1
|
da Silva EA, Faber J, Penitente AR, Fernandes J, Bertolucci PHF, Longo BM, Arida RM. Effects of resistance exercise on behavioral and molecular changes in transgenic female mice for Alzheimer's disease in early and advanced stages. Exp Neurol 2025; 388:115217. [PMID: 40089002 DOI: 10.1016/j.expneurol.2025.115217] [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: 06/27/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that affects memory and cognition, with a higher prevalence in women. Given the lack of effective treatment, physical activity stands out as a complementary approach to prevent or delay disease progression. While numerous studies on humans and animals indicate that aerobic exercise induces brain changes, the impact of resistance exercise (RE) on AD is not fully understood. OBJECTIVE This study aimed to investigate the behavioral and molecular changes induced by RE in female transgenic mice with AD at the early and advanced stages of the disease. MATERIALS AND METHODS Adult (initial phase - 7 to 8 months of age, n = 32) and adult/elderly (advanced phase - 22 to 23 months of age, n = 32) female mice (2xTg-AD) for the APPSWE/PS1dE9 mutation were subjected to a four-week RE protocol. Mobility, anxiety-like behavior, long-term memory (LTM), and depressive-like behavior were assessed. Beta-amyloid (βA) and cytokines were quantified using the ELISA technique. RESULTS There was a progressive increase in strength in both trained groups at different ages. RE reversed memory deficits only in adult AD animals and the anxiety-like behavior only in adult/elderly AD animals. RE reversed depressive-like behavior in adult and adult/elderly AD animals. RE reduced βA only in adult AD animals. RE modified the expression of several cytokines in animals in the early and advanced stage of AD. CONCLUSION RE can be a promising strategy to minimize the deleterious effects of AD; however, its effectiveness may be more limited to the early stages of the disease.
Collapse
Affiliation(s)
| | - Jean Faber
- Federal University of Sao Paulo - Neurology and Neurosurgery Department, Brazil
| | | | | | | | | | | |
Collapse
|
2
|
Lee MY, Campomayor NB, Kim HJ, Kim M. Hippocampal MCT4 as a key regulator in excessive exercise-induced cognitive impairment: involvement of neuroinflammation. Genes Genomics 2025:10.1007/s13258-025-01642-x. [PMID: 40244523 DOI: 10.1007/s13258-025-01642-x] [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: 03/13/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND As human life expectancy increases, maintaining a healthy lifestyle has become crucial. However, excessive exercise (EE) can lead to negative consequences such as muscle damage and exercise addiction. Recently, numerous reports have indicated that EE negatively impacts cognitive performance, although the exact mechanism remains unclear. OBJECTIVE This study aimed to investigate the specific mechanisms underlying cognitive alterations induced by EE. METHODS We conducted the Y-maze, Barnes maze, and Novel Object Recognition Test to assess both short-term and long-term memory, as well as object recognition ability. We then validated our findings using qRT-PCR to elucidate the underlying mechanisms. Additionally, Diclofenac (Dic), an anti-inflammatory drug, was administered to evaluate its effects on cognitive function and the results of the molecular experiments. RESULTS EE-induced mice exhibited cognitive impairments, along with elevated expression of inflammatory cytokines such as tumor necrosis factor-α, interleukin (IL) -6, and IL-1β, and downregulated monocarboxylate transporters (MCTs) like MCT4. However, animals pre-treated with Dic regained cognitive function, alongside restored levels of IL-6, IL-1β, and MCT4. CONCLUSION MCT4 plays may play a crucial role in EE-induced cognitive impairments.
Collapse
Affiliation(s)
- Min Yeong Lee
- Department of Convergence science, Sahmyook University, Hwarangro 815, Nowongu, Seoul, 01795, Republic of Korea
- Uimyung Research Institute for Neuroscience, Sahmyook University, Hwarangro 815, Nowongu, Seoul, 01795, Republic of Korea
| | - Nicole Bon Campomayor
- Department of Convergence science, Sahmyook University, Hwarangro 815, Nowongu, Seoul, 01795, Republic of Korea
- Uimyung Research Institute for Neuroscience, Sahmyook University, Hwarangro 815, Nowongu, Seoul, 01795, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Sahmyook University, Hwarangro 815, Nowongu, Seoul, 01795, Republic of Korea
| | - Mikyung Kim
- Uimyung Research Institute for Neuroscience, Sahmyook University, Hwarangro 815, Nowongu, Seoul, 01795, Republic of Korea.
- Department of Chemistry & Life science, Sahmyook University, Hwarangro 815, Nowongu, Seoul, 01795, Republic of Korea.
| |
Collapse
|
3
|
Gutierre RC, Rocha PR, Graciani AL, Coppi AA, Arida RM. Tau, amyloid, iron, oligodendrocytes ferroptosis, and inflammaging in the hippocampal formation of aged rats submitted to an aerobic exercise program. Brain Res 2025; 1850:149419. [PMID: 39725376 DOI: 10.1016/j.brainres.2024.149419] [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: 08/08/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease is a progressive neurodegenerative disease affecting memory, language, and thinking with no curative treatment. Symptoms appear gradually, and pathological brain changes may occur twenty years before the physical and psychological signs, pointing to the urgent development of preventive interventions. Physical activity has been investigated as a preventive tool to defeat the main biological features of AD: pathological amyloid protein plaques, tau tangles, myelin degeneration, and iron deposits in the brain. This work quantifies tau tangles, amyloid, iron, and ferroptosis in oligodendrocytes in the aged rat hippocampal formation and statistically correlates neuron-neuron, neuron-glia, and glia-glia crosstalk and the effect of physical exercise on it. Our results indicate that iron overload in the oligodendrocytes is an inducer of ferroptosis; physical exercise reduces inflammaging, and improves axon-myelin volume relations; tau, amyloid, iron, and hippocampal formation cells present statistical correlations. Our data suggest the beneficial effects of physical exercise in AD and a mathematical relationship between the hippocampal formation cells in sedentary and active individuals, which should be considered in human and animal studies as a guide to a better understanding of crosstalk physiology.
Collapse
Affiliation(s)
- R C Gutierre
- Almeria Institute of Integrative Science, São Paulo, Brazil.
| | - P R Rocha
- Federal University of São Paulo, Paulista School of Medicine, Department of Physiology, Laboratory of Neurophysiology, São Paulo, Brazil
| | - A L Graciani
- Federal University of São Paulo, Paulista School of Medicine, Department of Physiology, Laboratory of Neurophysiology, São Paulo, Brazil
| | - A A Coppi
- University of Bristol, Faculty of Health and Life Sciences, Bristol, United Kingdom
| | - R M Arida
- Federal University of São Paulo, Paulista School of Medicine, Department of Physiology, Laboratory of Neurophysiology, São Paulo, Brazil
| |
Collapse
|
4
|
Fuller OK, McLennan ED, Egan CL, Perera N, Terry LV, Pyun J, de Mendonca M, Telles GD, Smeuninx B, Burrows EL, Siddiqui G, Creek DJ, Scott JW, Pearen MA, Fonseka P, Nicolazzo JA, Mathivanan S, Hannan AJ, Ramm GA, Whitham M, Febbraio MA. Extracellular vesicles contribute to the beneficial effects of exercise training in APP/PS1 mice. iScience 2025; 28:111752. [PMID: 39898049 PMCID: PMC11787611 DOI: 10.1016/j.isci.2025.111752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/21/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Exercise improves cognitive function in Alzheimer's disease (AD) via mechanism that are not fully clear. Here, we first examined the effect of voluntary exercise training (VET) on energy metabolism and cognitive function in the APP/PS1 transgenic mouse (Tg) model of familial AD. Next, we profiled extracellular vesicles (EVs) and examined whether they may play a role in the protective effects of VET via intranasal administration of EVs, purified from the blood of sedentary (sEV) and/or acutely exercised (eEV) donor wild-type mice into APP/PS1Tg mice. We show that VET reduced resting energy expenditure (REE) and improved cognition in APP/PS1 Tg mice. Administration of eEV, but not sEV, also reduced REE, but had no effect on cognition. Taken together, these data show that exercise is effective intervention to improve symptoms of AD in APP/PS1Tg mice. In addition, eEVs mediate some of these effects, implicating EVs in the treatment of age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Oliver K. Fuller
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Emma D. McLennan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Casey L. Egan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Nimna Perera
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Lauren V. Terry
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Jae Pyun
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Mariana de Mendonca
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | | | - Benoit Smeuninx
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Emma L. Burrows
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Ghizal Siddiqui
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Darren J. Creek
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - John W. Scott
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | | | - Pamali Fonseka
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Joseph A. Nicolazzo
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Suresh Mathivanan
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Grant A. Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Martin Whitham
- School of Sport, Exercise & Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Mark A. Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Li D, Jia J, Zeng H, Zhong X, Chen H, Yi C. Efficacy of exercise rehabilitation for managing patients with Alzheimer's disease. Neural Regen Res 2024; 19:2175-2188. [PMID: 38488551 PMCID: PMC11034587 DOI: 10.4103/1673-5374.391308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/16/2023] [Accepted: 11/25/2023] [Indexed: 04/24/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive and degenerative neurological disease characterized by the deterioration of cognitive functions. While a definitive cure and optimal medication to impede disease progression are currently unavailable, a plethora of studies have highlighted the potential advantages of exercise rehabilitation for managing this condition. Those studies show that exercise rehabilitation can enhance cognitive function and improve the quality of life for individuals affected by AD. Therefore, exercise rehabilitation has been regarded as one of the most important strategies for managing patients with AD. Herein, we provide a comprehensive analysis of the currently available findings on exercise rehabilitation in patients with AD, with a focus on the exercise types which have shown efficacy when implemented alone or combined with other treatment methods, as well as the potential mechanisms underlying these positive effects. Specifically, we explain how exercise may improve the brain microenvironment and neuronal plasticity. In conclusion, exercise is a cost-effective intervention to enhance cognitive performance and improve quality of life in patients with mild to moderate cognitive dysfunction. Therefore, it can potentially become both a physical activity and a tailored intervention. This review may aid the development of more effective and individualized treatment strategies to address the challenges imposed by this debilitating disease, especially in low- and middle-income countries.
Collapse
Affiliation(s)
- Dan Li
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Jinning Jia
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Haibo Zeng
- Department of Pathology, Huichang County People’s Hospital, Ganzhou, Jiangxi Province, China
| | - Xiaoyan Zhong
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Chenju Yi
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong Province, China
| |
Collapse
|
6
|
Lanni I, Chiacchierini G, Papagno C, Santangelo V, Campolongo P. Treating Alzheimer's disease with brain stimulation: From preclinical models to non-invasive stimulation in humans. Neurosci Biobehav Rev 2024; 165:105831. [PMID: 39074672 DOI: 10.1016/j.neubiorev.2024.105831] [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: 05/20/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Alzheimer's disease (AD) is a severe and progressive neurodegenerative condition that exerts detrimental effects on brain function. As of now, there is no effective treatment for AD patients. This review explores two distinct avenues of research. The first revolves around the use of animal studies and preclinical models to gain insights into AD's underlying mechanisms and potential treatment strategies. Specifically, it delves into the effectiveness of interventions such as Optogenetics and Chemogenetics, shedding light on their implications for understanding pathophysiological mechanisms and potential therapeutic applications. The second avenue focuses on non-invasive brain stimulation (NiBS) techniques in the context of AD. Evidence suggests that NiBS can successfully modulate cognitive functions associated with various neurological and neuropsychiatric disorders, including AD, as demonstrated by promising findings. Here, we critically assessed recent findings in AD research belonging to these lines of research and discuss their potential impact on the clinical horizon of AD treatment. These multifaceted approaches offer hope for advancing our comprehension of AD pathology and developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Ilenia Lanni
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giulia Chiacchierini
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Costanza Papagno
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Valerio Santangelo
- Functional Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Philosophy, Social Sciences & Education, University of Perugia, Perugia, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy.
| |
Collapse
|
7
|
Yu L, Li Y, Lv Y, Gu B, Cai J, Liu QS, Zhao L. Treadmill Exercise Facilitates Synaptic Plasticity in APP/PS1 Mice by Regulating Hippocampal AMPAR Activity. Cells 2024; 13:1608. [PMID: 39404372 PMCID: PMC11475322 DOI: 10.3390/cells13191608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Accumulating evidence underscores exercise as a straightforward and cost-effective lifestyle intervention capable of mitigating the risk and slowing the emergence and progression of Alzheimer's disease (AD). However, the intricate cellular and molecular mechanisms mediating these exercise-induced benefits in AD remain elusive. The present study delved into the impact of treadmill exercise on memory retrieval performance, hippocampal synaptic plasticity, synaptic morphology, and the expression and activity of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors (AMPARs) in 6-month-old APP/PS1 mice. APP/PS1 mice (4-month-old males) were randomly assigned to either a treadmill exercise group or a sedentary group, with C57BL/6J mice (4-month-old males) as the control group (both exercise and sedentary). The exercise regimen spanned 8 weeks. Our findings revealed that 8-week treadmill exercise reversed memory retrieval impairment in step-down fear conditioning in 6-month-old APP/PS1 mice. Additionally, treadmill exercise enhanced basic synaptic strength, short-term potentiation (STP), and long-term potentiation (LTP) of the hippocampus in these mice. Moreover, treadmill exercise correlated with an augmentation in synapse numbers, refinement of synaptic structures, and heightened expression and activity of AMPARs. Our findings suggest that treadmill exercise improves behavioral performance and facilitates synaptic transmission by increasing structural synaptic plasticity and the activity of AMPARs in the hippocampus of 6-month-old APP/PS1 mice, which is involved in pre- and postsynaptic processes.
Collapse
Affiliation(s)
- Laikang Yu
- Department of Strength and Conditioning Assessment and Monitoring, Beijing Sport University, Beijing 100084, China;
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Yan Li
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Yuanyuan Lv
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
- China Institute of Sport and Health Science, Beijing Sport University, Beijing 100084, China
| | - Boya Gu
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Jiajia Cai
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Li Zhao
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing Sport University, Beijing 100084, China; (Y.L.); (Y.L.); (B.G.); (J.C.)
| |
Collapse
|
8
|
Fuller OK, McLennan ED, Egan CL, Burrows EL, Febbraio MA. Impact of voluntary exercise training on the metabolic and behavioral characteristics of the rTg4510 transgenic mouse model of frontotemporal dementia. Behav Brain Res 2024; 460:114810. [PMID: 38122903 DOI: 10.1016/j.bbr.2023.114810] [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: 07/06/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023]
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disorder that affects the frontal and temporal lobes of the brain, primarily in individuals under 65 years of age, and is the second most common form of dementia worldwide. There is no cure for FTD and current treatments offer limited symptomatic relief. Regular physical activity exhibits cognitive and neuroprotective benefits in healthy individuals and in various neurodegenerative diseases, such as Alzheimer's disease, but few studies have examined its efficacy in FTD. Accordingly, we investigated the impact of voluntary exercise training (VET) on the metabolic and behavioral characteristics of the rTg4510 transgenic mouse model of familial FTD. We show that regardless of genotype, VET increased energy expenditure, decreased sleep duration, and improved long-term memory in rTg4510 mice and WT littermates. Moreover, VET appeared to improve hyperactivity, a common feature of FTD, in rTg4510 mice. Although further work is required, these findings provide important insights into the potential benefits of physical activity in FTD.
Collapse
Affiliation(s)
- Oliver K Fuller
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Emma D McLennan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Casey L Egan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Emma L Burrows
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia.
| |
Collapse
|
9
|
Yang J, Yuan S, Jian Y, Lei Y, Hu Z, Yang Q, Yan X, Zheng L, Li J, Liu W. Aerobic exercise regulates GPR81 signal pathway and mediates complement- microglia axis homeostasis on synaptic protection in the early stage of Alzheimer's disease. Life Sci 2023; 331:122042. [PMID: 37634815 DOI: 10.1016/j.lfs.2023.122042] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
AIMS Memory impairment is a major clinical manifestation in Alzheimer's disease (AD) patients, while regular exercise may prevent and delay degenerative changes in memory functions, and our aim is to explore the influence and molecular mechanisms of aerobic exercise on the early stages of Alzheimer's disease. MAIN METHODS 3-month-old male APP/PS1 transgenic AD mice and C57BL/6J wild-type mice were randomly divided into four groups: wild-type and APP/PS1 mice with sedentary (WT-SED, AD-SED), and running (WT-RUN, AD-RUN) for 12-weeks. The spatial learning and memory function, RNA-sequencing, spine density, synaptic associated protein, mRNA and protein expression involved in G protein-coupled receptor 81 (GPR81) signaling pathway, and complement factors in brain were measured. KEY FINDINGS Aerobic exercise improved spatial learning and memory in APP/PS1 mice, potentially attributed to increased dendritic spine density. Subsequently, potential underlying mechanisms were identified through RNA sequencing: regular aerobic exercise could activate the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) cAMP/PKA signaling pathway and upregulate synaptic function-related proteins to promote synaptic growth, possibly by modulating GPR81. Notably, regular aerobic exercise inhibited microglial activation, reversed the microglial phenotype, reduced the production of initiation factor C1q and central factor C3 in the complement cascade in the brain, prevented the colocalization of microglia and PSD-95, and thus prevented synaptic loss. SIGNIFICANCE Physical exercise could play a critical role in improving cognitive function in AD by promoting synaptic growth and preventing synaptic loss, which may be related to the regulation of the GPR81/cAMP/PKA signaling pathway and inhibition of complement-mediated microglial phagocytosis of synapses.
Collapse
Affiliation(s)
- Jialun Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Shunling Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Ye Jian
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Yong Lei
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zelin Hu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Qiming Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Xinjun Yan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Lan Zheng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Jianghua Li
- College of Physical Education, Jiangxi Normal University, Nanchang 330022, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China; Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
10
|
Kose S, Kutlu MD, Kara S, Polat S, Akillioglu K. Investigation of the protective effect of long-term exercise on molecular pathways and behaviours in scopolamine induced alzheimer's disease-like condition. Brain Res 2023; 1814:148429. [PMID: 37269967 DOI: 10.1016/j.brainres.2023.148429] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/17/2023] [Accepted: 05/27/2023] [Indexed: 06/05/2023]
Abstract
Despite research, the role of exercise in treatment and prevention of neurodegenerative diseases remains unclear. Our study, investigated that protective effect of treadmill exercise on molecular pathways and cognitive behaviours in a scopolamine-induced model of Alzheimer's disease. For that purpose, male Balb/c mice subjected to exercise for 12 weeks. During the last 4 weeks of exercise, mice were given an injection of scopolamine (2 mg/kg). Following injection, open field test and Morris water maze test were used to assess emotional-cognitive behaviour. Hippocampus and prefrontal cortex of mice were isolated, and levels of BDNF, TrkB, and p-GSK3ßSer389 were assessed by western blotting, and levels of APP and Aß-40 were analysed by immunohistochemistry. In our study, scopolamine administration increased anxiety-like behaviour in open field test, while negatively affecting spatial learning and memory in Morris water maze test. We found that exercise had a protective effect against cognitive and emotional decline. Scopolamine decreased levels of p-GSK3ßSer389, BDNF in hippocampus and prefrontal cortex.Whereas TrkB decreased in hippocampus and increased in prefrontal cortex. There was an increase in p-GSK3ßSer389, BDNF, TrkB in the hippocampus, and p-GSK3ßSer389, BDNF in the prefrontal cortex in the exercise + scopolamine group. Immunohistochemical analysis showed that scopolamine administration increased APP and Aß-40 in hippocampus and prefrontal cortex in neuronal and perineuronal areas whereas Aß-40 and APP were reduced in exercise + scopolamine groups. In conclusion, long-term exercise may have a protective effect against scopolamine-induced impairments in cognitive-emotional behaviour. It can be suggested that this protective effect is mediated by increased BDNF levels and GSK3ßSer389 phosphorylation.
Collapse
Affiliation(s)
- Seda Kose
- Cukurova University Medical Faculty, Department of Physiology, Division of Neurophysiology, Adana 01330, Turkey.
| | - Meltem Donmez Kutlu
- Cukurova University Medical Faculty, Department of Physiology, Division of Neurophysiology, Adana 01330, Turkey
| | - Samet Kara
- Cukurova University Medical Faculty, Department of Histology and Embryology, Adana 01330, Turkey
| | - Sait Polat
- Cukurova University Medical Faculty, Department of Histology and Embryology, Adana 01330, Turkey
| | - Kubra Akillioglu
- Cukurova University Medical Faculty, Department of Physiology, Division of Neurophysiology, Adana 01330, Turkey
| |
Collapse
|
11
|
Félix-Soriano E, Sáinz N, Gil-Iturbe E, Castilla-Madrigal R, Celay J, Fernández-Galilea M, Pejenaute Á, Lostao MP, Martínez-Climent JA, Moreno-Aliaga MJ. Differential remodeling of subcutaneous white and interscapular brown adipose tissue by long-term exercise training in aged obese female mice. J Physiol Biochem 2023:10.1007/s13105-023-00964-2. [PMID: 37204588 DOI: 10.1007/s13105-023-00964-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 04/26/2023] [Indexed: 05/20/2023]
Abstract
Obesity exacerbates aging-induced adipose tissue dysfunction. This study aimed to investigate the effects of long-term exercise on inguinal white adipose tissue (iWAT) and interscapular brown adipose tissue (iBAT) of aged obese mice. Two-month-old female mice received a high-fat diet for 4 months. Then, six-month-old diet-induced obese animals were allocated to sedentarism (DIO) or to a long-term treadmill training (DIOEX) up to 18 months of age. In exercised mice, iWAT depot revealed more adaptability, with an increase in the expression of fatty acid oxidation genes (Cpt1a, Acox1), and an amelioration of the inflammatory status, with a favorable modulation of pro/antiinflammatory genes and lower macrophage infiltration. Additionally, iWAT of trained animals showed an increment in the expression of mitochondrial biogenesis (Pgc1a, Tfam, Nrf1), thermogenesis (Ucp1), and beige adipocytes genes (Cd137, Tbx1). In contrast, iBAT of aged obese mice was less responsive to exercise. Indeed, although an increase in functional brown adipocytes genes and proteins (Pgc1a, Prdm16 and UCP1) was observed, few changes were found on inflammation-related and fatty acid metabolism genes. The remodeling of iWAT and iBAT depots occurred along with an improvement in the HOMA index for insulin resistance and in glucose tolerance. In conclusion, long-term exercise effectively prevented the loss of iWAT and iBAT thermogenic properties during aging and obesity. In iWAT, the long-term exercise program also reduced the inflammatory status and stimulated a fat-oxidative gene profile. These exercise-induced adipose tissue adaptations could contribute to the beneficial effects on glucose homeostasis in aged obese mice.
Collapse
Affiliation(s)
- Elisa Félix-Soriano
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Neira Sáinz
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Eva Gil-Iturbe
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Rosa Castilla-Madrigal
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Jon Celay
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, University of Navarra, Pamplona, Spain
- CIBERONC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Marta Fernández-Galilea
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Álvaro Pejenaute
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - M Pilar Lostao
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - José A Martínez-Climent
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, University of Navarra, Pamplona, Spain
- CIBERONC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - María J Moreno-Aliaga
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
12
|
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: 1.0] [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
|
13
|
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
|
14
|
Guo L, Yang X, Zhang Y, Xu X, Li Y. Effect of exercise on cognitive function and synaptic plasticity in Alzheimer's disease models: A systematic review and meta-analysis. Front Aging Neurosci 2023; 14:1077732. [PMID: 36704501 PMCID: PMC9872519 DOI: 10.3389/fnagi.2022.1077732] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Cognitive decline is a central manifestation of Alzheimer's disease (AD), and its process is inseparable from changes in synaptic plasticity. The aim of this review was to summarize and evaluate the effectiveness of exercise on cognitive function and synaptic plasticity in AD animal models. Materials and methods Eligible studies were searched from PubMed, MEDLINE, EMBASE, Web of Science, and Cochrane Library from April to May 2022. The risk of bias was evaluated by Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE). The Morris water maze (MWM) test and synaptic plasticity were considered outcome measures. Data were analyzed using random-effects meta-analyses using the software Stata. Heterogeneity was examined by using I2 test. Sensitivity analysis and publication bias were also assessed. Results A total of 20 randomized controlled studies were eligible for study inclusion. Compared with controls, exercise decreased escape latency (SMD = -0.86, 95% CI: -1.21 to -0.50, P < 0.001), increased platform crossover numbers (SMD = 1.34, 95% CI: 0.57-2.11, P = 0.001) and time in the target quadrant (SMD = 1.65, 95% CI: 0.95-2.36, P < 0.001) and the expression of PSD95 (SMD = 0.73, 95% CI: 0.25-1.21, P = 0.003) in AD animals. The results of the subgroup analysis showed that exercise before AD had a greater effect on escape latency (SMD = -0.88, 95% CI: -1.25 to -0.52, P < 0.001), platform crossover numbers (SMD = 1.71, 95% CI: 1.23-2.18, P < 0.001), time in the target quadrant (SMD = 2.03, 95% CI: 1.19-2.87, P < 0.001) and the expression of PSD95 (SMD = 0.94, 95% CI: 0.19-1.69, P = 0.014) than exercise after AD. The results of the subgroup analysis also showed that treadmill running might be an appropriate exercise type. Conclusion Our findings suggested that exercise had a potential effect on improving cognitive function and synaptic plasticity. It can play a better neuroprotective role before AD. Systematic review registration PROSPERO, identifier: CRD42022328438.
Collapse
Affiliation(s)
- Linlin Guo
- College of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Xinxin Yang
- College of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Yuanyuan Zhang
- College of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Xinyi Xu
- College of Nursing, Hebei Medical University, Shijiazhuang, China,Postdoctoral Research Station in Basic Medicine, Hebei Medical University, Shijiazhuang, China,*Correspondence: Xinyi Xu ✉
| | - Yan Li
- College of Nursing, Hebei Medical University, Shijiazhuang, China,Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China,*Correspondence: Yan Li ✉
| |
Collapse
|
15
|
Azevedo CV, Hashiguchi D, Campos HC, Figueiredo EV, Otaviano SFSD, Penitente AR, Arida RM, Longo BM. The effects of resistance exercise on cognitive function, amyloidogenesis, and neuroinflammation in Alzheimer's disease. Front Neurosci 2023; 17:1131214. [PMID: 36937673 PMCID: PMC10017453 DOI: 10.3389/fnins.2023.1131214] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
With the increasing prevalence of Alzheimer's disease (AD) and difficulties in finding effective treatments, it is essential to discover alternative therapies through new approaches. In this regard, non-pharmacological therapies, such as physical exercise, have been proposed and explored for the treatment of AD. Recent studies have suggested that resistance exercise (RE) is an effective strategy for promoting benefits in memory and cognitive function, producing neuroprotective and anti-inflammatory effects, and reducing amyloid load and plaques, thereby reducing the risk, and alleviating the neurodegeneration process of AD and other types of dementia in the elderly. In addition, RE is the exercise recommended by the World Health Organization for the elderly due to its benefits in improving muscle strength and balance, and increasing autonomy and functional capacity, favoring improvements in the quality of life of the elderly population, who is more likely to develop AD and other types of dementia. In this mini-review, we discuss the impact of RE on humans affected by MCI and AD, and animal models of AD, and summarize the main findings regarding the effects of RE program on memory and cognitive functions, neurotrophic factors, Aβ deposition and plaque formation, as well as on neuroinflammation. Overall, the present review provides clinical and preclinical evidence that RE plays a role in alleviating AD symptoms and may help to understand the therapeutic potential of RE, thereby continuing the advances in AD therapies.
Collapse
Affiliation(s)
| | - Debora Hashiguchi
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte (UFRN), Natal, Brazil
| | | | | | | | - Arlete Rita Penitente
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- Escola de Medicina, Departamento de Ginecologia Obstetrícia e Propedêutica da, Universidade Federal de Ouro Preto (UFOP), Minas Gerais, Brazil
| | - Ricardo Mario Arida
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Beatriz Monteiro Longo
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- *Correspondence: Beatriz Monteiro Longo,
| |
Collapse
|
16
|
Mu L, Xia D, Cai J, Gu B, Liu X, Friedman V, Liu QS, Zhao L. Treadmill Exercise Reduces Neuroinflammation, Glial Cell Activation and Improves Synaptic Transmission in the Prefrontal Cortex in 3 × Tg-AD Mice. Int J Mol Sci 2022; 23:12655. [PMID: 36293516 PMCID: PMC9604030 DOI: 10.3390/ijms232012655] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
Physical exercise improves memory and cognition in physiological aging and Alzheimer's disease (AD), but the mechanisms remain poorly understood. Here, we test the hypothesis that Aβ oligomer accumulation, neuroinflammation, and glial cell activation may lead to disruption of synaptic transmission in the prefrontal cortex of 3 × Tg-AD Mice, resulting in impairment of learning and memory. On the other hand, treadmill exercise could prevent the pathogenesis and exert neuroprotective effects. Here, we used immunohistochemistry, western blotting, enzyme-linked immunosorbent assay, and slice electrophysiology to analyze the levels of GSK3β, Aβ oligomers (Aβ dimers and trimers), pro-inflammatory cytokines (IL-1β, IL-6, and TNFα), the phosphorylation of CRMP2 at Thr514, and synaptic currents in pyramidal neurons in the prefrontal cortex. We show that 12-week treadmill exercise beginning in three-month-old mice led to the inhibition of GSK3β kinase activity, decreases in the levels of Aβ oligomers, pro-inflammatory cytokines (IL-1β, IL-6, and TNFα), and the phosphorylation of CRMP2 at Thr514, reduction of microglial and astrocyte activation, and improvement of excitatory and inhibitory synaptic transmission of pyramidal neurons in the prefrontal cortex of 3 × Tg-AD Mice. Thus, treadmill exercise reduces neuroinflammation, glial cell activation and improves synaptic transmission in the prefrontal cortex in 3 × Tg-AD mice, possibly related to the inhibition of GSK3β kinase activity.
Collapse
Affiliation(s)
- Lianwei Mu
- Department of Exercise Physiology, Guangzhou Sport University, Guangzhou 510500, China
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Dongdong Xia
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| | - Jiajia Cai
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| | - Boya Gu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| | - Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Vladislav Friedman
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Li Zhao
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China
| |
Collapse
|
17
|
Aerobic Exercise Regulates Apoptosis through the PI3K/Akt/GSK-3β Signaling Pathway to Improve Cognitive Impairment in Alzheimer’s Disease Mice. Neural Plast 2022; 2022:1500710. [PMID: 36124291 PMCID: PMC9482542 DOI: 10.1155/2022/1500710] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/29/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Neuronal apoptosis is an important factor in the etiology of Alzheimer's disease (AD). Aerobic exercise (AE) enhances learning and memory, improves cognitive impairment, increases telomere binding protein expression, and decreases apoptosis regulators, but it remains unclear whether it can improve cognitive impairment caused by neuronal apoptosis in AD. Therefore, this study investigated whether an 8-week running table exercise intervention could reduce apoptosis and improve cognitive function in the hippocampal neurons of AD model mice. After the exercise intervention, we evaluated the learning memory ability (positioning, navigation, and spatial search) of mice using a Morris water labyrinth, Nissl staining, immunohistochemistry, and protein application to detect hippocampal PI3K/Akt/GSK-3β signaling pathway protein and hippocampal neuronal cell apoptosis protein B cell lymphoma 2 (Bcl-2) and apoptosis-promoting protein bcl-2-related X (Bax) protein expression. The results showed that aerobic exercise improved the location and spatial exploration ability of mice, increased the number of PI3K- and p-Akt-positive cells, increased the expression of PI3K, p-Akt, and bcl-2 proteins, decreased the expression of GSK-3β and Bax proteins, and increased the bcl-2/Bax ratio of mice. The results suggest that aerobic exercise can reduce apoptosis and improve cognitive function in AD mice. The molecular mechanism may involve activation of the PI3K/Akt/GSK-3β signaling pathway.
Collapse
|
18
|
Elsworthy RJ, Dunleavy C, Whitham M, Aldred S. Exercise for the prevention of Alzheimer's disease: Multiple pathways to promote non-amyloidogenic AβPP processing. AGING AND HEALTH RESEARCH 2022. [DOI: 10.1016/j.ahr.2022.100093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
19
|
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: 23] [Impact Index Per Article: 7.7] [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
|
20
|
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: 38] [Impact Index Per Article: 12.7] [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
|
21
|
Eggert S, Kins S, Endres K, Brigadski T. Brothers in arms: proBDNF/BDNF and sAPPα/Aβ-signaling and their common interplay with ADAM10, TrkB, p75NTR, sortilin, and sorLA in the progression of Alzheimer's disease. Biol Chem 2022; 403:43-71. [PMID: 34619027 DOI: 10.1515/hsz-2021-0330] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is an important modulator for a variety of functions in the central nervous system (CNS). A wealth of evidence, such as reduced mRNA and protein level in the brain, cerebrospinal fluid (CSF), and blood samples of Alzheimer's disease (AD) patients implicates a crucial role of BDNF in the progression of this disease. Especially, processing and subcellular localization of BDNF and its receptors TrkB and p75 are critical determinants for survival and death in neuronal cells. Similarly, the amyloid precursor protein (APP), a key player in Alzheimer's disease, and its cleavage fragments sAPPα and Aβ are known for their respective roles in neuroprotection and neuronal death. Common features of APP- and BDNF-signaling indicate a causal relationship in their mode of action. However, the interconnections of APP- and BDNF-signaling are not well understood. Therefore, we here discuss dimerization properties, localization, processing by α- and γ-secretase, relevance of the common interaction partners TrkB, p75, sorLA, and sortilin as well as shared signaling pathways of BDNF and sAPPα.
Collapse
Affiliation(s)
- Simone Eggert
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Tanja Brigadski
- Department of Informatics and Microsystem Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| |
Collapse
|
22
|
Cabral DF, Hinchman CA, Nunez C, Rice J, Loewenstein DA, Cahalin LP, Rundek T, Pascual-Leone A, Gomes-Osman J. Harnessing Neuroplasticity to Promote Brain Health in Aging Adults: Protocol for the MOVE-Cog Intervention Study. JMIR Res Protoc 2021; 10:e33589. [PMID: 34817393 PMCID: PMC8663452 DOI: 10.2196/33589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Background Extensive evidence supports a link between aerobic exercise and cognitive improvements in aging adults. A major limitation with existing research is the high variability in cognitive response to exercise. Our incomplete understanding of the mechanisms that influence this variability and the low adherence to exercise are critical knowledge gaps and major barriers for the systematic implementation of exercise for promoting cognitive health in aging. Objective We aimed to provide an in-person and remotely delivered intervention study protocol with the main goal of informing the knowledge gap on the mechanistic action of exercise on the brain by characterizing important mechanisms of neuroplasticity, cardiorespiratory fitness response, and genetics proposed to underlie cognitive response to exercise. Methods This is an open-label, 2-month, interventional study protocol in neurologically healthy sedentary adults. This study was delivered fully in-person and in remote options. Participants underwent a total of 30 sessions, including the screening session, 3 pretest (baseline) assessments, 24 moderate-to-vigorous aerobic exercise sessions, and 3 posttest assessments. We recruited participants aged 55 years and above, sedentary, and cognitively healthy. Primary outcomes were neuroplasticity, cognitive function, and cardiorespiratory fitness. Secondary outcomes included genetic factors, endothelium function, functional mobility and postural control, exercise questionnaires, depression, and sleep. We also explored study feasibility, exercise adherence, technology adaptability, and compliance of both in-person and remote protocols. Results The recruitment phase and data collection of this study have concluded. Results are expected to be published by the end of 2021 or in early 2022. Conclusions The data generated in these studies will introduce tangible parameters to guide the development of personalized exercise prescription models for maximal cognitive benefit in aging adults. Successful completion of the specific aims will enable researchers to acquire the appropriate expertise to design and conduct studies by testing personalized exercise interventions in person and remotely delivered, likely to be more effective at promoting cognitive health in aging adults. Trial Registration ClinicalTrials.gov NCT03804528; http://clinicaltrials.gov/ct2/show/NCT03804528 International Registered Report Identifier (IRRID) RR1-10.2196/33589
Collapse
Affiliation(s)
- Danylo F Cabral
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, United States
| | - Carrie A Hinchman
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Christina Nunez
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, United States
| | - Jordyn Rice
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, United States
| | - David A Loewenstein
- Center for Cognitive Neuroscience and Aging, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Lawrence P Cahalin
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, United States
| | - Tatjana Rundek
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States.,Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alvaro Pascual-Leone
- Department of Neurology, Harvard Medical School, Boston, MA, United States.,Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Rosindale, MA, United States.,Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Rosindale, MA, United States.,Guttmann Brain Health Institute, Barcelona, Spain
| | - Joyce Gomes-Osman
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, United States.,Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States.,Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
23
|
Kumar M, Bansal N. A Revisit to Etiopathogenesis and Therapeutic Strategies in Alzheimer's Disease. Curr Drug Targets 2021; 23:486-512. [PMID: 34792002 DOI: 10.2174/1389450122666211118125233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/05/2021] [Accepted: 09/13/2021] [Indexed: 11/22/2022]
Abstract
Dementia is a cluster of brain abnormalities that trigger progressive memory deficits and other cognitive abilities such as skills, language, or executive function. Alzheimer's disease (AD) is the foremost type of age-associated dementia that involves progressive neurodegeneration accompanied by profound cognitive deficits in advanced stages that severely hamper social or occupational abilities with or without the involvement of any other psychiatric condition. The last two decades witnessed a sharp increase (~123%) in mortality due to AD type dementia, typically owing to a very low disclosure rate (~45%) and hence, the prophylactic, as well as the therapeutic cure of AD, has been a huge challenge. Although understanding of AD pathogenesis has witnessed a remarkable growth (e.g., tauopathy, oxidative stress, lipid transport, glucose uptake, apoptosis, synaptic dysfunction, inflammation, and immune system), still a dearth of an effective therapeutic agent in the management of AD prompts the quest for newer pharmacological targets in the purview of its growing epidemiological status. Most of the current therapeutic strategies focus on modulation of a single target, e.g., inhibition of acetylcholinesterase, glutamate excitotoxicity (memantine), or nootropics (piracetam), even though AD is a multifaceted neurological disorder. There is an impedance urgency to find not only symptomatic but effective disease-modifying therapies. The present review focuses on the risk / protective factors and pathogenic mechanisms involved in AD. In addition to the existing symptomatic therapeutic approach, a diverse array of possible targets linked to pathogenic cascades have been re-investigated to envisage the pharmacotherapeutic strategies in AD.
Collapse
Affiliation(s)
- Manish Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Nitin Bansal
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University (CBLU), Bhiwani, Haryana 127021. India
| |
Collapse
|
24
|
Disentangling Mitochondria in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms222111520. [PMID: 34768950 PMCID: PMC8583788 DOI: 10.3390/ijms222111520] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a major cause of dementia in older adults and is fast becoming a major societal and economic burden due to an increase in life expectancy. Age seems to be the major factor driving AD, and currently, only symptomatic treatments are available. AD has a complex etiology, although mitochondrial dysfunction, oxidative stress, inflammation, and metabolic abnormalities have been widely and deeply investigated as plausible mechanisms for its neuropathology. Aβ plaques and hyperphosphorylated tau aggregates, along with cognitive deficits and behavioral problems, are the hallmarks of the disease. Restoration of mitochondrial bioenergetics, prevention of oxidative stress, and diet and exercise seem to be effective in reducing Aβ and in ameliorating learning and memory problems. Many mitochondria-targeted antioxidants have been tested in AD and are currently in development. However, larger streamlined clinical studies are needed to provide hard evidence of benefits in AD. This review discusses the causative factors, as well as potential therapeutics employed in the treatment of AD.
Collapse
|
25
|
Zhao N, Xu B. The beneficial effect of exercise against Alzheimer's disease may result from improved brain glucose metabolism. Neurosci Lett 2021; 763:136182. [PMID: 34418507 DOI: 10.1016/j.neulet.2021.136182] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023]
Abstract
The potential of physical exercise as an intervention for Alzheimer's disease (AD) has been extensively reported. In fact, a number of studies have highlighted improvements in β-amyloid (Aβ) peptide and hyperphosphorylated tau (p-tau) as critical mechanisms in exercise-induced beneficial neurological outcomes. However, no therapeutic management have been proven to be effective in humans. Recent evidence has shown that AD may be a metabolic disease related to glucose metabolic dysfunction in the brain. In this regard, some of the mechanisms responsible for the beneficial effects of physical exercise in the pathology of AD appear to be related to alterations in glucose metabolism. Therefore, we propose that the neuroprotective effect of physical exercise against AD through synergetic improvement in brain glucose metabolism and its pathophysiology. The novel perspective presented here partly explains the failure of Aβ/tau-based therapeutic approaches and provides evidence for brain glucose metabolism as a potential therapeutic target in AD.
Collapse
Affiliation(s)
- Na Zhao
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Bo Xu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China; College of Physical Education and Health, East China Normal University, Shanghai, China.
| |
Collapse
|
26
|
Zhu L, Fan JH, Chao FL, Zhou CN, Jiang L, Zhang Y, Luo YM, Zhang L, Xiao Q, Yang H, Zhang SS, Wu H, Tang Y. Running exercise protects spinophilin-immunoreactive puncta and neurons in the medial prefrontal cortex of APP/PS1 transgenic mice. J Comp Neurol 2021; 530:858-870. [PMID: 34585379 DOI: 10.1002/cne.25252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 11/06/2022]
Abstract
The medial prefrontal cortex (mPFC) is thought to be closely associated with emotional processes, decision making, and memory. Previous studies have identified the prefrontal cortex as one of the most vulnerable brain regions in Alzheimer's disease (AD). Running exercise has widely been recognized as a simple and effective method of physical activity that enhances brain function and slows the progression of AD. However, the effect of exercise on the mPFC of AD is unclear. To address these issues, we investigated the effects of 4 months of exercise on the numbers of spinophilin-immunoreactive puncta and neurons in the mPFC of 12-month-old APPswe/PSEN1dE9 (APP/PS1) transgenic AD model mice using stereological methods. The spatial learning and memory abilities of mice were tested using the Morris water maze. Four months of running exercise delayed declines in spatial learning and memory abilities. The stereological results showed significantly lower numbers of spinophilin-immunoreactive puncta and neurons in the mPFC of APP/PS1 mice than in the wild-type control group. The numbers of spinophilin-immunoreactive puncta and neurons in the mPFC of running APP/PS1 mice were significantly greater than those in the APP/PS1 control mice. In addition, running-induced improvements in spatial learning and memory were significantly associated with running-induced increases in spinophilin-immunoreactive puncta and neurons numbers in the mPFC. Running exercise could delay the loss of spinophilin-immunoreactive puncta and neurons in the mPFC of APP/PS1 mice. This finding might provide an important structural basis for exercise-induced improvements in the spatial learning and memory abilities of individuals with AD.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Histology and Embryology, Basic Medical College, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Jin-Hua Fan
- School of Life Sciences, Southwest University, Chongqing, P. R. China
| | - Feng-Lei Chao
- Department of Histology and Embryology, Basic Medical College, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Chun-Ni Zhou
- Department of Histology and Embryology, Basic Medical College, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Lin Jiang
- Lab Teaching & Management Center, Chongqing Medical University, Chongqing, P. R. China
| | - Yi Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Yan-Min Luo
- Department of Physiology, Chongqing Medical University, Chongqing, P. R. China
| | - Lei Zhang
- Department of Histology and Embryology, Basic Medical College, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Qian Xiao
- Department of Radioactive Medicine, Chongqing Medical University, Chongqing, P. R. China
| | - Hao Yang
- Department of Histology and Embryology, Basic Medical College, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Shan-Shan Zhang
- Department of Histology and Embryology, Basic Medical College, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Hong Wu
- Department of Histology and Embryology, Basic Medical College, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, Basic Medical College, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
27
|
Tan ZX, Dong F, Wu LY, Feng YS, Zhang F. The Beneficial Role of Exercise on Treating Alzheimer's Disease by Inhibiting β-Amyloid Peptide. Mol Neurobiol 2021; 58:5890-5906. [PMID: 34415486 DOI: 10.1007/s12035-021-02514-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is associated with a very large burden on global healthcare systems. Thus, it is imperative to find effective treatments of the disease. One feature of AD is the accumulation of neurotoxic β-amyloid peptide (Aβ). Aβ induces multiple pathological processes that are deleterious to nerve cells. Despite the development of medications that target the reduction of Aβ to treat AD, none has proven to be effective to date. Non-pharmacological interventions, such as physical exercise, are also being studied. The benefits of exercise on AD are widely recognized. Experimental and clinical studies have been performed to verify the role that exercise plays in reducing Aβ deposition to alleviate AD. This paper reviewed the various mechanisms involved in the exercise-induced reduction of Aβ, including the regulation of amyloid precursor protein cleaved proteases, the glymphatic system, brain-blood transport proteins, degrading enzymes and autophagy, which is beneficial to promote exercise therapy as a means of prevention and treatment of AD and indicates that exercise may provide new therapeutic targets for the treatment of AD.
Collapse
Affiliation(s)
- Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People's Republic of China
| | - Lin-Yu Wu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050051, People's Republic of China.
| |
Collapse
|
28
|
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
|
29
|
Mercerón-Martínez D, Ibaceta-González C, Salazar C, Almaguer-Melian W, Bergado-Rosado JA, Palacios AG. Alzheimer’s Disease, Neural Plasticity, and Functional Recovery. J Alzheimers Dis 2021; 82:S37-S50. [DOI: 10.3233/jad-201178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Alzheimer’s disease (AD) is the most common and devastating neurodegenerative condition worldwide, characterized by the aggregation of amyloid-β and phosphorylated tau protein, and is accompanied by a progressive loss of learning and memory. A healthy nervous system is endowed with synaptic plasticity, among others neural plasticity mechanisms, allowing structural and physiological adaptations to changes in the environment. This neural plasticity modification sustains learning and memory, and behavioral changes and is severely affected by pathological and aging conditions, leading to cognitive deterioration. This article reviews critical aspects of AD neurodegeneration as well as therapeutic approaches that restore neural plasticity to provide functional recoveries, including environmental enrichment, physical exercise, transcranial stimulation, neurotrophin involvement, and direct electrical stimulation of the amygdala. In addition, we report recent behavioral results in Octodon degus, a promising natural model for the study of AD that naturally reproduces the neuropathological alterations observed in AD patients during normal aging, including neuronal toxicity, deterioration of neural plasticity, and the decline of learning and memory.
Collapse
Affiliation(s)
- Daymara Mercerón-Martínez
- Experimental Electrophysiology Lab, International Center for Neurological Restoration (CIREN), Havana City, Cuba
| | | | - Claudia Salazar
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - William Almaguer-Melian
- Experimental Electrophysiology Lab, International Center for Neurological Restoration (CIREN), Havana City, Cuba
| | | | - Adrian G. Palacios
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
30
|
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: 2.5] [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
|
31
|
López-Ortiz S, Pinto-Fraga J, Valenzuela PL, Martín-Hernández J, Seisdedos MM, García-López O, Toschi N, Di Giuliano F, Garaci F, Mercuri NB, Nisticò R, Emanuele E, Lista S, Lucia A, Santos-Lozano A. Physical Exercise and Alzheimer's Disease: Effects on Pathophysiological Molecular Pathways of the Disease. Int J Mol Sci 2021; 22:ijms22062897. [PMID: 33809300 PMCID: PMC7999827 DOI: 10.3390/ijms22062897] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD), the most common form of neurodegenerative dementia in adults worldwide, is a multifactorial and heterogeneous disorder characterized by the interaction of genetic and epigenetic factors and the dysregulation of numerous intracellular signaling and cellular/molecular pathways. The introduction of the systems biology framework is revolutionizing the study of complex diseases by allowing the identification and integration of cellular/molecular pathways and networks of interaction. Here, we reviewed the relationship between physical activity and the next pathophysiological processes involved in the risk of developing AD, based on some crucial molecular pathways and biological process dysregulated in AD: (1) Immune system and inflammation; (2) Endothelial function and cerebrovascular insufficiency; (3) Apoptosis and cell death; (4) Intercellular communication; (5) Metabolism, oxidative stress and neurotoxicity; (6) DNA damage and repair; (7) Cytoskeleton and membrane proteins; (8) Synaptic plasticity. Moreover, we highlighted the increasingly relevant role played by advanced neuroimaging technologies, including structural/functional magnetic resonance imaging, diffusion tensor imaging, and arterial spin labelling, in exploring the link between AD and physical exercise. Regular physical exercise seems to have a protective effect against AD by inhibiting different pathophysiological molecular pathways implicated in AD.
Collapse
Affiliation(s)
- Susana López-Ortiz
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - Jose Pinto-Fraga
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - Pedro L. Valenzuela
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
| | - Juan Martín-Hernández
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - María M. Seisdedos
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
| | - Oscar García-López
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (N.T.); (F.G.)
- Department of Radiology, “Athinoula A. Martinos” Center for Biomedical Imaging, Boston, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Francesca Di Giuliano
- Neuroradiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (N.T.); (F.G.)
- Casa di Cura “San Raffaele Cassino”, 03043 Cassino, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy;
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Robert Nisticò
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, 00161 Rome, Italy;
- School of Pharmacy, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | | | - Simone Lista
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
- School of Pharmacy, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, 28670 Madrid, Spain; (P.L.V.); (O.G.-L.); (S.L.)
- Research Institute of the Hospital 12 de Octubre (“imas12”), 28041 Madrid, Spain
- Centro de Investigación Biomeédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), 28029 Madrid, Spain
- Correspondence:
| | - Alejandro Santos-Lozano
- i+HeALTH Research Group, Department of Health Sciences, European University Miguel de Cervantes, 47012 Valladolid, Spain; (S.L.-O.); (J.P.-F.); (J.M.-H.); (M.M.S.); (A.S.-L.)
- Research Institute of the Hospital 12 de Octubre (“imas12”), 28041 Madrid, Spain
| |
Collapse
|
32
|
Effects of Long-Term DHA Supplementation and Physical Exercise on Non-Alcoholic Fatty Liver Development in Obese Aged Female Mice. Nutrients 2021; 13:nu13020501. [PMID: 33546405 PMCID: PMC7913512 DOI: 10.3390/nu13020501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity and aging are associated to non-alcoholic fatty liver disease (NAFLD) development. Here, we investigate whether long-term feeding with a docosahexaenoic acid (DHA)-enriched diet and aerobic exercise, alone or in combination, are effective in ameliorating NAFLD in aged obese mice. Two-month-old female C57BL/6J mice received control or high fat diet (HFD) for 4 months. Then, the diet-induced obese (DIO) mice were distributed into four groups: DIO, DIO + DHA (15% dietary lipids replaced by a DHA-rich concentrate), DIO + EX (treadmill running), and DIO + DHA + EX up to 18 months. The DHA-rich diet reduced liver steatosis in DIO mice, decreasing lipogenic genes (Dgat2, Scd1, Srebp1c), and upregulated lipid catabolism genes (Hsl/Acox) expression. A similar pattern was observed in the DIO + EX group. The combination of DHA + exercise potentiated an increase in Cpt1a and Ppara genes, and AMPK activation, key regulators of fatty acid oxidation. Exercise, alone or in combination with DHA, significantly reversed the induction of proinflammatory genes (Mcp1, Il6, Tnfα, Tlr4) in DIO mice. DHA supplementation was effective in preventing the alterations induced by the HFD in endoplasmic reticulum stress-related genes (Ern1/Xbp1) and autophagy markers (LC3II/I ratio, p62, Atg7). In summary, long-term DHA supplementation and/or exercise could be helpful to delay NAFLD progression during aging in obesity.
Collapse
|
33
|
McGurran H, Glenn JM, Madero EN, Bott NT. Prevention and Treatment of Alzheimer's Disease: Biological Mechanisms of Exercise. J Alzheimers Dis 2020; 69:311-338. [PMID: 31104021 DOI: 10.3233/jad-180958] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. With an aging population and no disease modifying treatments available, AD is quickly becoming a global pandemic. A substantial body of research indicates that lifestyle behaviors contribute to the development of AD, and that it may be worthwhile to approach AD like other chronic diseases such as cardiovascular disease, in which prevention is paramount. Exercise is an important lifestyle behavior that may influence the course and pathology of AD, but the biological mechanisms underpinning these effects remain unclear. This review focuses on how exercise can modify four possible mechanisms which are involved with the pathology of AD: oxidative stress, inflammation, peripheral organ and metabolic health, and direct interaction with AD pathology. Exercise is just one of many lifestyle behaviors that may assist in preventing AD, but understanding the systemic and neurobiological mechanisms by which exercise affects AD could help guide the development of novel pharmaceutical agents and non-pharmacological personalized lifestyle interventions for at-risk populations.
Collapse
Affiliation(s)
- Hugo McGurran
- Research Master's Programme Brain and Cognitive Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Nicholas T Bott
- Neurotrack Technologies Inc., Redwood City, CA, USA.,Clinical Excellence Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Psychology, PGSP-Stanford Consortium, Palo Alto University, Palo Alto, CA, USA
| |
Collapse
|
34
|
Gil-Iturbe E, Félix-Soriano E, Sáinz N, Idoate-Bayón A, Castilla-Madrigal R, Moreno-Aliaga MJ, Lostao MP. Effect of aging and obesity on GLUT12 expression in small intestine, adipose tissue, muscle, and kidney and its regulation by docosahexaenoic acid and exercise in mice. Appl Physiol Nutr Metab 2020; 45:957-967. [PMID: 32176854 DOI: 10.1139/apnm-2019-0721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2023]
Abstract
Obesity is characterized by excessive fat accumulation and inflammation. Aging has also been characterized as an inflammatory condition, frequently accompanied by accumulation of visceral fat. Beneficial effects of exercise and n-3 long-chain polyunsaturated fatty acids in metabolic disorders have been described. Glucose transporter 12 (GLUT12) is one of the less investigated members of the GLUT family. Glucose, insulin, and tumor necrosis factor alpha (TNF-α) induce GLUT12 translocation to the membrane in muscle, adipose tissue, and intestine. We aimed to investigate GLUT12 expression in obesity and aging, and under diet supplementation with docosahexaenoic acid (DHA) alone or in combination with physical exercise in mice. Aging increased GLUT12 expression in intestine, kidney, and adipose tissue, whereas obesity reduced it. No changes on the transporter occurred in skeletal muscle. In obese 18-month-old mice, DHA further decreased GLUT12 in the 4 organs. Aerobic exercise alone did not modify GLUT12, but the changes triggered by exercise were able to prevent the DHA-diminishing effect, and almost restored GLUT12 basal levels. In conclusion, the downregulation of metabolism in aging would be a stimulus to upregulate GLUT12 expression. Contrary, obesity, an excessive energy condition, would induce GLUT12 downregulation. The combination of exercise and DHA would contribute to restore basal function of GLUT12. Novelty In small intestine, kidney and adipose tissue aging increases GLUT12 protein expression whereas obesity reduces it. Dietary DHA decreases GLUT12 in small intestine, kidney, adipose tissue and skeletal muscle. Exercise alone does not modify GLUT12 expression, nevertheless exercise prevents the DHA-diminishing effect on GLUT12.
Collapse
Affiliation(s)
- Eva Gil-Iturbe
- Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Navarra, Spain
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Navarra, Spain
| | - Elisa Félix-Soriano
- Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Navarra, Spain
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Navarra, Spain
| | - Neira Sáinz
- Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Navarra, Spain
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Navarra, Spain
| | - Adrián Idoate-Bayón
- Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Navarra, Spain
| | | | - María J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Navarra, Spain
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Navarra, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Navarra, Spain
- Institute of Health Carlos III (ISCIII), Biomedical Research Networking Center in Physiopathology of Obesity and Nutrition (CIBERobn), 28029 Madrid, Spain
| | - María Pilar Lostao
- Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Navarra, Spain
- Centre for Nutrition Research, University of Navarra, 31008 Pamplona, Navarra, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Navarra, Spain
| |
Collapse
|
35
|
Falkenhain K, Ruiz-Uribe NE, Haft-Javaherian M, Ali M, Stall Catchers, 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: 1.4] [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
|
36
|
Torromino G, Maggi A, De Leonibus E. Estrogen-dependent hippocampal wiring as a risk factor for age-related dementia in women. Prog Neurobiol 2020; 197:101895. [PMID: 32781107 DOI: 10.1016/j.pneurobio.2020.101895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
Women are more prone than men to develop age-related dementia, such as Alzheimer's disease (AD). This has been linked to the marked decrease in circulating estrogens during menopause. This review proposes to change this perspective and consider women's vulnerability to developing AD as a consequence of sex differences in the neurobiology of memory, focusing on the hippocampus. The hippocampus of cognitively impaired subjects tends to shrink with age; however, in many cases, this can be prevented by exercise or cognitive training, suggesting that if you do not use the hippocampus you lose it. We will review the developmental trajectory of sex steroids-regulated differences on the hippocampus, proposing that the overall shaping action of sex-steroids results in a lower usage of the hippocampus in females, which in turn makes them more vulnerable to the effects of ageing, the "network fragility hypothesis". To explain why women rely less on hippocampus-dependent strategies, we propose a "computational hypothesis" that is based on experimental evidence suggesting that the direct effects of estrogens on hippocampal synaptic and structural plasticity during the estrous-cycle confers instability to the memory-dependent hippocampal network. Finally, we propose to counteract AD with training and/or treatments, such as orienteering, which specifically favour the use of the hippocampus.
Collapse
Affiliation(s)
- Giulia Torromino
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy.
| |
Collapse
|
37
|
Chang LYL, Ardiles AO, Tapia-Rojas C, Araya J, Inestrosa NC, Palacios AG, Acosta ML. Evidence of Synaptic and Neurochemical Remodeling in the Retina of Aging Degus. Front Neurosci 2020; 14:161. [PMID: 32256305 PMCID: PMC7095275 DOI: 10.3389/fnins.2020.00161] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Accumulation of amyloid-beta (Aβ) peptides is regarded as the hallmark of neurodegenerative alterations in the brain of Alzheimer’s disease (AD) patients. In the eye, accumulation of Aβ peptides has also been suggested to be a trigger of retinal neurodegenerative mechanisms. Some pathological aspects associated with Aβ levels in the brain are synaptic dysfunction, neurochemical remodeling and glial activation, but these changes have not been established in the retina of animals with Aβ accumulation. We have employed the Octodon degus in which Aβ peptides accumulated in the brain and retina as a function of age. This current study investigated microglial morphology, expression of PSD95, synaptophysin, Iba-1 and choline acetyltransferase (ChAT) in the retina of juvenile, young and adult degus using immunolabeling methods. Neurotransmitters glutamate and gamma-aminobutyric acid (GABA) were detected using immunogold labeling and glutamate receptor subunits were quantified using Western blotting. There was an age-related increase in presynaptic and a decrease in post-synaptic retinal proteins in the retinal plexiform layers. Immunolabeling showed changes in microglial morphology characteristic of intermediate stages of activation around the optic nerve head (ONH) and decreasing activation toward the peripheral retina. Neurotransmitter expression pattern changed at juvenile ages but was similar in adults. Collectively, the results suggest that microglial activation, synaptic remodeling and neurotransmitter changes may be consequent to, or parallel to Aβ peptide and phosphorylated tau accumulation in the retina.
Collapse
Affiliation(s)
- Lily Y-L Chang
- School of Optometry and Vision Science, The University of Auckland, Auckland, New Zealand
| | - Alvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Cheril Tapia-Rojas
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Center for Aging and Regeneration, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Joaquin Araya
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Nibaldo C Inestrosa
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Center for Aging and Regeneration, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Adrian G Palacios
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Monica L Acosta
- School of Optometry and Vision Science, The University of Auckland, Auckland, New Zealand.,New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand.,Faculty of Medical and Health Sciences, Centre for Brain Research, The University of Auckland, Auckland, New Zealand.,Brain Research New Zealand - Rangahau Roro Aotearoa, Auckland, New Zealand
| |
Collapse
|
38
|
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: 13] [Impact Index Per Article: 2.6] [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
|
39
|
da Costa Daniele TM, de Bruin PFC, de Matos RS, de Bruin GS, Maia Chaves C, de Bruin VMS. Exercise effects on brain and behavior in healthy mice, Alzheimer's disease and Parkinson's disease model-A systematic review and meta-analysis. Behav Brain Res 2020; 383:112488. [PMID: 31991178 DOI: 10.1016/j.bbr.2020.112488] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/28/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
This systematic review and meta-analysis examines how exercise modifies brain and behavior in healthy mice, dementia (D) and Parkinson disease (PD) models. A search was performed on the Medline and Scopus electronic databases (2008-2019). Search terms were "mice", "brain", "treadmill", "exercise", "physical exercise". In the total, 430 were found but only 103 were included. Animals n = 1,172; exercised 4-8 weeks (Range 24 h to 32 weeks), 60 min/day (Range 8-120 min per day), and 10/12 m/min (Range 0.2 m/min to 36 m/min). Hippocampus, cerebral cortex, striatum and whole brain were more frequently investigated. Exercise improved learning and memory. Meta-analysis showed that exercise increased: cerebral BDNF in health (n = 150; z = 5.8, CI 3.43-12.05; p < 0.001 I2 = 94.3 %), D (n = 124; z = 4.18, CI = 2.22-9.12; p < 0.001; I2 = 93.7 %) and PD (n = 16 z = 4.26, CI 5.03-48.73 p < 0.001 I2 = 94.8 %). TrkB improved in health (n = 84 z = 5.49, CI 3.8-17.73 p < 0.001, I2 = 0.000) and PD (n = 22; z = 3.1, CI = 2.58-67.3, p < 0.002 I2 = 93.8 %). Neurogenesis increased in health (n = 68; z = 7.08, CI 5.65-21.25 p < 0.001; I2 17.58) and D model (n = 116; z = 4.18, CI 2.22-9.12 p < 0.001 I2 93.7 %). Exercise augmented amyloid clearance (n = 166; z = 7.51 CI = 4.86-14.85, p < 0.001 I2 = 58.72) and reduced amyloid plaques in D models (n = 49; z = 4.65, CI = 3.94-15.3 p < 0.001 I2 = 0.000). In conclusion, exercise improved brain and behavior, neurogenesis in healthy and dementia models, reduced toxicity and cerebral amyloid. Evidence regarding inflammation, oxidative stress and energy metabolism were scarce. Studies examining acute vs chronic exercise, extreme training and the durability of exercise benefit were rare. Vascular or glucose metabolism changes were seldom reported.
Collapse
Affiliation(s)
- Thiago Medeiros da Costa Daniele
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Ceará, Fortaleza, Brazil; Sleep and Biological Rhythms Laboratory, UFC, Brazil; Universidade Federal do Ceará (UFC), Brazil; Universidade de Fortaleza (UNIFOR).
| | - Pedro Felipe Carvalhedo de Bruin
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Ceará, Fortaleza, Brazil; Sleep and Biological Rhythms Laboratory, UFC, Brazil; Universidade Federal do Ceará (UFC), Brazil.
| | - Robson Salviano de Matos
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Ceará, Fortaleza, Brazil; Sleep and Biological Rhythms Laboratory, UFC, Brazil; Universidade Federal do Ceará (UFC), Brazil.
| | - Gabriela Sales de Bruin
- Universidade Federal do Ceará (UFC), Brazil; Department of Neurology, Washington University in St Louis, United States.
| | - Cauby Maia Chaves
- Universidade Federal do Ceará (UFC), Brazil; Departamento de Clínica Odontológica, UFC, Brazil.
| | - Veralice Meireles Sales de Bruin
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Ceará, Fortaleza, Brazil; Sleep and Biological Rhythms Laboratory, UFC, Brazil; Universidade Federal do Ceará (UFC), Brazil.
| |
Collapse
|
40
|
Martini F, Régis Leite M, Gonçalves Rosa S, Pregardier Klann I, Wayne Nogueira C. Strength exercise suppresses STZ-induced spatial memory impairment and modulates BDNF/ERK-CAMKII/CREB signalling pathway in the hippocampus of mice. Cell Biochem Funct 2020; 38:213-221. [PMID: 31978253 DOI: 10.1002/cbf.3470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 09/11/2019] [Accepted: 10/28/2019] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that has generated scientific interest because of its prevalence in the population. Studies indicate that physical exercise promotes neuroplasticity and improves cognitive function in animal models and in human beings. The aim of the present study was to investigate the effects of strength exercise on the hippocampal protein contents and memory performance in mice subjected to a model of sporadic AD induced by streptozotocin (STZ). Swiss mice received two injections of STZ (3 mg/kg, intracerebroventricular). After 21 days, they began physical training using a ladde. Mice performed this protocol for 4 weeks. After the last exercise training session, mice performed the Morris Water Maze test. The samples of hippocampus were excised and used to determine protein contents of brain-derived neurotrophic factor (BDNF), extracellular signal-regulated kinase-Ca2+ (ERK), calmodulin-dependent protein kinase (CAMKII) and cAMP-response element-binding protein (CREB) signalling pathway. Strength exercise was effective against the decrease in the time spent and distance travelled in the target quadrant by STZ-injected mice. Strength exercise was also effective against the reduction of mature BDNF, tropomyosin receptor kinase B and neuronal nuclear antigen (NeuN) hippocampal protein levels in STZ mice. The decrease in the hippocampal ratio of pERK/ERK, pCAMKII/CAMKII and pCREB/CREB induced by STZ was reversed by strength exercise. Strength exercise decreased Bax/Bcl2 ratio in the hippocampus of STZ-injected mice. The present study demonstrates that strength exercise modulated the hippocampal BDNF/ERK-CAMKII/CREB signalling pathway and suppressed STZ-induced spatial memory impairment in mice.
Collapse
Affiliation(s)
- Franciele Martini
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Marlon Régis Leite
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Suzan Gonçalves Rosa
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Isabella Pregardier Klann
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Cristina Wayne Nogueira
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| |
Collapse
|
41
|
Zeng B, Zhao G, Liu HL. The Differential Effect of Treadmill Exercise Intensity on Hippocampal Soluble Aβ and Lipid Metabolism in APP/PS1 Mice. Neuroscience 2020; 430:73-81. [PMID: 31954827 DOI: 10.1016/j.neuroscience.2020.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/02/2020] [Accepted: 01/06/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is characterized clinically by progressive impairments in learning and memory. Accumulating evidence suggests that regular exercise plays a neuroprotective role in aging-associated memory loss. Our previous study has confirmed that long-term treadmill exercise initiated either before or during the onset of β-amyloid (Aβ) pathology, was beneficial for reducing the levels of soluble Aβ and further improved cognition. In this study, in APP/PS1 mice, we assessed changes in soluble Aβ, and various blood biochemistry and molecular biological indices to assess whether exercise modulated lipid metabolism and thereby decelerated AD progression. Our results show that long-term treadmill exercise reduced the total cholesterol, triglyceride, and low-density lipoprotein cholesterol levels, and increased the level of high-density lipoprotein cholesterol. Exercise also decreased the levels of soluble Aβ1-40 and Aβ1-42, down-regulated retinoid X receptor expression, and up-regulated liver X receptor, Apolipoprotein E, Low density lipoprotein receptor, Low density lipoprotein receptor-related protein 1, and ATP-binding cassette transporter A1 expression. This indicates that long-term treadmill exercise alters the lipoprotein content, increases lipid metabolism and cholesterol transportation, reduces the soluble Aβ, and therein plays an important neuroprotective role and delays AD progression. We further show that medium exercise intensity (60%-70% of maximal oxygen uptake) was more efficacious in increasing lipid metabolism and reducing blood lipid levels and soluble Aβ levels, than low-intensity exercise (45-55% of maximal oxygen uptake). This research has broad prospects and implications, and offers a theoretical basis for the prevention of AD.
Collapse
Affiliation(s)
- B Zeng
- Department of Sports Medicine, China Medical University, Shenyang 110122, PR China
| | - G Zhao
- Department of Sports Medicine, China Medical University, Shenyang 110122, PR China
| | - H L Liu
- Department of Sports Medicine, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
42
|
Loprinzi PD. Effects of Exercise on Long-Term Potentiation in Neuropsychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1228:439-451. [PMID: 32342476 DOI: 10.1007/978-981-15-1792-1_30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Various neuropsychiatric conditions, such as depression, Alzheimer's disease, and Parkinson's disease, demonstrate evidence of impaired long-term potentiation, a cellular correlate of episodic memory function. This chapter discusses the mechanistic effects of these neuropsychiatric conditions on long-term potentiation and how exercise may help to attenuate these detrimental effects.
Collapse
Affiliation(s)
- Paul D Loprinzi
- Department of Health, Exercise Science, and Recreation Management, Exercise and Memory Laboratory, The University of Mississippi, Oxford, MS, USA.
| |
Collapse
|
43
|
Zhang L, Tang W, Chao FL, Zhou CN, Jiang L, Zhang Y, Liang X, Tang J, Qi YQ, Yang H, He Q, Zhang SS, Zhu L, Peng Y, Tang Y. Four-month treadmill exercise prevents the decline in spatial learning and memory abilities and the loss of spinophilin-immunoreactive puncta in the hippocampus of APP/PS1 transgenic mice. Neurobiol Dis 2019; 136:104723. [PMID: 31887353 DOI: 10.1016/j.nbd.2019.104723] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/20/2019] [Accepted: 12/26/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Previous studies have reported that exercise could improve the plasticity of hippocampal synapses. However, the effects of exercise on synapses in the hippocampus in Alzheimer's disease (AD) are not completely known. METHODS In this study, thirty 12-month-old male APP/PS1 double transgenic mice were randomly divided into a sedentary group (n = 15) and a running group (n = 15). Fifteen 12-month-old male wild-type littermates were assigned to the control group (n = 15). While running mice were assigned to treadmill running for four months, the control mice and sedentary mice did not run during the study period. After Morris water maze testing, five mice in each group were randomly selected for a stereological assessment of spinophilin-immunoreactive puncta in the CA1, CA2-3 and dentate gyrus (DG) of the hippocampus. RESULTS Morris water maze testing revealed that while the learning and memory abilities in sedentary APP/PS1 mice were significantly worse than those in wild-type control mice, the learning and memory abilities in running APP/PS1 mice were significantly better than those in sedentary APP/PS1 mice. The stereological results showed that the spinophilin-immunoreactive puncta numbers of the CA1, CA2-3 and DG in the hippocampus of sedentary APP/PS1 mice were significantly lower than those of wild-type control mice and that the numbers of these spines in the CA1, CA2-3 and DG in the hippocampus of running APP/PS1 mice were significantly higher than those of sedentary APP/PS1 mice. Moreover, a running-induced improvement in spatial learning and memory abilities was significantly correlated with running-induced increases in the spinophilin-immunoreactive puncta numbers in the CA1 and DG of the hippocampus. CONCLUSIONS Four-month treadmill exercise induced a significant improvement in spatial learning and memory abilities and a significant increase in the number of spinophilin-immunoreactive puncta of the CA1, CA2-3 and DG in the hippocampus of APP/PS1 mice. Running-induced improvements in spatial learning and memory abilities were significantly correlated with running-induced increases in the spinophilin-immunoreactive puncta numbers in the CA1 and DG of the hippocampus.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Wei Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Feng-Lei Chao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Chun-Ni Zhou
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Jiang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Yi Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Xin Liang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Ying-Qiang Qi
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Hao Yang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Qi He
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Shan-Shan Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Zhu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Yan Peng
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
44
|
Veronese N, Solmi M, Basso C, Smith L, Soysal P. Role of physical activity in ameliorating neuropsychiatric symptoms in Alzheimer disease: A narrative review. Int J Geriatr Psychiatry 2019; 34:1316-1325. [PMID: 30156330 DOI: 10.1002/gps.4962] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/31/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Neuropsychiatric symptoms (NPs) affect almost all patients with Alzheimer disease (AD). Because of the complications associated with the pharmacological treatment, nonpharmacological treatment (such as physical activity) can be considered as an additional complementary treatment option for NPs. The aim of this review is to evaluate the impact of physical activity on NPs in patients with AD. METHODS We searched Pubmed and Google Scholar for potential eligible articles until March 1, 2018. RESULTS Although there are contradictory results showing the impact of physical exercise on NPs, most of them reported that it had a significant effect on depression and sleep disturbances in patients with AD. The beneficial effects could be explained through several mechanisms, including modulated production of neurotransmitters; increasing neurotrophins, such as brain-derived neurotrophic factor; reduction of oxidative stress and inflammation; elevation of cerebral blood flow; hypothalamic pituitary adrenal axis regulation; and support of neurogenesis and synaptogenesis. Physical activity can also improve cardiovascular risk factors, which may exaggerate NPs. There is limited evidence for other NPs such as agitation, disinhibition, apathy, hallucinations, and anxiety. CONCLUSION Physical activity may ameliorate depression and sleep disturbances in patients with AD. Therefore, physical activity can be a "potential" add-on treatment to drugs to reduce or prevent these symptoms onset and recurrence in patients with AD. However, further studies are needed to focus on relationship between physical activity and other NPs.
Collapse
Affiliation(s)
- Nicola Veronese
- National Research Council, Neuroscience Institute, Aging Branch, Padova, Italy.,Geriatrics Unit, Department of Geriatric Care, Ortho Geriatrics and Rehabilitation, E.O. Galliera Hospital, National Relevance and High Specialization Hospital, Genoa, Italy
| | - Marco Solmi
- Department of Neurosciences, University of Padova, Padova, Italy.,Centro Neuroscienze Cognitive, University of Padua, Padua, Italy
| | | | - Lee Smith
- The Cambridge Centre for Sport and Exercise Sciences, Department of Life Sciences, Anglia Ruskin University, Cambridge, UK
| | - Pinar Soysal
- Department of Geriatric Medicine, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| |
Collapse
|
45
|
Zhang L, So KF. Exercise, spinogenesis and cognitive functions. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 147:323-360. [PMID: 31607360 DOI: 10.1016/bs.irn.2019.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exercise training improves mental and cognitive functions by enhancing neurogenesis and neuroprotection. Recent studies suggest the facilitation of spinogenesis across different brain regions including hippocampus and cerebral cortex by physical activity. In this article we will summarize major findings for exercise effects on synaptogenesis and spinogenesis, in order to provide mechanisms for exercise intervention of both psychiatric diseases and neurodegenerative disorders. We will also revisit major findings for molecular mechanism governing exercise-related spinogenesis, and will discuss the screening for novel factors, or exerkines, whose levels are correlated with endurance training and affect neural plasticity. We believe that further studies focusing on the molecular mechanism of exercise-mediate spinogenesis should benefit the optimization of exercise therapy in clinics and the evaluation of treatment efficiency using specific biomarkers.
Collapse
Affiliation(s)
- Li Zhang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, PR China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, PR China; Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, PR China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, PR China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, PR China; Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, PR China; State Key Laboratory of Brain and Cognitive Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, PR China.
| |
Collapse
|
46
|
Chuang CS, Chang JC, Soong BW, Chuang SF, Lin TT, Cheng WL, Orr HT, Liu CS. Treadmill training increases the motor activity and neuron survival of the cerebellum in a mouse model of spinocerebellar ataxia type 1. Kaohsiung J Med Sci 2019; 35:679-685. [PMID: 31271500 DOI: 10.1002/kjm2.12106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022] Open
Abstract
Spinocerebellar ataxia (SCA) type 1 (SCA1) is a rare autosomal dominant disorder that is characterized by worsening of disordered coordination, ataxia of the trunk, and other neurological symptoms. Physical activity improves both mobility and the daily living activities of patients with SCA. Intervention with daily regular treadmill exercise may slow the deterioration of cerebellar neurons in SCA1. Therefore, the signal changes and performance of cerebellar neurons after exercise in SCA1 was investigated in this study. We employed a transgenic mouse model of SCA1, generated by amplifying the cytosine-adenine-guanine trinucleotide repeat expansions, and the mice underwent 1 month of moderate daily treadmill exercise for 1 hour. The rotarod test revealed that the motor function of the SCA1 mice that underwent training was superior to that of the control SCA1 mice, which did not undergo training. Moreover, the cerebellar pathology revealed preserved Purkinje neurons stained by carbindin with an increase of the neuronal Per Arnt Sim domain protein 4, a key regulation in the structural and functional plasticity of neurons, in the excised SCA1 mice relative to the controls. The mechanism was related to an increase of phosphorylation of ribosomal protein S6, a downstream target of the mammalian target of rapamycin pathway, but not to autophagy activation. This study determined that regular treadmill exercise may play a crucial role in the viable support of cerebellar neurons in SCA1.
Collapse
Affiliation(s)
- Chieh-Sen Chuang
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan.,Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Jui-Chih Chang
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Bing-Wen Soong
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Institute of Neuroscience, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Fei Chuang
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ta-Tsung Lin
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Wen-Ling Cheng
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota
| | - Chin-San Liu
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan.,Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan.,School of Chinese Medicine, Graduate Institute of Integrated Medicine, Research Center for Chinese Medicine and Acupuncture, China Medical University, Taichung, Taiwan
| |
Collapse
|
47
|
Cabral DF, Rice J, Morris TP, Rundek T, Pascual-Leone A, Gomes-Osman J. Exercise for Brain Health: An Investigation into the Underlying Mechanisms Guided by Dose. Neurotherapeutics 2019; 16:580-599. [PMID: 31197642 PMCID: PMC6694330 DOI: 10.1007/s13311-019-00749-w] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There is a strong link between the practice of regular physical exercise and maintenance of cognitive brain health. Animal and human studies have shown that exercise exerts positive effects on cognition through a variety of mechanisms, such as changes in brain volume and connectivity, cerebral perfusion, synaptic plasticity, neurogenesis, and regulation of trophic factors. However, much of this data has been conducted in young humans and animals, raising questions regarding the generalizability of these findings to aging adults. Furthermore, it is not clear at which doses these effects might take place, and if effects would differ with varying exercise modes (such as aerobic, resistance training, combinations, or other). The purpose of this review is to summarize the evidence on the effects of exercise interventions on various mechanisms believed to support cognitive improvements: cerebral perfusion, synaptic neuroplasticity, brain volume and connectivity, neurogenesis, and regulation of trophic factors. We synthesized the findings according to exposure to exercise (short- [1 day-16 weeks], medium- [24-40 weeks], and long-term exercise [52 weeks and beyond]) and have limited our discussion of dose effects to studies in aging adults and aged animals (when human data was not available).
Collapse
Affiliation(s)
- Danylo F Cabral
- Department of Physical Therapy, University of Miami Miller School of Medicine, 5915 Ponce de Leon Boulevard, 5th Floor, Coral Gables, Florida, 33146, USA
- Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA
| | - Jordyn Rice
- Department of Physical Therapy, University of Miami Miller School of Medicine, 5915 Ponce de Leon Boulevard, 5th Floor, Coral Gables, Florida, 33146, USA
- Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA
| | - Timothy P Morris
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA
| | - Tatjana Rundek
- Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA
- Department of Neurology, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA
| | - Alvaro Pascual-Leone
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA
| | - Joyce Gomes-Osman
- Department of Physical Therapy, University of Miami Miller School of Medicine, 5915 Ponce de Leon Boulevard, 5th Floor, Coral Gables, Florida, 33146, USA.
- Evelyn McKnight Brain Institute, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA.
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA.
- Department of Neurology, University of Miami Miller School of Medicine, 1150 Northwest 14th Street, Suite 309, Miami, Florida, 33136, USA.
| |
Collapse
|
48
|
Li X, Inoue T, Hayashi M, Maejima H. Exercise enhances the expression of brain-derived neurotrophic factor in the hippocampus accompanied by epigenetic alterations in senescence-accelerated mice prone 8. Neurosci Lett 2019; 706:176-181. [DOI: 10.1016/j.neulet.2019.05.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/26/2019] [Accepted: 05/16/2019] [Indexed: 02/05/2023]
|
49
|
Frederiksen KS, Madsen K, Andersen BB, Beyer N, Garde E, Høgh P, Waldemar G, Hasselbalch SG, Law I. Moderate- to high-intensity exercise does not modify cortical β-amyloid in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2019; 5:208-215. [PMID: 31198839 PMCID: PMC6556817 DOI: 10.1016/j.trci.2019.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Animal models of Alzheimer's disease show that exercise may modify β-amyloid (Aβ) deposition. We examined the effect of a 16-week exercise intervention on cortical Aβ in patients with mild-to-moderate Alzheimer's disease. METHODS Thirty-six patients with Alzheimer's disease were randomized to either one hour of aerobic exercise three times weekly for 16 weeks or usual care. Pre and post intervention, 11Carbon-Pittsburgh compound B positron emission tomography was carried out to assess cortical Aβ, and quantified using standardized uptake value rations (SUVRs). RESULTS The intervention showed no effect on follow-up SUVRs in a covariance analysis with group allocation, baseline intervention SUVR, age, sex, and baseline Mini-Mental State Examination as predictors. Change in SUVRs did not correlate with changes in measures of physical or aerobic fitness. DISCUSSION The present findings do not support an effect of exercise on Aβ. However, the relatively short intervention period may account for a lack of efficacy. Further studies should test earlier and longer interventions.
Collapse
Affiliation(s)
- Kristian S. Frederiksen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Karine Madsen
- Neurobiology Research Unit, Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte B. Andersen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nina Beyer
- Musculoskeletal Rehabilitation Research Unit and Institute of Sports Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ellen Garde
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Public Health and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Peter Høgh
- Zealand University Hospital, Department of Neurology, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steen G. Hasselbalch
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Zhang X, He Q, Huang T, Zhao N, Liang F, Xu B, Chen X, Li T, Bi J. Treadmill Exercise Decreases Aβ Deposition and Counteracts Cognitive Decline in APP/PS1 Mice, Possibly via Hippocampal Microglia Modifications. Front Aging Neurosci 2019; 11:78. [PMID: 31024293 PMCID: PMC6461026 DOI: 10.3389/fnagi.2019.00078] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 03/19/2019] [Indexed: 12/31/2022] Open
Abstract
Recent studies have suggested that exercise may be beneficial for delaying or attenuating Alzheimer's disease (AD). However, the underlying mechanisms were not clear. Microglia-mediated neuroinflammation is suggested to play an important role in the pathology of AD. The present study investigated the beneficial effects of treadmill exercise on amyloid-β (Aβ) deposition and cognitive function in amyloid precursor protein (APP)/PS1 mice in the early stage of AD progression and microglia-mediated neuroinflammation was mainly analyzed. The results demonstrated that 12 weeks of treadmill exercise preserved hippocampal cognitive function in APP/PS1 mice and substantially suppressed Aβ accumulation in the hippocampus. Treadmill exercise significantly inhibited neuroinflammation, which was characterized by a remarkably reduced expression of pro-inflammatory factors and increased expression of anti-inflammatory mediators in the hippocampus, resulting from a shift in activated microglia from the M1 to M2 phenotype. Treadmill exercise also attenuated oxidative stress presented by a marked reduction in methane dicarboxylic aldehyde (MDA) level and dramatically elevated SOD and Mn-SOD activities in the hippocampus. These findings suggest that treadmill exercise can effectively prevent the decrease in hippocampal-dependent cognitive function and Aβ deposits in early AD progression possibly via modulating microglia-mediated neuroinflammation and oxidative stress.
Collapse
Affiliation(s)
| | - Qiang He
- College of Physical Education, Shandong Normal University, Jinan, China
| | - Tao Huang
- Department of Physical Education, Shanghai Jiao Tong University, Shanghai, China
| | - Na Zhao
- School of Physical Education & Health Care, East China Normal University, Shanghai, China
| | - Fei Liang
- School of Physical Education & Health Care, East China Normal University, Shanghai, China
| | - Bo Xu
- School of Physical Education & Health Care, East China Normal University, Shanghai, China
| | - Xianghe Chen
- College of Physical Education, Yangzhou University, Yangzhou, China
| | | | - Jianzhong Bi
- Department of Neurology Medicine, Shandong University, Jinan, China
| |
Collapse
|