
Collapse
|
7
|
Kimura N, Aota T, Aso Y, Yabuuchi K, Sasaki K, Masuda T, Eguchi A, Maeda Y, Aoshima K, Matsubara E. Predicting positron emission tomography brain amyloid positivity using interpretable machine learning models with wearable sensor data and lifestyle factors. Alzheimers Res Ther 2023; 15:212. [PMID: 38087316 PMCID: PMC10714506 DOI: 10.1186/s13195-023-01363-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Developing a screening method for identifying individuals at higher risk of elevated brain amyloid burden is important to reduce costs and burden to patients in clinical trials on Alzheimer's disease or the clinical setting. We developed machine learning models using objectively measured lifestyle factors to predict elevated brain amyloid burden on positron emission tomography. METHODS Our prospective cohort study of non-demented, community-dwelling older adults aged ≥ 65 years was conducted from August 2015 to September 2019 in Usuki, Oita Prefecture, Japan. One hundred and twenty-two individuals with mild cognitive impairment or subjective memory complaints (54 men and 68 women, median age: 75.50 years) wore wearable sensors and completed self-reported questionnaires, cognitive test, and positron emission tomography imaging at baseline. Moreover, 99 individuals in the second year and 61 individuals in the third year were followed up. In total, 282 eligible records with valid wearable sensors, cognitive test results, and amyloid imaging and data on demographic characteristics, living environments, and health behaviors were used in the machine learning models. Amyloid positivity was defined as a standardized uptake value ratio of ≥ 1.4. Models were constructed using kernel support vector machine, Elastic Net, and logistic regression for predicting amyloid positivity. The mean score among 10 times fivefold cross-validation repeats was utilized for evaluation. RESULTS In Elastic Net, the mean area under the receiver operating characteristic curve of the model using objectively measured lifestyle factors alone was 0.70, whereas that of the models using wearable sensors in combination with demographic characteristics and health and life environment questionnaires was 0.79. Moreover, 22 variables were common to all machine learning models. CONCLUSION Our machine learning models are useful for predicting elevated brain amyloid burden using readily-available and noninvasive variables without the need to visit a hospital. TRIAL REGISTRATION This prospective study was conducted in accordance with the Declaration of Helsinki and was approved by the local ethics committee of Oita University Hospital (UMIN000017442). A written informed consent was obtained from all participants. This research was performed based on the Strengthening the Reporting of Observational Studies in Epidemiology reporting guideline.
Collapse
Affiliation(s)
- Noriyuki Kimura
- Department of Neurology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama, Yufu, Oita, 879-5593, Japan.
| | - Tomoki Aota
- Microbes & Host Defense Domain Deep Human Biology Learning, Eisai Co., Ltd, 5-1-3, Tokodai, Tsukuba-Shi, Ibaraki, 300-2635, Japan
| | - Yasuhiro Aso
- Department of Neurology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama, Yufu, Oita, 879-5593, Japan
| | - Kenichi Yabuuchi
- Department of Neurology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama, Yufu, Oita, 879-5593, Japan
| | - Kotaro Sasaki
- Microbes & Host Defense Domain Deep Human Biology Learning, Eisai Co., Ltd, 5-1-3, Tokodai, Tsukuba-Shi, Ibaraki, 300-2635, Japan
| | - Teruaki Masuda
- Department of Neurology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama, Yufu, Oita, 879-5593, Japan
| | - Atsuko Eguchi
- Department of Neurology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama, Yufu, Oita, 879-5593, Japan
| | - Yoshitaka Maeda
- Microbes & Host Defense Domain Deep Human Biology Learning, Eisai Co., Ltd, 5-1-3, Tokodai, Tsukuba-Shi, Ibaraki, 300-2635, Japan
| | - Ken Aoshima
- Microbes & Host Defense Domain Deep Human Biology Learning, Eisai Co., Ltd, 5-1-3, Tokodai, Tsukuba-Shi, Ibaraki, 300-2635, Japan.
- School of Integrative and Global Majors, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Etsuro Matsubara
- Department of Neurology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama, Yufu, Oita, 879-5593, Japan
| |
Collapse
|
8
|
Morella I, Negro M, Dossena M, Brambilla R, D'Antona G. Gut-muscle-brain axis: Molecular mechanisms in neurodegenerative disorders and potential therapeutic efficacy of probiotic supplementation coupled with exercise. Neuropharmacology 2023; 240:109718. [PMID: 37774944 DOI: 10.1016/j.neuropharm.2023.109718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/13/2023] [Accepted: 09/16/2023] [Indexed: 10/01/2023]
Abstract
Increased longevity is often associated with age-related conditions. The most common neurodegenerative disorders in the older population are Alzheimer's disease (AD) and Parkinson's disease (PD), associated with progressive neuronal loss leading to functional and cognitive impairments. Although symptomatic treatments are available, there is currently no cure for these conditions. Gut dysbiosis has been involved in the pathogenesis of AD and PD, thus interventions targeting the "gut-brain axis" could potentially prevent or delay these pathologies. Recent evidence suggests that the skeletal muscle and the gut microbiota can affect each other via the "gut-muscle axis". Importantly, cognitive functions in AD and PD patients significantly benefit from physical activity. In this review, we aim to provide a comprehensive picture of the crosstalk between the brain, the skeletal muscle and the gut microbiota, introducing the concept of "gut-muscle-brain axis". Moreover, we discuss human and animal studies exploring the modulatory role of exercise and probiotics on cognition in AD and PD. Collectively, the findings presented here support the potential benefits of physical activity and probiotic supplementation in AD and PD. Further studies will be needed to develop targeted and multimodal strategies, including lifestyle changes, to prevent or delay the course of these pathologies.
Collapse
Affiliation(s)
- Ilaria Morella
- Neuroscience and Mental Health Innovation Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Massimo Negro
- Centro di Ricerca Interdipartimentale Nelle Attività Motorie e Sportive (CRIAMS)-Sport Medicine Centre, University of Pavia, Voghera, Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Riccardo Brambilla
- Neuroscience and Mental Health Innovation Institute, School of Biosciences, Cardiff University, Cardiff, UK; Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Giuseppe D'Antona
- Centro di Ricerca Interdipartimentale Nelle Attività Motorie e Sportive (CRIAMS)-Sport Medicine Centre, University of Pavia, Voghera, Italy; Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
9
|
Kim E, Kim H, Jedrychowski MP, Bakiasi G, Park J, Kruskop J, Choi Y, Kwak SS, Quinti L, Kim DY, Wrann CD, Spiegelman BM, Tanzi RE, Choi SH. Irisin reduces amyloid-β by inducing the release of neprilysin from astrocytes following downregulation of ERK-STAT3 signaling. Neuron 2023; 111:3619-3633.e8. [PMID: 37689059 PMCID: PMC10840702 DOI: 10.1016/j.neuron.2023.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/09/2023] [Accepted: 08/11/2023] [Indexed: 09/11/2023]
Abstract
A pathological hallmark of Alzheimer's disease (AD) is the deposition of amyloid-β (Aβ) protein in the brain. Physical exercise has been shown to reduce Aβ burden in various AD mouse models, but the underlying mechanisms have not been elucidated. Irisin, an exercise-induced hormone, is the secreted form of fibronectin type-III-domain-containing 5 (FNDC5). Here, using a three-dimensional (3D) cell culture model of AD, we show that irisin significantly reduces Aβ pathology by increasing astrocytic release of the Aβ-degrading enzyme neprilysin (NEP). This is mediated by downregulation of ERK-STAT3 signaling. Finally, we show that integrin αV/β5 acts as the irisin receptor on astrocytes required for irisin-induced release of astrocytic NEP, leading to clearance of Aβ. Our findings reveal for the first time a cellular and molecular mechanism by which exercise-induced irisin attenuates Aβ pathology, suggesting a new target pathway for therapies aimed at the prevention and treatment of AD.
Collapse
Affiliation(s)
- Eunhee Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hyeonwoo Kim
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA; Department of Biological Sciences, Korea Advanced Institute of Science & Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mark P Jedrychowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Grisilda Bakiasi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joseph Park
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jane Kruskop
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Younjung Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sang Su Kwak
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Luisa Quinti
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Christiane D Wrann
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
10
|
Li X, Zhang Y, Ding X, Jin Y, Wei C, Xu J. Mass Spectrometry Chromatography-Based Metabolomics: The Effect of Long-Term Aerobic Exercise on Learning Ability and the Metabolism of Intestinal Contents in Mice with Alzheimer's Disease. Metabolites 2023; 13:1150. [PMID: 37999246 PMCID: PMC10673277 DOI: 10.3390/metabo13111150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/02/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
This study aimed to investigate the effect of long-term aerobic exercise on the metabolism of intestinal contents in APP/PS1 mice was studied using a non-targeted metabolomics technique based on high-performance liquid chromatography-mass spectrometry (HPLC-MS) coupling, providing a theoretical basis for exercise to regulate the metabolism of Alzheimer's disease (AD) organisms. Three-month-old male C57BL/6JNju mice, six wild-type (NC, n = 6); 12 APP/PS1 double transgenic species in total, were randomly divided into AD model (AM, n = 6) and AD model exercise (AE, n = 6) groups. The mice in the NC group were fed naturally, the mice in the AM group were statically placed on a running platform, and the mice in the AE group received a 20-week long-term moderate intensity running platform exercise intervention. Following the exercise intervention, the cecum contents of the mice in each group were collected and analyzed using the HPLC-MS technique, with those meeting both variable important in projection (VIP)> 1.5 and p < 0.05 being screened as differential metabolites. A total of 32 different metabolites were detected between the AM and NC groups, with 19 up-regulated in the AM group such as phosphatidic acid (PA) (18:4(6Z,9Z,12Z,15Z)/21:0) and 13 down-regulated in the AM group, such as 4,8-dimethylnonanoyl, compared to the NC group; 98 different metabolites were found between the AM and AE groups, 41 of which were upregulated such as Lyso phosphatidylcholine (LysoPC) and 57 of which were downregulated compared to the AM group such as Phosphatidylinositol (PI). The regulation of linoleic acid metabolism, glycerophospholipid metabolism, bile secretion, phenylalanine metabolism, and other pathways was predominantly regulated by nine metabolites, which were subsequently identified as indicators of exercise intervention to enhance metabolism in AD mice. The metabolomic technique can identify the metabolic problems of intestinal contents in AD mice and initially screen the biomarkers of exercise to improve the metabolic disorders in AD. These findings can help us better understand the impact of aerobic exercise on AD metabolism.
Collapse
Affiliation(s)
- Xue Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (Y.Z.); (X.D.); (Y.J.); (C.W.); (J.X.)
| | | | | | | | | | | |
Collapse
|
11
|
Collins AM, Molina-Hidalgo C, Aghjayan SL, Fanning J, Erlenbach ED, Gothe NP, Velazquez-Diaz D, Erickson KI. Differentiating the influence of sedentary behavior and physical activity on brain health in late adulthood. Exp Gerontol 2023; 180:112246. [PMID: 37356467 DOI: 10.1016/j.exger.2023.112246] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/22/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
Public health messaging calls for individuals to be more physically active and less sedentary, yet these lifestyle behaviors have been historically studied independently. Both physical activity (PA) and sedentary behavior (SB) are linked through time-use in a 24-hour day and are related to health outcomes, such as neurocognition. While the benefits of PA on brain health in late adulthood have been well-documented, the influence of SB remains to be understood. The purpose of this paper was to critically review the evolving work on SB and brain health in late adulthood and emphasize key areas of consideration to inform potential research. Overall, the existing literature studying the impact of SB on the components and mechanisms of brain health are mixed and inconclusive, provided largely by cross-sectional and observational work employing a variety of measurement techniques of SB and brain health outcomes. Further, many studies did not conceptually or statistically account for the role of PA in the proposed relationships. Therefore, our understanding of the way in which SB may influence neurocognition in late adulthood is limited. Future efforts should include more prospective longitudinal and randomized clinical trials with intentional methodological approaches to better understand the relationships between SB and the brain in late adulthood, and how these potential links are differentiated from PA.
Collapse
Affiliation(s)
- Audrey M Collins
- AdventHealth Research Institute, Department of Neuroscience, AdventHealth, Orlando, FL, USA.
| | | | - Sarah L Aghjayan
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jason Fanning
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, NC, USA
| | - Emily D Erlenbach
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Neha P Gothe
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Daniel Velazquez-Diaz
- AdventHealth Research Institute, Department of Neuroscience, AdventHealth, Orlando, FL, USA; Exphy Research Group, Department of Physical Education, Faculty of Education Sciences, University Hospital, University of Cadiz, 11009 Cadiz, Spain; Biomedical Research and Innovation Institute of Cadiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain
| | - Kirk I Erickson
- AdventHealth Research Institute, Department of Neuroscience, AdventHealth, Orlando, FL, USA; Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Pinto-Hernandez P, Castilla-Silgado J, Coto-Vilcapoma A, Fernández-Sanjurjo M, Fernández-García B, Tomás-Zapico C, Iglesias-Gutiérrez E. Modulation of microRNAs through Lifestyle Changes in Alzheimer's Disease. Nutrients 2023; 15:3688. [PMID: 37686720 PMCID: PMC10490103 DOI: 10.3390/nu15173688] [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: 07/28/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Lifestyle factors, including diet and physical activity (PA), are known beneficial strategies to prevent and delay Alzheimer's disease (AD) development. Recently, microRNAs have emerged as potential biomarkers in multiple diseases, including AD. The aim of this review was to analyze the available information on the modulatory effect of lifestyle on microRNA expression in AD. Few studies have addressed this question, leaving important gaps and limitations: (1) in human studies, only circulating microRNAs were analyzed; (2) in mice studies, microRNA expression was only analyzed in brain tissue; (3) a limited number of microRNAs was analyzed; (4) no human nutritional intervention studies were conducted; and (5) PA interventions in humans and mice were poorly detailed and only included aerobic training. Despite this, some conclusions could be drawn. Circulating levels of let-7g-5p, miR-107, and miR-144-3p were associated with overall diet quality in mild cognitive impairment patients. In silico analysis showed that these microRNAs are implicated in synapse formation, microglia activation, amyloid beta accumulation, and pro-inflammatory pathways, the latter also being targeted by miR-129-5p and miR-192-5p, whose circulating levels are modified by PA in AD patients. PA also modifies miR-132, miR-15b-5p, miR-148b-3p, and miR-130a-5p expression in mice brains, which targets are related to the regulation of neuronal activity, ageing, and pro-inflammatory pathways. This supports the need to further explore lifestyle-related miRNA changes in AD, both as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Paola Pinto-Hernandez
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Juan Castilla-Silgado
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Almudena Coto-Vilcapoma
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Manuel Fernández-Sanjurjo
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Benjamín Fernández-García
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
- Department of Morphology and Cell Biology, Anatomy, University of Oviedo, 33006 Asturias, Spain
| | - Cristina Tomás-Zapico
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| | - Eduardo Iglesias-Gutiérrez
- Department of Functional Biology, Physiology, University of Oviedo, 33006 Asturias, Spain; (P.P.-H.); (J.C.-S.); (A.C.-V.); (M.F.-S.); (C.T.-Z.)
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Asturias, Spain;
| |
Collapse
|
13
|
Andrade-Guerrero J, Orta-Salazar E, Salinas-Lara C, Sánchez-Garibay C, Rodríguez-Hernández LD, Vargas-Rodríguez I, Barron-Leon N, Ledesma-Alonso C, Diaz-Cintra S, Soto-Rojas LO. Effects of Voluntary Physical Exercise on the Neurovascular Unit in a Mouse Model of Alzheimer's Disease. Int J Mol Sci 2023; 24:11134. [PMID: 37446312 DOI: 10.3390/ijms241311134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide. Histopathologically, AD presents two pathognomonic hallmarks: (1) neurofibrillary tangles, characterized by intracellular deposits of hyperphosphorylated tau protein, and (2) extracellular amyloid deposits (amyloid plaques) in the brain vasculature (cerebral amyloid angiopathy; CAA). It has been proposed that vascular amyloid deposits could trigger neurovascular unit (NVU) dysfunction in AD. The NVU is composed primarily of astrocytic feet, endothelial cells, pericytes, and basement membrane. Although physical exercise is hypothesized to have beneficial effects against AD, it is unknown whether its positive effects extend to ameliorating CAA and improving the physiology of the NVU. We used the triple transgenic animal model for AD (3xTg-AD) at 13 months old and analyzed through behavioral and histological assays, the effect of voluntary physical exercise on cognitive functions, amyloid angiopathy, and the NVU. Our results show that 3xTg-AD mice develop vascular amyloid deposits which correlate with cognitive deficits and NVU alteration. Interestingly, the physical exercise regimen decreases amyloid angiopathy and correlates with an improvement in cognitive function as well as in the underlying integrity of the NVU components. Physical exercise could represent a key therapeutic approach in cerebral amyloid angiopathy and NVU stability in AD patients.
Collapse
Affiliation(s)
- Jesús Andrade-Guerrero
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Erika Orta-Salazar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Citlaltepetl Salinas-Lara
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México 14269, Mexico
| | - Carlos Sánchez-Garibay
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México 14269, Mexico
| | - Luis Daniel Rodríguez-Hernández
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Isaac Vargas-Rodríguez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Nayeli Barron-Leon
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Carlos Ledesma-Alonso
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Sofía Diaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | - Luis O Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| |
Collapse
|
14
|
Xu L, Liu R, Qin Y, Wang T. Brain metabolism in Alzheimer's disease: biological mechanisms of exercise. Transl Neurodegener 2023; 12:33. [PMID: 37365651 DOI: 10.1186/s40035-023-00364-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
Alzheimer's disease (AD) is a major subtype of neurodegenerative dementia caused by long-term interactions and accumulation of multiple adverse factors, accompanied by dysregulation of numerous intracellular signaling and molecular pathways in the brain. At the cellular and molecular levels, the neuronal cellular milieu of the AD brain exhibits metabolic abnormalities, compromised bioenergetics, impaired lipid metabolism, and reduced overall metabolic capacity, which lead to abnormal neural network activity and impaired neuroplasticity, thus accelerating the formation of extracellular senile plaques and intracellular neurofibrillary tangles. The current absence of effective pharmacological therapies for AD points to the urgent need to investigate the benefits of non-pharmacological approaches such as physical exercise. Despite the evidence that regular physical activity can improve metabolic dysfunction in the AD state, inhibit different pathophysiological molecular pathways associated with AD, influence the pathological process of AD, and exert a protective effect, there is no clear consensus on the specific biological and molecular mechanisms underlying the advantages of physical exercise. Here, we review how physical exercise improves crucial molecular pathways and biological processes associated with metabolic disorders in AD, including glucose metabolism, lipid metabolism, Aβ metabolism and transport, iron metabolism and tau pathology. How metabolic states influence brain health is also presented. A better knowledge on the neurophysiological mechanisms by which exercise improves AD metabolism can contribute to the development of novel drugs and improvement of non-pharmacological interventions.
Collapse
Affiliation(s)
- Longfei Xu
- Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300050, China
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China
| | - Ran Liu
- Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300050, China
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China
| | - Yingkai Qin
- Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300050, China.
| | - Tianhui Wang
- Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300050, China.
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China.
| |
Collapse
|
15
|
Williams E, Mutlu-Smith M, Alex A, Chin XW, Spires-Jones T, Wang SH. Mid-Adulthood Cognitive Training Improves Performance in a Spatial Task but Does Not Ameliorate Hippocampal Pathology in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2023; 93:683-704. [PMID: 37066912 DOI: 10.3233/jad-221185] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND Prior experience in early life has been shown to improve performance in aging and mice with Alzheimer's disease (AD) pathology. However, whether cognitive training at a later life stage would benefit subsequent cognition and reduce pathology in AD mice needs to be better understood. OBJECTIVE This study aimed to verify if behavioral training in mid-adulthood would improve subsequent cognition and reduce AD pathology and astrogliosis. METHODS Mixed-sex APP/PS1 and wildtype littermate mice received a battery of behavioral training, composed of spontaneous alternation in the Y-maze, novel object recognition and location tasks, and spatial training in the water maze, or handling only at 7 months of age. The impact of AD genotype and prior training on subsequent learning and memory of aforementioned tasks were assessed at 9 months. RESULTS APP/PS1 mice made more errors than wildtype littermates in the radial-arm water maze (RAWM) task. Prior training prevented this impairment in APP/PS1 mice. Prior training also contributed to better efficiency in finding the escape platform in both APP/PS1 mice and wildtype littermates. Short-term and long-term memory of this RAWM task, of a reversal task, and of a transfer task were comparable among APP/PS1 and wildtype mice, with or without prior training. Amyloid pathology and astrogliosis in the hippocampus were also comparable between the APP/PS1 groups. CONCLUSION These data suggest that cognitive training in mid-adulthood improves subsequent accuracy in AD mice and efficiency in all mice in the spatial task. Cognitive training in mid-adulthood provides no clear benefit on memory or on amyloid pathology in midlife.
Collapse
Affiliation(s)
- Elizabeth Williams
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Menekşe Mutlu-Smith
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Ashli Alex
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Xi Wei Chin
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Tara Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Szu-Han Wang
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
16
|
Physical activity for cognitive health promotion: An overview of the underlying neurobiological mechanisms. Ageing Res Rev 2023; 86:101868. [PMID: 36736379 DOI: 10.1016/j.arr.2023.101868] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/13/2022] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Physical activity is one of the modifiable factors of cognitive decline and dementia with the strongest evidence. Although many influential reviews have illustrated the neurobiological mechanisms of the cognitive benefits of physical activity, none of them have linked the neurobiological mechanisms to normal exercise physiology to help the readers gain a more advanced, comprehensive understanding of the phenomenon. In this review, we address this issue and provide a synthesis of the literature by focusing on five most studied neurobiological mechanisms. We show that the body's adaptations to enhance exercise performance also benefit the brain and contribute to improved cognition. Specifically, these adaptations include, 1), the release of growth factors that are essential for the development and growth of neurons and for neurogenesis and angiogenesis, 2), the production of lactate that provides energy to the brain and is involved in the synthesis of glutamate and the maintenance of long-term potentiation, 3), the release of anti-inflammatory cytokines that reduce neuroinflammation, 4), the increase in mitochondrial biogenesis and antioxidant enzyme activity that reduce oxidative stress, and 5), the release of neurotransmitters such as dopamine and 5-HT that regulate neurogenesis and modulate cognition. We also discussed several issues relevant for prescribing physical activity, including what intensity and mode of physical activity brings the most cognitive benefits, based on their influence on the above five neurobiological mechanisms. We hope this review helps readers gain a general understanding of the state-of-the-art knowledge on the neurobiological mechanisms of the cognitive benefits of physical activity and guide them in designing new studies to further advance the field.
Collapse
|
17
|
Rangasamy SB, Jana M, Dasarathi S, Kundu M, Pahan K. Treadmill workout activates PPARα in the hippocampus to upregulate ADAM10, decrease plaques and improve cognitive functions in 5XFAD mouse model of Alzheimer's disease. Brain Behav Immun 2023; 109:204-218. [PMID: 36682514 PMCID: PMC10023420 DOI: 10.1016/j.bbi.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 12/26/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Although liver is rich in peroxisome proliferator-activated receptor α (PPARα), recently we have described the presence of PPARα in hippocampus where it is involved in non-amyloidogenic metabolism of amyloid precursor protein (APP) via ADAM10, decreasing amyloid plaques and improving memory and learning. However, mechanisms to upregulate PPARα in vivo in the hippocampus are poorly understood. Regular exercise has multiple beneficial effects on human health and here, we describe the importance of regular mild treadmill exercise in upregulating PPARα in vivo in the hippocampus of 5XFAD mouse model of Alzheimer's disease. We also demonstrate that treadmill exercise remained unable to stimulate ADAM10, reduce plaque pathology and improve cognitive functions in 5XFADΔPPARα mice (5XFAD mice lacking PPARα). On the other hand, treadmill workout increased ADAM10, decreased plaque pathology and protected memory and learning in 5XFADΔPPARβ mice (5XFAD mice lacking PPARβ). Moreover, the other PPAR (PPARγ) also did not play any role in the transcription of ADAM10 in vivo in the hippocampus of treadmill exercised 5XFAD mice. These results underline an important role of PPARα in which treadmill exercise remains unable to exhibit neuroprotection in the hippocampus in the absence of PPARα.
Collapse
Affiliation(s)
- Suresh B Rangasamy
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| | - Malabendu Jana
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| | - Sridevi Dasarathi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Madhuchhanda Kundu
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| | - Kalipada Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, USA.
| |
Collapse
|
18
|
de Frutos Lucas J, Sewell KR, García-Colomo A, Markovic S, Erickson KI, Brown BM. How does apolipoprotein E genotype influence the relationship between physical activity and Alzheimer's disease risk? A novel integrative model. Alzheimers Res Ther 2023; 15:22. [PMID: 36707869 PMCID: PMC9881295 DOI: 10.1186/s13195-023-01170-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/15/2023] [Indexed: 01/29/2023]
Abstract
BACKGROUND Wide evidence suggests that physical activity (PA) confers protection against Alzheimer's disease (AD). On the other hand, the apolipoprotein E gene (APOE) ε4 allele represents the greatest genetic risk factor for developing AD. Extensive research has been conducted to determine whether frequent PA can mitigate the increased AD risk associated with APOE ε4. However, thus far, these attempts have produced inconclusive results. In this context, one possible explanation could be that the influence of the combined effect of PA and APOE ε4 carriage might be dependent on the specific outcome measure utilised. MAIN BODY In order to bridge these discrepancies, the aim of this theoretical article is to propose a novel model on the interactive effects of PA and APOE ε4 carriage on well-established mechanisms underlying AD. Available literature was searched to investigate how PA and APOE ε4 carriage, independently and in combination, may alter several molecular pathways involved in AD pathogenesis. The reviewed mechanisms include amyloid beta (Aβ) and tau deposition and clearance, neuronal resilience and neurogenesis, lipid function and cerebrovascular alterations, brain immune response and glucose metabolism. Finally, combining all this information, we have built an integrative model, which includes evidence-based and theoretical synergistic interactions across mechanisms. Moreover, we have identified key knowledge gaps in the literature, providing a list of testable hypotheses that future studies need to address. CONCLUSIONS We conclude that PA influences a wide array of molecular targets involved in AD neuropathology. A deeper understanding of where, when and, most importantly, how PA decreases AD risk even in the presence of the APOE ε4 allele will enable the creation of new protocols using exercise along pharmaceuticals in combined therapeutic approaches.
Collapse
Affiliation(s)
- Jaisalmer de Frutos Lucas
- Experimental Psychology, Cognitive Processes and Logopedia Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcón, Spain.
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, 6027, Australia.
- Departamento de PsicologíaFacultad de Ciencias de la Vida y de la Naturaleza, Universidad Antonio de Nebrija, 28015, Madrid, Spain.
| | - Kelsey R Sewell
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Alejandra García-Colomo
- Experimental Psychology, Cognitive Processes and Logopedia Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcón, Spain
| | - Shaun Markovic
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
- PROFITH "PROmoting FITness and Health Through Physical Activity" Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, 18071, Granada, Spain
- AdventHealth Research Institute, Orlando, FL, 32804, USA
| | - Belinda M Brown
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
| |
Collapse
|
19
|
Solis-Urra P, Rodriguez-Ayllon M, Álvarez-Ortega M, Molina-Hidalgo C, Molina-Garcia P, Arroyo-Ávila C, García-Hermoso A, Collins AM, Jain S, Gispert JD, Liu-Ambrose T, Ortega FB, Erickson KI, Esteban-Cornejo I. Physical Performance and Amyloid-β in Humans: A Systematic Review and Meta-Analysis of Observational Studies. J Alzheimers Dis 2023; 96:1427-1439. [PMID: 38007656 DOI: 10.3233/jad-230586] [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] [Indexed: 11/27/2023]
Abstract
BACKGROUND Accumulation of amyloid-β (Aβ) plaques is one of the main features of Alzheimer's disease (AD). Physical performance has been related to dementia risk and Aβ, and it has been hypothesized as one of the mechanisms leading to greater accumulation of Aβ. Yet, no evidence synthesis has been performed in humans. OBJECTIVE To investigate the association of physical performance with Aβ in humans, including Aβ accumulation on brain, and Aβ abnormalities measured in cerebrospinal fluid (CSF) and blood. METHODS A systematic review with multilevel meta-analysis was performed from inception to June 16th, 2022. Studies were eligible if they examined the association of physical performance with Aβ levels, including the measure of physical performance as a predictor and the measure of Aβ as an outcome in humans. RESULTS 7 articles including 2,619 participants were included in the meta-analysis. The results showed that physical performance was not associated with accumulation of Aβ in the brain (ES = 0.01; 95% CI -0.21 to 0.24; I2 = 69.9%), in the CSF (ES = -0.28; 95% CI -0.98 to 0.41; I2 = 91.0%) or in the blood (ES = -0.19; 95% CI -0.61 to 0.24; I2 = 99.75%). Significant heterogeneity was found across the results , which posed challenges in arriving at consistent conclusions; and the limited number of studies hindered the opportunity to conduct a moderation analysis. CONCLUSIONS The association between physical performance and Aβ is inconclusive. This uncertainly arises from the limited number of studies, study design limitations, and heterogeneity of measurement approaches. More studies are needed to determine whether physical performance is related to Aβ levels in humans.
Collapse
Affiliation(s)
- Patricio Solis-Urra
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
- Nuclear Medicine Services, "Virgen de Las Nieves", University Hospital, Granada, Spain
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar, Chile
| | - María Rodriguez-Ayllon
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Miriam Álvarez-Ortega
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Cristina Molina-Hidalgo
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
- AdventHealth Research Institute, Neuroscience, Orlando, FL, USA
| | - Pablo Molina-Garcia
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
- Physical Medicine and Rehabilitation Service, Virgen de las Nieves University Hospital, Instituto de Investigacion Biosanitaria ibs.GRANADA, Granada, Spain
| | - Cristina Arroyo-Ávila
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Antonio García-Hermoso
- Navarrabiomed, Hospital Universitario de Navarra, IdiSNA, Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | | | - Shivangi Jain
- AdventHealth Research Institute, Neuroscience, Orlando, FL, USA
| | - Juan Domingo Gispert
- BarcelonaBeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Teresa Liu-Ambrose
- Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
- Aging, Mobility, and Cognitive Health Laboratory, Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Francisco B Ortega
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
- AdventHealth Research Institute, Neuroscience, Orlando, FL, USA
| | - Irene Esteban-Cornejo
- Department of Physical Education and Sports, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- ibs.GRANADA Instituto de Investigación Biosanitaria, Granada, Spain
| |
Collapse
|
20
|
Abstract
This review deals with an unwelcome reality about several forms of dementia, including Alzheimer's disease- that these dementias are caused, in part or whole, by the aging of the vasculature. Since the vasculature ages in us all, dementia is our fate, sealed by the realit!ies of the circulation; it is not a disease with a cure pending. Empirically, cognitive impairment before our 7th decade is uncommon and considered early, while a diagnosis in our 11th decade is late but common in that cohort (>40%). Projections from earlier ages suggest that the prevalence of dementia in people surviving into their 12th decade exceeds 80%. We address the question why so few of many interventions known to delay dementia are recognized as therapy; and we try to resolve this few-and-many paradox, identifying opportunities for better treatment, especially pre-diagnosis. The idea of dementia as a fate is resisted, we argue, because it negates the hope of a cure. But the price of that hope is lost opportunity. An approach more in line with the evidence, and more likely to limit suffering, is to understand the damage that accumulates with age in the cerebral vasculature and therefore in the brain, and which eventually gives rise to cognitive symptoms in late life, too often leading to dementia. We argue that hope should be redirected to delaying that damage and with it the onset of cognitive loss; and, for each individual, it should be redirected to a life-long defense of their brain.
Collapse
Affiliation(s)
- Marcus J Andersson
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Jonathan Stone
- School of Medical Sciences and Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
21
|
Environmental stimulation in Huntington disease patients and animal models. Neurobiol Dis 2022; 171:105725. [DOI: 10.1016/j.nbd.2022.105725] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 01/07/2023] Open
|
22
|
Pedrini S, Chatterjee P, Nakamura A, Tegg M, Hone E, Rainey-Smith SR, Rowe CC, Dore V, Villemagne VL, Ames D, Kaneko N, Gardener SL, Taddei K, Fernando B, Martins I, Bharadwaj P, Sohrabi HR, Masters CL, Brown B, Martins RN. The Association Between Alzheimer's Disease-Related Markers and Physical Activity in Cognitively Normal Older Adults. Front Aging Neurosci 2022; 14:771214. [PMID: 35418852 PMCID: PMC8996810 DOI: 10.3389/fnagi.2022.771214] [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: 09/06/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies have indicated that physical activity may be beneficial in reducing the risk for Alzheimer's disease (AD), although the underlying mechanisms are not fully understood. The goal of this study was to evaluate the relationship between habitual physical activity levels and brain amyloid deposition and AD-related blood biomarkers (i.e., measured using a novel high-performance mass spectrometry-based assay), in apolipoprotein E (APOE) ε4 carriers and noncarriers. We evaluated 143 cognitively normal older adults, all of whom had brain amyloid deposition assessed using positron emission tomography and had their physical activity levels measured using the International Physical Activity Questionnaire (IPAQ). We observed an inverse correlation between brain amyloidosis and plasma beta-amyloid (Aβ)1−42 but found no association between brain amyloid and plasma Aβ1−40 and amyloid precursor protein (APP)669−711. Additionally, higher levels of physical activity were associated with lower plasma Aβ1−40, Aβ1−42, and APP669−711 levels in APOE ε4 noncarriers. The ratios of Aβ1−40/Aβ1−42 and APP669−711/Aβ1−42, which have been associated with higher brain amyloidosis in previous studies, differed between APOE ε4 carriers and non-carriers. Taken together, these data indicate a complex relationship between physical activity and brain amyloid deposition and potential blood-based AD biomarkers in cognitively normal older adults. In addition, the role of APOE ε4 is still unclear, and more studies are necessary to bring further clarification.
Collapse
Affiliation(s)
- Steve Pedrini
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
| | - Pratishtha Chatterjee
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
- Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Akinori Nakamura
- Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Michelle Tegg
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
| | - Eugene Hone
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
| | - Stephanie R. Rainey-Smith
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Christopher C. Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia
| | - Vincent Dore
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia
| | - Victor L. Villemagne
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - David Ames
- National Ageing Research Institute, Parkville, VIC, Australia
- Academic Unit for Psychiatry of Old Age, St George's Hospital, University of Melbourne, Kew, VIC, Australia
| | - Naoki Kaneko
- Koichi Tanaka Mass Spectrometry Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Sam L. Gardener
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
| | - Kevin Taddei
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
| | - Binosha Fernando
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
| | - Ian Martins
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
| | - Prashant Bharadwaj
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
| | - Hamid R. Sohrabi
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Colin L. Masters
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Belinda Brown
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Ralph N. Martins
- School of Medical Sciences, Sarich Neuroscience Research Institute, Edith Cowan University, Nedlands, WA, Australia
- Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, WA, Australia
- *Correspondence: Ralph N. Martins
| |
Collapse
|
23
|
Gonneaud J, Moreau I, Felisatti F, Arenaza‐Urquijo E, Ourry V, Touron E, de la Sayette V, Vivien D, Chételat G. Men and women show partly distinct effects of physical activity on brain integrity. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12302. [PMID: 35382233 PMCID: PMC8959639 DOI: 10.1002/dad2.12302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/04/2022] [Accepted: 01/16/2022] [Indexed: 12/14/2022]
Abstract
Introduction Physical inactivity and female sex are independently associated with increased Alzheimer's disease (AD) lifetime risk. This study investigates the possible interactions between sex and physical activity on neuroimaging biomarkers. Methods In 134 cognitively unimpaired older adults (≥65 years, 82 women) from the Age-Well randomized controlled trial (baseline data), we investigated the association between physical activity and multimodal neuroimaging (gray matter volume, glucose metabolism, perfusion, and amyloid burden), and how sex modulates these associations. Results The anterior cingulate cortex volume was independently associated with sex and physical activity. Sex and physical activity interacted on perfusion and amyloid deposition in medial parietal regions, such that physical activity was related to perfusion only in women, and to amyloid burden only in men. Discussion Physical activity has both sex-dependent and sex-independent associations with brain integrity. Our findings highlight partly distinct reserve mechanisms in men and women, which might in turn influence their risk of AD. Highlights Sex and physical activity have been linked to Alzheimer's disease (AD) progression.The association of sex and physical activity with brain health is partly independent.Different reserve mechanisms exist in men and women.
Collapse
Affiliation(s)
- Julie Gonneaud
- Normandie UniversitéUniversité de CaenInstitut National de la Santé et de la Recherche MédicaleUnité 1237 "Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain@ Caen‐NormandieGIP CyceronCaenFrance
| | - Ilana Moreau
- Normandie UniversitéUniversité de CaenInstitut National de la Santé et de la Recherche MédicaleUnité 1237 "Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain@ Caen‐NormandieGIP CyceronCaenFrance
| | - Francesca Felisatti
- Normandie UniversitéUniversité de CaenInstitut National de la Santé et de la Recherche MédicaleUnité 1237 "Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain@ Caen‐NormandieGIP CyceronCaenFrance
| | - Eider Arenaza‐Urquijo
- Normandie UniversitéUniversité de CaenInstitut National de la Santé et de la Recherche MédicaleUnité 1237 "Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain@ Caen‐NormandieGIP CyceronCaenFrance
- Barcelonabeta Brain Research CenterFundación Pasqual MaragallBarcelonaSpain
| | - Valentin Ourry
- Normandie UniversitéUniversité de CaenInstitut National de la Santé et de la Recherche MédicaleUnité 1237 "Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain@ Caen‐NormandieGIP CyceronCaenFrance
- Normandie UnivUNICAEN, PSL UniversitéEPHE, INSERM, U1077CHU de CaenGIP CyceronNIMHCaenFrance
| | - Edelweiss Touron
- Normandie UniversitéUniversité de CaenInstitut National de la Santé et de la Recherche MédicaleUnité 1237 "Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain@ Caen‐NormandieGIP CyceronCaenFrance
| | - Vincent de la Sayette
- Normandie UnivUNICAEN, PSL UniversitéEPHE, INSERM, U1077CHU de CaenGIP CyceronNIMHCaenFrance
- Service de NeurologieCentre Hospitalier Universitaire de CaenCaenFrance
| | - Denis Vivien
- Normandie UniversitéUniversité de CaenInstitut National de la Santé et de la Recherche MédicaleUnité 1237 "Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain@ Caen‐NormandieGIP CyceronCaenFrance
| | - Gaël Chételat
- Normandie UniversitéUniversité de CaenInstitut National de la Santé et de la Recherche MédicaleUnité 1237 "Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain@ Caen‐NormandieGIP CyceronCaenFrance
| |
Collapse
|
24
|
Ribarič S. Physical Exercise, a Potential Non-Pharmacological Intervention for Attenuating Neuroinflammation and Cognitive Decline in Alzheimer's Disease Patients. Int J Mol Sci 2022; 23:ijms23063245. [PMID: 35328666 PMCID: PMC8952567 DOI: 10.3390/ijms23063245] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/14/2022] Open
Abstract
This narrative review summarises the evidence for considering physical exercise (PE) as a non-pharmacological intervention for delaying cognitive decline in patients with Alzheimer’s disease (AD) not only by improving cardiovascular fitness but also by attenuating neuroinflammation. Ageing is the most important risk factor for AD. A hallmark of the ageing process is a systemic low-grade chronic inflammation that also contributes to neuroinflammation. Neuroinflammation is associated with AD, Parkinson’s disease, late-onset epilepsy, amyotrophic lateral sclerosis and anxiety disorders. Pharmacological treatment of AD is currently limited to mitigating the symptoms and attenuating progression of the disease. AD animal model studies and human studies on patients with a clinical diagnosis of different stages of AD have concluded that PE attenuates cognitive decline not only by improving cardiovascular fitness but possibly also by attenuating neuroinflammation. Therefore, low-grade chronic inflammation and neuroinflammation should be considered potential modifiable risk factors for AD that can be attenuated by PE. This opens the possibility for personalised attenuation of neuroinflammation that could also have important health benefits for patients with other inflammation associated brain disorders (i.e., Parkinson’s disease, late-onset epilepsy, amyotrophic lateral sclerosis and anxiety disorders). In summary, life-long, regular, structured PE should be considered as a supplemental intervention for attenuating the progression of AD in human. Further studies in human are necessary to develop optimal, personalised protocols, adapted to the progression of AD and the individual’s mental and physical limitations, to take full advantage of the beneficial effects of PE that include improved cardiovascular fitness, attenuated systemic inflammation and neuroinflammation, stimulated brain Aβ peptides brain catabolism and brain clearance.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
25
|
Musaeus CS, Johansen LB, Hasselbalch S, Beyer N, Høgh P, Siebner HR, Frederiksen KS. Sixteen weeks of aerobic exercise does not alter resting-state connectivity of the precuneus in patients with Alzheimer's disease. Curr Alzheimer Res 2022; 19:171-177. [PMID: 35249488 DOI: 10.2174/1567205019666220304091241] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/27/2021] [Accepted: 12/08/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION In healthy elderly persons and patients with mild cognitive impairment, physical exercise can increase functional brain connectivity in the default mode network (DMN) measured by restingstate functional magnetic resonance imaging (rs-fMRI). However, no studies have so far investigated the effect of physical exercise on functional resting-state connectivity in the DMN in patients with Alzheimer's disease (AD). OBJECTIVE In a single-blinded randomized controlled trial, we assessed the effects of an aerobic exercise intervention of 16 weeks of physical exercise on DMN connectivity using rs-fMRI in patients with AD. METHODS Forty-five patients were randomly assigned to either a control or exercise group. The exercise group performed 60-min of aerobic exercise three times per week for 16 weeks. All the patients underwent whole-brain rs-fMRI at 3 T, at baseline, and after 16 weeks. Since the posterior cingulate cortex (PCC) and adjacent precuneus constitute a central hub of the DMN, this parietal region was defined as region-ofinterest and used as the seed region for functional connectivity analysis of the rs-fMRI data treating age and gender as covariates. RESULTS Neither seed-based analysis, seeded in the PCC/precuneus region nor ICA-based analyses, focusing on components of the DMN network, showed any exercise-induced changes in functional resting-state connectivity from baseline to follow-up. CONCLUSION 16 weeks of aerobic exercise does not modify functional connectivity of the PCC/precuneus region in patients with AD. A longer intervention may be needed to show the effect of exercise on brain connectivity.
Collapse
Affiliation(s)
- Christian Sandøe Musaeus
- Department of Neurology, Danish Dementia Research Centre (DDRC), University of Copenhagen, Rigshospitalet, Denmark
| | - Louise Baruël Johansen
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Steen Hasselbalch
- Department of Neurology, Danish Dementia Research Centre (DDRC), University of Copenhagen, Rigshospitalet, Denmark
| | - Nina Beyer
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Høgh
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Regional Dementia Research Centre, Zealand University Hospital, Roskilde, Denmark
| | - Hartwig Roman Siebner
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University, Hospital Bispebjerg, Copenhagen, Denmark
| | - Kristian Steen Frederiksen
- Department of Neurology, Danish Dementia Research Centre (DDRC), University of Copenhagen, Rigshospitalet, Denmark
| |
Collapse
|
26
|
Huuha AM, Norevik CS, Moreira JBN, Kobro-Flatmoen A, Scrimgeour N, Kivipelto M, Van Praag H, Ziaei M, Sando SB, Wisløff U, Tari AR. Can exercise training teach us how to treat Alzheimer's disease? Ageing Res Rev 2022; 75:101559. [PMID: 34999248 DOI: 10.1016/j.arr.2022.101559] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 01/02/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and there is currently no cure. Novel approaches to treat AD and curb the rapidly increasing worldwide prevalence and costs of dementia are needed. Physical inactivity is a significant modifiable risk factor for AD, estimated to contribute to 12.7% of AD cases worldwide. Exercise interventions in humans and animals have shown beneficial effects of exercise on brain plasticity and cognitive functions. In animal studies, exercise also improved AD pathology. The mechanisms underlying these effects of exercise seem to be associated mainly with exercise performance or cardiorespiratory fitness. In addition, exercise-induced molecules of peripheral origin seem to play an important role. Since exercise affects the whole body, there likely is no single therapeutic target that could mimic all the benefits of exercise. However, systemic strategies may be a viable means to convey broad therapeutic effects in AD patients. Here, we review the potential of physical activity and exercise training in AD prevention and treatment, shining light on recently discovered underlying mechanisms and concluding with a view on future development of exercise-free treatment strategies for AD.
Collapse
Affiliation(s)
- Aleksi M Huuha
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Cecilie S Norevik
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - José Bianco N Moreira
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Asgeir Kobro-Flatmoen
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, and Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway; K.G. Jebsen Centre for Alzheimer's Disease, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nathan Scrimgeour
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Miia Kivipelto
- Karolinska Institute, Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Stockholm, Sweden; Karolinska University Hospital, Theme Aging and Inflammation, Stockholm, Sweden
| | - Henriette Van Praag
- Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, United States
| | - Maryam Ziaei
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, and Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway; Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Sigrid Botne Sando
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ulrik Wisløff
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Atefe R Tari
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| |
Collapse
|
27
|
Erickson KI, Donofry SD, Sewell KR, Brown BM, Stillman CM. Cognitive Aging and the Promise of Physical Activity. Annu Rev Clin Psychol 2022; 18:417-442. [PMID: 35044793 DOI: 10.1146/annurev-clinpsy-072720-014213] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Is the field of cognitive aging irretrievably concerned with decline and deficits, or is it shifting to emphasize the hope of preservation and enhancement of cognitive function in late life? A fragment of an answer comes from research attempting to understand the reasons for individual variability in the extent and rate of cognitive decline. This body of work has created a sense of optimism based on evidence that there are some health behaviors that amplify cognitive performance or mitigate the rate of age-related cognitive decline. In this context, we discuss the role of physical activity on neurocognitive function in late adulthood and summarize how it can be conceptualized as a constructive approach both for the maintenance of cognitive function and as a therapeutic for enhancing or optimizing cognitive function in late life. In this way, physical activity research can be used to shape perceptions of cognitive aging. Expected final online publication date for the Annual Review of Clinical Psychology, Volume 18 is May 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kirk I Erickson
- Department of Psychology and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; .,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia.,PROFITH "PROmoting FITness and Health through physical activity" Research Group, Department of Physical Education and Sports, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - Shannon D Donofry
- Department of Psychology and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; .,Psychiatry and Behavioral Health Institute, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Kelsey R Sewell
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Belinda M Brown
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Chelsea M Stillman
- Department of Psychology and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| |
Collapse
|
28
|
Ma C, Lin M, Gao J, Xu S, Huang L, Zhu J, Huang J, Tao J, Chen L. The impact of physical activity on blood inflammatory cytokines and neuroprotective factors in individuals with mild cognitive impairment: a systematic review and meta-analysis of randomized-controlled trials. Aging Clin Exp Res 2022; 34:1471-1484. [PMID: 35025094 DOI: 10.1007/s40520-021-02069-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/27/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Accumulated evidence has proved that both neuroinflammation and neuroprotection existing at the stage of mild cognitive impairment (MCI) may mediate its progression, which can conversely be modulated by physical activity (PA). However, further research is needed to clarify which factors are involved in that process. OBJECTIVES To identify the impact of PA on inflammatory cytokines and neuroprotective factors in individuals with MCI. METHODS Four databases [PubMed, Cochrane Library, Cochrane Library (Trials), Embase and Web of Science Core Collection] were searched from their inception to October 2021 for randomized-controlled trials (RCTs) assessing the biochemical effect of PA on biomarkers in participants with MCI. Pooled effect size was calculated by the standardized mean difference (SMD). RESULTS A total of 13 RCTs involving 514 participants by reporting 8 inflammatory cytokines [tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, -6, -8, -10, -15, C-reactive protein (CRP) and interferon-γ (IFN-γ) and 5 neuroprotective factors (brain-derived neurotrophic factor (BDNF), insulin-like growth factor (IGF-1), vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2), irisin] were included. The meta-analysis showed that PA had positive effects on decreasing TNF-α (SMD = - 0.32, 95% CI - 0.58 to 0.07, p = 0.01; I2 = 32%) and CRP (SMD = - 0.68, 95% CI - 1.05 to 0.32, p = 0.0002; I2 = 18%), while significantly improving BDNF (SMD = 0.32, 95% CI 0.09-0.56, p = 0.007; I2 = 42%) and IGF-1 (SMD = 0.42, 95% CI 0.03-0.81, p = 0.03; I2 = 0%). CONCLUSION PA had a certain effect on inhibiting inflammatory cytokines but promoting neuroprotective factors in individuals with MCI which may provide a possible explanation for the potential molecular mechanism of PA on cognitive improvement.
Collapse
Affiliation(s)
- Chuyi Ma
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Miaoran Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jiahui Gao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Shurui Xu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Li Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jingfang Zhu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jia Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Lidian Chen
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology & Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| |
Collapse
|
29
|
Multiple Applications of Different Exercise Modalities with Rodents. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3898710. [PMID: 34868454 PMCID: PMC8639251 DOI: 10.1155/2021/3898710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/14/2021] [Accepted: 11/12/2021] [Indexed: 12/29/2022]
Abstract
A large proportion of chronic diseases can be derived from a sedentary lifestyle. Raising physical activity awareness is indispensable, as lack of exercise is the fourth most common cause of death worldwide. Animal models in different research fields serve as important tools in the study of acute or chronic noncommunicable disorders. With the help of animal-based exercise research, exercise-mediated complex antioxidant and inflammatory pathways can be explored, which knowledge can be transferred to human studies. Whereas sustained physical activity has an enormous number of beneficial effects on many organ systems, these animal models are easily applicable in several research areas. This review is aimed at providing an overall picture of scientific research studies using animal models with a focus on different training modalities. Without wishing to be exhaustive, the most commonly used forms of exercise are presented.
Collapse
|
30
|
Chu-Tan JA, Kirkby M, Natoli R. Running to save sight: The effects of exercise on retinal health and function. Clin Exp Ophthalmol 2021; 50:74-90. [PMID: 34741489 DOI: 10.1111/ceo.14023] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/29/2022]
Abstract
The benefits of exercise to human health have long been recognised. However, only in the past decade have researchers started to discover the molecular benefits that exercise confers, especially to the central nervous system (CNS). These discoveries include the magnitude of molecular messages that are communicated from skeletal muscle to the CNS. Despite these advances in understanding, very limited studies have been conducted to decipher the molecular benefits of exercise in retinal health and disease. Here, we review the latest work on the effects of exercise on the retina and discuss its effects on the wider CNS, with a focus on demonstrating the potential applicability and comparative molecular mechanisms that may be occurring in the retina. This review covers the key molecular pathways where exercise exerts its effects: oxidative stress and mitochondrial health; inflammation; protein aggregation; neuronal health; and tissue crosstalk via extracellular vesicles. Further research on the benefits of exercise to the retina and its molecular messages within extracellular vesicles is highly topical in this field.
Collapse
Affiliation(s)
- Joshua A Chu-Tan
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia.,The Australian National University Medical School, The Australian National University, Acton, Australia
| | - Max Kirkby
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Acton, Australia.,The Australian National University Medical School, The Australian National University, Acton, Australia
| |
Collapse
|
31
|
Memel M, Buchman AS, Bennett DA, Casaletto K. Relationship between objectively measured physical activity on neuropathology and cognitive outcomes in older adults: Resistance versus resilience? ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12245. [PMID: 34692982 PMCID: PMC8515358 DOI: 10.1002/dad2.12245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Physical activity (PA) is associated with better cognitive and brain health. However, it remains unclear whether PA relates to accumulation of disease pathology ("resistance") or indirectly moderates adverse effects of pathology on cognition ("cognitive resilience"). METHODS Five hundred thirteen Rush Memory and Aging Project (MAP) decedents completed longitudinal actigraphy monitoring, cognitive testing, and neuropathological examination. Cross-sectional models tested the relationship between average PA and pathology, and the moderating effect of baseline PA on the association between pathology and cognition. Longitudinal models examined whether changes in PA moderated associations between pathology and cognition. RESULTS PA was negatively associated with Lewy body disease (LBD), but positively associated with Alzheimer's disease (AD) burdens. Baseline PA attenuated the association between cerebrovascular pathology and cognition, whereas longitudinal change in PA attenuated associations between AD, cerebral amyloid angiopathy, TAR DNA-binding protein 43, and atherosclerosis on cognitive decline. DISCUSSION Whereas PA relates to "cognitive resilience" against cerebrovascular disease, AD, and other neuropathologies, "resistance" effects were limited.
Collapse
Affiliation(s)
- Molly Memel
- San Francisco VA Medical CenterSan FranciscoCaliforniaUSA
- UCSF Memory and Aging CenterSan FranciscoCaliforniaUSA
| | - Aron S. Buchman
- Rush University Medical Center–Rush Alzheimer's Disease CenterChicagoIllinoisUSA
| | - David A. Bennett
- Rush University Medical Center–Rush Alzheimer's Disease CenterChicagoIllinoisUSA
| | | |
Collapse
|
32
|
Vasconcelos-Filho FSL, da Rocha Oliveira LC, de Freitas TBC, de Pontes PADS, Rocha-E-Silva RCD, Godinho WDN, Chaves EMC, da Silva CGL, Soares PM, Ceccatto VM. Effect of involuntary chronic physical exercise on beta-amyloid protein in experimental models of Alzheimer's disease: Systematic review and meta-analysis. Exp Gerontol 2021; 153:111502. [PMID: 34339821 DOI: 10.1016/j.exger.2021.111502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 07/03/2021] [Accepted: 07/21/2021] [Indexed: 11/19/2022]
Abstract
The excessive deposition of β-amyloid proteins (Aβ) is directly correlated with the establishment and development of Alzheimer's Disease (AD). Current treatments for AD only reduce symptoms instead of acting on Aβ, the primary etiological agent. Hence, the anti-amyloid effect of regular exercise has been widely investigated as an alternative therapy. This systematic review and meta-analysis examined the anti-amyloid effect of regular physical exercise in animal models of AD. The search was conducted on the electronic databases Pubmed, Embase, Scopus and Web of Science without data limitation and using the following describers: "amyloid beta" (OR senile plaque OR amyloid plaque) and "exercise" (OR physical activity OR training). The risk of bias was evaluated using the SYRCLE's tool. Meta-analyses were conducted using models of random continuous effects. A total of 36 studies were selected and most used: transgenic mice (n = 29), treadmill training, duration of 12 weeks (interval of 4 to 28 weeks), rate of 60 min/day (interval of 30 min and up until free access) and speed of 12 m/min (interval of 3.2 to 32 m/min). The hippocampus and cortex were the most frequently investigated regions. Meta-analysis demonstrated a decrease in Aβ with greater effect in unspecified isoforms Meta-analysis demonstrated a decrease in Aβ with greater effect in unspecified isoforms (N = 4; SMD = -2.71, IC 95%: -3.59, -1.84, p < 0.00001, Q2 = 3.38, I2 = 11%) and Aβ1-42 (N = 21; SMD = -1.94, IC 95%: -2.37, -1.51, p < 0.00001, Q2 = 33,37, I2 = 40%). Concerning training, greater effect was found with: 1) swimming (N = 4; SMD = -1.98, IC 95%: -3,28 - -0,68, p = 0.003, Q2 = 9.74, I2 = 69%), 2) moderate intensity (N = 4; SMD = -2.03, IC 95%: -3.31 - -0.75, p < 0.005, Q2 = 12.68, I2 = 76%); 3) duration up to six weeks (N = 6; N = 6; SMD = -2.35, IC 95%: -3.15 - -1.55, p < 0.00001, Q2 = 8.38, I2 = 40%); 4) young animals (SMD = -2.00, IC 95%: -2.59 - -1.42, p < 0.00001, Q2 = 24.90, I2 = 52%); 5) in the amygdala region (N = 1; SMD = -8.56, IC 95%: -12.88 - -4.23, p = 0.0001) and females (N = 4; SMD = -2.14, IC 95%: -3.48 - -0.79, p = 0.002, Q2 = 10.31, I2 = 71%). However, the reduction of Aβ was associated with decrease of amyloidogenic pathway and increase of non-amyloidogenic. Hence, regular physical exercise demonstrated anti-amyloid effect in experimental models of AD through positive alterations in APP processing through different signaling pathways.
Collapse
Affiliation(s)
- Francisco Sérgio Lopes Vasconcelos-Filho
- Pró-reitoria de Cultura, Universidade Federal do Cariri, Juazeiro do Norte, Ceará, Brazil; Laboratório de Bioquímica e Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil.
| | - Lucas Christyan da Rocha Oliveira
- Departamento de Ciências da Saúde, Faculdade de Medicina, Universidade Federal Rural do Semi-árido, Mossoró, Rio Grande do Norte, Brazil
| | | | | | | | - Welton Daniel Nogueira Godinho
- Laboratório de Bioquímica e Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Edna Maria Camelo Chaves
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | | | - Paula Matias Soares
- Laboratório de Bioquímica e Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Vânia Marilande Ceccatto
- Laboratório de Bioquímica e Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
33
|
Markovic SJ, Fitzgerald M, Peiffer JJ, Scott BR, Rainey-Smith SR, Sohrabi HR, Brown BM. The impact of exercise, sleep, and diet on neurocognitive recovery from mild traumatic brain injury in older adults: A narrative review. Ageing Res Rev 2021; 68:101322. [PMID: 33737117 DOI: 10.1016/j.arr.2021.101322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/06/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023]
Abstract
Mild traumatic brain injury (mTBI) accounts for a large majority of traumatic brain injuries sustained globally each year. Older adults, who are already susceptible to age-related declines to neurocognitive health, appear to be at an increased risk of both sustaining an mTBI and experiencing slower or impaired recovery. There is also growing evidence that mTBI is a potential risk factor for accelerated cognitive decline and neurodegeneration. Lifestyle-based interventions are gaining prominence as a cost-effective means of maintaining cognition and brain health with age. Consequently, inter-individual variations in exercise, sleep, and dietary patterns could influence the trajectory of post-mTBI neurocognitive recovery, particularly in older adults. This review synthesises the current animal and human literature centred on the mechanisms through which lifestyle-related habits and behaviours could influence acute and longer-term neurocognitive functioning following mTBI. Numerous neuroprotective processes which are impacted by lifestyle factors have been established in animal models of TBI. However, the literature is characterised by a lack of translation to human samples and limited appraisal of the interaction between ageing and brain injury. Further research is needed to better establish the therapeutic utility of applying lifestyle-based modifications to improve post-mTBI neurocognitive outcomes in older adults.
Collapse
Affiliation(s)
- Shaun J Markovic
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South St, Murdoch, Western Australia, Australia; Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute Building, 8 Verdun St, Nedlands, Western Australia, Australia.
| | - Melinda Fitzgerald
- Curtin Health Innovation Research Institute, Sarich Neuroscience Research Institute Building, 8 Verdun St, Nedlands, Western Australia, Australia; Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute Building, 8 Verdun St, Nedlands, Western Australia, Australia; School of Biological Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, Western Australia, Australia
| | - Jeremiah J Peiffer
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South St, Murdoch, Western Australia, Australia; Centre for Healthy Ageing, Murdoch University, 90 South St, Murdoch, Western Australia, Australia; Murdoch Applied Sports Science Laboratory, Murdoch University, 90 South St, Murdoch, Western Australia, Australia
| | - Brendan R Scott
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South St, Murdoch, Western Australia, Australia; Centre for Healthy Ageing, Murdoch University, 90 South St, Murdoch, Western Australia, Australia; Murdoch Applied Sports Science Laboratory, Murdoch University, 90 South St, Murdoch, Western Australia, Australia
| | - Stephanie R Rainey-Smith
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute Building, 8 Verdun St, Nedlands, Western Australia, Australia; Centre for Healthy Ageing, Murdoch University, 90 South St, Murdoch, Western Australia, Australia; School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Dr, Joondalup, Western Australia, Australia; School of Psychological Science, University of Western Australia, 35 Stirling Hwy, Crawley, Western Australia, Australia
| | - Hamid R Sohrabi
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute Building, 8 Verdun St, Nedlands, Western Australia, Australia; Centre for Healthy Ageing, Murdoch University, 90 South St, Murdoch, Western Australia, Australia; School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Dr, Joondalup, Western Australia, Australia; Department of Biomedical Sciences, Macquarie University, Balaclava Rd, Macquarie Park, New South Wales, Australia
| | - Belinda M Brown
- Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, 90 South St, Murdoch, Western Australia, Australia; Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute Building, 8 Verdun St, Nedlands, Western Australia, Australia; Centre for Healthy Ageing, Murdoch University, 90 South St, Murdoch, Western Australia, Australia
| |
Collapse
|
34
|
High-intensity interval training and moderate-intensity continuous training alleviate β-amyloid deposition by inhibiting NLRP3 inflammasome activation in APPswe/PS1dE9 mice. Neuroreport 2021; 31:425-432. [PMID: 32150150 DOI: 10.1097/wnr.0000000000001429] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Recent study has demonstrated that high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) have the same effect to alleviate β-amyloid pathology in the hippocampus of APPswe/PS1dE9 (APP/PS1) mice. Activation of nucleotide binding and oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is pivotal and has been demonstrated to accelerate β-amyloid accumulation. The present study aimed to examine whether the exercise-induced β-amyloid reduction was associated with changes in NLRP3 inflammasome activation. APP/PS1 transgenic mice were randomly assigned to a transgenic sedentary group, HIIT group and MICT group. Nontransgenic littermates were used as wild-type sedentary group. Mice in HIIT and MICT groups were subjected to treadmill exercise for 12 weeks, 5 days/week. The results showed that compared with transgenic sedentary group, β-amyloid deposition in the hippocampus of HIIT and MICT groups were significantly reduced. Moreover, both HIIT and MICT groups displayed significant increases in the expression of microglial phagocytic receptors triggering receptor expressed on myeloid cells 2, CD36 and scavenger receptor class A compared with transgenic sedentary group. In addition, HIIT and MICT had the same effect to inhibit NLRP3 inflammasome activation, as evidenced by significant reduction in IL-1β, active caspase-1p20, NLRP3 and apoptosis-associated speck-like protein containing a caspase activating and recruitment domain (ASC) levels as well as decreased NLRP3/ASC colocalization. These findings indicate that HIIT appears to be an effective intervention as MICT to reduced β-amyloid deposition by regulating NLRP3 inflammasome-controlled microglial phagocytosis.
Collapse
|
35
|
Vasconcelos-Filho FSL, da Rocha Oliveira LC, de Freitas TBC, de Pontes PADS, da Rocha-E-Silva RC, Chaves EMC, da Silva CGL, Soares PM, Ceccatto VM. Neuroprotective mechanisms of chronic physical exercise via reduction of β-amyloid protein in experimental models of Alzheimer's disease: A systematic review. Life Sci 2021; 275:119372. [PMID: 33745893 DOI: 10.1016/j.lfs.2021.119372] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/27/2021] [Accepted: 03/06/2021] [Indexed: 12/09/2022]
Abstract
AIMS Alzheimer's disease (AD) is the most common irreversible chronic neurodegenerative disease. It is characterized by the abnormal accumulation of β-amyloid protein (Aβ), which triggers homeostatic breakage in several physiological systems. However, the effect of chronic exercise on the formation of Aβ as an alternative therapy has been investigated. This systematic review examines the antiamyloid effect of different types and intensities of exercise, seeking to elucidate its neuroprotective mechanisms. MAIN METHODS The research was conducted in the electronic databases Pubmed, Embase, Scopus and Web of Science, using the following descriptors: "amyloid beta" (OR senile plaque OR amyloid plaque) and "exercise" (OR physical activity OR training). The risk of bias was evaluated through SYRCLE's Risk of Bias for experimental studies. KEY FINDINGS 2268 articles were found, being 36 included in the study. A higher frequency of use of mice with genetic alterations was identified for the Alzheimer's disease (AD) model (n = 29). It was used as chronic training: treadmill running (n = 24), voluntary running wheel (n = 7), swimming (n = 4) and climbing (n = 2). The hippocampus and the cortex were the most investigated regions. However, physiological changes accompanied by the reduction of Aβ and associated with AD progression were verified. It is concluded that exercise reduces the production of Aβ in models of animals with AD. SIGNIFICANCE Nevertheless, this effect contributes to the improvement of several physiological aspects related to Aβ and that contribute to neurological impairment in AD.
Collapse
Affiliation(s)
- Francisco Sérgio Lopes Vasconcelos-Filho
- Pró-reitoria de Cultura, Universidade Federal do Cariri, Juazeiro do Norte, Ceará, Brazil; Laboratório de Bioquímica e Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil.
| | - Lucas Christyan da Rocha Oliveira
- Departamento de Ciências da Saúde, Faculdade de Medicina, Universidade Federal Rural do Semi-árido, Mossoró, Rio Grande do Norte, Brazil
| | | | | | | | - Edna Maria Camelo Chaves
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | | | - Paula Matias Soares
- Laboratório de Bioquímica e Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| | - Vânia Marilande Ceccatto
- Laboratório de Bioquímica e Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
36
|
Lambertus M, Øverberg LT, Andersson KA, Hjelden MS, Hadzic A, Haugen ØP, Storm‐Mathisen J, Bergersen LH, Geiseler S, Morland C. L-lactate induces neurogenesis in the mouse ventricular-subventricular zone via the lactate receptor HCA 1. Acta Physiol (Oxf) 2021; 231:e13587. [PMID: 33244894 DOI: 10.1111/apha.13587] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 12/17/2022]
Abstract
AIM Adult neurogenesis occurs in two major niches in the brain: the subgranular zone of the hippocampal formation and the ventricular-subventricular zone. Neurogenesis in both niches is reduced in ageing and neurological disease involving dementia. Exercise can rescue memory by enhancing hippocampal neurogenesis, but whether exercise affects adult neurogenesis in the ventricular-subventricular zone remains unresolved. Previously, we reported that exercise induces angiogenesis through activation of the lactate receptor HCA1. The aim of the present study is to investigate HCA1 -dependent effects on neurogenesis in the two main neurogenic niches. METHODS Wild-type and HCA1 knock-out mice received high intensity interval exercise, subcutaneous injections of L-lactate, or saline injections, five days per week for seven weeks. Well-established markers for proliferating cells (Ki-67) and immature neurons (doublecortin), were used to investigate neurogenesis in the subgranular zone and the ventricular-subventricular zone. RESULTS We demonstrated that neurogenesis in the ventricular-subventricular zone is enhanced by HCA1 activation: Treatment with exercise or lactate resulted in increased neurogenesis in wild-type, but not in HCA1 knock-out mice. In the subgranular zone, neurogenesis was induced by exercise in both genotypes, but unaffected by lactate treatment. CONCLUSION Our study demonstrates that neurogenesis in the two main neurogenic niches in the brain is regulated differently: Neurogenesis in both niches was induced by exercise, but only in the ventricular-subventricular zone was neurogenesis induced by lactate through HCA1 activation. This opens for a role of HCA1 in the physiological control of neurogenesis, and potentially in counteracting age-related cognitive decline.
Collapse
Affiliation(s)
- Marvin Lambertus
- Section for Pharmacology and Pharmaceutical Biosciences Department of Pharmacy The Faculty of Mathematics and Natural Sciences University of Oslo Oslo Norway
| | - Linda Thøring Øverberg
- Section for Pharmacology and Pharmaceutical Biosciences Department of Pharmacy The Faculty of Mathematics and Natural Sciences University of Oslo Oslo Norway
- Institute for Behavioural Sciences Faculty of Health Sciences OsloMet—Oslo Metropolitan University Oslo Norway
| | - Krister A. Andersson
- The Brain and Muscle Energy Group, Electron Microscopy Laboratory Institute of Oral Biology Faculty of Dentistry University of Oslo Oslo Norway
- Division of Anatomy Department of Molecular Medicine Institute of Basic Medical Sciences University of Oslo Oslo Norway
| | - Malin S. Hjelden
- Section for Pharmacology and Pharmaceutical Biosciences Department of Pharmacy The Faculty of Mathematics and Natural Sciences University of Oslo Oslo Norway
| | - Alena Hadzic
- Section for Pharmacology and Pharmaceutical Biosciences Department of Pharmacy The Faculty of Mathematics and Natural Sciences University of Oslo Oslo Norway
| | - Øyvind P. Haugen
- The Brain and Muscle Energy Group, Electron Microscopy Laboratory Institute of Oral Biology Faculty of Dentistry University of Oslo Oslo Norway
| | - Jon Storm‐Mathisen
- Division of Anatomy Department of Molecular Medicine Institute of Basic Medical Sciences University of Oslo Oslo Norway
| | - Linda Hildegard Bergersen
- The Brain and Muscle Energy Group, Electron Microscopy Laboratory Institute of Oral Biology Faculty of Dentistry University of Oslo Oslo Norway
- Center for Healthy Aging Department of Neuroscience and Pharmacology Faculty of Health Sciences University of Copenhagen Copenhagen Denmark
| | - Samuel Geiseler
- Section for Pharmacology and Pharmaceutical Biosciences Department of Pharmacy The Faculty of Mathematics and Natural Sciences University of Oslo Oslo Norway
| | - Cecilie Morland
- Section for Pharmacology and Pharmaceutical Biosciences Department of Pharmacy The Faculty of Mathematics and Natural Sciences University of Oslo Oslo Norway
- Institute for Behavioural Sciences Faculty of Health Sciences OsloMet—Oslo Metropolitan University Oslo Norway
| |
Collapse
|
37
|
McCarty MF, DiNicolantonio JJ, Lerner A. A Fundamental Role for Oxidants and Intracellular Calcium Signals in Alzheimer's Pathogenesis-And How a Comprehensive Antioxidant Strategy May Aid Prevention of This Disorder. Int J Mol Sci 2021; 22:2140. [PMID: 33669995 PMCID: PMC7926325 DOI: 10.3390/ijms22042140] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress and increased cytoplasmic calcium are key mediators of the detrimental effects on neuronal function and survival in Alzheimer's disease (AD). Pathways whereby these perturbations arise, and then prevent dendritic spine formation, promote tau hyperphosphorylation, further amplify amyloid β generation, and induce neuronal apoptosis, are described. A comprehensive program of nutraceutical supplementation, comprised of the NADPH oxidase inhibitor phycocyanobilin, phase two inducers, the mitochondrial antioxidant astaxanthin, and the glutathione precursor N-acetylcysteine, may have important potential for antagonizing the toxic effects of amyloid β on neurons and thereby aiding prevention of AD. Moreover, nutraceutical antioxidant strategies may oppose the adverse impact of amyloid β oligomers on astrocyte clearance of glutamate, and on the ability of brain capillaries to export amyloid β monomers/oligomers from the brain. Antioxidants, docosahexaenoic acid (DHA), and vitamin D, have potential for suppressing microglial production of interleukin-1β, which potentiates the neurotoxicity of amyloid β. Epidemiology suggests that a health-promoting lifestyle, incorporating a prudent diet, regular vigorous exercise, and other feasible measures, can cut the high risk for AD among the elderly by up to 60%. Conceivably, complementing such lifestyle measures with long-term adherence to the sort of nutraceutical regimen outlined here may drive down risk for AD even further.
Collapse
Affiliation(s)
| | | | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer 5262000, Israel
| |
Collapse
|
38
|
Seifi-Nahavandi B, Yaghmaei P, Ahmadian S, Ghobeh M, Ebrahim-Habibi A. Cymene consumption and physical activity effect in Alzheimer's disease model: an in vivo and in vitro study. J Diabetes Metab Disord 2021; 19:1381-1389. [PMID: 33520841 DOI: 10.1007/s40200-020-00658-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 10/07/2020] [Indexed: 12/17/2022]
Abstract
Purpose Alzheimer's disease (AD) is one of the most important neurodegenerative diseases and accompanied by the production of free radicals and inflammatory factors. Studies have shown that p-cymene has anti-inflammatory and anti-oxidant effects. Here, the effects of this compound were investigated on a rat model of AD. Methods In order to create Alzheimer's rat model, bilateral injection of Amyloid β1-42 (Aβ1-42) into rats hippocampus was performed. Both therapeutic (post-AD induction) and preventive effects of p-cymene consumption with doses of 50 and 100 mg/kg were investigated. In addition, the effects of adding short-term exercise to the process were also observed. In vitro, Aβ1-42 peptide was driven toward fibril formation and effect of p-cymene was observed on the resulting fibrils. Results Learning and memory indices in the AD rats were significantly reduced compared to the Sham group, while p-cymene consumption with both doses, as well as performing exercise counteracted AD consequences. Moreover, increased neurogenesis and reduced amyloid plaques counts were observed in treated rats. In vitro formed fibrils of Aβ1-42 were partially disaggregated in the presence of p-cymene. Discussion p-Cymene could act on this AD model via antioxidant and anti-inflammatory properties as well as direct anti-fibril effect. Conclusion p-cymene can improve AD-related disorders including memory impairment.
Collapse
Affiliation(s)
| | - Parichehreh Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shahin Ahmadian
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Maryam Ghobeh
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azadeh Ebrahim-Habibi
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Vidoni ED, Morris JK, Watts A, Perry M, Clutton J, Van Sciver A, Kamat AS, Mahnken J, Hunt SL, Townley R, Honea R, Shaw AR, Johnson DK, Vacek J, Burns JM. Effect of aerobic exercise on amyloid accumulation in preclinical Alzheimer's: A 1-year randomized controlled trial. PLoS One 2021; 16:e0244893. [PMID: 33444359 PMCID: PMC7808620 DOI: 10.1371/journal.pone.0244893] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Our goal was to investigate the role of physical exercise to protect brain health as we age, including the potential to mitigate Alzheimer's-related pathology. We assessed the effect of 52 weeks of a supervised aerobic exercise program on amyloid accumulation, cognitive performance, and brain volume in cognitively normal older adults with elevated and sub-threshold levels of cerebral amyloid as measured by amyloid PET imaging. METHODS AND FINDINGS This 52-week randomized controlled trial compared the effects of 150 minutes per week of aerobic exercise vs. education control intervention. A total of 117 underactive older adults (mean age 72.9 [7.7]) without evidence of cognitive impairment, with elevated (n = 79) or subthreshold (n = 38) levels of cerebral amyloid were randomized, and 110 participants completed the study. Exercise was conducted with supervision and monitoring by trained exercise specialists. We conducted 18F-AV45 PET imaging of cerebral amyloid and anatomical MRI for whole brain and hippocampal volume at baseline and Week 52 follow-up to index brain health. Neuropsychological tests were conducted at baseline, Week 26, and Week 52 to assess executive function, verbal memory, and visuospatial cognitive domains. Cardiorespiratory fitness testing was performed at baseline and Week 52 to assess response to exercise. The aerobic exercise group significantly improved cardiorespiratory fitness (11% vs. 1% in the control group) but there were no differences in change measures of amyloid, brain volume, or cognitive performance compared to control. CONCLUSIONS Aerobic exercise was not associated with reduced amyloid accumulation in cognitively normal older adults with cerebral amyloid. In spite of strong systemic cardiorespiratory effects of the intervention, the observed lack of cognitive or brain structure benefits suggests brain benefits of exercise reported in other studies are likely to be related to non-amyloid effects. TRIAL REGISTRATION NCT02000583; ClinicalTrials.gov.
Collapse
Affiliation(s)
- Eric D. Vidoni
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Jill K. Morris
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Amber Watts
- Department of Psychology, University of Kansas, Lawrence, KS, United States of America
| | - Mark Perry
- Department of Radiology, University of Kansas Health System, Kansas City, KS, United States of America
| | - Jon Clutton
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Angela Van Sciver
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Ashwini S. Kamat
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Jonathan Mahnken
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Suzanne L. Hunt
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Ryan Townley
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Robyn Honea
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - Ashley R. Shaw
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| | - David K. Johnson
- Department of Neurology, University of California–Davis, Sacramento, CA, United States of America
| | - James Vacek
- Department of Cardiovascular Medicine, University of Kansas Health System, Kansas City, KS, United States of America
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, United States of America
| |
Collapse
|
40
|
Exercise as Potential Therapeutic Target to Modulate Alzheimer's Disease Pathology in APOE ε4 Carriers: A Systematic Review. Cardiol Ther 2021; 10:67-88. [PMID: 33403644 PMCID: PMC8126521 DOI: 10.1007/s40119-020-00209-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease for which no effective treatment exists at present. Previous research has found that exercise reduces the risk of AD. Since the apolipoprotein E (APOE) ε4 allele increases the risk of AD and is associated with faster disease progression than the other isoforms, we aimed to highlight the impact of exercise on AD pathology in APOE ε4 carriers. This review focuses on the effect of exercise on cognitive function, dementia risk, amyloid-β (Aβ) metabolism, lipid metabolism, neuroinflammation, neurotrophic factors and vascularization in APOE ε4 carriers. We searched the literature in the PubMed electronic database using the following search terms: physical activity, exercise, aerobic fitness, training, sport, APOE4, Alzheimer's disease, AD and dementia. By cross-referencing, additional publications were identified. Selected studies required older adults to take part in an exercise intervention or to make use of self-reported physical activity questionnaires. All included studies were written and published in English between 2000 and 2020. From these studies, we conclude that exercise is a non-pharmacological treatment option for high-risk APOE ε4 carriers to ameliorate the AD pathological processes including reducing Aβ load, protecting against hippocampal atrophy, improving cognitive function, stabilizing cholesterol levels and lowering pro-inflammatory signals. Variation in study design related to age, cognitive outcomes and the type of intervention explained the differences in study outcomes. However, exercise seems to be effective in delaying the onset of AD and may improve the quality of life of AD patients.
Collapse
|
41
|
De Sousa RAL, Rodrigues CM, Mendes BF, Improta-Caria AC, Peixoto MFD, Cassilhas RC. Physical exercise protocols in animal models of Alzheimer's disease: a systematic review. Metab Brain Dis 2021; 36:85-95. [PMID: 33095371 DOI: 10.1007/s11011-020-00633-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 10/14/2020] [Indexed: 12/31/2022]
Abstract
Several animal studies have showed the beneficial effects of physical exercise (PE) on brain function and health. Alzheimer's Disease (AD) is the most common type of dementia, characterized by the presence of aggregated extracellular amyloid-beta (Aβ) and neurofibrillary tangles, with progressive cognitive decline. Therapeutic approaches such as PE showed to be effective in halting AD progression. Here, we present a systematic review about PE and AD. The search was carried out using the PubMed and LILACS databases. The following keywords were used: Alzheimer; PE; animal model. All found studies adopted aerobic exercise training as the PE protocol (100%). We identified running on treadmill as the most commonly used PE routine (62.5%). The duration of each session, intensity, frequency, and period of training most used were 60 min/day (62.5%), moderate intensity (87.5%), 5 days/week (62.5%), and 4 (37.5%) or 12 (37.5%) weeks, respectively. The AD animal models most used were the Tg APP/PS1ΔE9 (25%), models based on i.c.v. infusion of AβOs (25%) and streptozotocin (25%). All protocols used rodents to their experiments (100%), but mice were the most common (62.5%). Finally, the main results presented in all studies were capable to reduce significantly AD consequences, such as reducing Aβ or pro-inflammatory proteins levels (100%). The lack of resistance training protocols in animal models of AD indicates a huge gap that should be investigated in future studies. We suggest that PE protocols must be adapted according to the specie, lineage and life span of the animal.
Collapse
Affiliation(s)
- Ricardo Augusto Leoni De Sousa
- Multicenter Post Graduation Program in Physiological Sciences, Brazilian Society of Physiology, Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, Minas Gerais, Brazil.
- Neuroscience and Exercise Study Group (Grupo de Estudos em Neurociências e Exercício - GENE), Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, Minas Gerais, Brazil.
| | - Cíntia Maria Rodrigues
- Multicenter Post Graduation Program in Physiological Sciences, Brazilian Society of Physiology, Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, Minas Gerais, Brazil
- Fudamental Nursing Post Graduation Program, São Paulo State University, Ribeirão Preto, São Paulo, Brazil
| | - Bruno Ferreira Mendes
- Multicenter Post Graduation Program in Physiological Sciences, Brazilian Society of Physiology, Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, Minas Gerais, Brazil
| | | | - Marco Fabrício Dias Peixoto
- Multicenter Post Graduation Program in Physiological Sciences, Brazilian Society of Physiology, Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, Minas Gerais, Brazil
- Post-Graduate Program in Health Science, Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, Minas Gerais, Brazil
| | - Ricardo Cardoso Cassilhas
- Multicenter Post Graduation Program in Physiological Sciences, Brazilian Society of Physiology, Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, Minas Gerais, Brazil
- Neuroscience and Exercise Study Group (Grupo de Estudos em Neurociências e Exercício - GENE), Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, Minas Gerais, Brazil
- Post-Graduate Program in Health Science, Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, Minas Gerais, Brazil
| |
Collapse
|
42
|
Does APOE genotype moderate the relationship between physical activity, brain health and dementia risk? A systematic review. Ageing Res Rev 2020; 64:101173. [PMID: 32961338 DOI: 10.1016/j.arr.2020.101173] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/09/2020] [Accepted: 08/31/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION For decades, researchers have tried to understand the moderating effect of APOE ε4 carriage on the relationship between physical activity (PA), brain health and dementia risk. However, this field has produced inconsistent findings. METHOD We conducted a systematic review of the literature, searching for observational and interventional studies examining the effect of APOE ε4 carriage on the relationships between PA, dementia risk and different markers of brain health. RESULTS Observational studies using dementia risk as a primary outcome measure generally found that in shorter follow-up periods (up to 10 years) both APOE ε4 carriers and non-carriers benefit from PA, although longer follow-ups showed mixed results. In neuroimaging studies, mainly carriers or both groups showed benefits. Additionally, the association between PA and amyloid burden was more evident among carriers. Overall, studies with greater samples of active APOE ε4 carriers are more likely to report benefits within this group in terms of lower dementia risk and reduced brain pathology. DISCUSSION Although we have identified some patterns for the modulating effect of APOE ε4 on PA and dementia or brain pathology, the available data is, overall, inconclusive. Heterogeneity in study design, methodology, and outcomes blur the ability to detect clear associations.
Collapse
|
43
|
Dincer A, Gordon BA, Hari-Raj A, Keefe SJ, Flores S, McKay NS, Paulick AM, Shady Lewis KE, Feldman RL, Hornbeck RC, Allegri R, Ances BM, Berman SB, Brickman AM, Brooks WS, Cash DM, Chhatwal JP, Farlow MR, la Fougère C, Fox NC, Fulham MJ, Jack CR, Joseph-Mathurin N, Karch CM, Lee A, Levin J, Masters CL, McDade EM, Oh H, Perrin RJ, Raji C, Salloway SP, Schofield PR, Su Y, Villemagne VL, Wang Q, Weiner MW, Xiong C, Yakushev I, Morris JC, Bateman RJ, L S Benzinger T. Comparing cortical signatures of atrophy between late-onset and autosomal dominant Alzheimer disease. NEUROIMAGE-CLINICAL 2020; 28:102491. [PMID: 33395982 PMCID: PMC7689410 DOI: 10.1016/j.nicl.2020.102491] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/18/2020] [Accepted: 10/30/2020] [Indexed: 11/30/2022]
Abstract
Cortical signatures selective to AD could provide an early MRI biomarker. Autosomal dominant Alzheimer disease (ADAD) may model an ideal AD signature. ADAD and late-onset maps overlap in parietal cortex but contain unique features. Signatures predicted increasing amyloid within their own, but not across cohorts. These results indicate atrophy in AD can take multiple spatial patterns.
Defining a signature of cortical regions of interest preferentially affected by Alzheimer disease (AD) pathology may offer improved sensitivity to early AD compared to hippocampal volume or mesial temporal lobe alone. Since late-onset Alzheimer disease (LOAD) participants tend to have age-related comorbidities, the younger-onset age in autosomal dominant AD (ADAD) may provide a more idealized model of cortical thinning in AD. To test this, the goals of this study were to compare the degree of overlap between the ADAD and LOAD cortical thinning maps and to evaluate the ability of the ADAD cortical signature regions to predict early pathological changes in cognitively normal individuals. We defined and analyzed the LOAD cortical maps of cortical thickness in 588 participants from the Knight Alzheimer Disease Research Center (Knight ADRC) and the ADAD cortical maps in 269 participants from the Dominantly Inherited Alzheimer Network (DIAN) observational study. Both cohorts were divided into three groups: cognitively normal controls (nADRC = 381; nDIAN = 145), preclinical (nADRC = 153; nDIAN = 76), and cognitively impaired (nADRC = 54; nDIAN = 48). Both cohorts underwent clinical assessments, 3T MRI, and amyloid PET imaging with either 11C-Pittsburgh compound B or 18F-florbetapir. To generate cortical signature maps of cortical thickness, we performed a vertex-wise analysis between the cognitively normal controls and impaired groups within each cohort using six increasingly conservative statistical thresholds to determine significance. The optimal cortical map among the six statistical thresholds was determined from a receiver operating characteristic analysis testing the performance of each map in discriminating between the cognitively normal controls and preclinical groups. We then performed within-cohort and cross-cohort (e.g. ADAD maps evaluated in the Knight ADRC cohort) analyses to examine the sensitivity of the optimal cortical signature maps to the amyloid levels using only the cognitively normal individuals (cognitively normal controls and preclinical groups) in comparison to hippocampal volume. We found the optimal cortical signature maps were sensitive to early increases in amyloid for the asymptomatic individuals within their respective cohorts and were significant beyond the inclusion of hippocampus volume, but the cortical signature maps performed poorly when analyzing across cohorts. These results suggest the cortical signature maps are a useful MRI biomarker of early AD-related neurodegeneration in preclinical individuals and the pattern of decline differs between LOAD and ADAD.
Collapse
Affiliation(s)
- Aylin Dincer
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Amrita Hari-Raj
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Sarah J Keefe
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nicole S McKay
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Angela M Paulick
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kristine E Shady Lewis
- Sanders Brown Center on Aging & Alzheimer's, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Rebecca L Feldman
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ricardo Allegri
- Department of Cognitive Neurology, Neuropsychology and Neuropsychiatry, FLENI, Buenos Aires, Argentina
| | - Beau M Ances
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sarah B Berman
- Department of Neurology and Clinical & Translational Science, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain and Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - William S Brooks
- Neuroscience Research Australia, Sydney, NSW, Australia; Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - David M Cash
- Dementia Research Centre and UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martin R Farlow
- Department of Neurology, Department of Radiology and Imaging Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christian la Fougère
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital of Tübingen, Tübingen, Germany
| | - Nick C Fox
- Dementia Research Centre and UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Michael J Fulham
- Department of Molecular Imaging, Royal Prince Alfred Hospital and University of Sydney, Sydney, NSW, Australia
| | | | - Nelly Joseph-Mathurin
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Celeste M Karch
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Athene Lee
- Department of Psychiatry and Human Behavior, Department of Neurology, Butler Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Eric M McDade
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hwamee Oh
- Department of Psychiatry and Human Behavior, Department of Neurology, Butler Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Richard J Perrin
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Cyrus Raji
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stephen P Salloway
- Department of Psychiatry and Human Behavior, Department of Neurology, Butler Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Victor L Villemagne
- Department of Molecular Imaging and Therapy, Department of Medicine, Austin Health, University of Melbourne, Melbourne, VIC, Australia
| | - Qing Wang
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael W Weiner
- Department of Radiology and Biomedical Imaging, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Chengjie Xiong
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Igor Yakushev
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - John C Morris
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Randall J Bateman
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Department of Neurology, Department of Psychiatry, Department of Pathology and Immunology, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, USA.
| | | |
Collapse
|
44
|
Falkenhain K, Ruiz-Uribe NE, Haft-Javaherian M, Ali M, Michelucci PE, Schaffer CB, Bracko O. A pilot study investigating the effects of voluntary exercise on capillary stalling and cerebral blood flow in the APP/PS1 mouse model of Alzheimer's disease. PLoS One 2020; 15:e0235691. [PMID: 32857763 PMCID: PMC7455035 DOI: 10.1371/journal.pone.0235691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/19/2020] [Indexed: 11/18/2022] Open
Abstract
Exercise exerts a beneficial effect on the major pathological and clinical symptoms associated with Alzheimer’s disease in humans and mouse models of the disease. While numerous mechanisms for such benefits from exercise have been proposed, a clear understanding of the causal links remains elusive. Recent studies also suggest that cerebral blood flow in the brain of both Alzheimer’s patients and mouse models of the disease is decreased and that the cognitive symptoms can be improved when blood flow is restored. We therefore hypothesized that the mitigating effect of exercise on the development and progression of Alzheimer’s disease may be mediated through an increase in the otherwise reduced brain blood flow. To test this idea, we performed a pilot study to examine the impact of three months of voluntary wheel running in a small cohort of ~1-year-old APP/PS1 mice on short-term memory function, brain inflammation, amyloid deposition, and baseline cerebral blood flow. Our findings that exercise led to a trend toward improved spatial short-term memory, reduced brain inflammation, markedly increased neurogenesis in the dentate gyrus, and a reduction in hippocampal amyloid-beta deposits are consistent with other reports on the impact of exercise on the progression of Alzheimer’s related symptoms in mouse models. Notably, we did not observe any impact of wheel running on overall baseline blood flow nor on the incidence of non-flowing capillaries, a mechanism we recently identified as one contributing factor to cerebral blood flow deficits in mouse models of Alzheimer’s disease. Overall, our findings add to the emerging picture of differential effects of exercise on cognition and blood flow in Alzheimer’s disease pathology by showing that capillary stalling is not decreased following exercise.
Collapse
Affiliation(s)
- Kaja Falkenhain
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
- Institute of Cognitive Science, Osnabrück University, Osnabrück, Germany
| | - Nancy E. Ruiz-Uribe
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Mohammad Haft-Javaherian
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Muhammad Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | | | | | - Chris B. Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
- * E-mail:
| |
Collapse
|
45
|
Ewers M, Biechele G, Suárez-Calvet M, Sacher C, Blume T, Morenas-Rodriguez E, Deming Y, Piccio L, Cruchaga C, Kleinberger G, Shaw L, Trojanowski JQ, Herms J, Dichgans M, Brendel M, Haass C, Franzmeier N. Higher CSF sTREM2 and microglia activation are associated with slower rates of beta-amyloid accumulation. EMBO Mol Med 2020; 12:e12308. [PMID: 32790063 PMCID: PMC7507349 DOI: 10.15252/emmm.202012308] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/03/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Microglia activation is the brain's major immune response to amyloid plaques in Alzheimer's disease (AD). Both cerebrospinal fluid (CSF) levels of soluble TREM2 (sTREM2), a biomarker of microglia activation, and microglia PET are increased in AD; however, whether an increase in these biomarkers is associated with reduced amyloid-beta (Aβ) accumulation remains unclear. To address this question, we pursued a two-pronged translational approach. Firstly, in non-demented and demented individuals, we tested CSF sTREM2 at baseline to predict (i) amyloid PET changes over ∼2 years and (ii) tau PET cross-sectionally assessed in a subset of patients. We found higher CSF sTREM2 associated with attenuated amyloid PET increase and lower tau PET. Secondly, in the AppNL-G-F mouse model of amyloidosis, we studied baseline 18 F-GE180 microglia PET and longitudinal amyloid PET to test the microglia vs. Aβ association, without any confounding co-pathologies often present in AD patients. Higher microglia PET at age 5 months was associated with a slower amyloid PET increase between ages 5-to-10 months. In conclusion, higher microglia activation as determined by CSF sTREM2 or microglia PET shows protective effects on subsequent amyloid accumulation.
Collapse
Affiliation(s)
- Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Christian Sacher
- Department of Nuclear Medicine, University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
| | - Tanja Blume
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Estrella Morenas-Rodriguez
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Yuetiva Deming
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Carlos Cruchaga
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Gernot Kleinberger
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,ISAR Bioscience GmbH, Planegg, Germany
| | - Leslie Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital Munich, Ludwig Maximilian University Munich, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany
| |
Collapse
|
46
|
Frederiksen KS, Gjerum L, Waldemar G, Hasselbalch SG. Physical Activity as a Moderator of Alzheimer Pathology: A Systematic Review of Observational Studies. Curr Alzheimer Res 2020; 16:362-378. [PMID: 30873924 DOI: 10.2174/1567205016666190315095151] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 02/12/2019] [Accepted: 03/13/2019] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Observational studies have found that physical activity is associated with a reduced risk of cognitive decline and dementia. Whether physical activity may also reduce the level of AD pathology, remains undetermined. OBJECTIVE To examine the relationship between physical activity and AD biomarkers (beta-amyloid1- 42, total tau and phosphorylated tau in CSF, amyloid PET, hippocampal atrophy on MRI and parietotemporal hypometabolism on brain 18F-FDG-PET). METHODS We carried out a systematic review of the observational studies of physical activity and AD biomarkers in healthy subjects, subjective cognitive complaints, mild cognitive impairment (MCI) and AD dementia. RESULTS We identified a total of 40 papers, which were eligible for inclusion. Thirty-four studies were conducted on healthy subjects, 3 on MCI and healthy subjects, 1 on MCI, and 2 on AD and healthy controls. Six studies reported on CSF biomarkers, 9 on amyloid PET, 29 on MRI and 4 on brain 18FFDG- PET. The majority of studies did not find a significant association between physical activity and AD biomarkers. CONCLUSION The quality of included studies with only a few longitudinal studies, limits the conclusions which may be drawn from the present findings especially regarding the biomarkers other than hippocampal volume. However, the majority of the identified studies did not find a significant association.
Collapse
Affiliation(s)
- Kristian Steen Frederiksen
- Danish Dementia Research Centre, Section 6911, Department of Neurology, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark
| | - Le Gjerum
- Danish Dementia Research Centre, Section 6911, Department of Neurology, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Section 6911, Department of Neurology, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Danish Dementia Research Centre, Section 6911, Department of Neurology, Rigshospitalet, University of Copenhagen, 9 Blegdamsvej, DK-2100 Copenhagen, Denmark
| |
Collapse
|
47
|
Smith TO, Mistry D, Lee H, Dosanji S, Finnegan S, Fordham B, Nichols VP, Sheehan B, Lamb SE. Moderators of Cognitive Outcomes from an Exercise Program in People with Mild to Moderate Dementia. J Am Geriatr Soc 2020; 68:2095-2100. [PMID: 32441331 DOI: 10.1111/jgs.16552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/26/2020] [Accepted: 04/29/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND/OBJECTIVES Our aim was to estimate whether baseline participant variables were able to moderate the effect of an exercise intervention on cognition in patients with mild to moderate dementia. DESIGN Subgroup analysis of a multicenter pragmatic randomized controlled trial. SETTING Community-based gym/rehabilitation centers. PARTICIPANTS A total of 494 community-dwelling participants with mild to moderate dementia. INTERVENTION Participants were randomized to a moderate- to high-intensity aerobic and strength exercise program or a usual care control group. Experimental group participants attended twice weekly 60- to 90-minute gym sessions for 4 months. Participants were prescribed home exercises for an additional hour per week during the supervised period and 150 minutes each week after the supervised period. MEASUREMENTS Multilevel regression model analyses were undertaken to identify individual moderators of cognitive function measured through the Alzheimer Disease Assessment Scale-Cognitive Subscale score at 12 months. RESULTS When tested for a formal interaction effect, only cognitive function assessed by the baseline number cancellation test demonstrated a statistically significant interaction effect (-2.7 points; 95% confidence interval = -5.14 to -0.21). CONCLUSION People with worse number cancellation test scores may experience greater progression of cognitive decline in response to a moderate- to high-intensity exercise program. Further analyses to examine whether these findings can be replicated in planned sufficiently powered analyses are indicated.
Collapse
Affiliation(s)
- Toby O Smith
- Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Dipesh Mistry
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Hopin Lee
- Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | | | - Susanne Finnegan
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Bethany Fordham
- Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Vivien P Nichols
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Bart Sheehan
- Arden Mental health Acute Team, Coventry and Warwickshire Partnership Trust, Coventry, UK
| | - Sarah E Lamb
- Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK.,College of Medicine and Health Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
48
|
Thomas R, Zimmerman SD, Yuede KM, Cirrito JR, Tai LM, Timson BF, Yuede CM. Exercise Training Results in Lower Amyloid Plaque Load and Greater Cognitive Function in an Intensity Dependent Manner in the Tg2576 Mouse Model of Alzheimer's Disease. Brain Sci 2020; 10:brainsci10020088. [PMID: 32046299 PMCID: PMC7071605 DOI: 10.3390/brainsci10020088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/27/2020] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Three months of exercise training (ET) decreases soluble Aβ40 and Aβ42 levels in an intensity dependent manner early in life in Tg2576 mice (Moore et al., 2016). Here, we examined the effects of 12 months of low- and high- intensity exercise training on cognitive function and amyloid plaque load in the cortex and hippocampus of 15-month-old Tg2576 mice. Low- (LOW) and high- (HI) intensity ET animals ran at speeds of 15 m/min on a level treadmill and 32 m/min at a 10% grade, respectively, for 60 min/day, five days/week, from 3 to 15 months of age. Sedentary mice (SED) were placed on a level, non-moving, treadmill for the same duration. ET mice demonstrated a significantly lower amyloid plaque load in the cortex and hippocampus that was intensity dependent. Improvement in cognitive function, assessed by Morris Water Maze and Novel Object Recognition tests, was greater in the HI group compared to the LOW and SED groups. LOW mice performed better in the initial latency to the platform location during the probe trial of the Morris Water Maze (MWM) test than SED, but not in any other aspect of MWM or the Novel Object Recognition test. The results of this study indicate that exercise training decreases amyloid plaque load in an intensity dependent manner and that high-intensity exercise training improves cognitive function relative to SED mice, but the intensity of the LOW group was below the threshold to demonstrate robust improvement in cognitive function in Tg2576 mice.
Collapse
Affiliation(s)
- Riya Thomas
- Department of Biomedical Sciences, Missouri State University, Springfield, MO 65897, USA; (R.T.); (S.D.Z.); (B.F.T.)
| | - Scott D. Zimmerman
- Department of Biomedical Sciences, Missouri State University, Springfield, MO 65897, USA; (R.T.); (S.D.Z.); (B.F.T.)
| | - Kayla M. Yuede
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; (K.M.Y.); (J.R.C.)
| | - John R. Cirrito
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; (K.M.Y.); (J.R.C.)
- Hope Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Benjamin F. Timson
- Department of Biomedical Sciences, Missouri State University, Springfield, MO 65897, USA; (R.T.); (S.D.Z.); (B.F.T.)
| | - Carla M. Yuede
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; (K.M.Y.); (J.R.C.)
- Hope Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Correspondence: ; Tel.: +1-314-362-9001
| |
Collapse
|
49
|
Hippocampal Growth Factor and Myokine Cathepsin B Expression following Aerobic and Resistance Training in 3xTg-AD Mice. Int J Chronic Dis 2020; 2020:5919501. [PMID: 32090058 PMCID: PMC7011393 DOI: 10.1155/2020/5919501] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 11/18/2022] Open
Abstract
Aerobic training (AT) can support brain health in Alzheimer's disease (AD); however, the role of resistance training (RT) in AD is not well established. Aside from direct effects on the brain, exercise may also regulate brain function through secretion of muscle-derived myokines. Aims. This study examined the effects of AT and RT on hippocampal BDNF and IGF-1 signaling, β-amyloid expression, and myokine cathepsin B in the triple transgenic (3xTg-AD) model of AD. 3xTg-AD mice were assigned to one of the following groups: sedentary (Tg), aerobic trained (Tg+AT, 9 wks treadmill running), or resistance trained (Tg+RT, 9 wks weighted ladder climbing) (n = 10/group). Rotarod latency and strength were assessed pre- and posttraining. Hippocampus and skeletal muscle were collected after training and analyzed by high-resolution respirometry, ELISA, and immunoblotting. Tg+RT showed greater grip strength than Tg and Tg+AT at posttraining (p < 0.01). Only Tg+AT improved rotarod peak latency (p < 0.01). Hippocampal IGF-1 concentration was ~15% greater in Tg+AT and Tg+RT compared to Tg (p < 0.05); however, downstream signals of p-IGF-1R, p-Akt, p-MAPK, and p-GSK3β were not altered. Cathepsin B, hippocampal p-CREB and BDNF, and hippocampal mitochondrial respiration were not affected by AT or RT. β-Amyloid was ~30% lower in Tg+RT compared to Tg (p < 0.05). This data suggests that regular resistance training reduces β-amyloid in the hippocampus concurrent with increased concentrations of IGF-1. Both types of training offered distinct benefits, either by improving physical function or by modifying signals in the hippocampus. Therefore, inclusion of both training modalities may address central defects, as well as peripheral comorbidities in AD.
Collapse
|
50
|
Svensson M, Andersson E, Manouchehrian O, Yang Y, Deierborg T. Voluntary running does not reduce neuroinflammation or improve non-cognitive behavior in the 5xFAD mouse model of Alzheimer's disease. Sci Rep 2020; 10:1346. [PMID: 31992814 PMCID: PMC6987124 DOI: 10.1038/s41598-020-58309-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/10/2020] [Indexed: 11/29/2022] Open
Abstract
Physical exercise has been suggested to reduce the risk of developing Alzheimer’s disease (AD) as well as ameliorate the progression of the disease. However, we recently published results from two large epidemiological studies showing no such beneficial effects on the development of AD. In addition, long-term, voluntary running in the 5xFAD mouse model of AD did not affect levels of soluble amyloid beta (Aβ), synaptic proteins or cognitive function. In this follow-up study, we investigate whether running could impact other pathological aspects of the disease, such as insoluble Aβ levels, the neuroinflammatory response and non-cognitive behavioral impairments. We investigated the effects of 24 weeks of voluntary wheel running in female 5xFAD mice (n = 30) starting at 2–3 months of age, before substantial extracellular plaque formation. Running mice developed hindlimb clasping earlier (p = 0.009) compared to sedentary controls. Further, running exacerbated the exploratory behavior in Elevated plus maze (p = 0.001) and anxiety in Open field (p = 0.024) tests. Additionally, microglia, cytokines and insoluble Aβ levels were not affected. Taken together, our findings suggest that voluntary wheel running is not a beneficial intervention to halt disease progression in 5xFAD mice.
Collapse
Affiliation(s)
- Martina Svensson
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, BMC B11, 22184, Lund, Sweden.
| | - Emelie Andersson
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, BMC B11, 22184, Lund, Sweden
| | - Oscar Manouchehrian
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, BMC B11, 22184, Lund, Sweden
| | - Yiyi Yang
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, BMC B11, 22184, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Lund University, BMC B11, 22184, Lund, Sweden.
| |
Collapse
|