Acessibilidade / Reportar erro

Interaction between physical exercise and APOE gene polymorphism on cognitive function in older people

Abstract

We aimed to present an overview of the literature regarding the interaction between physical exercise and APOE gene polymorphism on cognitive function, particularly in patients with Alzheimer's disease (AD). Firstly, this review focused on the effect of the physical exercise on cognitive function, regardless of APOE gene polymorphism. Some studies have shown that a high level of cardiorespiratory fitness is associated with less neuronal damage with an improvement in memory score tests whereas other studies failed to detect any association between physical exercise and cognitive improvement either in healthy individuals or patients with AD. Taken together, standardized protocols and more longitudinal studies are required to provide a better insight into the effects of physical exercise on cognitive function. Although there is no agreement in the literature regarding the effects of physical exercise on cognitive function, it is well established that it improves social interaction and the feeling of well-being, thereby positively contributing to the quality of life of the elderly. Regarding the influence of physical exercise on cognitive function in APOE ε4 allele carriers, the data trend shows that the carriers of allele ε4 for APOE gene were more responsive to the beneficial effects of physical exercise on cognitive function compared with non-carriers. Nevertheless, studies with larger sample sizes will provide more accuracy about this relationship.

Alzheimer’s disease; Exercise; Cognition; Apolipoproteins E; Genetic polymorphism; Aged


Introduction

Advances in biomedical research have resulted in higher life expectancy (11. World health organization. World health organization (WHO). Available at https://www.who.int/ageing/sdgs/en/. accessed March 15, 2020.
https://www.who.int/ageing/sdgs/en/...
). On the other hand, increased longevity has led to a rise in the prevalence of neurodegenerative diseases, which are associated with high costs in the health care system. In addition, the diagnosis of some of these diseases is very difficult and several causal factors are involved (22. Reitz C, Mayeux R. Alzheimer disease: epidemiology, diagnostic criteria, risk factors, and biomarkers. Biochem Pharmacol 2014; 88: 640–651, doi: 10.1016/j.bcp.2013.12.024.
https://doi.org/10.1016/j.bcp.2013.12.02...
). Several types of dementia exist and Alzheimer's disease (AD) is the most prevalent one (approximately 60-80% of the cases) followed by vascular dementia (20-30%) and frontal-temporal dementia (10%) (33. Ferretti C, Sarti FM, Nitrini R, Ferreira FF, Brucki SMD. An assessment of direct and indirect costs of dementia in Brazil. PLoS One 2018; 13: e0193209, doi: 10.1371/journal.pone.0193209.
https://doi.org/10.1371/journal.pone.019...
). Although many studies have revealed the neurobiology of AD, much is yet to be discovered related to the underlying mechanisms by which this disease affects neural regulation and its function.

Evidence has shown that APOE-ε4 carriers have an increased risk for AD, with heterozygous carriers of one ε4 allele being 3-4 times more likely to develop this neurodegenerative disorder than non-carriers, and with the risk for homozygous carriers being even higher (44. Najm R, Jones EA, Huang Y. Apolipoprotein E4, inhibitory network dysfunction, and Alzheimer's disease. Mol Neurodegener 2019; 14: 24, doi: 10.1186/s13024-019-0324-6.
https://doi.org/10.1186/s13024-019-0324-...
).

The practice of physical exercise is considered a useful intervention for preventing the decline in cognitive function or for attenuating the risk factors for AD such as arterial hypertension, dyslipidemias, type II diabetes mellitus, and metabolic syndrome (33. Ferretti C, Sarti FM, Nitrini R, Ferreira FF, Brucki SMD. An assessment of direct and indirect costs of dementia in Brazil. PLoS One 2018; 13: e0193209, doi: 10.1371/journal.pone.0193209.
https://doi.org/10.1371/journal.pone.019...
). However, the beneficial effect of physical exercise on AD is not clear. Given that pharmacological treatments have been ineffective both in the prevention and attenuation of the progression of AD (55. Reiss AB, Arain HA, Stecker MM, Siegart NM, Kasselman LJ. Amyloid toxicity in Alzheimer's disease. Rev Neurosci 2018; 29: 613–627, doi: 10.1515/revneuro-2017-0063.
https://doi.org/10.1515/revneuro-2017-00...
), it is important to examine how a lifestyle intervention such as physical exercise would mitigate the deleterious effects of AD on daily activities in an attempt to improve the quality of life of the elderly. Therefore, this review aimed to address the interaction between physical exercise and the presence of polymorphism in APOE on cognitive function in older people.

In order to provide a clinical basis for AD, we summarized some concepts that are currently accepted, and subsequently, we focused on the current knowledge about the possible effects of physical exercise on cognitive function in APOΕε4 allele carriers.

Material and Methods

Search strategy

The methodological design of this study consisted of the search and analysis of articles that verified the effects of physical exercise on cognitive function in APOEε4 allele carriers, particularly in patients with AD. The bibliographic search was conducted on the following databases: Web of Science, Pubmed, Biological Abstracts, PsycINFO, and Scopus, from 2000 to 2020.

The following keywords and Boolean operators were used: physical exercise OR physical activity OR exercise OR training AND cognition OR genetic polymorphism OR apolipoprotein E AND Alzheimer’s disease OR aged OR older OR elderly. Besides the database search, a manual search was carried out on the references from the selected articles. The search was conducted in January of 2020. Afterward, articles were analyzed in the following order: 1) title analysis; 2) abstract analysis; 3) whole article analysis; and 4) selection of articles.

Inclusion criteria

Inclusion criteria were i) human studies and ii) interventions with physical exercise/physical activity, excluding any diet intervention and/or supplementation.

Results and Discussion

After a search based on the established criteria, 1,548 articles were found. In the first analysis, 828 articles were excluded as the title was not related to the aim of this study, and 664 articles were excluded by their abstracts. Thus, the reading of the 56 selected articles was performed for this review.

Pathophysiology of AD

AD, accounting for 60-80% of all cases of dementia, is characterized by progressive loss of cognitive function, changes in behavior, and loss of motor capacity, thereby affecting the patient's quality of life (66. Lehert P, Villaseca P, Hogervorst E, Maki PM, Henderson VW. Individually modifiable risk factors to ameliorate cognitive aging: a systematic review and meta-analysis. Climacteric 2015; 18: 678–689, doi: 10.3109/13697137.2015.1078106.
https://doi.org/10.3109/13697137.2015.10...
,77. Raz L, Knoefel J, Bhaskar K. The neuropathology and cerebrovascular mechanisms of dementia. J Cereb Blood Flow Metab 2016; 36: 172–186, doi: 10.1038/jcbfm.2015.164.
https://doi.org/10.1038/jcbfm.2015.164...
). The causes of AD are unknown, and its etiology is multifactorial involving genetic, epigenetic, and environmental factors (88. Bertram L, Lill CM, Tanzi RE. The genetics of Alzheimer's disease: back to the future. Neuron 2010; 68: 270–281, doi: 10.1016/j.neuron.2010.10.013.
https://doi.org/10.1016/j.neuron.2010.10...
,99. Chouliaras L, Rutten BP, Kenis G, Peerbooms O, Visser PJ, Verhey F, et al. Epigenetic regulation in the pathophysiology of Alzheimer's disease. Prog Neurobiol 2010; 90: 498–510, doi: 10.1016/j.pneurobio.2010.01.002.
https://doi.org/10.1016/j.pneurobio.2010...
). The most widely accepted hypothesis for the pathogenesis of AD is that the accumulation of β-amyloid peptide (βA) in the brain leads to neurotoxicity. βA is derived from the amyloid precursor protein (APP) via two pathways - one involving the enzyme α-secretase that produces SAPPα, which plays a neuroprotective role, and another involving γ-secretase that produces βA - and its degradation is catalyzed by the enzyme neprilysin. Under physiological conditions, the balance between βA production and degradation is maintained by the action of this enzyme and an imbalance leads to its accumulation and a decline in cognitive function and neuronal death (1010. Turner AJ, Fisk L, Nalivaeva NN. Targeting amyloid-degrading enzymes as therapeutic strategies in neurodegeneration. Ann NY Acad Sci 2004; 1035: 1–20, doi: 10.1196/annals.1332.001.
https://doi.org/10.1196/annals.1332.001...
). Accumulation of hyperphosphorylated neurofibrillary tangles of tau proteins was observed parallel to the accumulation of βA. Tau, a protein found in the axons, binds to microtubules and promotes their assembly and stability, and its phosphorylation is regulated by the balanced action of multiple enzymes. In AD, hyperphosphorylation of tau results in the formation of neurofibrillary tangles that compromise axonal transport and neuronal function. Accumulation of extracellular βA and tau hyperphosphorylation in the neuron result in progressive cerebral atrophy, with neuronal degeneration and dementia (1111. Iqbal K, Liu F, Gong CX. Recent developments with tau-based drug discovery. Expert Opin Drug Discov 2018; 13: 399–410, doi: 10.1080/17460441.2018.1445084.
https://doi.org/10.1080/17460441.2018.14...
). Please see Figure 1 for more details.

Figure 1
Current hypothesis of the physiopathological mechanisms involved in Alzheimer's disease.

AD can be classified as familial (1-5% of cases) or sporadic (95% of cases) (77. Raz L, Knoefel J, Bhaskar K. The neuropathology and cerebrovascular mechanisms of dementia. J Cereb Blood Flow Metab 2016; 36: 172–186, doi: 10.1038/jcbfm.2015.164.
https://doi.org/10.1038/jcbfm.2015.164...
). Inherited dominant mutations in the APP and neprilysin genes are implicated in the pathophysiology of early-onset AD (1212. Dubey H, Gulati K, Ray A. Recent studies on cellular and molecular mechanisms in Alzheimer's disease: focus on epigenetic factors and histone deacetylase. Rev Neurosci 2018; 29: 241–260, doi: 10.1515/revneuro-2017-0049.
https://doi.org/10.1515/revneuro-2017-00...
). Consequently, these genes are considered biomarkers for the disease. On the other hand, the etiology of the sporadic form, which shows a late onset (after 65 years), has not yet been elucidated (22. Reitz C, Mayeux R. Alzheimer disease: epidemiology, diagnostic criteria, risk factors, and biomarkers. Biochem Pharmacol 2014; 88: 640–651, doi: 10.1016/j.bcp.2013.12.024.
https://doi.org/10.1016/j.bcp.2013.12.02...
,1313. Karch CM, Ezerskiy LA, Bertelsen S, Alzheimer's disease genetics consortium (ADGC), Goate AM. Alzheimer's disease risk polymorphisms regulate gene expression in the ZCWPW1 and the CELF1 Loci. PLoS One 2016; 11: e0148717, doi: 10.1371/journal.pone.0148717.
https://doi.org/10.1371/journal.pone.014...
). Therefore, the analysis of polymorphisms in candidate genes has contributed to risk assessment and the primary prevention of several chronic degenerative diseases, including diabetes mellitus, myocardial infarction (1414. Relvas WGM, Izar MCO, Helfenstein T, Fonseca MIH, Colovati M, Oliveira A, et al. Relationship between gene polymorphisms and prevalence of myocardial infarction among diabetic and non-diabetic subjects. Atherosclerosis 2005; 178: 101–105, doi: 10.1016/j.atherosclerosis.2004.05.025.
https://doi.org/10.1016/j.atherosclerosi...
), and AD (1515. Huang Y, Mucke L. Alzheimer's mechanisms and therapeutic strategies. Cell 2012; 148: 1204–1222, doi: 10.1016/j.cell.2012.02.040.
https://doi.org/10.1016/j.cell.2012.02.0...
).

Apolipoprotein E (APOE)

APOE plays a regulatory role in the central nervous system (CNS), acting in the distribution and transport of cholesterol to the nerve tissues for the maintenance of their integrity (neuroplasticity) as well as in the immune response and mitochondrial function of neurons (1616. Huang Y, Mahley RW. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer's diseases. Neurobiol Dis 2014; 72: 3–12, doi: 10.1016/j.nbd.2014.08.025.
https://doi.org/10.1016/j.nbd.2014.08.02...
). APOE, one of the main CNS lipoproteins, is synthesized peripherally in the liver and in the CNS by the astrocytes and has three isoforms - APOEε2, APOEε3, and APOEε4 - with two important functional domains: the N-terminal domain that contains its receptor binding sites and the C-terminal domain that binds to βA14 (1717. Driscoll I, Snively BM, Espeland MA, Shumaker SA, Rapp SR, Goveas JS, et al. A candidate gene study of risk for dementia in older, postmenopausal women: results from the women's health initiative memory study. Int J Geriatr Psychiatry 2019; 34: 692–699, doi: 10.1002/gps.5068.
https://doi.org/10.1002/gps.5068...
). APOE is located in chromosome 19q13.2 and codes for the three common alleles (ε2, ε3, ε4). Carriers of the homozygous or heterozygous ε4 allele have increased genetic risk for late onset AD (1717. Driscoll I, Snively BM, Espeland MA, Shumaker SA, Rapp SR, Goveas JS, et al. A candidate gene study of risk for dementia in older, postmenopausal women: results from the women's health initiative memory study. Int J Geriatr Psychiatry 2019; 34: 692–699, doi: 10.1002/gps.5068.
https://doi.org/10.1002/gps.5068...
). A recent study of neuroimaging and neuropathology has shown that APOEε4 carriers have abundant and accelerated βA deposition compared with non-carriers (1818. Lyall DM, Cox SR, Lyall LM, Celis-Morales C, Cullen B, Mackay DF, et al. Association between APOE e4 and white matter hyperintensity volume, but not total brain volume or white matter integrity. Brain Imaging Behav 2019, doi: 10.1007/s11682-019-00069-9.
https://doi.org/10.1007/s11682-019-00069...
).

AD and physical exercise

The beneficial effects in both experimental models and humans of physical exercise on hypertension, type 2 diabetes mellitus, and dyslipidemia, which are considered risk factors for AD, are well established (1919. Zanesco A, Antunes E. Effects of exercise training on the cardiovascular system: pharmacological approaches. Pharmacol Ther 2007; 114: 307–317, doi: 10.1016/j.pharmthera.2007.03.010.
https://doi.org/10.1016/j.pharmthera.200...
20. Esposti RD, Sponton CH, Malagrino PA, Carvalho FC, Peres E, Puga GM, et al. Influence of eNOS gene polymorphism on cardiometabolic parameters in response to physical training in postmenopausal women. Braz J Med Biol Res 2011; 44: 855–863, doi: 10.1590/S0100-879X2011007500106.
https://doi.org/10.1590/S0100-879X201100...
2121. Trask AJ, Delbin MA, Katz PS, Zanesco A, Lucchesi PA. Differential coronary resistance microvessel remodeling between type 1 and type 2 diabetic mice: impact of exercise training. Vascul Pharmacol 2012; 57: 187–193, doi: 10.1016/j.vph.2012.07.007.
https://doi.org/10.1016/j.vph.2012.07.00...
). However, some studies have failed to clearly show whether physical exercise prevents AD or mitigates its progression, either due to methodological difficulties or differences in physical exercise protocols (2222. Aguera Sánchez MÁ, Barbancho Ma MÁ, García-Casares N. Effect of physical exercise on Alzheimer's disease. A systematic review [in Spanish]. Aten Primaria 2020; 52: 307–318, doi: 10.1016/j.aprim.2018.09.010.
https://doi.org/10.1016/j.aprim.2018.09....
,2323. Li XY, Zhang M, Xu W, Li JQ, Cao XP, Yu JT, et al. Midlife modifiable risk factors for dementia: a systematic review and meta-analysis of 34 prospective cohort studies. Curr Alzheimer Res 2019; 16: 1254–1268, doi: 10.2174/1567205017666200103111253.
https://doi.org/10.2174/1567205017666200...
). It is believed that the beneficial effects of physical exercise on cognitive function are indirect (2424. Beckett MW, Ardern CI, Rotondi MA. A meta-analysis of prospective studies on the role of physical activity and the prevention of Alzheimer's disease in older adults. BMC Geriatr 2015; 15: 9, doi: 10.1186/s12877-015-0007-2.
https://doi.org/10.1186/s12877-015-0007-...
). A classic example is the fact that individuals who experienced ischemic events are more likely to develop AD, with a higher production of reactive oxygen species resulting in the accumulation of βA, and physical exercise was shown to act as an antioxidant and to improve some types of AD (2525. Desmond DW, Moroney JT, Sano M, Stern Y. Incidence of dementia after ischemic stroke: results of a longitudinal study. Stroke 2002; 33: 2254–2260, doi: 10.1161/01.STR.0000028235.91778.95.
https://doi.org/10.1161/01.STR.000002823...
,2626. Cotman CW, Berchtold NC. Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci 2002; 25: 295–301, doi: 10.1016/S0166-2236(02)02143-4.
https://doi.org/10.1016/S0166-2236(02)02...
27. Lazarov O, Mattson MP, Peterson DA, Pimplikar SW, van Praag H. When neurogenesis encounters aging and disease. Trends Neurosci 2010; 33: 569–579, doi: 10.1016/j.tins.2010.09.003.
https://doi.org/10.1016/j.tins.2010.09.0...
).

Studies that have shown the beneficial effect of physical exercise on cognitive function, as well as those in which no changes were detected, are described below, and we subsequently address the interaction between physical exercise, APOE polymorphism, and cognitive function.

Beneficial effects of physical exercise

Studies have shown that physical exercise has systemic positive effects on AD via the induction of angiogenesis and anti-inflammatory and antioxidant activities (2626. Cotman CW, Berchtold NC. Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci 2002; 25: 295–301, doi: 10.1016/S0166-2236(02)02143-4.
https://doi.org/10.1016/S0166-2236(02)02...
2828. Kobilo T, Liu QR, Gandhi K, Mughal M, Shaham Y, van Praag H. Running is the neurogenic and neurotrophic stimulus in environmental enrichment. Learn Mem 2011; 18: 605–609, doi: 10.1101/lm.2283011.
https://doi.org/10.1101/lm.2283011...
), whereas other studies have shown that physical exercise promotes neurogenesis as well as reduces tau protein hyperphosphorylation and excessive Aβ production (2929. Lourenco MV, Frozza RL, de Freitas GB, Zhang H, Kincheski GC, Ribeiro FC, et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer's models. Nat Med 2019; 25: 165–175, doi: 10.1038/s41591-018-0275-4.
https://doi.org/10.1038/s41591-018-0275-...
). However, several studies have been conducted with animal models and it is known that experimental models of AD do not reflect the changes observed in humans. Considering this context, we present details of some clinical studies.

A previous study including healthy men showed that exercise training for approximately 2 years without direct supervision improved cerebral blood flow compared with physically inactive individuals (3030. Ainslie PN, Cotter JD, George KP, Lucas S, Murrell C, Shave R, et al. Elevation in cerebral blood flow velocity with aerobic fitness throughout healthy human ageing. J Physiol 2008; 586: 4005–4010, doi: 10.1113/jphysiol.2008.158279.
https://doi.org/10.1113/jphysiol.2008.15...
). Another study evaluated elderly individuals with normal cognitive function, mild dysfunction, or AD. The practice of physical exercise was associated with higher brain volume in all groups and inversely correlated with body mass index (3131. Boyle CP, Raji CA, Erickson KI, Lopez OL, Becker JT, Gach HM, et al. Physical activity, body mass index, and brain atrophy in Alzheimer's disease. Neurobiol Aging 2015; 36 Suppl 1: S194–S202, doi: 10.1016/j.neurobiolaging.2014.05.036.
https://doi.org/10.1016/j.neurobiolaging...
). A similar study showed a positive association between cardiorespiratory fitness and white brain mass integrity in elderly individuals (3232. Perea RD, Vidoni ED, Morris JK, Graves RS, Burns JM, Honea RA. Cardiorespiratory fitness and white matter integrity in Alzheimer's disease. Brain Imaging Behav 2016; 10: 660–668, doi: 10.1007/s11682-015-9431-3.
https://doi.org/10.1007/s11682-015-9431-...
). Recently, it was observed that elderly individuals with mild cognitive dysfunction who practiced physical exercise exhibited improvement in the memory score that was associated with reduced atrophy of the hippocampus (3333. Morris JK, Vidoni ED, Johnson DK, Van Sciver A, Mahnken JD, Honea RA, et al. Aerobic exercise for Alzheimer's disease: A randomized controlled pilot trial. PLoS One 2017; 12: e0170547, doi: 10.1371/journal.pone.0170547.
https://doi.org/10.1371/journal.pone.017...
). Consistent with these findings, an inverse relationship was observed between βA42 concentration and cognitive dysfunction in individuals with high cardiorespiratory fitness (3434. Schultz SA, Boots EA, Almeida RP, Oh JM, Einerson J, Korcarz CE, et al. Cardiorespiratory fitness attenuates the influence of amyloid on cognition. J Int Neuropsychol Soc 2015; 21: 841–850, doi: 10.1017/S1355617715000843.
https://doi.org/10.1017/S135561771500084...
,3535. Law LL, Rol RN, Schultz SA, Dougherty RJ, Edwards DF, Koscik RL, et al. Moderate-intensity physical activity associates with CSF biomarkers in a cohort at risk for Alzheimer's disease. Alzheimers Dement (Amst) 2018; 10: 188–195, doi: 10.1016/j.dadm.2018.01.001.
https://doi.org/10.1016/j.dadm.2018.01.0...
).

Studies involving the direct application of an exercise training program or direct supervision of physical activity in the elderly are scarce and the greatest limitation is adherence. A study to evaluate patients with AD undergoing 12 weeks of physical activity showed a positive association between aerobic capacity and cognitive test scores (3636. Holthoff VA, Marschner K, Scharf M, Steding J, Meyer S, Koch R, et al. Effects of physical activity training in patients with Alzheimer's dementia: results of a pilot RCT study. PLoS One 2015; 10: e0121478, doi: 10.1371/journal.pone.0121478.
https://doi.org/10.1371/journal.pone.012...
). Another study showed improvement in cognitive test scores of patients with AD and in the peak value of VO2 (3737. Sobol NA, Dall CH, Høgh P, Hoffmann K, Frederiksen KS, Vogel A, et al. Change in fitness and the relation to change in cognition and neuropsychiatric symptoms after aerobic exercise in patients with mild Alzheimer's disease. J Alzheimers Dis 2018; 65: 137–145, doi: 10.3233/JAD-180253.
https://doi.org/10.3233/JAD-180253...
). Further, a recent review showed that improvement in cardiorespiratory fitness promotes the release of systemic mediators that potentially act on the CNS, thereby preventing the neuronal damage of AD (3838. Tari AR, Norevik CS, Scrimgeour NR, Kobro-Flatmoen A, Storm-Mathisen J, Bergersen LH, et al. Are the neuroprotective effects of exercise training systemically mediated? Prog Cardiovasc Dis 2019; 62: 94–101, doi: 10.1016/j.pcad.2019.02.003.
https://doi.org/10.1016/j.pcad.2019.02.0...
). Table 1 summarizes these studies.

Table 1
Studies showing beneficial effects of physical exercise on cognitive function.

Moreover, neurotrophic factors (NTFs) have been evaluated in the response to physical exercise because they play an important role in memory formation, cognitive capacity, and neuron survival, and physical exercise improves neuroplasticity. The most studied NTFs are neural growth factor, brain-derived neurotrophic factor (BDNF), and neurotrophins (2626. Cotman CW, Berchtold NC. Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci 2002; 25: 295–301, doi: 10.1016/S0166-2236(02)02143-4.
https://doi.org/10.1016/S0166-2236(02)02...
,3939. Seifert T, Brassard P, Wissenberg M, Rasmussen P, Nordby P, Stallknecht B, et al. Endurance training enhances BDNF release from the human brain. Am J Physiol Regul Integr Comp Physiol 2010; 298: R372–R377, doi: 10.1152/ajpregu.00525.2009.
https://doi.org/10.1152/ajpregu.00525.20...
,4040. Cassilhas RC, Tufik S, de Mello MT. Physical exercise, neuroplasticity, spatial learning, and memory. Cell Mol Life Sci 2016; 73: 975–983, doi: 10.1007/s00018-015-2102-0.
https://doi.org/10.1007/s00018-015-2102-...
). However, the mechanisms by which physical exercise/training would promote these effects in AD remain unclear.

Absence of beneficial effects of physical exercise

Contrary to the above-mentioned studies, a systematic review showed that a conclusion about beneficial effects of physical exercise cannot be drawn from the studies conducted. However, a multi-domain intervention that associates physical activity, diet, and cognitive training was shown to improve cognitive abilities among the elderly (4141. Brasure M, Desai P, Davila H, Nelson VA, Calvert C, Jutkowitz E, et al. Physical activity interventions in preventing cognitive decline and Alzheimer-type dementia: a systematic review. Ann Intern Med 2018; 168: 30–38, doi: 10.7326/M17-1528.
https://doi.org/10.7326/M17-1528...
). With regard to cerebral blood flow, exercise training did not modify the Mini-Mental State Exam scores and did not lead to changes in cerebral blood flow (4242. van der Kleij LA, Petersen ET, Siebner HR, Hendrikse J, Frederiksen KS, Sobol NA, et al. The effect of physical exercise on cerebral blood flow in Alzheimer's disease. Neuroimage Clin 2018; 20: 650–654, doi: 10.1016/j.nicl.2018.09.003.
https://doi.org/10.1016/j.nicl.2018.09.0...
). In fact, a recent study evaluating brain damage biomarkers or inflammatory mediators in cerebrospinal fluid in patients with AD who trained found no changes after the intervention (4343. Jensen CS, Portelius E, Høgh P, Wermuth L, Blennow K, Zetterberg H, et al. Effect of physical exercise on markers of neuronal dysfunction in cerebrospinal fluid in patients with Alzheimer's disease. Alzheimers Dement (NY) 2017; 3: 284–290, doi: 10.1016/j.trci.2017.03.007.
https://doi.org/10.1016/j.trci.2017.03.0...
). A recent systematic review examining the relationship between physical activity and several biomarkers for AD concluded that the quality of the studies does not facilitate a definitive conclusion and that most of the studies analyzed showed no association between physical exercise and cognitive improvement (4444. Frederiksen KS, Gjerum L, Waldemar G, Hasselbalch SG. Physical activity as a moderator of Alzheimer pathology: a systematic review of observational studies. Curr Alzheimer Res 2019; 16: 362–378, doi: 10.2174/1567205016666190315095151.
https://doi.org/10.2174/1567205016666190...
).

Therefore, standardized protocols and larger and more detailed randomized controlled clinical trials with long-term follow-up are required to provide a better insight into the effects of physical exercise on cognitive function (4545. Brett L, Traynor V, Stapley P. Effects of physical exercise on health and well-being of individuals living with a dementia in nursing homes: a systematic review. J Am Med Dir Assoc 2016; 17: 104–116, doi: 10.1016/j.jamda.2015.08.016.
https://doi.org/10.1016/j.jamda.2015.08....
46. Guure CB, Ibrahim NA, Adam MB, Said SM. Impact of physical activity on cognitive decline, dementia, and its subtypes: meta-analysis of prospective studies. Biomed Res Int 2017; e9016924, doi: 10.1155/2017/9016924.
https://doi.org/10.1155/2017/9016924...
4747. Cammisuli DM, Innocenti A, Fusi J, Franzoni F, Pruneti C. Aerobic exercise effects upon cognition in Alzheimer's disease: a systematic review of randomized controlled trials. Arch Ital Biol 2018; 156: 54–63, doi: 10.12871/00039829201816.
https://doi.org/10.12871/00039829201816...
).

It should also be emphasized that the beneficial effects in the elderly have been observed more consistently in participants without dementia, whereas the results are controversial in patients with AD, suggesting that the practice of physical exercise has preventive effects on brain activity and that the potential existence of various forms of AD generates disparate results (4848. Vidoni ED, Gayed MR, Honea RA, Savage CR, Hobbs D, Burns JM. Alzheimer's disease alters the relationship of cardiorespiratory fitness with brain activity during the stroop task. Phys Ther 2013; 93: 993–1002, doi: 10.2522/ptj.20120465.
https://doi.org/10.2522/ptj.20120465...
). Although there is no agreement in the literature regarding the effects of physical exercise on cognitive function, it is well established that it improves social interaction and the feeling of well-being, thereby positively contributing to the quality of life of the elderly. Therefore, public policies to encourage the practice of physical exercise are fundamental to minimize the prevalence of AD or delay its onset. Table 2 summarizes these studies.

Table 2
Studies showing absence of beneficial effects of physical exercise on cognitive function.

Interaction between APOE genetics and physical exercise on cognitive function

A previous study showed that physically active individuals who were APOEε4 allele carriers exhibited a stable course of cognitive performance and protection against hippocampal atrophy during an 18-month follow-up period compared with physically inactive alleloε4 carriers. This suggests that physical exercise offers protection against AD neurodegeneration even in carriers of the APOΕε4 allele (4949. Smith JC, Lancaster MA, Nielson KA, Woodard JL, Seidenberg M, Durgerian S, et al. Interactive effects of physical activity and APOE-ε4 on white matter tract diffusivity in healthy elders. Neuroimage 2016; 131: 102–112, doi: 10.1016/j.neuroimage.2015.08.007.
https://doi.org/10.1016/j.neuroimage.201...
). In a recent study evaluating participants aged ≥65 years (362 carriers of the APOΕε4 allele and 747 non-carriers), it was concluded that multi-domain interventions (diet, physical exercise, cognitive training, and vascular risk monitoring) were beneficial for cognitive function, even in patients with the APOΕε4 allele. However, patient adherence remains a challenge (5050. Solomon A, Turunen H, Ngandu T, Peltonen M, Levälahti E, Helisalmi S, et al. Effect of the apolipoprotein e genotype on cognitive change during a multidomain lifestyle intervention: a subgroup analysis of a randomized clinical trial. JAMA Neurol 2018; 75: 462–470, doi: 10.1001/jamaneurol.2017.4365.
https://doi.org/10.1001/jamaneurol.2017....
). Another study evaluating 1,438 elderly individuals for 6.5 years showed that physical exercise was able to mitigate dementia risk in participants with the APOΕε4 allele compared with physically inactive elderly individuals (5151. Shih IF, Paul K, Haan M, Yu Y, Ritz B. Physical activity modifies the influence of apolipoprotein E ε4 allele and type 2 diabetes on dementia and cognitive impairment among older Mexican Americans. Alzheimers Dement 2018; 14: 1–9, doi: 10.1016/j.jalz.2017.05.005.
https://doi.org/10.1016/j.jalz.2017.05.0...
). The association between cardiovascular risk, educational level, and leisure activity has also been studied in patients with the APOEε4 allele and the findings show that those with low vascular risk, high educational level, and who practiced leisure activities have a low dementia risk (5252. Ferrari C, Xu WL, Wang HX, Winblad B, Sorbi S, Qiu C, et al. How can elderly apolipoprotein E ε4 carriers remain free from dementia? Neurobiol Aging 2013; 34: 13–21, doi: 10.1016/j.neurobiolaging.2012.03.003.
https://doi.org/10.1016/j.neurobiolaging...
). A recent study showed that APOEε4 allele carriers are more responsive to exercise training, with better performance in motor and cognitive tests compared with non-carriers of the allele (5353. Jensen CS, Bahl JM, Ostergaard LB, Hogh P, Wermuth L, Heslegrave A, et al. 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, doi: 10.1016/j.exger.2019.04.003.
https://doi.org/10.1016/j.exger.2019.04....
).

In contrast, other studies have failed to detect any effect of physical exercise on dementia in patients who carry ε4 allele. One study showed that the likelihood of developing dementia is not significantly different between those who exercise and those who do not (5454. Fenesi B, Fang H, Kovacevic A, Oremus M, Raina P, Heisz JJ. Physical exercise moderates the relationship of Apolipoprotein E (APOE) genotype and dementia risk: a population-based study. J Alzheimer Dis 2017; 56: 297–303, doi: 10.3233/JAD-160424.
https://doi.org/10.3233/JAD-160424...
). In agreement with that, it has been shown that exercise training did not modify the cognitive function in patients with the APOEε4 allele (5555. Stern Y, MacKay-Brandt A, Lee S, McKinley P, McIntyre K, Razlighi Q, et al. Effect of aerobic exercise on cognition in younger adults: A randomized clinical trial. Neurology 2019; 92: e905–e916, doi: 10.1212/WNL.0000000000007003.
https://doi.org/10.1212/WNL.000000000000...
). Moreover, the APOEε4 allele carriers have a lower plasma concentration of BDNF, and exercise training does not promote changes in the levels of this neurotrophic factor (5656. Allard JS, Ntekim O, Johnson SP, Ngwa JS, Bond V, Pinder D, et al. APOEε4 impacts up-regulation of a brain-derived neurotrophic factor after a six-month stretch and aerobic exercise intervention in mild cognitively impaired elderly African Americans: a pilot study. Exp Gerontol 2017; 87: 129–136, doi: 10.1016/j.exger.2016.11.001.
https://doi.org/10.1016/j.exger.2016.11....
). Table 3 summarizes these studies.

Table 3
Studies showing interaction between APOE gene polymorphism, physical exercise, and cognitive function.

Conclusion

In general, physical exercise might be effective in ameliorating the score of cognitive tests and likely prevent neurodegeneration in individuals who have genetic risk factors, such as APOEε4 allele carriers. Nevertheless, studies with larger sample sizes will provide more accuracy in this relationship.

Acknowledgments

The authors thank São Paulo Research Foundation (FAPESP) for financial support (Grant: #2019/13343-0).

References

  • 1
    World health organization. World health organization (WHO). Available at https://www.who.int/ageing/sdgs/en/ accessed March 15, 2020.
    » https://www.who.int/ageing/sdgs/en/
  • 2
    Reitz C, Mayeux R. Alzheimer disease: epidemiology, diagnostic criteria, risk factors, and biomarkers. Biochem Pharmacol 2014; 88: 640–651, doi: 10.1016/j.bcp.2013.12.024.
    » https://doi.org/10.1016/j.bcp.2013.12.024
  • 3
    Ferretti C, Sarti FM, Nitrini R, Ferreira FF, Brucki SMD. An assessment of direct and indirect costs of dementia in Brazil. PLoS One 2018; 13: e0193209, doi: 10.1371/journal.pone.0193209.
    » https://doi.org/10.1371/journal.pone.0193209
  • 4
    Najm R, Jones EA, Huang Y. Apolipoprotein E4, inhibitory network dysfunction, and Alzheimer's disease. Mol Neurodegener 2019; 14: 24, doi: 10.1186/s13024-019-0324-6.
    » https://doi.org/10.1186/s13024-019-0324-6
  • 5
    Reiss AB, Arain HA, Stecker MM, Siegart NM, Kasselman LJ. Amyloid toxicity in Alzheimer's disease. Rev Neurosci 2018; 29: 613–627, doi: 10.1515/revneuro-2017-0063.
    » https://doi.org/10.1515/revneuro-2017-0063
  • 6
    Lehert P, Villaseca P, Hogervorst E, Maki PM, Henderson VW. Individually modifiable risk factors to ameliorate cognitive aging: a systematic review and meta-analysis. Climacteric 2015; 18: 678–689, doi: 10.3109/13697137.2015.1078106.
    » https://doi.org/10.3109/13697137.2015.1078106
  • 7
    Raz L, Knoefel J, Bhaskar K. The neuropathology and cerebrovascular mechanisms of dementia. J Cereb Blood Flow Metab 2016; 36: 172–186, doi: 10.1038/jcbfm.2015.164.
    » https://doi.org/10.1038/jcbfm.2015.164
  • 8
    Bertram L, Lill CM, Tanzi RE. The genetics of Alzheimer's disease: back to the future. Neuron 2010; 68: 270–281, doi: 10.1016/j.neuron.2010.10.013.
    » https://doi.org/10.1016/j.neuron.2010.10.013
  • 9
    Chouliaras L, Rutten BP, Kenis G, Peerbooms O, Visser PJ, Verhey F, et al. Epigenetic regulation in the pathophysiology of Alzheimer's disease. Prog Neurobiol 2010; 90: 498–510, doi: 10.1016/j.pneurobio.2010.01.002.
    » https://doi.org/10.1016/j.pneurobio.2010.01.002
  • 10
    Turner AJ, Fisk L, Nalivaeva NN. Targeting amyloid-degrading enzymes as therapeutic strategies in neurodegeneration. Ann NY Acad Sci 2004; 1035: 1–20, doi: 10.1196/annals.1332.001.
    » https://doi.org/10.1196/annals.1332.001
  • 11
    Iqbal K, Liu F, Gong CX. Recent developments with tau-based drug discovery. Expert Opin Drug Discov 2018; 13: 399–410, doi: 10.1080/17460441.2018.1445084.
    » https://doi.org/10.1080/17460441.2018.1445084
  • 12
    Dubey H, Gulati K, Ray A. Recent studies on cellular and molecular mechanisms in Alzheimer's disease: focus on epigenetic factors and histone deacetylase. Rev Neurosci 2018; 29: 241–260, doi: 10.1515/revneuro-2017-0049.
    » https://doi.org/10.1515/revneuro-2017-0049
  • 13
    Karch CM, Ezerskiy LA, Bertelsen S, Alzheimer's disease genetics consortium (ADGC), Goate AM. Alzheimer's disease risk polymorphisms regulate gene expression in the ZCWPW1 and the CELF1 Loci. PLoS One 2016; 11: e0148717, doi: 10.1371/journal.pone.0148717.
    » https://doi.org/10.1371/journal.pone.0148717
  • 14
    Relvas WGM, Izar MCO, Helfenstein T, Fonseca MIH, Colovati M, Oliveira A, et al. Relationship between gene polymorphisms and prevalence of myocardial infarction among diabetic and non-diabetic subjects. Atherosclerosis 2005; 178: 101–105, doi: 10.1016/j.atherosclerosis.2004.05.025.
    » https://doi.org/10.1016/j.atherosclerosis.2004.05.025
  • 15
    Huang Y, Mucke L. Alzheimer's mechanisms and therapeutic strategies. Cell 2012; 148: 1204–1222, doi: 10.1016/j.cell.2012.02.040.
    » https://doi.org/10.1016/j.cell.2012.02.040
  • 16
    Huang Y, Mahley RW. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer's diseases. Neurobiol Dis 2014; 72: 3–12, doi: 10.1016/j.nbd.2014.08.025.
    » https://doi.org/10.1016/j.nbd.2014.08.025
  • 17
    Driscoll I, Snively BM, Espeland MA, Shumaker SA, Rapp SR, Goveas JS, et al. A candidate gene study of risk for dementia in older, postmenopausal women: results from the women's health initiative memory study. Int J Geriatr Psychiatry 2019; 34: 692–699, doi: 10.1002/gps.5068.
    » https://doi.org/10.1002/gps.5068
  • 18
    Lyall DM, Cox SR, Lyall LM, Celis-Morales C, Cullen B, Mackay DF, et al. Association between APOE e4 and white matter hyperintensity volume, but not total brain volume or white matter integrity. Brain Imaging Behav 2019, doi: 10.1007/s11682-019-00069-9.
    » https://doi.org/10.1007/s11682-019-00069-9
  • 19
    Zanesco A, Antunes E. Effects of exercise training on the cardiovascular system: pharmacological approaches. Pharmacol Ther 2007; 114: 307–317, doi: 10.1016/j.pharmthera.2007.03.010.
    » https://doi.org/10.1016/j.pharmthera.2007.03.010
  • 20
    Esposti RD, Sponton CH, Malagrino PA, Carvalho FC, Peres E, Puga GM, et al. Influence of eNOS gene polymorphism on cardiometabolic parameters in response to physical training in postmenopausal women. Braz J Med Biol Res 2011; 44: 855–863, doi: 10.1590/S0100-879X2011007500106.
    » https://doi.org/10.1590/S0100-879X2011007500106
  • 21
    Trask AJ, Delbin MA, Katz PS, Zanesco A, Lucchesi PA. Differential coronary resistance microvessel remodeling between type 1 and type 2 diabetic mice: impact of exercise training. Vascul Pharmacol 2012; 57: 187–193, doi: 10.1016/j.vph.2012.07.007.
    » https://doi.org/10.1016/j.vph.2012.07.007
  • 22
    Aguera Sánchez MÁ, Barbancho Ma MÁ, García-Casares N. Effect of physical exercise on Alzheimer's disease. A systematic review [in Spanish]. Aten Primaria 2020; 52: 307–318, doi: 10.1016/j.aprim.2018.09.010.
    » https://doi.org/10.1016/j.aprim.2018.09.010
  • 23
    Li XY, Zhang M, Xu W, Li JQ, Cao XP, Yu JT, et al. Midlife modifiable risk factors for dementia: a systematic review and meta-analysis of 34 prospective cohort studies. Curr Alzheimer Res 2019; 16: 1254–1268, doi: 10.2174/1567205017666200103111253.
    » https://doi.org/10.2174/1567205017666200103111253
  • 24
    Beckett MW, Ardern CI, Rotondi MA. A meta-analysis of prospective studies on the role of physical activity and the prevention of Alzheimer's disease in older adults. BMC Geriatr 2015; 15: 9, doi: 10.1186/s12877-015-0007-2.
    » https://doi.org/10.1186/s12877-015-0007-2
  • 25
    Desmond DW, Moroney JT, Sano M, Stern Y. Incidence of dementia after ischemic stroke: results of a longitudinal study. Stroke 2002; 33: 2254–2260, doi: 10.1161/01.STR.0000028235.91778.95.
    » https://doi.org/10.1161/01.STR.0000028235.91778.95
  • 26
    Cotman CW, Berchtold NC. Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci 2002; 25: 295–301, doi: 10.1016/S0166-2236(02)02143-4.
    » https://doi.org/10.1016/S0166-2236(02)02143-4
  • 27
    Lazarov O, Mattson MP, Peterson DA, Pimplikar SW, van Praag H. When neurogenesis encounters aging and disease. Trends Neurosci 2010; 33: 569–579, doi: 10.1016/j.tins.2010.09.003.
    » https://doi.org/10.1016/j.tins.2010.09.003
  • 28
    Kobilo T, Liu QR, Gandhi K, Mughal M, Shaham Y, van Praag H. Running is the neurogenic and neurotrophic stimulus in environmental enrichment. Learn Mem 2011; 18: 605–609, doi: 10.1101/lm.2283011.
    » https://doi.org/10.1101/lm.2283011
  • 29
    Lourenco MV, Frozza RL, de Freitas GB, Zhang H, Kincheski GC, Ribeiro FC, et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer's models. Nat Med 2019; 25: 165–175, doi: 10.1038/s41591-018-0275-4.
    » https://doi.org/10.1038/s41591-018-0275-4
  • 30
    Ainslie PN, Cotter JD, George KP, Lucas S, Murrell C, Shave R, et al. Elevation in cerebral blood flow velocity with aerobic fitness throughout healthy human ageing. J Physiol 2008; 586: 4005–4010, doi: 10.1113/jphysiol.2008.158279.
    » https://doi.org/10.1113/jphysiol.2008.158279
  • 31
    Boyle CP, Raji CA, Erickson KI, Lopez OL, Becker JT, Gach HM, et al. Physical activity, body mass index, and brain atrophy in Alzheimer's disease. Neurobiol Aging 2015; 36 Suppl 1: S194–S202, doi: 10.1016/j.neurobiolaging.2014.05.036.
    » https://doi.org/10.1016/j.neurobiolaging.2014.05.036
  • 32
    Perea RD, Vidoni ED, Morris JK, Graves RS, Burns JM, Honea RA. Cardiorespiratory fitness and white matter integrity in Alzheimer's disease. Brain Imaging Behav 2016; 10: 660–668, doi: 10.1007/s11682-015-9431-3.
    » https://doi.org/10.1007/s11682-015-9431-3
  • 33
    Morris JK, Vidoni ED, Johnson DK, Van Sciver A, Mahnken JD, Honea RA, et al. Aerobic exercise for Alzheimer's disease: A randomized controlled pilot trial. PLoS One 2017; 12: e0170547, doi: 10.1371/journal.pone.0170547.
    » https://doi.org/10.1371/journal.pone.0170547
  • 34
    Schultz SA, Boots EA, Almeida RP, Oh JM, Einerson J, Korcarz CE, et al. Cardiorespiratory fitness attenuates the influence of amyloid on cognition. J Int Neuropsychol Soc 2015; 21: 841–850, doi: 10.1017/S1355617715000843.
    » https://doi.org/10.1017/S1355617715000843
  • 35
    Law LL, Rol RN, Schultz SA, Dougherty RJ, Edwards DF, Koscik RL, et al. Moderate-intensity physical activity associates with CSF biomarkers in a cohort at risk for Alzheimer's disease. Alzheimers Dement (Amst) 2018; 10: 188–195, doi: 10.1016/j.dadm.2018.01.001.
    » https://doi.org/10.1016/j.dadm.2018.01.001
  • 36
    Holthoff VA, Marschner K, Scharf M, Steding J, Meyer S, Koch R, et al. Effects of physical activity training in patients with Alzheimer's dementia: results of a pilot RCT study. PLoS One 2015; 10: e0121478, doi: 10.1371/journal.pone.0121478.
    » https://doi.org/10.1371/journal.pone.0121478
  • 37
    Sobol NA, Dall CH, Høgh P, Hoffmann K, Frederiksen KS, Vogel A, et al. Change in fitness and the relation to change in cognition and neuropsychiatric symptoms after aerobic exercise in patients with mild Alzheimer's disease. J Alzheimers Dis 2018; 65: 137–145, doi: 10.3233/JAD-180253.
    » https://doi.org/10.3233/JAD-180253
  • 38
    Tari AR, Norevik CS, Scrimgeour NR, Kobro-Flatmoen A, Storm-Mathisen J, Bergersen LH, et al. Are the neuroprotective effects of exercise training systemically mediated? Prog Cardiovasc Dis 2019; 62: 94–101, doi: 10.1016/j.pcad.2019.02.003.
    » https://doi.org/10.1016/j.pcad.2019.02.003
  • 39
    Seifert T, Brassard P, Wissenberg M, Rasmussen P, Nordby P, Stallknecht B, et al. Endurance training enhances BDNF release from the human brain. Am J Physiol Regul Integr Comp Physiol 2010; 298: R372–R377, doi: 10.1152/ajpregu.00525.2009.
    » https://doi.org/10.1152/ajpregu.00525.2009
  • 40
    Cassilhas RC, Tufik S, de Mello MT. Physical exercise, neuroplasticity, spatial learning, and memory. Cell Mol Life Sci 2016; 73: 975–983, doi: 10.1007/s00018-015-2102-0.
    » https://doi.org/10.1007/s00018-015-2102-0
  • 41
    Brasure M, Desai P, Davila H, Nelson VA, Calvert C, Jutkowitz E, et al. Physical activity interventions in preventing cognitive decline and Alzheimer-type dementia: a systematic review. Ann Intern Med 2018; 168: 30–38, doi: 10.7326/M17-1528.
    » https://doi.org/10.7326/M17-1528
  • 42
    van der Kleij LA, Petersen ET, Siebner HR, Hendrikse J, Frederiksen KS, Sobol NA, et al. The effect of physical exercise on cerebral blood flow in Alzheimer's disease. Neuroimage Clin 2018; 20: 650–654, doi: 10.1016/j.nicl.2018.09.003.
    » https://doi.org/10.1016/j.nicl.2018.09.003
  • 43
    Jensen CS, Portelius E, Høgh P, Wermuth L, Blennow K, Zetterberg H, et al. Effect of physical exercise on markers of neuronal dysfunction in cerebrospinal fluid in patients with Alzheimer's disease. Alzheimers Dement (NY) 2017; 3: 284–290, doi: 10.1016/j.trci.2017.03.007.
    » https://doi.org/10.1016/j.trci.2017.03.007
  • 44
    Frederiksen KS, Gjerum L, Waldemar G, Hasselbalch SG. Physical activity as a moderator of Alzheimer pathology: a systematic review of observational studies. Curr Alzheimer Res 2019; 16: 362–378, doi: 10.2174/1567205016666190315095151.
    » https://doi.org/10.2174/1567205016666190315095151
  • 45
    Brett L, Traynor V, Stapley P. Effects of physical exercise on health and well-being of individuals living with a dementia in nursing homes: a systematic review. J Am Med Dir Assoc 2016; 17: 104–116, doi: 10.1016/j.jamda.2015.08.016.
    » https://doi.org/10.1016/j.jamda.2015.08.016
  • 46
    Guure CB, Ibrahim NA, Adam MB, Said SM. Impact of physical activity on cognitive decline, dementia, and its subtypes: meta-analysis of prospective studies. Biomed Res Int 2017; e9016924, doi: 10.1155/2017/9016924.
    » https://doi.org/10.1155/2017/9016924
  • 47
    Cammisuli DM, Innocenti A, Fusi J, Franzoni F, Pruneti C. Aerobic exercise effects upon cognition in Alzheimer's disease: a systematic review of randomized controlled trials. Arch Ital Biol 2018; 156: 54–63, doi: 10.12871/00039829201816.
    » https://doi.org/10.12871/00039829201816
  • 48
    Vidoni ED, Gayed MR, Honea RA, Savage CR, Hobbs D, Burns JM. Alzheimer's disease alters the relationship of cardiorespiratory fitness with brain activity during the stroop task. Phys Ther 2013; 93: 993–1002, doi: 10.2522/ptj.20120465.
    » https://doi.org/10.2522/ptj.20120465
  • 49
    Smith JC, Lancaster MA, Nielson KA, Woodard JL, Seidenberg M, Durgerian S, et al. Interactive effects of physical activity and APOE-ε4 on white matter tract diffusivity in healthy elders. Neuroimage 2016; 131: 102–112, doi: 10.1016/j.neuroimage.2015.08.007.
    » https://doi.org/10.1016/j.neuroimage.2015.08.007
  • 50
    Solomon A, Turunen H, Ngandu T, Peltonen M, Levälahti E, Helisalmi S, et al. Effect of the apolipoprotein e genotype on cognitive change during a multidomain lifestyle intervention: a subgroup analysis of a randomized clinical trial. JAMA Neurol 2018; 75: 462–470, doi: 10.1001/jamaneurol.2017.4365.
    » https://doi.org/10.1001/jamaneurol.2017.4365
  • 51
    Shih IF, Paul K, Haan M, Yu Y, Ritz B. Physical activity modifies the influence of apolipoprotein E ε4 allele and type 2 diabetes on dementia and cognitive impairment among older Mexican Americans. Alzheimers Dement 2018; 14: 1–9, doi: 10.1016/j.jalz.2017.05.005.
    » https://doi.org/10.1016/j.jalz.2017.05.005
  • 52
    Ferrari C, Xu WL, Wang HX, Winblad B, Sorbi S, Qiu C, et al. How can elderly apolipoprotein E ε4 carriers remain free from dementia? Neurobiol Aging 2013; 34: 13–21, doi: 10.1016/j.neurobiolaging.2012.03.003.
    » https://doi.org/10.1016/j.neurobiolaging.2012.03.003
  • 53
    Jensen CS, Bahl JM, Ostergaard LB, Hogh P, Wermuth L, Heslegrave A, et al. 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, doi: 10.1016/j.exger.2019.04.003.
    » https://doi.org/10.1016/j.exger.2019.04.003
  • 54
    Fenesi B, Fang H, Kovacevic A, Oremus M, Raina P, Heisz JJ. Physical exercise moderates the relationship of Apolipoprotein E (APOE) genotype and dementia risk: a population-based study. J Alzheimer Dis 2017; 56: 297–303, doi: 10.3233/JAD-160424.
    » https://doi.org/10.3233/JAD-160424
  • 55
    Stern Y, MacKay-Brandt A, Lee S, McKinley P, McIntyre K, Razlighi Q, et al. Effect of aerobic exercise on cognition in younger adults: A randomized clinical trial. Neurology 2019; 92: e905–e916, doi: 10.1212/WNL.0000000000007003.
    » https://doi.org/10.1212/WNL.0000000000007003
  • 56
    Allard JS, Ntekim O, Johnson SP, Ngwa JS, Bond V, Pinder D, et al. APOEε4 impacts up-regulation of a brain-derived neurotrophic factor after a six-month stretch and aerobic exercise intervention in mild cognitively impaired elderly African Americans: a pilot study. Exp Gerontol 2017; 87: 129–136, doi: 10.1016/j.exger.2016.11.001.
    » https://doi.org/10.1016/j.exger.2016.11.001

Publication Dates

  • Publication in this collection
    09 Dec 2020
  • Date of issue
    2021

History

  • Received
    10 Apr 2020
  • Accepted
    18 Sept 2020
Associação Brasileira de Divulgação Científica Av. Bandeirantes, 3900, 14049-900 Ribeirão Preto SP Brazil, Tel. / Fax: +55 16 3315-9120 - Ribeirão Preto - SP - Brazil
E-mail: bjournal@terra.com.br