Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Dementia is one of the greatest global challenges for health and social care in the 21st century. Alzheimer's disease (AD), the most common type of dementia, is by no means an inevitable consequence of growing old. Several lifestyle factors may increase, or reduce, an individual's risk of developing AD. Much has been written over the ages about the benefits of exercise and physical activity. Among the risk factors associated with AD is a low level of physical activity. The relationship between physical and mental health was established several years ago. In this review, we discuss the role of exercise (aerobic and resistance) training as a therapeutic strategy for the treatment and prevention of AD. Older adults who exercise are more likely to maintain cognition. We address the main protective mechanism on brain function modulated by physical exercise by examining both human and animal studies. We will pay especial attention to the potential role of exercise in the modulation of amyloid β turnover, inflammation, synthesis and release of neurotrophins, and improvements in cerebral blood flow. Promoting changes in lifestyle in presymptomatic and predementia disease stages may have the potential for delaying one-third of dementias worldwide. Multimodal interventions that include the adoption of an active lifestyle should be recommended for older populations.

Free full text 


Logo of jsporthsLink to Publisher's site
J Sport Health Sci. 2020 Sep; 9(5): 394–404.
Published online 2020 Feb 4. https://doi.org/10.1016/j.jshs.2020.01.004
PMCID: PMC7498620
PMID: 32780691

Physical exercise in the prevention and treatment of Alzheimer's disease

Abstract

Dementia is one of the greatest global challenges for health and social care in the 21st century. Alzheimer's disease (AD), the most common type of dementia, is by no means an inevitable consequence of growing old. Several lifestyle factors may increase, or reduce, an individual's risk of developing AD. Much has been written over the ages about the benefits of exercise and physical activity. Among the risk factors associated with AD is a low level of physical activity. The relationship between physical and mental health was established several years ago. In this review, we discuss the role of exercise (aerobic and resistance) training as a therapeutic strategy for the treatment and prevention of AD. Older adults who exercise are more likely to maintain cognition. We address the main protective mechanism on brain function modulated by physical exercise by examining both human and animal studies. We will pay especial attention to the potential role of exercise in the modulation of amyloid β turnover, inflammation, synthesis and release of neurotrophins, and improvements in cerebral blood flow. Promoting changes in lifestyle in presymptomatic and predementia disease stages may have the potential for delaying one-third of dementias worldwide. Multimodal interventions that include the adoption of an active lifestyle should be recommended for older populations.

Keywords: Aerobic exercise, Dementia, Exercise training, Lifestyle factors, Multidomain interventions, Resistance exercise

1. Introduction

Alzheimer's disease (AD) is the most common type of dementia and one of the most frequent neurodegenerative pathologies in elderly people, constituting about 90% of the cases of dementia in this population. AD is characterized by its irreversibility and progressive functional, cognitive, and behavioral loss. It is usually accompanied by various brain disorders such as amnesia, agnosia, apraxia, and aphasia.1

Twenty-four million people in the world suffer from AD, and it is expected that by 2050 this number will be 4 times greater.2 These alarming numbers are explained by the fact that the main risk factor for this disease is age,3 and by 2050 it is estimated that the population over 65 years will be 3 times larger than it was in 2010, reaching close to 1.5 billion people.

AD is a devastating disease. The social and human cost for patients and caregivers is overwhelming, and the economic cost of AD is unaffordable. For instance, in the United States in 2017, the annual cost of care per patient over 65 with AD or another type of dementia was USD 48,000, or 3.5 times more than for a person without any type of dementia.4,5

AD is characterized by the loss of neuronal synapses and pyramidal neurons accompanied by progressive cognitive neurodegeneration.6 The regions associated with more complex brain functions, such as the hippocampus and the neocortex, are the most affected.

Although there are several hypotheses to explain the origin of AD, the 2 main neuropathological features include the extracellular accumulation of senile plaques around the neurons and the glia, and the intracellular formation of neurofibrillary tangles (NFTs). Senile plaques are formed mainly by the accumulation of insoluble amyloid β (Aβ) protein, especially its isoform Aβ42, which is formed by the enzymatic cleavage of the transmembrane amyloid precursor protein (APP).7,8 NFTs are formed from the hyperphosphorylation of the tau protein. The main function of this protein is to stabilize the microtubules so that they connect with other elements of the neuronal cytoskeleton, such as neurofilaments and actin.9

Another feature in AD is the inflammatory response at the brain level. In the early stages of the disease, the immune response results in a removal of Aβ, ameliorating its symptoms.10,11 To generate a balance between the deposition and the clearance of Aβ, microglia plays a fundamental role by engulfing insoluble deposits of this peptide through the activation of receptors that promote phagocytic action, such as cluster of differentiation 36 (CD36), and receptors for advanced-glycosylation end products.12,13 Microglia also activates extracellular proteases such as neprilysin, the insulin degrading enzyme, endothelin converting enzyme 1, angiotensin converting enzyme, matrix metalloprotease, and the presequence peptidase.14, 15, 16 During advanced stages of the disease there is a prolonged stimulation of the immune response caused by Aβ, which leads to a decrease in the efficiency of the microglia to degrade it.17 In turn, this leads to the release of several proinflammatory products, such as cytokines, reactive oxygen, and nitrogen species, leading to an increase in the production of Aβ and the hyperphosphorylation of tau, generating more damage and neuronal death.18, 19, 20 This state contributes to an increase in cognitive decline and the development of the disease.

The risk factors associated with AD, apart from age, are low educational level, diabetes, genetics, belonging to certain minority ethnic groups (prevalence of dementia is higher in British African–Caribbean people),21 diet, previous brain injuries, sleep disorders, hypertension, hearing loss, and low levels of physical activity.22 Physical activity (“any movement of the body”) and exercise (“physical activity that is typified by specific and purposeful training”) constitute 2 different concepts.23 However, for the sake of clarity we refer to both these concepts interchangeably since some of the reports we refer to in this review use these expressions as synonyms. Older adults are considered physically active when they perform moderate-intensity aerobic physical activities for a minimum of 30 min, 5 days/week, or vigorous-intensity aerobic activity for a minimum of 20 min, 3 days/week.23 Also, combinations of moderate- and vigorous-intensity activity can be performed to meet this recommendation. Moderate-intensity aerobic activity involves a moderate level of effort relative to an individual's aerobic fitness. On a 10-point scale, where sitting is 0 and all-out effort is 10, moderate-intensity activity is rated as a 5 or 6 and produces noticeable increases in heart rate and breathing. Using the same scale, vigorous-intensity activity is rated as a 7 or 8 and produces large increases in heart rate and breathing. Thus, physical inactivity, a concept that has also been included in this review, is defined as not meeting any of the following 2 criteria: 30 min of moderate-intensity physical activity on at least 5 days/week or 20 min of vigorous-intensity physical activity on at least 3 days/week.

Gender is also considered a risk factor for AD; two-thirds of AD patients are women.5 This proportion is mainly attributed to the greater longevity of women over men, which makes them more susceptible to this and other age-associated diseases and syndromes.24 Apart from this fact, if there is a real relationship between sex and AD risk, it is not clear. In the literature, epidemiological studies with controversial results can be found.25, 26, 27 Several results point to sex hormones as responsible, at least partially, for the higher incidence of AD in women. The important changes in sex hormone concentrations associated with a woman's age, the role of sexual dimorphism in the neuronal structures development, the gene-by-sex interactions, and the increased specific immune response of women, have all been associated with an increased risk of AD.26

In this review, we mainly discuss the role of physical exercise as a therapeutic strategy for the treatment and prevention of AD.

2. Physical exercise and physiologic brain aging

Physical inactivity contributes to about 5 million deaths in the world each year from noncommunicable diseases.28 The inverse relationship between a physically active life and the risk of suffering cognitive decline is widely documented,29 and aerobic exercise training has been the most extensively used option for the study of the effect of physical activity in alleviating the negative impact of aging in cognitive function.29,30 The first evidence was published in the 1970s, when it was shown that middle-aged sports practitioners had better response in cognitive tasks that involved a psychomotor component when compared with sedentary aged-matched subjects.31 More recently, 1 study found a significant improvement in memory among middle-aged trained individuals, compared to sedentary subjects, when the Free and Cued Immediate Recall test was administered to both groups.32 The positive impact of long-term exercise training by delaying the onset of physiologic memory loss suggests the effectiveness of exercise as a preventive strategy against age-related memory loss and neurodegeneration. However, it is worth mentioning that late-onset exercise interventions have also shown positive results in the delay of brain aging. For example, in a ground-breaking paper published in 2011, the authors found that 1 year of moderate-intensity exercise (40 min duration, 3 days/week) increased the size of the hippocampus, as well as increasing spatial memory, in healthy older individuals.33 The anatomic changes induced by aerobic exercise have also been confirmed by other research groups. For instance, it has been found that 6 months of exercise (60 min duration, 3 days/week) is sufficient to increase both the gray and white matter in the anterior cingulate cortex, as measured by magnetic resonance imaging, in older, cognitively healthy subjects.34 Longer aerobic exercise training protocols (3 years’ duration) in sedentary older women have shown improvements in reaction time, motor function, and cognitive processing speed, indicating that exercise is effective in reversing or at least slowing the age-related declines in motor performance and in speed of cognitive processing.35 However, some evidence shows that aerobic exercise interventions do not always induce improvements in cognitive function in older subjects (60−80 years).36, 37, 38 These contradictory results may be explained by the training duration, frequency, and/or intensity of the interventions.

The impact of resistance exercise on cognitive function has gained notice in the past few years. In a recent meta-analysis, more than 24 studies were reviewed that investigated the effects of weight lifting on different cognitive outcomes in older individuals. According to the results, resistance training has positive effects on measurements related to the detection of cognitive impairment and executive functions. However, resistance training has no effect on working memory measurements.39 Although a high heterogeneity was observed in all analyses, the authors concluded that resistance training seemed to have positive effects on cognition. However, future research will need to determine why the effects are so variable.

From this first part of the review, we can conclude that exercise programs with components of both aerobic and resistance training, of moderate intensity, and lasting at least 45 min per session on as many days of the week as possible, are beneficial in terms of cognitive function in older healthy adults.40

3. Physical exercise and multimodal interventions for the treatment and prevention of AD

Dementia is by no means an inevitable consequence of reaching retirement age. There are lifestyle factors that may decrease, or increase, an individual's risk of developing dementia.22 Around 35% of dementia is attributable to a combination of 9 risk factors: low education level, midlife hypertension, midlife obesity, hearing loss, later-life depression, diabetes, smoking, social isolation, and, of course, low physical activity.41

A meta-analysis that included 16 studies with more than 160,000 participants found a 45% reduction in the risk of developing AD due to the regular practice of physical activity (hazard ratio = 0.55, 95% confidence interval: 0.36−0.84, p = 0.006).42 In a sample of 716 older subjects followed for 3.5 years, similar results were found when assessing the risk of suffering AD. Those individuals with low daily physical activity levels were 53% more likely to suffer AD than those who reported more active lives (hazard ratio = 0.477, 95% confidence interval: 0.273−0.832).43

Although there are discrepancies,36,44, 45, 46, 47, 48, 49 the benefits of engaging in physical activities have also been seen in populations diagnosed with mild cognitive impairment (MCI) and AD. Several studies have found improvements in executive functions, memory, and cognitive tests in individuals with MCI who engaged in an aerobic exercise program.50, 51, 52, 53, 54, 55 Improvements in the cognitive function of individuals with MCI through physical activity are important in preventing the progression of the disease and in decreasing the number of AD in older people. In individuals diagnosed with AD, and, in general, at early stages of the disease, aerobic exercise alone, or accompanied with cognitive stimulation, induces improvements in some aspects of brain function,56, 57, 58, 59, 60, 61, 62, 63, 64, 65 although in some studies exercise was accompanied with cognitive situation.

Few studies have been published on the effects of resistance training in people with MCI or AD.44,66, 67, 68 Prominent among them is the randomized, double-blind trial that included 100 people between 55 and 86 years of age with MCI.69 In this study it was shown that 6 months of resistance exercise induced improvements in memory, attention, and executive functions. Moreover, those benefits persisted 12 months after the end of the intervention period.

Prevention through the promotion of changes in lifestyle in presymptomatic and predementia disease stages may have the potential for delaying one-third of dementias.70 Engaging in physical exercise programs seems to be effective, but it is clearly not enough. Studies that examine multimodal interventions hypothesize that an integrated approach to addressing multiple risk factors for AD may be more successful than single-component interventions in producing benefits. Multimodal interventions often include changes in diet, physical activity, and cognitive training (Fig. 1).

Fig 1

Brain function in normal aging and Alzheimer's disease is influenced by lifestyle factors. The usual trajectory for brain aging (orange line) indicates that, years before death, an individual shows signs of mild cognitive impairment. In some cases, that cognitive damage takes place many years before death and even leads to dementia (red line). By modifying lifestyle factors through multimodal interventions, an individual can delay the onset of cognitive impairment until a very advanced age (green line).

Several studies attribute to the diet a key role in delaying the progression of AD. Some of these studies have tested diets considered as heart-healthy, such as the Dietary Approaches to Stop Hypertension diet (DASH), Mediterranean–DASH Intervention for Neurodegenerative Delay diet (MIND), or the Mediterranean diet.71 The inflammatory component of AD has been treated by supplementation with fatty acids, vitamins, flavonoids, polyphenols, probiotics, and dietary advanced glycation end products.72 Ketogenic diets, which are protective in other neurological disorders, also seem to have great potential in the prevention and treatment of the disease.73 In humans, few clinical trials have been carried out using ketogenic diets; instead, ketogenic agents have been used.74 In animal models of AD, the increase in ketone levels in the body through intermittent fasting or supplementation with ketone esters have shown promising results.75,76

The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER), a multidomain intervention consisting of diet, exercise, cognitive training, and vascular risk monitoring, resulted in an improvement or maintenance of cognitive functioning in at-risk elderly people from the general population.77 This trial was the first methodologically robust study showing that with a 2-year intervention, it was possible to reduce the risk of cognitive decline among the general elderly population, rather than solely with patients in a clinical setting. More recently, it has been found that engaging in physical activity and lowering vascular risk may have additive protective effects on delaying the progression of AD.22 Greater physical activity and lower vascular risk independently attenuated the negative association of Aβ burden with cognitive decline and neurodegeneration in asymptomatic individuals.78

4. Potential protective mechanisms of physical exercise on brain aging, from mice to humans

The beneficial effects of physical activity go beyond skeletal muscle and involve adaptations in other organs. Exercise causes changes in the brain at the anatomic, cellular, and molecular levels by inducing a cascade of cellular and molecular processes that promote different physiologic phenomena, including angiogenesis, neurogenesis, synaptogenesis, and stimulation of neurotrophic factors that enhance learning, memory, and brain plasticity (Fig. 2).53,60,79, 80, 81, 82, 83, 84 The availability of transgenic murine models that simulate the main neuropathologic characteristics of AD has enabled the study of the main protective mechanisms of exercise on brain aging. Among these mechanisms is an increase in cerebral blood flow (CBF) in several cortical and subcortical areas,85 together with the decrease in the formation of Aβ and the phosphorylation of the tau protein.86,87

Fig 2

Potential protective mechanisms of exercise on brain aging.

5. Summary of the main mechanisms that exercise can activate at the brain level

5.1. β-Amyloid peptide, tau protein, and exercise

In transgenic mice models for AD, a decrease in Aβ plaques and NFTs after both voluntary and forced exercise interventions has been found. In some cases, these findings have been accompanied by improvements in learning and memory.86, 87, 88, 89 APP/PS1 are double transgenic mice expressing a chimeric mouse/human amyloid precursor protein (APP) (Mo/HuAPP695swe) and a mutant human presenilin 1 (PS1-dE9), both directed to central nervous system (CNS) neurons. In a recent study in double transgenic APPswe/PSEN1ΔE9 mice, 10 weeks of voluntary exercise decreased both the load and the size of the Aβ plaques, as well as the phosphorylation of tau in the hippocampus. Improvements in spatial memory, a decrease in hippocampal neuronal loss, and an increase in neurogenesis in the Cornu Ammonis 3 (CA3) zone and the dentate gyrus were also reported in the study.90 These results have been supported by studies in other AD transgenic mice models such as hPS2m (overexpress human Presenilin 2 mutant),89 Tg-NSE/htau23 (overexpress human tau23 under the control of the NSE promoter),88 Thy-Tau22 (human 4-repeat tau mutated at sites G272V and P301S under the control of Thy1.2 promoter),91 P301S-tau (overexpress the P301S-mutant human tau gene),87 and Tg2576 (overexpress human APP 695).92 It is assumed that a successful therapy for AD would both remove the pathologic hallmarks of the disease and provide some functional recovery. However, there are studies that have not found a decrease in Aβ load and/or tau phosphorylation despite improvements in learning tasks and/or memory in the exercised mice.93, 94, 95 A key factor with exercise interventions is to start them in an early pathologic state. Exercise cannot counteract the formation of plaques and the cognitive deterioration if the intervention is carried out after their appearance.86 Moreover, it has been shown in mice models that voluntary exercise may be superior to forced exercise if certain aspects of the disease, such as plaque deposition and memory impairment, are to be reduced.92

Studies in humans have also found an inverse correlation between physical activity levels and the plasma and brain Aβ load in elderly people without cognitive disorders.96,97 Moreover, it has been found that a 6-month aerobic exercise intervention in individuals with MCI induced a 24% decrease in the plasma Aβ1-42 levels when compared with the levels found in the control group.50 Jointly, all of these results indicate a potential role of exercise in modulating the Aβ turnover.

5.2. Exercise and inflammation

The term “inflammaging” characterizes a widely accepted hypothesis that aging is accompanied by a low-grade chronic upregulation of certain proinflammatory responses. It is characterized by a relative decline in adaptive immunity and T-helper 2 responses and is associated with increased cell-mediated responses.98 Inflammaging has been considered to be a “prodrome” to AD and microglial malfunction, a common feature during aging.99

Exercise has important modulatory effects on immune function.100 Systemically, physical exercise has been shown to have a positive effect on markers of inflammation, and recently those effects have been extended to the CNS.101

In the Tg2576 AD animal model, it was shown that 3 weeks of voluntary exercise were sufficient to decrease the concentration of Aβ and the proinflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). These changes were accompanied by an increase in proteins related to the immune response, such as interferon-γ and macrophage inflammatory protein-1α, in the hippocampus.102

The anti-inflammatory effects of exercise on the CNS have also been evaluated in older animals. Higher levels of hippocampal IL-10 (the main anti-inflammatory cytokine) and lower IL-1β/IL-10, IL-6/IL-10, and TNF-α/IL-10 ratios have been found in rats exercised for 10 days.103 Barrientos et al.104 found that the long-term memory impairment in older rats injected with Escherichia coli (E. coli)intraperitoneally could be prevented when they had free access to running wheels. Along with these findings, voluntary exercise also prevented the reduction in brain-derived neurotrophic factor (BDNF) expression in the CA1 zone of the hippocampus. Finally, the microglia of the exercised rats had a lower inflammatory response than the control rats, as measured by the expression of TNF-α, IL-1β, and IL-6. These findings highlight the capacity of exercise to modulate inflammatory responses on CNS and reinforce its potential in minimizing the risk of suffering diseases related to neuroinflammation, such as AD.

Particularly in elderly people, regular physical activity has been shown to have a positive effect in the reduction of inflammatory markers such as Creaction protein, IL-6, and TNF-α105, 106, 107, 108, 109, 110 at the systemic level. In many cases, this has been associated with better performance in cognitive tests in both cross-sectional111, 112, 113 and longitudinal studies.114, 115, 116, 117

Aerobic exercise lasting more than 2 weeks118, 119, 120, 121 has been shown to improve the immune system in healthy elderly people by increasing the activity of the natural killer cells, the proliferation of T lymphocytes, hematopoietic stem cells, and endothelial progenitor cells. However, it is noteworthy that the results achieved with resistance exercise interventions have been more heterogeneous.122

5.3. Neurotrophins, cognitive function, and exercise

Neurotrophins are growth factors belonging to the family of neurotrophic factors, which regulate axonal growth, synaptic plasticity, neurotransmission, hippocampal neurogenesis, synaptic protein expression, and long-term potentiation.123, 124, 125, 126, 127, 128

Among neurotrophins, BDNF has been extensively studied, and its relationship with exercise and cognitive function has been widely established.32,50,129, 130, 131, 132, 133, 134, 135 The brain synthesizes about 75% of BDNF under normal conditions. During prolonged exercise, this synthesis is increased by 2- to 3-fold, and there is evidence indicating that circulating BDNF levels correlate with brain tissue BDNF levels.132,136

Exercise provides cognitive benefit in 5xFAD mice (express human APP and PSEN1 transgenes with a total of 5 AD-linked mutations: the Swedish (K670N/M671L), Florida (I716V), and London (V717I) mutations in APP, and the M146L and L286V mutations in PSEN1), a mouse model of AD, by inducing both adult hippocampal neurogenesis and by elevating the levels of BDNF.137 This combination seems to generate an adequate brain environment for the maintenance and survival of the new neurons in regions of the brain affected by AD. The induction of adult hippocampal neurogenesis in combination with the elevation of BDNF levels successfully mimic the beneficial effects of exercise in AD mice.137 However, the induction of hippocampal neurogenesis alone did not benefit cognition or AD markers. Exercise has also shown its effectiveness on cognitive function and BDNF levels in mice models of accelerated aging.130 Four weeks of moderate exercise were sufficient to improve recognition memory and increased hippocampal BDNF expression in 13-month-old mice.

Low levels of BDNF have been found in brains of people with AD,138,139 which supports the hypothesis that improving its production may be an effective alternative that delays the onset of the disease. In humans, several studies indicate that both acute and chronic exercise140, 141, 142, 143, 144, 145, 146 contribute to an increase in the peripheral levels of BDNF, and, in some cases, it is related to gains in cognitive function.131,147, 148, 149 Moreover, aerobic exercise-induced increments in hippocampal volume are associated with greater serum levels of BDNF in cognitively healthy older individuals,33 and high serum levels of BDNF have been associated with low risk of AD.150

The pathways responsible for the synthesis and release of BDNF in AD patients seem to be susceptible to stimulation with exercise. In fact, one bout of acute aerobic exercise increased the BDNF plasma levels in patients with AD and in healthy controls. Additionally, BDNF was associated with their physical activity levels.151

5.4. AD, CBF, and exercise

Aging has been associated with progressive losses in cognitive function and CBF.152, 153, 154, 155, 156 It has been shown that the rate of decrease with age of the CBF is 0.35%−0.45% per year in middle-aged and elderly subjects (Fig. 3).157,158 This leads to an accelerated decline in cognitive function and an increased risk of suffering dementia in the general population.159

Fig 3

CBF by age groups (at an interval of 5 years), measured by MRI (De Vis et al.,160 used with permission of John Wiley and Sons). CBF = cerebral blood flow; MRI = magnetic resonance imaging.

Although the cause of the CBF decrease in aging is not entirely clear, it has been postulated that changes in the density and elasticity of cerebral blood vessels, as well as the degeneration of pericytes and a reduction in the activity and number of neurons, could explain it.161,162

Compared with healthy people, individuals with AD show a decrease of up to 40% in the CBF. Regions such as the precuneus, the hippocampus, the posterior cingulate gyrus, and the temporal, occipital, and parietal lobes are mainly affected.163, 164, 165, 166, 167

In animal models, it has been shown that the soluble form of Aβ protein can accumulate in cerebral blood vessels (which is known as amyloid angiopathy), generating a decrease in CBF and a vasoconstriction effect, possibly affecting neuronal activity.168,169 Intra-arterially injected solubilized Aβ1-40 in rats of 7−8 months of age decreased CBF and increased vascular resistance in the cerebral cortex of the animals, indicating that Aβ could contribute to the hypoperfusion observed during AD.170

In older adults, the regular practice of exercise has a positive effect both on peripheral endothelial function171 and on the increase of CBF in areas such as the hippocampus.156,172,173 In middle-aged, cognitively healthy subjects, even short-term exercise programs, such as 3 h of aerobic exercise per week for 12 weeks, are sufficient to improve CBF at rest in regions such as the anterior cingulate and the hippocampus.174,175 However, even in physically active people, short periods of inactivity can decrease CBF in up to 8 regions of the brain, including the hippocampus.176 Thus, CBF is sensitive to small changes in people's lifestyle, thus making it necessary to maintain a regular practice of exercise to preserve brain health.

Regarding AD patients and exercise, the results from studies have not been consistent.177 For instance, in a study, exercise did not lead to any improvements in CBF,178 concentrations of Aβ, total tau, or phosphorylated tau in the cerebrospinal fluid of people with AD.179 This indicates that, although functional parameters are still likely to be positively affected in people with AD, changes in brain function may require an adjustment in the different components of the load, such as the duration of exercise sessions and the frequency, intensity, and type of exercise during the intervention.

Finding mimetics of physical exercise for those patients who have very compromised functionality is not easy, because physical exercise is a very systemic intervention and targets at several potential causes of AD. The possibility of using molecular mimetics, such as 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), is known to only improve brain function transiently180 and does not achieve the same positive effects as, for instance, running. Another option would be to use neuromuscular electrical stimulation or vibration in people who are unable to perform physical exercise, but this has not yet been tested in AD patients.181

6. Conclusion

Promoting changes in lifestyle in presymptomatic and predementia disease stages may have the potential for delaying one-third of dementias worldwide. Exercise modulates Aβ turnover, inflammation, the synthesis and release of neurotrophins, and CBF. Multimodal interventions that include the adoption of an active lifestyle should be recommended for older populations.

Acknowledgments

This work was supported by the following grants: Instituto de Salud Carlos III and co-funded by FEDER (Grant number PIE15/00013); SAF2016-75508-R from the Spanish Ministry of Education and Science (MEC); CB16/10/00435 (CIBERFES); PROMETEOII2014/056 from Conselleria, de Sanitat de la Generalitat Valenciana and EU Funded CM1001 and FRAILOMIC-HEALTH.2012.2.1.1-2; and ADVANTAGE-724099 Join Action (HP-JA) 3rd EU Health Programme and DIALBFRAIL-LATAM (825546 H2020-SC1-BHC).

Authors’ contributions

ADR and GOG performed the literature search and review and wrote the manuscript; ASP, CAC, FM, CGL, AGC, CBL, AC, and EGD analyzed and discussed the data and reviewed the manuscript; MCGC and JV designed and supervised the review, secured funding, and wrote the manuscript. All authors have read and approved the final version of the manuscript, and agree with the order of presentation of the authors.

Competing interests

The authors declare that they have no competing interests.

Footnotes

Peer review under responsibility of Shanghai University of Sport.

References

1. Yesavage J.A., Brooks J.O., Taylor J., Tinklenberg J. Development of aphasia, apraxia, and agnosia and decline in Alzheimer's disease. Am J Psychiatry. 1993;150:742–747. [Abstract] [Google Scholar]
2. Dos Santos Picanco L.C., Ozela P.F., de Fatima de Brito Brito M., Pinheiro A.A., Padilha E.C., Braga F.S. Alzheimer's disease: a review from the pathophysiology to diagnosis, new perspectives for pharmacological treatment. Curr Med Chem. 2018;25:3141–3159. [Abstract] [Google Scholar]
3. World Health Organization. Global health and aging. Available at: https://www.who.int/ageing/publications/global_health.pdf. [accessed 15.01.2019].
4. Alzheimer's Association. 2018 Alzheimer's disease facts and figures: includes a special report on the financial and personal benefits of early diagnosis. Available at: https://www.alz.org/media/HomeOffice/FactsandFigures/facts-and-figures.pdf. [accessed 18.11.2018].
5. Hebert L.E., Weuve J., Scherr P.A., Evans D.A. Alzheimer disease in the United States (2010−2050) estimated using the 2010 census. Neurology. 2013;80:1778–1783. [Europe PMC free article] [Abstract] [Google Scholar]
6. Francis P.T., Palmer A.M., Snape M., Wilcock G.K. The cholinergic hypothesis of Alzheimer's disease: a review of progress. J Neurol Neurosurg Psychiatry. 1999;66:137–147. [Europe PMC free article] [Abstract] [Google Scholar]
7. Sun X., Jin L., Ling P. Review of drugs for Alzheimer's disease. Drug Discov Ther. 2012;6:285–290. [Abstract] [Google Scholar]
8. Hardy J.A., Higgins G.A. Alzheimer's disease: the amyloid cascade hypothesis. Science. 1992;256:184–185. [Abstract] [Google Scholar]
9. Aamodt E.J., Williams R.C., Jr. Microtubule-associated proteins connect microtubules and neurofilaments in vitro. Biochemistry. 1984;23:6023–6031. [Abstract] [Google Scholar]
10. Chakrabarty P., Jansen-West K., Beccard A., Ceballos-Diaz C., Levites Y., Verbeeck C. Massive gliosis induced by interleukin-6 suppresses Aβ deposition in vivo: evidence against inflammation as a driving force for amyloid deposition. FASEB J. 2010;24:548–559. [Europe PMC free article] [Abstract] [Google Scholar]
11. Shaftel S.S., Kyrkanides S., Olschowka J.A., Miller J.N., Johnson R.E., O'Banion M.K. Sustained hippocampal IL-1β overexpression mediates chronic neuroinflammation and ameliorates Alzheimer plaque pathology. J Clin Invest. 2007;117:1595–1604. [Europe PMC free article] [Abstract] [Google Scholar]
12. El Khoury J., Hickman S.E., Thomas C.A., Loike J.D., Silverstein S.C. Microglia, scavenger receptors, and the pathogenesis of Alzheimer's disease. Neurobiol Aging. 1998;19(Suppl. 1):S81–S84. [Abstract] [Google Scholar]
13. Yan S.D., Chen X., Fu J., Chen M., Zhu H., Roher A. RAGE and amyloid-β peptide neurotoxicity in Alzheimer's disease. Nature. 1996;382:685–691. [Abstract] [Google Scholar]
14. Marr R.A., Hafez D.M. Amyloid-beta and Alzheimer's disease: the role of neprilysin-2 in amyloid-beta clearance. Front Aging Neurosci. 2014;6:187. 10.3389/fnagi.2014.00187. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
15. Vitek M.P., Bhattacharya K., Glendening J.M., Stopa E., Vlassara H., Bucala R. Advanced glycation end products contribute to amyloidosis in Alzheimer disease. Proc Natl Acad Sci U S A. 1994;91:4766–4770. [Europe PMC free article] [Abstract] [Google Scholar]
16. Lee C.Y., Landreth G.E. The role of microglia in amyloid clearance from the AD brain. J Neural Transm (Vienna) 2010;117:949–960. [Europe PMC free article] [Abstract] [Google Scholar]
17. Hickman S.E., Allison E.K., El Khoury J. Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer's disease mice. J Neurosci. 2008;28:8354–8360. [Europe PMC free article] [Abstract] [Google Scholar]
18. Hsieh H.L., Yang C.M. Role of redox signaling in neuroinflammation and neurodegenerative diseases. Biomed Res Int. 2013;2013 10.1155/2013/484613. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
19. Wee Yong V. Inflammation in neurological disorders: a help or a hindrance? Neuroscientist. 2010;16:408–420. [Abstract] [Google Scholar]
20. Meda L., Cassatella M.A., Szendrei G.I., Otvos L., Jr., Baron P., Villalba M. Activation of microglial cells by β-amyloid protein and interferon-γ Nature. 1995;374:647–650. [Abstract] [Google Scholar]
21. Adelman S., Blanchard M., Livingston G. A systematic review of the prevalence and covariates of dementia or relative cognitive impairment in the older African-Caribbean population in Britain. Int J Geriatr Psychiatry. 2009;24:657–665. [Abstract] [Google Scholar]
22. Livingston G., Sommerlad A., Orgeta V., Costafreda S.G., Huntley J., Ames D. Dementia prevention, intervention, and care. The Lancet. 2017;390:2673–2734. [Abstract] [Google Scholar]
23. Vina J., Borras C., Sanchis-Gomar F., Martinez-Bello V.E., Olaso-Gonzalez G., Gambini J. Pharmacological properties of physical exercise in the elderly. Curr Pharm Des. 2014;20:3019–3029. [Abstract] [Google Scholar]
24. Plassman B.L., Langa K.M., Fisher G.G., Heeringa S.G., Weir D.R., Ofstedal M.B. Prevalence of dementia in the United States: the aging, demographics, and memory study. Neuroepidemiology. 2007;29:125–132. [Europe PMC free article] [Abstract] [Google Scholar]
25. Fisher D.W., Bennett D.A., Dong H. Sexual dimorphism in predisposition to Alzheimer's disease. Neurobiol Aging. 2018;70:308–324. [Europe PMC free article] [Abstract] [Google Scholar]
26. Fiest K.M., Roberts J.I., Maxwell C.J., Hogan D.B., Smith E.E., Frolkis A. The prevalence and incidence of dementia due to Alzheimer's disease: a systematic review and meta-analysis. Can J Neurol Sci. 2016;43(Suppl. 1):S51–S82. [Abstract] [Google Scholar]
27. Irvine K., Laws K.R., Gale T.M., Kondel T.K. Greater cognitive deterioration in women than men with Alzheimer's disease: a meta analysis. J Clin Exp Neuropsychol. 2012;34:989–998. [Abstract] [Google Scholar]
28. Lee I.M., Shiroma E.J., Lobelo F., Puska P., Blair S.N., Katzmarzyk P.T. Effect of physical inactivity on major non-communicable diseases worldwide: an analysis of burden of disease and life expectancy. The Lancet. 2012;380:219–229. [Europe PMC free article] [Abstract] [Google Scholar]
29. Laurin D., Verreault R., Lindsay J., MacPherson K., Rockwood K. Physical activity and risk of cognitive impairment and dementia in elderly persons. Arch Neurol. 2001;58:127–144. [Abstract] [Google Scholar]
30. Sofi F., Valecchi D., Bacci D., Abbate R., Gensini G.F., Casini A. Physical activity and risk of cognitive decline: a meta-analysis of prospective studies. J Intern Med. 2011;269:107–117. [Abstract] [Google Scholar]
31. Spirduso W.W., Clifford P. Replication of age and physical activity effects on reaction and movement time. J Gerontol. 1978;33:26–30. [Abstract] [Google Scholar]
32. De la Rosa A., Solana E., Corpas R., Bartrés-Faz D., Pallàs M., Vina J. Long-term exercise training improves memory in middle-aged men and modulates peripheral levels of BDNF and Cathepsin B. Sci Rep. 2019;9:3337. 10.1038/s41598-019-40040-8. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
33. Erickson K.I., Voss M.W., Prakash R.S., Basak C., Szabo A., Chaddock L. Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci U S A. 2011;108:3017–3022. [Europe PMC free article] [Abstract] [Google Scholar]
34. Colcombe S.J., Erickson K.I., Scalf P.E., Kim J.S., Prakash R., McAuley E. Aerobic exercise training increases brain volume in aging humans. J Gerontol A Biol Sci Med Sci. 2006;61:1166–1170. [Abstract] [Google Scholar]
35. Rikli R.E., Edwards D.J. Effects of a three-year exercise program on motor function and cognitive processing speed in older women. Res Q Exerc Sport. 1991;62:61–67. [Abstract] [Google Scholar]
36. Blumenthal J.A., Emery C.F., Madden D.J., Schniebolk S., Walsh-Riddle M., George L.K. Long-term effects of exercise on psychological functioning in older men and women. J Gerontol. 1991;46:P352–P361. [Abstract] [Google Scholar]
37. Hill R.D., Storandt M., Malley M. The impact of long-term exercise training on psychological function in older adults. J Gerontol. 1993;48:P12–P17. [Abstract] [Google Scholar]
38. Panton L.B., Graves J.E., Pollock M.L., Hagberg J.M., Chen W. Effect of aerobic and resistance training on fractionated reaction time and speed of movement. J Gerontol. 1990;45:M26–M31. [Abstract] [Google Scholar]
39. Landrigan J.F., Bell T., Crowe M., Clay O.J., Mirman D. Lifting cognition: a meta-analysis of effects of resistance exercise on cognition. Psychol Res. 2020;84:1167–1183. [Abstract] [Google Scholar]
40. Northey J.M., Cherbuin N., Pumpa K.L., Smee D.J., Rattray B. Exercise interventions for cognitive function in adults older than 50: a systematic review with meta-analysis. Br J Sport Med. 2018;52:154–160. [Abstract] [Google Scholar]
41. Orgeta V., Mukadam N., Sommerlad A., Livingston G. The Lancet Commission on dementia prevention, intervention, and care: a call for action. Ir J Psychol Med. 2019;36:85–88. [Abstract] [Google Scholar]
42. Hamer M., Chida Y. Physical activity and risk of neurodegenerative disease: a systematic review of prospective evidence. Psychol Med. 2009;39:3–11. [Abstract] [Google Scholar]
43. Buchman A.S., Boyle P.A., Yu L., Shah R.C., Wilson R.S., Bennett D.A. Total daily physical activity and the risk of AD and cognitive decline in older adults. Neurology. 2012;78:1323–1329. [Europe PMC free article] [Abstract] [Google Scholar]
44. Vital T.M., Hernández S.S.S., Pedroso R.V., Teixeira C.V.L., Garuffi M., Stein A.M. Effects of weight training on cognitive functions in elderly with Alzheimer's disease. Dement Neuropsychol. 2012;6:253–259. [Europe PMC free article] [Abstract] [Google Scholar]
45. Cott C.A., Dawson P., Sidani S., Wells D. The effects of a walking/talking program on communication, ambulation, and functional status in residents with Alzheimer disease. Alzheimer Dis Assoc Disord. 2002;16:81–87. [Abstract] [Google Scholar]
46. Toots A., Littbrand H., Boström G., Hörnsten C., Holmberg H., Lundin-Olsson L. Effects of exercise on cognitive function in older people with dementia: a randomized controlled trial. J Alzheimers Dis. 2017;60:323–332. [Europe PMC free article] [Abstract] [Google Scholar]
47. Oken B.S., Zajdel D., Kishiyama S., Flegal K., Dehen C., Haas M. Randomized, controlled, six-month trial of yoga in healthy seniors: effects on cognition and quality of life. Altern Ther Health Med. 2006;12:40–47. [Europe PMC free article] [Abstract] [Google Scholar]
48. Madden D.J., Blumenthal J.A., Allen P.A., Emery C.F. Improving aerobic capacity in healthy older adults does not necessarily lead to improved cognitive performance. Psychol Aging. 1989;4:307–320. [Abstract] [Google Scholar]
49. van Uffelen J.G.Z., Chin A., Paw M.J.M., Hopman-Rock M., van Mechelen W. The effects of exercise on cognition in older adults with and without cognitive decline: a systematic review. Clin J Sport Med. 2008;18:486–500. [Abstract] [Google Scholar]
50. Baker L.D., Frank L.L., Foster-Schubert K., Green P.S., Wilkinson C.W., McTiernan A. Effects of aerobic exercise on mild cognitive impairment: a controlled trial. Arch Neurol. 2010;67:71–79. [Europe PMC free article] [Abstract] [Google Scholar]
51. Nagamatsu L.S., Handy T.C., Hsu C.L., Voss M., Liu-Ambrose T. Resistance training promotes cognitive and functional brain plasticity in seniors with probable mild cognitive impairment. Arch Intern Med. 2012;172:666–668. [Europe PMC free article] [Abstract] [Google Scholar]
52. Blumenthal J.A., Smith P.J., Mabe S., Hinderliter A., Lin P.H., Liao L. Lifestyle and neurocognition in older adults with cognitive impairments: a randomized trial. Neurology. 2019;92:e212–e223. [Europe PMC free article] [Abstract] [Google Scholar]
53. Weuve J., Kang J.H., Manson J.E., Breteler M.M., Ware J.H., Grodstein F. Physical activity, including walking, and cognitive function in older women. JAMA. 2004;292:1454–1461. [Abstract] [Google Scholar]
54. Zheng G., Xia R., Zhou W., Tao J., Chen L. Aerobic exercise ameliorates cognitive function in older adults with mild cognitive impairment: a systematic review and meta-analysis of randomised controlled trials. Br J Sports Med. 2016;50:1443–1450. [Abstract] [Google Scholar]
55. Scherder E.J., Van Paasschen J., Deijen J.B., Van Der Knokke S., Orlebeke J.F., Burgers I. Physical activity and executive functions in the elderly with mild cognitive impairment. Aging Ment Health. 2005;9:272–280. [Abstract] [Google Scholar]
56. Coelho F.G., Santos-Galduroz R.F., Gobbi S., Stella F. Systematized physical activity and cognitive performance in elderly with Alzheimer's dementia: a systematic review. Braz J Psychiatry. 2009;31:163–170. [in Portuguese] [Abstract] [Google Scholar]
57. Friedman R., Tappen R.M. The effect of planned walking on communication in Alzheimer's disease. J Am Geriatr Soc. 1991;39:650–654. [Abstract] [Google Scholar]
58. Rolland Y., Rival L., Pillard F., Lafont C., Rivére D., Albaréde J. Feasibility [corrected] of regular physical exercise for patients with moderate to severe Alzheimer disease. J Nutr Health Aging. 2000;4:109–113. [Abstract] [Google Scholar]
59. Öhman H., Savikko N., Strandberg T.E., Kautiainen H., Raivio M.M., Laakkonen M.L. Effects of exercise on cognition: the Finnish Alzheimer Disease Exercise Trial: a randomized, controlled trial. J Am Geriatr Soc. 2016;64:731–738. [Abstract] [Google Scholar]
60. Heyn P., Abreu B.C., Ottenbacher K.J. The effects of exercise training on elderly persons with cognitive impairment and dementia: a meta-analysis. Arch Phys Med Rehabil. 2004;85:1694–1704. [Abstract] [Google Scholar]
61. Yu F., Kolanowski A.M., Strumpf N.E., Eslinger P.J. Improving cognition and function through exercise intervention in Alzheimer's disease. J Nurs Scholarsh. 2006;38:358–365. [Abstract] [Google Scholar]
62. Vreugdenhil A., Cannell J., Davies A., Razay G. A community-based exercise programme to improve functional ability in people with Alzheimer's disease: a randomized controlled trial. Scand J Caring Sci. 2012;26:12–19. [Abstract] [Google Scholar]
63. Panza G.A., Taylor B.A., MacDonald H V., Johnson B.T., Zaleski A.L., Livingston J. Can exercise improve cognitive symptoms of Alzheimer's disease? J Am Geriatr Soc. 2018;66:487–495. [Abstract] [Google Scholar]
64. Morris J.K., Vidoni E.D., Johnson D.K., Van Sciver A., Mahnken J.D., Honea R.A. Aerobic exercise for Alzheimer's disease: a randomized controlled pilot trial. PLoS One. 2017;12 10.1371/journal.pone.0170547. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
65. Palleschi L., Vetta F., De Gennaro E., Idone G., Sottosanti G., Gianni W. Effect of aerobic training on the cognitive performance of elderly patients with senile dementia of Alzheimer type. Arch Gerontol Geriatr. 1996;22(Suppl. 1):S47–S50. [Abstract] [Google Scholar]
66. Hong S.G., Kim J.H., Jun T.W. Effects of 12-Week resistance exercise on electroencephalogram patterns and cognitive function in the elderly with mild cognitive impairment: a randomized controlled trial. Clin J Sport Med. 2018;28:500–508. [Abstract] [Google Scholar]
67. Yoon D.H., Kang D., Kim H., Kim J.S., Song H.S., Song W. Effect of elastic band-based high-speed power training on cognitive function, physical performance and muscle strength in older women with mild cognitive impairment. Geriatr Gerontol Int. 2017;17:765–772. [Abstract] [Google Scholar]
68. Garuffi M., Costa J.L., Hernández S.S., Vital T.M., Stein A.M., dos Santos J.G. Effects of resistance training on the performance of activities of daily living in patients with Alzheimer's disease. Geriatr Gerontol Int. 2013;13:322–328. [Abstract] [Google Scholar]
69. Fiatarone Singh M.A., Gates N., Saigal N., Wilson G.C., Meiklejohn J., Brodaty H. The study of mental and resistance training (SMART) study—resistance training and/or cognitive training in mild cognitive impairment: a randomized, double-blind, double-sham controlled trial. J Am Med Dir Assoc. 2014;15:873–880. [Abstract] [Google Scholar]
70. Viña J., Sanz-Ros J. Alzheimer's disease: only prevention makes sense. Eur J Clin Invest. 2018;48:e13005. 10.1111/eci.13005. [Abstract] [CrossRef] [Google Scholar]
71. Morris M.C., Tangney C.C., Wang Y., Sacks F.M., Bennett D.A., Aggarwal N.T. MIND diet associated with reduced incidence of Alzheimer's disease. Alzheimers Dement. 2015;11:1007–1014. [Europe PMC free article] [Abstract] [Google Scholar]
72. Szczechowiak K., Diniz B.S., Leszek J. Diet and Alzheimer's dementia – nutritional approach to modulate inflammation. Pharmacol Biochem Behav. 2019;184 10.1016/j.pbb.2019.172743. [Abstract] [CrossRef] [Google Scholar]
73. Broom G.M., Shaw I.C., Rucklidge J.J. The ketogenic diet as a potential treatment and prevention strategy for Alzheimer's disease. Nutrition. 2019;60:118–121. [Abstract] [Google Scholar]
74. Henderson S.T., Vogel J.L., Barr L.J., Garvin F., Jones J.J., Costantini L.C. Study of the ketogenic agent AC-1202 in mild to moderate Alzheimer's disease: a randomized, double-blind, placebo-controlled, multicenter trial. Nutr Metab (Lond) 2009;6:31. 10.1186/1743-7075-6-31. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
75. Pawlosky R.J., Kemper M.F., Kashiwaya Y., King M.T., Mattson M.P., Veech R.L. Effects of a dietary ketone ester on hippocampal glycolytic and tricarboxylic acid cycle intermediates and amino acids in a 3xTgAD mouse model of Alzheimer's disease. J Neurochem. 2017;141:195–207. [Europe PMC free article] [Abstract] [Google Scholar]
76. Halagappa V.K., Guo Z., Pearson M., Matsuoka Y., Cutler R.G., Laferla F.M. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease. Neurobiol Dis. 2007;26:212–220. [Abstract] [Google Scholar]
77. Ngandu T., Lehtisalo J., Solomon A., Levälahti E., Ahtiluoto S., Antikainen R. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): a randomised controlled trial. The Lancet. 2015;385:2255–2263. [Abstract] [Google Scholar]
78. Rabin J.S., Klein H., Kirn D.R., Schultz A.P., Yang H.S., Hampton O. Associations of physical activity and β-amyloid with longitudinal cognition and neurodegeneration in clinically normal older adults. JAMA Neurol. 2019;76:1203–1210. [Europe PMC free article] [Abstract] [Google Scholar]
79. Deslandes A., Moraes H., Ferreira C., Veiga H., Silveira H., Mouta R. Exercise and mental health: many reasons to move. Neuropsychobiology. 2009;59:191–198. [Abstract] [Google Scholar]
80. Eggermont L., Swaab D., Luiten P., Scherder E. Exercise, cognition and Alzheimer's disease: more is not necessarily better. Neurosci Biobehav Rev. 2006;30:562–575. [Abstract] [Google Scholar]
81. van Praag H., Christie B.R., Sejnowski T.J., Gage F.H. Running enhances neurogenesis, learning, and long-term potentiation in mice. Proc Natl Acad Sci U S A. 1999;96:13427–13431. [Europe PMC free article] [Abstract] [Google Scholar]
82. Farmer J., Zhao X., van Praag H., Wodtke K., Gage F.H., Christie B.R. Effects of voluntary exercise on synaptic plasticity and gene expression in the dentate gyrus of adult male Sprague-Dawley rats in vivo. Neuroscience. 2004;124:71–79. [Abstract] [Google Scholar]
83. Cotman C.W., Berchtold N.C. Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci. 2002;25:295–301. [Abstract] [Google Scholar]
84. Colcombe S., Kramer A.F. Fitness effects on the cognitive function of older adults: a meta-analytic study. Psychol Sci. 2003;14:125–130. [Abstract] [Google Scholar]
85. Gligoroska J.P., Manchevska S. The effect of physical activity on cognition- physiological mechanisms. Mater Sociomed. 2012;24:198–202. [Europe PMC free article] [Abstract] [Google Scholar]
86. Adlard P.A., Perreau V.M., Pop V., Cotman C.W. Voluntary exercise decreases amyloid load in a transgenic model of Alzheimer's disease. J Neurosci. 2005;25:4217–4221. [Europe PMC free article] [Abstract] [Google Scholar]
87. Ohia-Nwoko O., Montazari S., Lau Y.S., Eriksen J.L. Long-term treadmill exercise attenuates tau pathology in P301S tau transgenic mice. Mol Neurodegener. 2014;9:54. 10.1186/1750-1326-9-54. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
88. Leem Y.H., Lim H.J., Shim S.B., Cho J.Y., Kim B.S., Han P.L. Repression of tau hyperphosphorylation by chronic endurance exercise in aged transgenic mouse model of tauopathies. J Neurosci Res. 2009;87:2561–2570. [Abstract] [Google Scholar]
89. Um H.S., Kang E.B., Koo J.H., Kim H.T., Lee Jin, Kim E.J. Treadmill exercise represses neuronal cell death in an aged transgenic mouse model of Alzheimer's disease. Neurosci Res. 2011;69:161–173. [Abstract] [Google Scholar]
90. Tapia-Rojas C., Aranguiz F., Varela-Nallar L., Inestrosa N.C. Voluntary running attenuates memory loss, decreases neuropathological changes and induces neurogenesis in a mouse model of Alzheimer's disease. Brain Pathol. 2016;26:62–74. [Abstract] [Google Scholar]
91. Belarbi K., Burnouf S., Fernandez-Gomez F.J., Laurent C., Lestavel S., Figeac M. Beneficial effects of exercise in a transgenic mouse model of Alzheimer's disease-like Tau pathology. Neurobiol Dis. 2011;43:486–494. [Abstract] [Google Scholar]
92. Yuede C.M., Zimmerman S.D., Dong H., Kling M.J., Bero A.W., Holtzman D.M. Effects of voluntary and forced exercise on plaque deposition, hippocampal volume, and behavior in the Tg2576 mouse model of Alzheimer's disease. Neurobiol Dis. 2009;35:426–432. [Europe PMC free article] [Abstract] [Google Scholar]
93. Richter H., Ambrée O., Lewejohann L., Herring A., Keyvani K., Paulus W. Wheel-running in a transgenic mouse model of Alzheimer's disease: protection or symptom? Behav Brain Res. 2008;190:74–84. [Abstract] [Google Scholar]
94. García-Mesa Y., López-Ramos J.C., Giménez-Llort L., Revilla S., Guerra R., Gruart A. Physical exercise protects against Alzheimer's disease in 3xTg-AD mice. J Alzheimer's Dis. 2011;24:421–454. [Abstract] [Google Scholar]
95. Wolf S.A., Kronenberg G., Lehmann K., Blankenship A., Overall R., Staufenbiel M. Cognitive and physical activity differently modulate disease progression in the amyloid precursor protein (APP)-23 model of Alzheimer's disease. Biol Psychiatry. 2006;60:1314–1323. [Abstract] [Google Scholar]
96. Liang K.Y., Mintun M.A., Fagan A.M., Goate A.M., Bugg J.M., Holtzman D.M. Exercise and Alzheimer's disease biomarkers in cognitively normal older adults. Ann Neurol. 2010;68:311–318. [Europe PMC free article] [Abstract] [Google Scholar]
97. Stillman C.M., Lopez O.L., Becker J.T., Kuller L.H., Mehta P.D., Tracy R.P. Physical activity predicts reduced plasma β amyloid in the Cardiovascular Health Study. Ann Clin Transl Neurol. 2017;4:284–291. [Europe PMC free article] [Abstract] [Google Scholar]
98. Franceschi C., Capri M., Monti D., Giunta S., Olivieri F., Sevini F. Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mech Ageing Dev. 2007;128:92–105. [Abstract] [Google Scholar]
99. Giunta B., Fernandez F., Nikolic W.V., Obregon D., Rrapo E., Town T. Inflammaging as a prodrome to Alzheimer's disease. J Neuroinflammation. 2008;5:51. 10.1186/1742-2094-5-51. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
100. Gomez-Cabrera M.C., Viña J., Ji L.L. Role of redox signaling and inflammation in skeletal muscle adaptations to training. Antioxidants(Basel) 2016;5:E48. 10.3390/antiox5040048. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
101. Jensen C.S., Bahl J.M., Østergaard L.B., Høgh P., Wermuth L., Heslegrave A. Exercise as a potential modulator of inflammation in patients with Alzheimer's disease measured in cerebrospinal fluid and plasma. Exp Gerontol. 2019;121:91–98. [Abstract] [Google Scholar]
102. Nichol K.E., Poon W.W., Parachikova A.I., Cribbs D.H., Glabe C.G., Cotman C.W. Exercise alters the immune profile in Tg2576 Alzheimer mice toward a response coincident with improved cognitive performance and decreased amyloid. J Neuroinflammation. 2008;5:13. 10.1186/1742-2094-5-13. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
103. Gomes da Silva S., Simões P.S.R., Mortara R.A., Scorza F.A., Cavalheiro E.A., da Graça Naffah-Mazzacoratti M. Exercise-induced hippocampal anti-inflammatory response in aged rats. J Neuroinflammation. 2013;10:61. 10.1186/1742-2094-10-61. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
104. Barrientos R.M., Frank M.G., Crysdale N.Y., Chapman T.R., Ahrendsen J.T., Day H.E. Little exercise, big effects: reversing aging and infection-induced memory deficits, and underlying processes. J Neurosci. 2011;31:11578–11586. [Europe PMC free article] [Abstract] [Google Scholar]
105. Yu Z., Ye X., Wang J., Qi Q., Franco O.H., Rennie K.L. Associations of physical activity with inflammatory factors, adipocytokines, and metabolic syndrome in middle-aged and older Chinese people. Circulation. 2009;119:2969–2977. [Abstract] [Google Scholar]
106. Reuben D.B., Judd-Hamilton L., Harris T.B., Seeman T.E. The associations between physical activity and inflammatory markers in high-functioning older persons: MacArthur studies of successful aging. J Am Geriatr Soc. 2003;51:1125–1130. [Abstract] [Google Scholar]
107. Colbert L.H., Visser M., Simonsick E.M., Tracy R.P., Newman A.B., Kritchevsky S.B. Physical activity, exercise, and inflammatory markers in older adults: findings from the Health, Aging and Body Composition Study. J Am Geriatr Soc. 2004;52:1098–1104. [Abstract] [Google Scholar]
108. Wannamethee S.G., Lowe G.D., Whincup P.H., Rumley A., Walker M., Lennon L. Physical activity and hemostatic and inflammatory variables in elderly men. Circulation. 2002;105:1785–1790. [Abstract] [Google Scholar]
109. Taaffe D.R., Harris T.B., Ferrucci L., Rowe J., Seeman T.E. Cross-sectional and prospective relationships of interleukin-6 and C-reactive protein with physical performance in elderly persons: MacArthur studies of successful aging. J Gerontol A Biol Sci Med Sci. 2000;55:M709–M715. [Abstract] [Google Scholar]
110. Elosua R., Bartali B., Ordovas J.M., Corsi A.M., Lauretani F., Ferrucci L. Association between physical activity, physical performance, and inflammatory biomarkers in an elderly population: the InCHIANTI study. J Gerontol A Biol Sci Med Sci. 2005;60:760–767. [Abstract] [Google Scholar]
111. Ravaglia G., Forti P., Maioli F., Arnone G., Pantieri G., Cocci C. The clock-drawing test in elderly Italian community dwellers: associations with sociodemographic status and risk factors for vascular cognitive impairment. Dement Geriatr Cogn Disord. 2003;16:287–295. [Abstract] [Google Scholar]
112. Baune B.T., Ponath G., Golledge J., Varga G., Arolt V., Rothermundt M. Association between IL-8 cytokine and cognitive performance in an elderly general population—the MEMO-Study. Neurobiol Aging. 2008;29:937–944. [Abstract] [Google Scholar]
113. Alley D.E., Crimmins E.M., Karlamangla A., Hu P., Seeman T.E. Inflammation and rate of cognitive change in high-functioning older adults. J Gerontol A Biol Sci Med Sci. 2008;63:50–55. [Europe PMC free article] [Abstract] [Google Scholar]
114. Yaffe K., Lindquist K., Penninx B.W., Simonsick E.M., Pahor M., Kritchevsky S. Inflammatory markers and cognition in well-functioning African-American and white elders. Neurology. 2003;61:76–80. [Abstract] [Google Scholar]
115. Teunissen C.E., van Boxtel M.P., Bosma H., Bosmans E., Delanghe J., De Bruijn C. Inflammation markers in relation to cognition in a healthy aging population. J Neuroimmunol. 2003;134:142–150. [Abstract] [Google Scholar]
116. Weaver J.D., Huang M.H., Albert M., Harris T., Rowe J.W., Seeman T.E. Interleukin-6 and risk of cognitive decline: MacArthur studies of successful aging. Neurology. 2002;59:371–378. [Abstract] [Google Scholar]
117. Cobb J.L., Wolf P.A., Au R., White R., D'Agostino R.B. The effect of education on the incidence of dementia and Alzheimer's disease in the Framingham Study. Neurology. 1995;45:1707–1712. [Abstract] [Google Scholar]
118. Yan H., Kuroiwa A., Tanaka H., Shindo M., Kiyonaga A., Nagayama A. Effect of moderate exercise on immune senescence in men. Eur J Appl Physiol. 2001;86:105–111. [Abstract] [Google Scholar]
119. Thijssen D.H.J., Vos J.B., Verseyden C., van Zonneveld A.J., Smits P., Sweep F.C. Haematopoietic stem cells and endothelial progenitor cells in healthy men: effect of aging and training. Aging Cell. 2006;5:495–503. [Abstract] [Google Scholar]
120. Woods J.A., Ceddia M.A., Wolters B.W., Evans J.K., Lu Q., McAuley E. Effects of 6 months of moderate aerobic exercise training on immune function in the elderly. Mech Ageing Dev. 1999;109:1–19. [Abstract] [Google Scholar]
121. Fairey A.S., Courneya K.S., Field C.J., Bell G.J., Jones L.W., Mackey J.R. Randomized controlled trial of exercise and blood immune function in postmenopausal breast cancer survivors. J Appl Physiol(1985) 2005;98:1534–1540. [Abstract] [Google Scholar]
122. Sellami M., Gasmi M., Denham J., Hayes L.D., Stratton D., Padulo J. Effects of acute and chronic exercise on immunological parameters in the elderly aged: can physical activity counteract the effects of aging? Front Immunol. 2018;9:2187. 10.3389/fimmu.2018.02187. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
123. Birling M.C., Price J. Influence of growth factors on neuronal differentiation. Curr Opin Cell Biol. 1995;7:878–884. [Abstract] [Google Scholar]
124. Thoenen H. Neurotrophins and neuronal plasticity. Science. 1995;270:593–598. [Abstract] [Google Scholar]
125. Lewin G.R., Barde Y.A. Physiology of the neurotrophins. Annu Rev Neurosci. 1996;19:289–317. [Abstract] [Google Scholar]
126. Huang E.J., Reichardt L.F. Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci. 2001;24:677–736. [Europe PMC free article] [Abstract] [Google Scholar]
127. Tartaglia N., Du J., Tyler W.J., Neale E., Pozzo-Miller L., Lu B. Protein synthesis-dependent and -independent regulation of hippocampal synapses by brain-derived neurotrophic factor. J Biol Chem. 2001;276:37585–37593. [Abstract] [Google Scholar]
128. Dinoff A., Herrmann N., Swardfager W., Liu C.S., Sherman C., Chan S. The effect of exercise training on resting concentrations of peripheral brain-cerived neurotrophic factor (BDNF): a meta-analysis. PLoS One. 2016;11 10.1371/journal.pone.0163037. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
129. Berchtold N.C., Chinn G., Chou M., Kesslak J.P., Cotman C.W. Exerciseprimes a molecular memory for brain-derived neurotrophic factor protein induction in the rat hippocampus. Neuroscience. 2005;133:853–861. [Abstract] [Google Scholar]
130. Maejima H., Kanemura N., Kokubun T., Murata K., Takayanagi K. Exercise enhances cognitive function and neurotrophin expression in the hippocampus accompanied by changes in epigenetic programming in senescence-accelerated mice. Neurosci Lett. 2018;665:67–73. [Abstract] [Google Scholar]
131. Winter B., Breitenstein C., Mooren F.C., Voelker K., Fobker M., Lechtermann A. High impact running improves learning. Neurobiol Learn Mem. 2007;87:597–609. [Abstract] [Google Scholar]
132. Rasmussen P., Brassard P., Adser H., Pedersen M.V., Leick L., Hart E. Evidence for a release of brain-derived neurotrophic factor from the brain during exercise. Exp Physiol. 2009;94:1062–1069. [Abstract] [Google Scholar]
133. Kim T.W., Choi H.H., Chung Y.R. Treadmill exercise alleviates impairment of cognitive function by enhancing hippocampal neuroplasticity in the high-fat diet-induced obese mice. J Exerc Rehabil. 2016;12:156–162. [Europe PMC free article] [Abstract] [Google Scholar]
134. García-Mesa Y., Pareja-Galeano H., Bonet-Costa V., Revilla S., Gómez-Cabrera M.C., Gambini J. Physical exercise neuroprotects ovariectomized 3xTg-AD mice through BDNF mechanisms. Psychoneuroendocrinology. 2014;45:154–166. [Abstract] [Google Scholar]
135. Pareja-Galeano H., Brioche T., Sanchis-Gomar F., Montal A., Jovaní C., Martínez-Costa C. Impact of exercise training on neuroplasticity-related growth factors in adolescents. J Musculoskelet Neuronal Interact. 2013;13:368–371. [Abstract] [Google Scholar]
136. Klein A.B., Williamson R., Santini M.A., Clemmensen C., Ettrup A., Rios M. Blood BDNF concentrations reflect brain-tissue BDNF levels across species. Int J Neuropsychopharmacol. 2011;14:347–353. [Abstract] [Google Scholar]
137. Choi S.H., Bylykbashi E., Chatila Z.K., Lee S.W., Pulli B., Clemenson G.D. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer's mouse model. Science. 2018;361:eaan8821. 10.1126/science.aan8821. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
138. Holsinger R.M., Schnarr J., Henry P., Castelo V.T., Fahnestock M. Quantitation of BDNF mRNA in human parietal cortex by competitive reverse transcription-polymerase chain reaction: decreased levels in Alzheimer's disease. Brain Res Mol Brain Res. 2000;76:347–354. [Abstract] [Google Scholar]
139. Phillips H.S., Hains J.M., Armanini M., Laramee G.R., Johnson S.A., Winslow J.W. BDNF mRNA is decreased in the hippocampus of individuals with Alzheimer's disease. Neuron. 1991;7:695–702. [Abstract] [Google Scholar]
140. Gold S.M., Schulz K.H., Hartmann S., Mladek M., Lang U.E., Hellweg R. Basal serum levels and reactivity of nerve growth factor and brain-derived neurotrophic factor to standardized acute exercise in multiple sclerosis and controls. J Neuroimmunol. 2003;138:99–105. [Abstract] [Google Scholar]
141. Zoladz J.A., Pilc A., Majerczak J., Grandys M., Zapart-Bukowska J., Duda K. Endurance training increases plasma brain-derived neurotrophic factor concentration in young healthy men. J Physiol Pharmacol. 2008;59(Suppl. 7):S119–S132. [Abstract] [Google Scholar]
142. Tang S.W., Chu E., Hui T., Helmeste D., Law C. Influence of exercise on serum brain-derived neurotrophic factor concentrations in healthy human subjects. Neurosci Lett. 2008;431:62–65. [Abstract] [Google Scholar]
143. Seifert T., Brassard P., Wissenberg M., Rasmussen P., Nordby P., Stallknecht B. Endurance training enhances BDNF release from the human brain. Am J Physiol Regul Integr Comp Physiol. 2010;298:R372–R377. [Abstract] [Google Scholar]
144. Rojas Vega S., Strüder H.K., Vera Wahrmann B., Schmidt A., Bloch W., Hollmann W. Acute BDNF and cortisol response to low intensity exercise and following ramp incremental exercise to exhaustion in humans. Brain Res. 2006;1121:59–65. [Abstract] [Google Scholar]
145. Cho H.C., Kim J., Kim S., Son Y.H., Lee N., Jung S.H. The concentrations of serum, plasma and platelet BDNF are all increased by treadmill VO₂max performance in healthy college men. Neurosci Lett. 2012;519:78–83. [Abstract] [Google Scholar]
146. Brunelli A., Dimauro I., Sgrò P., Emerenziani G.P., Magi F., Baldari C. Acute exercise modulates BDNF and pro-BDNF protein content in immune cells. Med Sci Sport Exerc. 2012;44:1871–1880. [Abstract] [Google Scholar]
147. Griffin É.W., Mullally S., Foley C., Warmington S.A., O'Mara S.M., Kelly A.M. Aerobic exercise improves hippocampal function and increases BDNF in the serum of young adult males. Physiol Behav. 2011;104:934–941. [Abstract] [Google Scholar]
148. Ferris L.T., Williams J.S., Shen C.L. The effect of acute exercise on serum brain-derived neurotrophic factor levels and cognitive function. Med Sci Sports Exerc. 2007;39:728–734. [Abstract] [Google Scholar]
149. Vaughan S., Wallis M., Polit D., Steele M., Shum D., Morris N. The effects of multimodal exercise on cognitive and physical functioning and brain-derived neurotrophic factor in older women: a randomised controlled trial. Age Ageing. 2014;43:623–629. [Abstract] [Google Scholar]
150. Weinstein G., Beiser A.S., Choi S.H., Preis S.R., Chen T.C., Vorgas D. Serum brain-derived neurotrophic factor and the risk for dementia: the Framingham heart study. JAMA Neurol. 2014;71:55. 10.1001/jamaneurol.2013.4781. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
151. Coelho F.G., Vital T.M., Stein A.M., Arantes F.J., Rueda A.V., Camarini R. Acute aerobic exercise increases brain-derived neurotrophic factor levels in elderly with Alzheimer's disease. J Alzheimers Dis. 2014;39:401–408. [Abstract] [Google Scholar]
152. Bertsch K., Hagemann D., Hermes M., Walter C., Khan R., Naumann E. Resting cerebral blood flow, attention, and aging. Brain Res. 2009;1267:77–88. [Abstract] [Google Scholar]
153. Hagstadius S., Risberg J. Regional cerebral blood flow characteristics and variations with age in resting normal subjects. Brain Cogn. 1989;10:28–43. [Abstract] [Google Scholar]
154. Harada C.N., Natelson Love M.C., Triebel K.L. Normal cognitive aging. Clin Geriatr Med. 2013;29:737–752. [Europe PMC free article] [Abstract] [Google Scholar]
155. Park D.C., Polk T.A., Mikels J.A., Taylor S.F., Marshuetz C. Cerebral aging: integration of brain and behavioral models of cognitive function. Dialogues Clin Neurosci. 2001;3:151–165. [Europe PMC free article] [Abstract] [Google Scholar]
156. Heo S., Prakash R.S., Voss M.W., Erickson K.I., Ouyang C., Sutton B.P. Resting hippocampal blood flow, spatial memory and aging. Brain Res. 2010;1315:119–127. [Europe PMC free article] [Abstract] [Google Scholar]
157. Parkes L.M., Rashid W., Chard D.T., Tofts P.S. Normal cerebral perfusion measurements using arterial spin labeling: reproducibility, stability, and age and gender effects. Magn Reson Med. 2004;51:736–743. [Abstract] [Google Scholar]
158. Leenders K.L., Perani D., Lammertsma A.A., Heather J.D., Buckingham P., Healy M.J. Cerebral blood flow, blood volume and oxygen utilization. Normal values and effect of age. Brain. 1990;113:27–47. [Abstract] [Google Scholar]
159. Wolters F.J., Zonneveld H.I., Hofman A., van der Lugt A., Koudstaal P.J., Vernooij M.W. Cerebral perfusion and the risk of dementia: a population-based study. Circulation. 2017;136:719–728. [Abstract] [Google Scholar]
160. De Vis J.B., Peng S.L., Chen X., Li Y., Liu P., Sur S. Arterial-spin-labeling (ASL) perfusion MRI predicts cognitive function in elderly individuals: a 4-year longitudinal study. J Magn Reson Imaging. 2018;48:449–458. [Europe PMC free article] [Abstract] [Google Scholar]
161. Zhang N., Gordon M.L., Goldberg T.E. Cerebral blood flow measured by arterial spin labeling MRI at resting state in normal aging and Alzheimer's disease. Neurosci Biobehav Rev. 2017;72:168–175. [Abstract] [Google Scholar]
162. Kalaria R.N. Cerebral vessels in ageing and Alzheimer's disease. Pharmacol Ther. 1996;72:193–214. [Abstract] [Google Scholar]
163. Austin B.P., Nair V.A., Meier T.B., Xu G., Rowley H.A., Carlsson C.M. Effects of hypoperfusion in Alzheimer's disease. J Alzheimers Dis. 2011;26:123–133. [Europe PMC free article] [Abstract] [Google Scholar]
164. Du A.T., Jahng G.H., Hayasaka S., Kramer J.H., Rosen H.J., Gorno-Tempini M.L. Hypoperfusion in frontotemporal dementia and Alzheimer disease by arterial spin labeling MRI. Neurology. 2006;67:1215–1220. [Europe PMC free article] [Abstract] [Google Scholar]
165. Asllani I., Habeck C., Scarmeas N., Borogovac A., Brown T.R., Stern Y. Multivariate and univariate analysis of continuous arterial spin labeling perfusion MRI in Alzheimer's disease. J Cereb Blood Flow Metab. 2008;28:725–736. [Europe PMC free article] [Abstract] [Google Scholar]
166. Binnewijzend M.A., Kuijer J.P., Benedictus M.R., van der Flier W.M., Wink A.M., Wattjes M.P. Cerebral blood flow measured with 3D pseudocontinuous arterial spin-labeling MR imaging in Alzheimer disease and mild cognitive impairment: a marker for disease severity. Radiology. 2013;267:221–230. [Abstract] [Google Scholar]
167. Johnson N.A., Jahng G.H., Weiner M.W., Miller B.L., Chui H.C., Jagust W.J. Pattern of cerebral hypoperfusion in Alzheimer disease and mild cognitive impairment measured with arterial spin-labeling MR imaging: initial experience. Radiology. 2005;234:851–859. [Europe PMC free article] [Abstract] [Google Scholar]
168. Dietrich H.H., Xiang C., Han B.H., Zipfel G.J., Holtzman D.M. Soluble amyloid-beta, effect on cerebral arteriolar regulation and vascular cells. Mol Neurodegener. 2010;5:15. 10.1186/1750-1326-5-15. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
169. Niwa K., Younkin L., Ebeling C., Turner S.K., Westaway D., Younkin S. Aβ1-40-related reduction in functional hyperemia in mouse neocortex during somatosensory activation. Proc Natl Acad Sci U S A. 2000;97:9735–9740. [Europe PMC free article] [Abstract] [Google Scholar]
170. Suo Z., Humphrey J., Kundtz A., Sethi F., Placzek A., Crawford F. Soluble Alzheimers β-amyloid constricts the cerebral vasculature in vivo. Neurosci Lett. 1998;257:77–80. [Abstract] [Google Scholar]
171. Taddei S., Galetta F., Virdis A., Ghiadoni L., Salvetti G., Franzoni F. Physical activity prevents age-related impairment in nitric oxide availability in elderly athletes. Circulation. 2000;101:2896–2901. [Abstract] [Google Scholar]
172. Burdette J.H., Laurienti P.J., Espeland M.A., Morgan A., Telesford Q., Vechlekar C.D. Using network science to evaluate exercise-associated brain changes in older adults. Front Aging Neurosci. 2010;2:23. 10.3389/fnagi.2010.00023. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
173. Ainslie P.N., Cotter J.D., George K.P., Lucas S., Murrell C., Shave R. Elevation in cerebral blood flow velocity with aerobic fitness throughout healthy human ageing. J Physiol. 2008;586:4005–4010. [Abstract] [Google Scholar]
174. Chapman S.B., Aslan S., Spence J.S., Defina L.F., Keebler M.W., Didehbani N. Shorter term aerobic exercise improves brain, cognition, and cardiovascular fitness in aging. Front Aging Neurosci. 2013;5:75. 10.3389/fnagi.2013.00075. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
175. Pereira A.C., Huddleston D.E., Brickman A.M., Sosunov A.A., Hen R., McKhann G.M. An in vivo correlate of exercise-induced neurogenesis in the adult dentate gyrus. Proc Natl Acad Sci U S A. 2007;104:5638–5643. [Europe PMC free article] [Abstract] [Google Scholar]
176. Alfini A.J., Weiss L.R., Leitner B.P., Smith T.J., Hagberg J.M., Smith J.C. Hippocampal and cerebral blood flow after exercise cessation in master athletes. Front Aging Neurosci. 2016;8:184. 10.3389/fnagi.2016.00184. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
177. Sobol N.A., Hoffmann K., Frederiksen K.S., Vogel A., Vestergaard K., Brændgaard H. Effect of aerobic exercise on physical performance in patients with Alzheimer's disease. Alzheimer's Dement. 2016;12:1207–1215. [Abstract] [Google Scholar]
178. van der Kleij L.A., Petersen E.T., Siebner H.R., Hendrikse J., Frederiksen K.S., Sobol N.A. The effect of physical exercise on cerebral blood flow in Alzheimer's disease. NeuroImage Clin. 2018;20:650–654. [Europe PMC free article] [Abstract] [Google Scholar]
179. Steen Jensen C., Portelius E., Siersma V., Høgh P., Wermuth L., Blennow K. Cerebrospinal fluid amyloid Beta and tau concentrations are not modulated by 16 weeks of moderate- to high-intensity physical exercise in patients with Alzheimer disease. Dement Geriatr Cogn Disord. 2016;42:146–158. [Abstract] [Google Scholar]
180. Guerrieri D., van Praag H. Exercise-mimetic AICAR transiently benefits brain function. Oncotarget. 2015;6:18293–18313. [Europe PMC free article] [Abstract] [Google Scholar]
181. Valenzuela P.L., Castillo-García A., Morales J.S., Izquierdo M., Serra-Rexach J.A., Santos-Lozano A. Physical exercise in the oldest old. Compr Physiol. 2019;9:1281–1304. [Abstract] [Google Scholar]

Articles from Journal of Sport and Health Science are provided here courtesy of Shanghai University of Sport

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/75498171
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/75498171

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1016/j.jshs.2020.01.004

Supporting
Mentioning
Contrasting
3
170
0

Article citations


Go to all (174) article citations

Funding 


Funders who supported this work.

Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (1)

Conselleria de Sanitat Universal i Salut Pública

    European Commission (4)

    Federación Española de Enfermedades Raras (2)

    Instituto de Salud Carlos III

      Ministerio de Educación, Cultura y Deporte (1)