Abstract
Inactivity, obesity, and insulin resistance are significant risk factors for the development of Alzheimer’s disease (AD). Several studies have demonstrated that diet-induced obesity, inactivity, and insulin resistance exacerbate the neuropathological hallmarks of AD. The aggregation of β-amyloid peptides is one of these hallmarks. β-Site amyloid precursor protein-cleaving enzyme 1 (BACE1) is the rate-limiting enzyme in amyloid precursor protein (APP) processing, leading to β-amyloid peptide formation. Understanding how BACE1 content and activity are regulated is essential for establishing therapies aimed at reducing and/or slowing the progression of AD. Exercise training has been proven to reduce the risk of AD as well as decrease β-amyloid production and BACE1 content and/or activity. However, these long-term interventions also result in improvements in adiposity, circulating metabolites, glucose tolerance, and insulin sensitivity making it difficult to determine the direct effects of exercise on brain APP processing. This review highlights this large void in our knowledge and discusses our current understanding of the direct of effect of exercise on β-amyloid production. We have concentrated on the central role that brain-derived neurotrophic factor (BDNF) may play in mediating the direct effects of exercise on reducing brain BACE1 content and activity as well as β-amyloid production. Future studies should aim to generate a greater understanding of how obesity and exercise can directly alter APP processing and AD-related pathologies. This knowledge could provide evidence-based hypotheses for designing therapies to reduce the risk of AD and dementia.
Keywords: acute exercise, Alzheimer’s disease, β-amyloid, β-secretase
the world’s population is aging at an alarming rate with projections that the number of Alzheimer’s disease (AD) patients in the United States will reach 16 million by 2050 (7) and over 100 million people worldwide (8). In addition to this rapidly greying population, worldwide obesity has nearly doubled since 1980 (27). Both human research and animal research have established that inactivity and a high caloric intake resulting in obesity is a significant risk factor for cognitive impairment and the development of sporadic AD (28, 51, 63, 72, 119). This is alarming and highlights the need for identifying and developing strategies for the prevention and treatment of AD.
Currently, therapies are limited to temporary relief of the symptoms of AD and few are targeted at the underlying disease mechanisms. Lifestyle factors, such as increased exercise or physical activity, are known to reduce the risk of AD as well as slow the progression of the disease (22, 29, 55, 60, 95). However, the cellular and molecular mechanisms for such benefits have not yet been identified. Understanding these molecular mechanisms will have a significant impact on future therapeutic targets aimed at reducing AD.
The accumulation of senile plaques and accumulation of neurofibrillary tangles are neuropathologic hallmarks of AD. Senile plaques are extracellular aggregates of small peptides known as β-amyloid peptides, and neurofibrillary tangles are intracellular aggregates of the hyperphosphorylated microtubule-associated protein τ (40, 85). The excess accumulation and aggregation of β-amyloid peptides are detrimental to neuronal networks (34) and play a pivotal role as an upstream molecule in the process of neurodegeneration (38), thus identifying β-amyloid peptides as key players in the molecular mechanisms of early disease progression (33, 96). Previous work has shown that exercise training can reduce β-amyloid production. However, long-term exercise interventions also result in improvements in whole body health making it difficult to determine the direct effects of exercise on brain amyloid precursor protein (APP) processing. This leaves us with a large void in understanding the exact mechanism(s) behind exercise-induced changes in APP processing in the brain. This review will discuss our current understanding of APP processing and how exercise can alter the production of β-amyloid peptides. Furthermore, this review will summarize published evidence for a role of exercise-induced brain-derived neurotrophic factor (BDNF) on reducing β-amyloid peptide production.
APP Processing
The accumulation of β-amyloid peptides is central to the pathogenesis of AD (52, 69, 97) and can be observed almost a decade before cognitive impairment is seen in AD patients (9, 71, 101). This is known as preclinical AD where the pathological processes are active for several years before clinically detectable impairments (71). While the most reliable detection of β-amyloid plaques is by post mortem analysis, advances in imaging techniques such as positron emission tomography (PET) and analysis of cerebral spinal fluid (CSF) can now detect β-amyloid in vivo. Both PET and CSF β-amyloid highly correlate with brain biopsy findings (98, 118). Previous work has reported that preclinical AD, detected either by the CSF signature for AD (23, 42) or by PET, predicts symptomatic AD (71). Furthermore, the two methods were directly compared with two different assays in a clinical cohort of consecutive patients with mild cognitive impairment who later developed AD dementia. The study concluded that both CSF analysis and amyloid PET perform equally well and that either method can be used in the clinical workup of AD for increased diagnostic accuracy (83). Given this information, an understanding of the how β-amyloid production is regulated is vital to developing strategies aimed at the prevention and treatment of AD.
β-Amyloid peptides are proteolytic products of a type I transmembrane protein known as APP (87, 105). APP is synthesized at the endoplasmic reticulum and enters intracellular transport along the secretory, endocytic, and recycling routes in both the soma and neuronal processes (for review, see Ref. 75). APP processing occurs via one of two pathways, the nonamyloidogenic pathway and amyloidogenic pathway, which is characteristic of AD (Fig. 1). The nonamyloidogenic pathway is initiated by α-secretase, which cleaves APP releasing a large soluble fragment of APP (sAPPα) leaving the COOH-terminal fragment (APP-CTFα or C83) containing P3 and the APP intracellular domain (AICD) in the membrane. Subsequent cleavage of APP-CTFα by γ-secretase releases the nontoxic P3 fragment as well as AICD (81).
Fig. 1.
Amyloid precursor processing (APP). The nonamyloidogenic pathway (left) is initiated by α secretase, which cleaves APP releasing a large soluble fragment of APP (sAPPα) leaving the COOH-terminal fragment (APP-CTFα or C83) containing P3 and the APP intracellular domain (AICD) in the membrane. Subsequent cleavage of APP-CTFα by γ-secretase releases the nontoxic P3 fragment as well as AICD. The first step in the amyloidogenic pathway of APP processing (right) is the cleavage of APP by the β-secretase enzyme, also known as β-site APP-cleaving enzyme 1 (BACE1). BACE1 cleaves APP at the extra-membrane domain, producing a soluble fragment of APP (sAPPβ) and the COOH-terminal fragment (APP-CTFβ or C99). The sAPPβ peptide is released into the extracellular space. γ-Secretase subsequently cleaves APP-CTFβ at the intramembrane domain, producing β-amyloid and the AICD (31, 36, 56). The β-amyloid peptides then accumulate and aggregate forming β-amyloid plaques.
The first step in the amyloidogenic pathway of APP processing is the cleavage of APP by the β-secretase enzyme, also known as β-site APP-cleaving enzyme 1 (BACE1) (89). BACE1 is the rate-limiting enzyme in the production of β-amyloid peptides, and a significant increase in BACE1 activity has been reported in sporadic AD brains (108, 110, 121). Thus BACE1 can be an important therapeutic and/or preventive target in AD research. BACE1 cleaves APP at the extra-membrane domain, producing a soluble fragment of APP (sAPPβ) and the COOH-terminal fragment (APP-CTFβ or C99). The sAPPβ peptide is released into the extracellular space through endosome recycling (39). γ-Secretase subsequently cleaves APP-CTFβ at the intramembrane domain, producing β-amyloid and the AICD (31, 36, 56). The exact site of the γ-secretase cleavage of APP-CTFβ can vary, which results in the release of different β-amyloid peptide lengths (56). The most common β-amyloid peptide lengths are 40 and 42 amino acids, of which β-amyloid 42 is considered to be the most pathogenic in the development of AD (19) (Fig. 1).
Because of its role in β-amyloid peptide production, BACE1 is considered a biomarker for early detection, prediction, and progression of AD (37, 38, 109). Understanding how BACE1 content and activity are regulated holds potential for establishing therapies aimed at reducing and/or slowing the progression of AD. Although the level of BACE mRNA does not appear to be altered in human AD patients or transgenic mouse models of AD (48, 53, 123), BACE1 enzymatic activity is elevated in the aged brain (31) and in AD brains (30, 45, 122). In addition, humans with an APP mutation that effects the β-cleavage site (50) and mice with a targeted deletion of BACE1 (64, 94) are protected from β-amyloid peptide formation, thus providing strong evidence in support of the concept that BACE1 reduction or inhibition could be efficacious for the treatment of AD. Given this, BACE1 has become a key therapeutic target with several BACE1 inhibitors undergoing clinical trials (as reviewed in Refs. 108, 120). Currently, there are five companies (Merck, Biogen/Eisai, AstraZeneca/Eli Lilly, Johnson & Johnson, and Novartis) with BACE1 inhibitors in various phases (II-III) of clinical trials and various patient populations. With Eli Lilly having two BACE1 inhibitors fail in clinical trials (LY2811376 and LY2886721) and the recently halted clinical trial by Merck of the BACE1 inhibitor verubecestat (MK-8931) (74), the results from other ongoing trials using BACE1 inhibitor drugs are highly anticipated.
Obesity, Insulin Resistance, and APP Processing
Inactivity, obesity, and insulin resistance are significant risk factors in the development of AD (12, 66–68, 72, 88, 116). It is known that diet-induced obesity results in impaired cognitive function in rodents (43, 76) and humans (21, 119). Moreover, a link between insulin resistance and type 2 diabetes and increased β-amyloid deposition has been demonstrated in several transgenic mouse models of AD (12, 43, 51, 66–68, 91) as well as in humans (82). Together, this provides evidence that APP processing may be directly altered with obesity and insulin resistance. Several researchers have demonstrated that high-fat feeding increases the expression of BACE1 (104, 115, 124). In agreement with this hypothesis, rodent models of type 2 diabetes [diet-induced, streptozotocin (STZ)-induced, and genetic models] result in increased BACE1, APP, and β-amyloid production in the brain (61, 124). However, this is not always the case. Others have demonstrated that high-fat feeding increases the level of APP-CTFβ without changing APP or BACE1 content, indicating that a high-fat diet may increase BACE1 activity, followed by promotion of APP cleavage (67). In agreement with this, we have demonstrated that high-fat feeding of male wild-type C57BL6/J mice results in an increase in BACE1 activity with no change in BACE1 protein content or mRNA expression (65). The exact mechanisms leading to the changes in BACE1 content and/or activity are likely multifaceted; however, increased inflammation, cellular stress, and impaired energy metabolism represent early abnormalities that precede or accompany the initial stages of neurodegeneration, all of which may be directly altered by exercise (65) (Fig. 2A).
Fig. 2.
Potential effects of obesity and exercise on amyloid precursor processing. A: inactivity, obesity, and insulin resistance can result in increased β-site APP-cleaving enzyme 1 (BACE1) activity resulting in increased production of the soluble fragment of APP (sAPPβ), the COOH-terminal fragment (APP-CTFβ or C99) and β-amyloid peptides. B: potential role for exercise-induced BDNF signaling through TrKB on reducing β-amyloid production through increased α-secretase activity and reduced BACE1 content and/or activity.
Exercise and APP Processing
Evidence from epidemiological and experimental studies clearly indicates that regular physical exercise not only combats obesity and obesity-related comorbidities but that it also prevents cognitive decline due to aging and neurodegenerative diseases (17, 29, 41, 55, 60, 102, 114). Epidemiological studies have estimated that reduced physical activity (classified by total number of activities, hours per month, and percent intensity) was associated with a 250% increase in the risk of developing AD (29), while individuals who engage in physical activity [assessed from questionnaires and categorized as high (exercise 3 times/wk or more and an intensity higher than walking), moderate (exercise 3 times/wk or more and an intensity equal to walking), and low] have a significantly reduced risk of developing AD (60). Together, this provides evidence that exercise is an effective strategy to improve brain health and reduce and/or slow the progression of AD; however, the mechanisms driving these beneficial changes remain unknown. Several studies in animal models support existing epidemiological studies demonstrating the benefit of exercise in improving the pathology of AD (1, 14, 103, 106). The cognitive benefits of exercise have been shown in rodents submitted to long-term voluntary exercise in running wheels (79, 107), as well as after 3- to 12-wk training periods on a treadmill (4, 5, 10). Further work has demonstrated that endurance exercise training is an effective strategy for improving high-fat diet-induced cognitive impairment, which is more closely associated with the pathogenesis of AD in today’s society (13, 80). Treadmill training has been reported to improve spatial memory in different animal models of AD, such as lesion induced (46) or STZ induced (93), while long-term physical activity has been shown to slow and prevent high-fat diet-induced impairments in neurogenesis (18, 20, 57) and memory (70).
Evidence from animal studies suggests that exercise training could reduce the development of neurodegenerative processes by preventing β-amyloid peptide production. Specifically, Adlard et al. (1) demonstrated that increased physical activity decreases β-amyloid peptide levels in the TgCRND8 mouse model of AD. Using 5 mo of voluntary exercise, they found a reduction in β-amyloid 40 and β-amyloid 42 in both the cortical and hippocampal regions of the brain (1). This reduction in β-amyloid peptides was comparable to effects seen with β-amyloid immunization interventions where there is a 50% reduction in β-amyloid in this same transgenic line (49). In agreement with this work, Maesako et al. (67) demonstrated in APP transgenic mice that voluntary exercise inhibits high-fat diet-induced β-amyloid deposition and memory deficit. In a follow-up study, they demonstrated the importance of voluntary exercise over diet control in reducing β-amyloid accumulation and memory deficit (68). Studies utilizing controlled treadmill training have also demonstrated an exercise-induced reduction of β-amyloid peptides (62, 125). The mechanisms linking exercise to a reduction in β-amyloid peptides have not been elucidated; however, studies indicate that exercise can alter APP processing. Adlard et al. (1) found no change in mRNA or protein levels of neprilyin or insulin-degrading enzyme, both classic β-amyloid degradation pathways (1). Interestingly, 10 wk of voluntary exercise resulted in a downregulation of BACE1 activity in high-fat fed APP transgenic mice (67).
It is clear that long-term exercise or physical activity has beneficial effects in improving cognition and in reducing β-amyloid production. However, with exercise training and long-term physical activity there are changes external the brain that may result in indirect effects on brain health and signaling, such as reductions in adiposity as well as improved glucose homeostasis (65). This makes it difficult to determine if changes in the brain are due to direct effects of exercise or are secondary to changes in adiposity and/or improved insulin sensitivity/metabolic health (65). It is important to determine if exercise has a direct effect on the brain, without the long-term adaptations in other tissues, if we are to determine the mechanisms underlying the exercise-induced decreases in BACE1 content and/or activity and β-amyloid peptides. Understanding the direct effects of exercise on the brain is key to our understanding of the long-term changes induced by exercise and in developing effective lifestyle interventions or therapies aimed at reducing AD.
The use of acute (one bout) exercise provides an ideal model to examine the direct effects of exercise on brain APP processing and BACE1. To address this question, we recently examined the ability of one acute bout of exercise to reduce BACE1 content and activity in obese, insulin-resistant mice. We made the novel finding that one bout of exercise reduced BACE1 content and activity and reversed high-fat diet-induced markers of energetic stress in the cortex of obese male mice fed (65). These findings occurred in the absence of alterations in adiposity and circulating metabolites in the obese mice. This highlights the fact that exercise has a direct effect on brain BACE1 content and activity and provides further evidence in support of the therapeutic potential of exercise regardless of alterations in adiposity. However, the exercise-induced signaling cascades leading to reduced BACE1 content and activity remain to be determined. This knowledge concerning the underlying mechanisms is needed to develop efficient exercise programs and pharmaceutical targets aimed at reducing β-amyloid load.
Exercise and BDNF
A number of studies suggest that the synthesis and release of trophic factors, particularly BDNF, play a crucial role mediating the effects of exercise on the brain (77, 111). BDNF is a member of the neurotrophin family of growth factors and promotes neuronal survival, neurite outgrowth, and synaptic plasticity through its interactions with its receptor, tyrosine kinase B (TrkB) (84, 86). Furthermore, BDNF is vital for cognitive performance in the short term and for adaptations in brain morphology in the long term (15, 73). Growing evidence suggests that a decrease in BDNF levels could be associated with the pathogenesis of AD (25, 32). Two cross-sectional studies examining plasma BDNF concentrations between older adults with AD and healthy controls found that concentrations of BDNF were significantly lower in those with AD compared with controls providing evidence in support of a link between BDNF and AD pathology (58, 59). In addition to circulating BDNF, in brains from AD patients, BDNF expression is decreased in the hippocampus and some cortical areas such as the temporal and frontal cortex (24, 99).
Exercise is known to increase both circulating BDNF as well as BDNF content in the brain, yet surprisingly no studies have assessed the mechanistic link between acute exercise-induced increases in BDNF and decreased BACE1 content or activity. In humans, acute exercise stimulates a peripheral increase of BDNF from sites including the liver, muscles, and blood cells (161, 162). A recent review by Knaepen et al. (54) described the positive relationship between exercise intensity and plasma BDNF concentrations. The study suggested a dose-response relationship between acute exercise and plasma BDNF concentrations, with high-intensity and graded exercise tests eliciting the greatest exercise-induced increases in plasma BDNF concentration in healthy participants. In addition, there is also evidence that increases in plasma BDNF concentrations can be observed in response to a variety of different exercise protocols and modalities (e.g., step tests, V̇o2max tests, submaximal endurance exercise, and submaximal sprints) (26, 35, 90, 117). Importantly, BDNF can cross the blood-brain barrier and then function to stimulate central neurotrophin increases, especially in the hippocampus (16).
Binding of BDNF to TrkB receptors leads to the autophosphorylation of the intracellular tyrosine kinase domain of these receptors. This results in the activation of several downstream signaling pathways (47, 92). The downstream pathways activated by TrkB signaling include mitogen-activated protein kinase (MAPK), phospholipase C-c (PLC-c), phosphatidylinositol-3-kinase (PI3K), protein kinase C (PKC) (47), and cAMP-response element binding (CREB) protein (112, 113). It remains elusive if or how these pathways may be related to APP processing. In neuronal cultures, BDNF has specific and dose-dependent protective effect on neuronal toxicity induced by β-amyloid 42. When treated with a selective inhibitor of the tyrosine protein kinase activity of the Trk family (K252a) this protective effect was inhibited (6). These results demonstrate a direct link between BDNF binding to its TrkB receptor and reduced β-amyloid production. In an in vivo model, Vayman et al. (112, 113) demonstrated that exercise-induced BDNF promotes synaptic plasticity through downstream targets, CREB protein, synapsin I, and synaptophysin, while simultaneously increasing its own mRNA and its receptor TrkB. Whether this exercise-induced increase in BDNF and TrkB signaling is directly involved in β-amyloid production in AD requires further study. Supporting evidence for a role of BDNF and TrkB in AD comes from a cell culture model where TrkB signaling can modulate APP content and processing. In SH-SY5Y cells, retinoic acid can increase expression of TrkB and concomitant treatment with BDNF can increase APP promoter transcription and promote accumulation of sAPP-α and AICD by shifting APP processing toward the α-secretase or nonamyloidogenic pathway (44). Furthermore, recently published work from Nigam et al. (78) demonstrated that 3 wk of voluntary wheel running reduced β-amyloid levels and increased sAPPα in the hippocampus of a transgenic mouse model of AD. This was accompanied by a significant increase in hippocampal BDNF of the runner mice. These results lead the authors to hypothesize that exercise-induced BDNF may alter α-secretase activity. To investigate this, SH-SY5Y cells were treated with a α-secretase inhibitor, BDNF, or the combination of BDNF and the inhibitor. From these experiments, the authors concluded that BDNF reduces β-amyloid levels through a mechanisms involving increased α-secretase activity (78). Interestingly, cell lysates that were treated with the α-secretase inhibitor and BDNF displayed higher levels of sAPPα compared with cells that were treated only with the inhibitor. This indicates that BDNF may alter β-amyloid production through another pathway. It remains to be determined if BDNF has a direct effect on BACE1 in response to an acute bout of exercise remains.
Both chronic voluntary wheel running and moderate to high-intensity treadmill exercise training of rodents result in the upregulation of BDNF mRNA and protein content in the hippocampus (2, 3, 11, 100, 111). Similar to the human studies described above, studies investigating the effects of acute exercise on brain BDNF levels in rodents have similar findings where brain BDNF expression is increased. A study comparing low- and moderate-intensity acute treadmill exercise (30 min at 15 m/min vs. 30 min at 25 m/min) demonstrated that exercise resulted in elevations of BDNF mRNA in the hippocampus (specifically the CA1, CA3, and dentate gyrus regions) with the highest levels occurring ~1.5 h postexercise (179). This evidence showing that exercise increases circulating and brain BDNF content as well as signaling is strong; however, a direct link between BDNF signaling and BACE1 activity has yet to be determined.
Conclusions
There is a clear void in our understanding of the mechanisms underlying the direct effects of exercise on changes in BACE1 content and activity. This review has aimed to highlight a potential role for exercise-induced BDNF in reducing β-amyloid production through alterations in BACE1 content and/or activity. We suggest that BDNF signaling is directly involved in the exercise-induced reduction in BACE1 activity and β-amyloid production. However, more evidence is needed to support this idea and to elucidate the underlying signaling cascades linking the two. Future studies need to assess the extent to which BDNF actually mediates the effects of acute exercise on BACE1 and β-amyloid production.
Perspectives and Significance
The importance of BACE1 as the rate-limiting enzyme involved in the production of β-amyloid peptides is clear. Novel information gained from future studies exploring the direct effect of exercise-induced BDNF on BACE1 content and activity will enhance our understanding of the underlying mechanisms regulating APP processing and will set the foundation for therapeutic targets and drug development designed to improve approaches to prevent and treat AD. This information is valuable both in terms of understanding the underlying cellular mechanisms leading to decline in BACE1 and in terms of designing evidence-based preventative or therapeutic interventions for individuals with an elevated risk for AD.
GRANTS
This paper was supported by Canadian Network for Research and Innovation in Machining Technology, Natural Sciences and Engineering Research Council of Canada Grant RGPIN-2017-03904.
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the author.
AUTHOR CONTRIBUTIONS
R.E.M. conceived and designed research; R.E.M. prepared figures; R.E.M. drafted manuscript; R.E.M. edited and revised manuscript; R.E.M. approved final version of manuscript.
REFERENCES
- 1.Adlard PA, Perreau VM, Pop V, Cotman CW. Voluntary exercise decreases amyloid load in a transgenic model of Alzheimer’s disease. J Neurosci 25: 4217–4221, 2005. doi: 10.1523/JNEUROSCI.0496-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Aguiar AS Jr, Castro AA, Moreira EL, Glaser V, Santos AR, Tasca CI, Latini A, Prediger RD. Short bouts of mild-intensity physical exercise improve spatial learning and memory in aging rats: involvement of hippocampal plasticity via AKT, CREB and BDNF signaling. Mech Ageing Dev 132: 560–567, 2011. doi: 10.1016/j.mad.2011.09.005. [DOI] [PubMed] [Google Scholar]
- 3.Aguiar AS Jr, Tuon T, Pinho CA, Silva LA, Andreazza AC, Kapczinski F, Quevedo J, Streck EL, Pinho RA. Intense exercise induces mitochondrial dysfunction in mice brain. Neurochem Res 33: 51–58, 2008. doi: 10.1007/s11064-007-9406-x. [DOI] [PubMed] [Google Scholar]
- 4.Albeck DS, Sano K, Prewitt GE, Dalton L. Mild forced treadmill exercise enhances spatial learning in the aged rat. Behav Brain Res 168: 345–348, 2006. doi: 10.1016/j.bbr.2005.11.008. [DOI] [PubMed] [Google Scholar]
- 5.Ang ET, Dawe GS, Wong PT, Moochhala S, Ng YK. Alterations in spatial learning and memory after forced exercise. Brain Res 1113: 186–193, 2006. doi: 10.1016/j.brainres.2006.07.023. [DOI] [PubMed] [Google Scholar]
- 6.Arancibia S, Silhol M, Moulière F, Meffre J, Höllinger I, Maurice T, Tapia-Arancibia L. Protective effect of BDNF against beta-amyloid induced neurotoxicity in vitro and in vivo in rats. Neurobiol Dis 31: 316–326, 2008. doi: 10.1016/j.nbd.2008.05.012. [DOI] [PubMed] [Google Scholar]
- 7.Alzheimer’s Association 2012 Alzheimer’s disease facts and figures. Alzheimers Dement 8: 131–168, 2012. doi: 10.1016/j.jalz.2012.02.001. [DOI] [PubMed] [Google Scholar]
- 8.Barnes DE, Yaffe K. Accuracy of summary risk score for prediction of Alzheimer disease: better than demographics alone? Arch Neurol 68: 264, 2011. doi: 10.1001/archneurol.2011.4. [DOI] [PubMed] [Google Scholar]
- 9.Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, Marcus DS, Cairns NJ, Xie X, Blazey TM, Holtzman DM, Santacruz A, Buckles V, Oliver A, Moulder K, Aisen PS, Ghetti B, Klunk WE, McDade E, Martins RN, Masters CL, Mayeux R, Ringman JM, Rossor MN, Schofield PR, Sperling RA, Salloway S, Morris JC; Dominantly Inherited Alzheimer Network . Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med 367: 795–804, 2012. doi: 10.1056/NEJMoa1202753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Berchtold NC, Castello N, Cotman CW. Exercise and time-dependent benefits to learning and memory. Neuroscience 167: 588–597, 2010. doi: 10.1016/j.neuroscience.2010.02.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Berchtold NC, Chinn G, Chou M, Kesslak JP, Cotman CW. Exercise primes a molecular memory for brain-derived neurotrophic factor protein induction in the rat hippocampus. Neuroscience 133: 853–861, 2005. doi: 10.1016/j.neuroscience.2005.03.026. [DOI] [PubMed] [Google Scholar]
- 12.Bhat NR, Thirumangalakudi L. Increased tau phosphorylation and impaired brain insulin/IGF signaling in mice fed a high fat/high cholesterol diet. J Alzheimers Dis 36: 781–789, 2013. doi: 10.3233/JAD-2012-121030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Cheng J, Chen L, Han S, Qin L, Chen N, Wan Z. Treadmill running and rutin reverse high fat diet induced cognitive impairment in diet induced obese mice. J Nutr Health Aging 20: 503–508, 2016. doi: 10.1007/s12603-015-0616-7. [DOI] [PubMed] [Google Scholar]
- 14.Cho JY, Hwang DY, Kang TS, Shin DH, Hwang JH, Lim CH, Lee SH, Lim HJ, Min SH, Seo SJ, Song YS, Nam KT, Lee KS, Cho JS, Kim YK. Use of NSE/PS2m-transgenic mice in the study of the protective effect of exercise on Alzheimer’s disease. J Sports Sci 21: 943–951, 2003. doi: 10.1080/0264041031000140365. [DOI] [PubMed] [Google Scholar]
- 15.Cirulli F, Berry A, Chiarotti F, Alleva E. Intrahippocampal administration of BDNF in adult rats affects short-term behavioral plasticity in the Morris water maze and performance in the elevated plus-maze. Hippocampus 14: 802–807, 2004. doi: 10.1002/hipo.10220. [DOI] [PubMed] [Google Scholar]
- 16.Cotman CW, Berchtold NC, Christie LA. Exercise builds brain health: key roles of growth factor cascades and inflammation. Trends Neurosci 30: 464–472, 2007. doi: 10.1016/j.tins.2007.06.011. [DOI] [PubMed] [Google Scholar]
- 17.Cotman CW, Engesser-Cesar C. Exercise enhances and protects brain function. Exerc Sport Sci Rev 30: 75–79, 2002. doi: 10.1097/00003677-200204000-00006. [DOI] [PubMed] [Google Scholar]
- 18.Curlik DM 2nd, Shors TJ. Training your brain: do mental and physical (MAP) training enhance cognition through the process of neurogenesis in the hippocampus? Neuropharmacology 64: 506–514, 2013. doi: 10.1016/j.neuropharm.2012.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Duering M, Grimm MO, Grimm HS, Schröder J, Hartmann T. Mean age of onset in familial Alzheimer’s disease is determined by amyloid beta 42. Neurobiol Aging 26: 785–788, 2005. doi: 10.1016/j.neurobiolaging.2004.08.002. [DOI] [PubMed] [Google Scholar]
- 20.Eadie BD, Redila VA, Christie BR. Voluntary exercise alters the cytoarchitecture of the adult dentate gyrus by increasing cellular proliferation, dendritic complexity, and spine density. J Comp Neurol 486: 39–47, 2005. doi: 10.1002/cne.20493. [DOI] [PubMed] [Google Scholar]
- 21.Edwards LM, Murray AJ, Holloway CJ, Carter EE, Kemp GJ, Codreanu I, Brooker H, Tyler DJ, Robbins PA, Clarke K. Short-term consumption of a high-fat diet impairs whole-body efficiency and cognitive function in sedentary men. FASEB J 25: 1088–1096, 2011. doi: 10.1096/fj.10-171983. [DOI] [PubMed] [Google Scholar]
- 22.Erickson KI, Miller DL, Roecklein KA. The aging hippocampus: interactions between exercise, depression, and BDNF. Neuroscientist 18: 82–97, 2012. doi: 10.1177/1073858410397054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Fagan AM, Roe CM, Xiong C, Mintun MA, Morris JC, Holtzman DM. Cerebrospinal fluid tau/beta-amyloid(42) ratio as a prediction of cognitive decline in nondemented older adults. Arch Neurol 64: 343–349, 2007. doi: 10.1001/archneur.64.3.noc60123. [DOI] [PubMed] [Google Scholar]
- 24.Fahnestock M, Garzon D, Holsinger RM, Michalski B. Neurotrophic factors and Alzheimer’s disease: are we focusing on the wrong molecule? J Neural Transm Suppl 62: 241–252, 2002. doi: 10.1007/978-3-7091-6139-5_22. [DOI] [PubMed] [Google Scholar]
- 25.Ferrer I, Marín C, Rey MJ, Ribalta T, Goutan E, Blanco R, Tolosa E, Martí E. BDNF and full-length and truncated TrkB expression in Alzheimer disease. Implications in therapeutic strategies. J Neuropathol Exp Neurol 58: 729–739, 1999. doi: 10.1097/00005072-199907000-00007. [DOI] [PubMed] [Google Scholar]
- 26.Ferris LT, Williams JS, Shen CL. The effect of acute exercise on serum brain-derived neurotrophic factor levels and cognitive function. Med Sci Sports Exerc 39: 728–734, 2007. doi: 10.1249/mss.0b013e31802f04c7. [DOI] [PubMed] [Google Scholar]
- 27.Finucane MM, Stevens GA, Cowan MJ, Danaei G, Lin JK, Paciorek CJ, Singh GM, Gutierrez HR, Lu Y, Bahalim AN, Farzadfar F, Riley LM, Ezzati M; Global Burden of Metabolic Risk Factors of Chronic Diseases Collaborating Group (Body Mass Index) . National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9·1 million participants. Lancet 377: 557–567, 2011. doi: 10.1016/S0140-6736(10)62037-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Francis H, Stevenson R. The longer-term impacts of Western diet on human cognition and the brain. Appetite 63: 119–128, 2013. doi: 10.1016/j.appet.2012.12.018. [DOI] [PubMed] [Google Scholar]
- 29.Friedland RP, Fritsch T, Smyth KA, Koss E, Lerner AJ, Chen CH, Petot GJ, Debanne SM. Patients with Alzheimer’s disease have reduced activities in midlife compared with healthy control-group members. Proc Natl Acad Sci USA 98: 3440–3445, 2001. doi: 10.1073/pnas.061002998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Fukumoto H, Cheung BS, Hyman BT, Irizarry MC. Beta-secretase protein and activity are increased in the neocortex in Alzheimer disease. Arch Neurol 59: 1381–1389, 2002. doi: 10.1001/archneur.59.9.1381. [DOI] [PubMed] [Google Scholar]
- 31.Fukumoto H, Rosene DL, Moss MB, Raju S, Hyman BT, Irizarry MC. Beta-secretase activity increases with aging in human, monkey, and mouse brain. Am J Pathol 164: 719–725, 2004. doi: 10.1016/S0002-9440(10)63159-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Fumagalli F, Racagni G, Riva MA. The expanding role of BDNF: a therapeutic target for Alzheimer’s disease? Pharmacogenomics J 6: 8–15, 2006. doi: 10.1038/sj.tpj.6500337. [DOI] [PubMed] [Google Scholar]
- 33.Gandy S. The role of cerebral amyloid beta accumulation in common forms of Alzheimer disease. J Clin Invest 115: 1121–1129, 2005. doi: 10.1172/JCI25100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gouras GK, Almeida CG, Takahashi RH. Intraneuronal Abeta accumulation and origin of plaques in Alzheimer’s disease. Neurobiol Aging 26: 1235–1244, 2005. doi: 10.1016/j.neurobiolaging.2005.05.022. [DOI] [PubMed] [Google Scholar]
- 35.Griffin EW, Mullally S, Foley C, Warmington SA, O’Mara SM, Kelly AM. Aerobic exercise improves hippocampal function and increases BDNF in the serum of young adult males. Physiol Behav 104: 934–941, 2011. doi: 10.1016/j.physbeh.2011.06.005. [DOI] [PubMed] [Google Scholar]
- 36.Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat Rev Mol Cell Biol 8: 101–112, 2007. doi: 10.1038/nrm2101. [DOI] [PubMed] [Google Scholar]
- 37.Hampel H, Shen Y. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) as a biological candidate marker of Alzheimer’s disease. Scand J Clin Lab Invest 69: 8–12, 2009. doi: 10.1080/00365510701864610. [DOI] [PubMed] [Google Scholar]
- 38.Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science 256: 184–185, 1992. doi: 10.1126/science.1566067. [DOI] [PubMed] [Google Scholar]
- 39.Hasebe N, Fujita Y, Ueno M, Yoshimura K, Fujino Y, Yamashita T. Soluble β-amyloid Precursor Protein Alpha binds to p75 neurotrophin receptor to promote neurite outgrowth. PLoS One 8: e82321, 2013. doi: 10.1371/journal.pone.0082321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hauw JJ, Verny M, Delaère P, Cervera P, He Y, Duyckaerts C. Constant neurofibrillary changes in the neocortex in progressive supranuclear palsy. Basic differences with Alzheimer’s disease and aging. Neurosci Lett 119: 182–186, 1990. doi: 10.1016/0304-3940(90)90829-X. [DOI] [PubMed] [Google Scholar]
- 41.Hayes SM, Alosco ML, Forman DE. The effects of aerobic exercise on cognitive and neural decline in aging and cardiovascular disease. Curr Geriatr Rep 3: 282–290, 2014. doi: 10.1007/s13670-014-0101-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Herukka SK, Hallikainen M, Soininen H, Pirttilä T. CSF Abeta42 and tau or phosphorylated tau and prediction of progressive mild cognitive impairment. Neurology 64: 1294–1297, 2005. doi: 10.1212/01.WNL.0000156914.16988.56. [DOI] [PubMed] [Google Scholar]
- 43.Ho L, Qin W, Pompl PN, Xiang Z, Wang J, Zhao Z, Peng Y, Cambareri G, Rocher A, Mobbs CV, Hof PR, Pasinetti GM. Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s disease. FASEB J 18: 902–904, 2004. doi: 10.1096/fj.03-0978fje. [DOI] [PubMed] [Google Scholar]
- 44.Holback S, Adlerz L, Iverfeldt K. Increased processing of APLP2 and APP with concomitant formation of APP intracellular domains in BDNF and retinoic acid-differentiated human neuroblastoma cells. J Neurochem 95: 1059–1068, 2005. doi: 10.1111/j.1471-4159.2005.03440.x. [DOI] [PubMed] [Google Scholar]
- 45.Holsinger RM, McLean CA, Beyreuther K, Masters CL, Evin G. Increased expression of the amyloid precursor beta-secretase in Alzheimer’s disease. Ann Neurol 51: 783–786, 2002. doi: 10.1002/ana.10208. [DOI] [PubMed] [Google Scholar]
- 46.Hoveida R, Alaei H, Oryan S, Parivar K, Reisi P. Treadmill running improves spatial memory in an animal model of Alzheimer’s disease. Behav Brain Res 216: 270–274, 2011. doi: 10.1016/j.bbr.2010.08.003. [DOI] [PubMed] [Google Scholar]
- 47.Huang EJ, Reichardt LF. Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci 24: 677–736, 2001. doi: 10.1146/annurev.neuro.24.1.677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Irizarry MC, Locascio JJ, Hyman BT. β-Site APP cleaving enzyme mRNA expression in APP transgenic mice: anatomical overlap with transgene expression and static levels with aging. Am J Pathol 158: 173–177, 2001. doi: 10.1016/S0002-9440(10)63955-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Janus C, Pearson J, McLaurin J, Mathews PM, Jiang Y, Schmidt SD, Chishti MA, Horne P, Heslin D, French J, Mount HT, Nixon RA, Mercken M, Bergeron C, Fraser PE, St George-Hyslop P, Westaway D. A beta peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer’s disease. Nature 408: 979–982, 2000. doi: 10.1038/35050110. [DOI] [PubMed] [Google Scholar]
- 50.Jonsson T, Atwal JK, Steinberg S, Snaedal J, Jonsson PV, Bjornsson S, Stefansson H, Sulem P, Gudbjartsson D, Maloney J, Hoyte K, Gustafson A, Liu Y, Lu Y, Bhangale T, Graham RR, Huttenlocher J, Bjornsdottir G, Andreassen OA, Jönsson EG, Palotie A, Behrens TW, Magnusson OT, Kong A, Thorsteinsdottir U, Watts RJ, Stefansson K. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature 488: 96–99, 2012. doi: 10.1038/nature11283. [DOI] [PubMed] [Google Scholar]
- 51.Julien C, Tremblay C, Phivilay A, Berthiaume L, Emond V, Julien P, Calon F. High-fat diet aggravates amyloid-beta and tau pathologies in the 3xTg-AD mouse model. Neurobiol Aging 31: 1516–1531, 2010. doi: 10.1016/j.neurobiolaging.2008.08.022. [DOI] [PubMed] [Google Scholar]
- 52.Kim D, Tsai LH. Bridging physiology and pathology in AD. Cell 137: 997–1000, 2009. doi: 10.1016/j.cell.2009.05.042. [DOI] [PubMed] [Google Scholar]
- 53.Kimura N, Nakamura SI, Honda T, Takashima A, Nakayama H, Ono F, Sakakibara I, Doi K, Kawamura S, Yoshikawa Y. Age-related changes in the localization of presenilin-1 in cynomolgus monkey brain. Brain Res 922: 30–41, 2001. doi: 10.1016/S0006-8993(01)03146-8. [DOI] [PubMed] [Google Scholar]
- 54.Knaepen K, Goekint M, Heyman EM, Meeusen R. Neuroplasticity–exercise-induced response of peripheral brain-derived neurotrophic factor: a systematic review of experimental studies in human subjects. Sports Med 40: 765–801, 2010. doi: 10.2165/11534530-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 55.Kramer AF, Erickson KI, Colcombe SJ. Exercise, cognition, and the aging brain. J Appl Physiol (1985) 101: 1237–1242, 2006. doi: 10.1152/japplphysiol.00500.2006. [DOI] [PubMed] [Google Scholar]
- 56.Kummer MP, Heneka MT. Truncated and modified amyloid-beta species. Alzheimers Res Ther 6: 28, 2014. doi: 10.1186/alzrt258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lafenêtre P, Leske O, Ma-Högemeie Z, Haghikia A, Bichler Z, Wahle P, Heumann R. Exercise can rescue recognition memory impairment in a model with reduced adult hippocampal neurogenesis. Front Behav Neurosci 3: 34, 2010. 10.3389/neuro.08.034.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Laske C, Stransky E, Leyhe T, Eschweiler GW, Maetzler W, Wittorf A, Soekadar S, Richartz E, Koehler N, Bartels M, Buchkremer G, Schott K. BDNF serum and CSF concentrations in Alzheimer’s disease, normal pressure hydrocephalus and healthy controls. J Psychiatr Res 41: 387–394, 2007. doi: 10.1016/j.jpsychires.2006.01.014. [DOI] [PubMed] [Google Scholar]
- 59.Laske C, Stransky E, Leyhe T, Eschweiler GW, Schott K, Langer H, Gawaz M. Decreased brain-derived neurotrophic factor (BDNF)- and β-thromboglobulin (β-TG)- blood levels in Alzheimer’s disease. Thromb Haemost 96: 102–103, 2006. doi: 10.1160/TH06-03-0173. [DOI] [PubMed] [Google Scholar]
- 60.Laurin D, Verreault R, Lindsay J, MacPherson K, Rockwood K. Physical activity and risk of cognitive impairment and dementia in elderly persons. Arch Neurol 58: 498–504, 2001. doi: 10.1001/archneur.58.3.498. [DOI] [PubMed] [Google Scholar]
- 61.Li ZG, Zhang W, Sima AA. Alzheimer-like changes in rat models of spontaneous diabetes. Diabetes 56: 1817–1824, 2007. doi: 10.2337/db07-0171. [DOI] [PubMed] [Google Scholar]
- 62.Liu HL, Zhao G, Zhang H, Shi LD. Long-term treadmill exercise inhibits the progression of Alzheimer’s disease-like neuropathology in the hippocampus of APP/PS1 transgenic mice. Behav Brain Res 256: 261–272, 2013. doi: 10.1016/j.bbr.2013.08.008. [DOI] [PubMed] [Google Scholar]
- 63.Luchsinger JA, Tang MX, Shea S, Mayeux R. Caloric intake and the risk of Alzheimer disease. Arch Neurol 59: 1258–1263, 2002. doi: 10.1001/archneur.59.8.1258. [DOI] [PubMed] [Google Scholar]
- 64.Luo Y, Bolon B, Kahn S, Bennett BD, Babu-Khan S, Denis P, Fan W, Kha H, Zhang J, Gong Y, Martin L, Louis JC, Yan Q, Richards WG, Citron M, Vassar R. Mice deficient in BACE1, the Alzheimer’s beta-secretase, have normal phenotype and abolished beta-amyloid generation. Nat Neurosci 4: 231–232, 2001. doi: 10.1038/85059. [DOI] [PubMed] [Google Scholar]
- 65.MacPherson RE, Baumeister P, Peppler WT, Wright DC, Little JP. Reduced cortical BACE1 content with one bout of exercise is accompanied by declines in AMPK, AKT, and MAPK signaling in obese, glucose intolerant mice. J Appl Physiol (1985) 119: 1097–1104, 2015. doi: 10.1152/japplphysiol.00299.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Maesako M, Uemura K, Iwata A, Kubota M, Watanabe K, Uemura M, Noda Y, Asada-Utsugi M, Kihara T, Takahashi R, Shimohama S, Kinoshita A. Continuation of exercise is necessary to inhibit high fat diet-induced β-amyloid deposition and memory deficit in amyloid precursor protein transgenic mice. PLoS One 8: e72796, 2013. doi: 10.1371/journal.pone.0072796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Maesako M, Uemura K, Kubota M, Kuzuya A, Sasaki K, Asada M, Watanabe K, Hayashida N, Ihara M, Ito H, Shimohama S, Kihara T, and Kinoshita A. Environmental enrichment ameliorated high-fat diet-induced Aβ deposition and memory deficit in APP transgenic mice. Neurobiol Aging 33: 1011 e1011–1023, 2012. doi: 10.1016/j.neurobiolaging.2011.10.028. [DOI] [PubMed] [Google Scholar]
- 68.Maesako M, Uemura K, Kubota M, Kuzuya A, Sasaki K, Hayashida N, Asada-Utsugi M, Watanabe K, Uemura M, Kihara T, Takahashi R, Shimohama S, Kinoshita A. Exercise is more effective than diet control in preventing high fat diet-induced β-amyloid deposition and memory deficit in amyloid precursor protein transgenic mice. J Biol Chem 287: 23024–23033, 2012. doi: 10.1074/jbc.M112.367011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Marcello E, Epis R, Di Luca M. Amyloid flirting with synaptic failure: towards a comprehensive view of Alzheimer’s disease pathogenesis. Eur J Pharmacol 585: 109–118, 2008. doi: 10.1016/j.ejphar.2007.11.083. [DOI] [PubMed] [Google Scholar]
- 70.Molteni R, Wu A, Vaynman S, Ying Z, Barnard RJ, Gómez-Pinilla F. Exercise reverses the harmful effects of consumption of a high-fat diet on synaptic and behavioral plasticity associated to the action of brain-derived neurotrophic factor. Neuroscience 123: 429–440, 2004. doi: 10.1016/j.neuroscience.2003.09.020. [DOI] [PubMed] [Google Scholar]
- 71.Morris JC, Roe CM, Grant EA, Head D, Storandt M, Goate AM, Fagan AM, Holtzman DM, Mintun MA. Pittsburgh compound B imaging and prediction of progression from cognitive normality to symptomatic Alzheimer disease. Arch Neurol 66: 1469–1475, 2009. doi: 10.1001/archneurol.2009.269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Morris MC, Evans DA, Bienias JL, Tangney CC, Bennett DA, Aggarwal N, Schneider J, Wilson RS. Dietary fats and the risk of incident Alzheimer disease. Arch Neurol 60: 194–200, 2003. doi: 10.1001/archneur.60.2.194. [DOI] [PubMed] [Google Scholar]
- 73.Mu JS, Li WP, Yao ZB, Zhou XF. Deprivation of endogenous brain-derived neurotrophic factor results in impairment of spatial learning and memory in adult rats. Brain Res 835: 259–265, 1999. doi: 10.1016/S0006-8993(99)01592-9. [DOI] [PubMed] [Google Scholar]
- 74.Mullard A. BACE inhibitor bust in Alzheimer trial. Nat Rev Drug Discov 16: 155, 2017. 10.1038/nrd.2017.43. [DOI] [PubMed] [Google Scholar]
- 75.Muresan V, Ladescu Muresan Z. Amyloid-β precursor protein: Multiple fragments, numerous transport routes and mechanisms. Exp Cell Res 334: 45–53, 2015. doi: 10.1016/j.yexcr.2014.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Murray AJ, Knight NS, Cochlin LE, McAleese S, Deacon RM, Rawlins JN, Clarke K. Deterioration of physical performance and cognitive function in rats with short-term high-fat feeding. FASEB J 23: 4353–4360, 2009. doi: 10.1096/fj.09-139691. [DOI] [PubMed] [Google Scholar]
- 77.Nascimento CM, Pereira JR, Pires de Andrade L, Garuffi M, Ayan C, Kerr DS, Talib LL, Cominetti MR, Stella F. Physical exercise improves peripheral BDNF levels and cognitive functions in mild cognitive impairment elderly with different bdnf Val66Met genotypes. J Alzheimers Dis 43: 81–91, 2015. doi: 10.3233/JAD-140576. [DOI] [PubMed] [Google Scholar]
- 78.Nigam SM, Xu S, Kritikou JS, Marosi K, Brodin L, Mattson MP. Exercise and BDNF reduce Aβ production by enhancing α-secretase processing of APP. J Neurochem 142: 286–296, 2017. doi: 10.1111/jnc.14034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Nithianantharajah J, Hannan AJ. The neurobiology of brain and cognitive reserve: mental and physical activity as modulators of brain disorders. Prog Neurobiol 89: 369–382, 2009. doi: 10.1016/j.pneurobio.2009.10.001. [DOI] [PubMed] [Google Scholar]
- 80.Noble EE, Mavanji V, Little MR, Billington CJ, Kotz CM, Wang C. Exercise reduces diet-induced cognitive decline and increases hippocampal brain-derived neurotrophic factor in CA3 neurons. Neurobiol Learn Mem 114: 40–50, 2014. doi: 10.1016/j.nlm.2014.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Nunan J, Small DH. Regulation of APP cleavage by alpha-, beta- and gamma-secretases. FEBS Lett 483: 6–10, 2000. doi: 10.1016/S0014-5793(00)02076-7. [DOI] [PubMed] [Google Scholar]
- 82.Ohara T, Doi Y, Ninomiya T, Hirakawa Y, Hata J, Iwaki T, Kanba S, Kiyohara Y. Glucose tolerance status and risk of dementia in the community: the Hisayama study. Neurology 77: 1126–1134, 2011. doi: 10.1212/WNL.0b013e31822f0435. [DOI] [PubMed] [Google Scholar]
- 83.Palmqvist S, Zetterberg H, Mattsson N, Johansson P, Minthon L, Blennow K, Olsson M, Hansson O; Alzheimer’s Disease Neuroimaging Initiative; Swedish BioFINDER Study Group . Detailed comparison of amyloid PET and CSF biomarkers for identifying early Alzheimer disease. Neurology 85: 1240–1249, 2015. doi: 10.1212/WNL.0000000000001991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Patapoutian A, Reichardt LF. Trk receptors: mediators of neurotrophin action. Curr Opin Neurobiol 11: 272–280, 2001. doi: 10.1016/S0959-4388(00)00208-7. [DOI] [PubMed] [Google Scholar]
- 85.Pitt J, Thorner M, Brautigan D, Larner J, Klein WL. Protection against the synaptic targeting and toxicity of Alzheimer’s-associated Aβ oligomers by insulin mimetic chiro-inositols. FASEB J 27: 199–207, 2013. doi: 10.1096/fj.12-211896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Poo MM. Neurotrophins as synaptic modulators. Nat Rev Neurosci 2: 24–32, 2001. doi: 10.1038/35049004. [DOI] [PubMed] [Google Scholar]
- 87.Priller C, Bauer T, Mitteregger G, Krebs B, Kretzschmar HA, Herms J. Synapse formation and function is modulated by the amyloid precursor protein. J Neurosci 26: 7212–7221, 2006. doi: 10.1523/JNEUROSCI.1450-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Profenno LA, Porsteinsson AP, Faraone SV. Meta-analysis of Alzheimer’s disease risk with obesity, diabetes, and related disorders. Biol Psychiatry 67: 505–512, 2010. doi: 10.1016/j.biopsych.2009.02.013. [DOI] [PubMed] [Google Scholar]
- 89.Rajendran L, Honsho M, Zahn TR, Keller P, Geiger KD, Verkade P, Simons K. Alzheimer’s disease beta-amyloid peptides are released in association with exosomes. Proc Natl Acad Sci USA 103: 11172–11177, 2006. doi: 10.1073/pnas.0603838103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Rasmussen P, Brassard P, Adser H, Pedersen MV, Leick L, Hart E, Secher NH, Pedersen BK, Pilegaard H. Evidence for a release of brain-derived neurotrophic factor from the brain during exercise. Exp Physiol 94: 1062–1069, 2009. doi: 10.1113/expphysiol.2009.048512. [DOI] [PubMed] [Google Scholar]
- 91.Refolo LM, Pappolla MA, Malester B, LaFrancois J, Bryant-Thomas T, Wang R, Tint GS, Sambamurti K, Duff K. Hypercholesterolemia accelerates the Alzheimer’s amyloid pathology in a transgenic mouse model. Neurobiol Dis 7: 321–331, 2000. doi: 10.1006/nbdi.2000.0304. [DOI] [PubMed] [Google Scholar]
- 92.Reichardt LF. Neurotrophin-regulated signalling pathways. Philos Trans R Soc Lond B Biol Sci 361: 1545–1564, 2006. doi: 10.1098/rstb.2006.1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Reisi P, Alaei H, Babri S, Sharifi MR, Mohaddes G. Effects of treadmill running on spatial learning and memory in streptozotocin-induced diabetic rats. Neurosci Lett 455: 79–83, 2009. doi: 10.1016/j.neulet.2009.03.052. [DOI] [PubMed] [Google Scholar]
- 94.Roberds SL, Anderson J, Basi G, Bienkowski MJ, Branstetter DG, Chen KS, Freedman SB, Frigon NL, Games D, Hu K, Johnson-Wood K, Kappenman KE, Kawabe TT, Kola I, Kuehn R, Lee M, Liu W, Motter R, Nichols NF, Power M, Robertson DW, Schenk D, Schoor M, Shopp GM, Shuck ME, Sinha S, Svensson KA, Tatsuno G, Tintrup H, Wijsman J, Wright S, McConlogue L. BACE knockout mice are healthy despite lacking the primary beta-secretase activity in brain: implications for Alzheimer’s disease therapeutics. Hum Mol Genet 10: 1317–1324, 2001. doi: 10.1093/hmg/10.12.1317. [DOI] [PubMed] [Google Scholar]
- 95.Scarmeas N, Luchsinger JA, Schupf N, Brickman AM, Cosentino S, Tang MX, Stern Y. Physical activity, diet, and risk of Alzheimer disease. JAMA 302: 627–637, 2009. doi: 10.1001/jama.2009.1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Selkoe DJ; American College of Physicians; American Physiological Society . Alzheimer disease: mechanistic understanding predicts novel therapies. Ann Intern Med 140: 627–638, 2004. doi: 10.7326/0003-4819-140-8-200404200-00047. [DOI] [PubMed] [Google Scholar]
- 97.Selkoe DJ. Soluble oligomers of the amyloid beta-protein impair synaptic plasticity and behavior. Behav Brain Res 192: 106–113, 2008. doi: 10.1016/j.bbr.2008.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Seppälä TT, Nerg O, Koivisto AM, Rummukainen J, Puli L, Zetterberg H, Pyykkö OT, Helisalmi S, Alafuzoff I, Hiltunen M, Jääskeläinen JE, Rinne J, Soininen H, Leinonen V, Herukka SK. CSF biomarkers for Alzheimer disease correlate with cortical brain biopsy findings. Neurology 78: 1568–1575, 2012. doi: 10.1212/WNL.0b013e3182563bd0. [DOI] [PubMed] [Google Scholar]
- 99.Siegel GJ, Chauhan NB. Neurotrophic factors in Alzheimer’s and Parkinson’s disease brain. Brain Res Rev 33: 199–227, 2000. doi: 10.1016/S0165-0173(00)00030-8. [DOI] [PubMed] [Google Scholar]
- 100.Soya H, Nakamura T, Deocaris CC, Kimpara A, Iimura M, Fujikawa T, Chang H, McEwen BS, Nishijima T. BDNF induction with mild exercise in the rat hippocampus. Biochem Biophys Res Commun 358: 961–967, 2007. doi: 10.1016/j.bbrc.2007.04.173. [DOI] [PubMed] [Google Scholar]
- 101.Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, Iwatsubo T, Jack CR Jr., Kaye J, Montine TJ, Park DC, Reiman EM, Rowe CC, Siemers E, Stern Y, Yaffe K, Carrillo MC, Thies B, Morrison-Bogorad M, Wagster MV, Phelps CH. Toward defining the preclinical stages of Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement 7: 280–292, 2011. doi: 10.1016/j.jalz.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Svensson M, Lexell J, Deierborg T. Effects of physical exercise on neuroinflammation, neuroplasticity, neurodegeneration, and behavior: what we can learn from animal models in clinical settings. Neurorehabil Neural Repair 29: 577–589, 2015. doi: 10.1177/1545968314562108. [DOI] [PubMed] [Google Scholar]
- 103.Tapia-Rojas C, Aranguiz F, Varela-Nallar L, Inestrosa NC. Voluntary running attenuates memory loss, decreases neuropathological changes and induces neurogenesis in a mouse model of Alzheimer’s disease. Brain Pathol 26: 62–74, 2016. doi: 10.1111/bpa.12255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Thirumangalakudi L, Prakasam A, Zhang R, Bimonte-Nelson H, Sambamurti K, Kindy MS, Bhat NR. High cholesterol-induced neuroinflammation and amyloid precursor protein processing correlate with loss of working memory in mice. J Neurochem 106: 475–485, 2008. doi: 10.1111/j.1471-4159.2008.05415.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Turner PR, O’Connor K, Tate WP, Abraham WC. Roles of amyloid precursor protein and its fragments in regulating neural activity, plasticity and memory. Prog Neurobiol 70: 1–32, 2003. doi: 10.1016/S0301-0082(03)00089-3. [DOI] [PubMed] [Google Scholar]
- 106.Um HS, Kang EB, Leem YH, Cho IH, Yang CH, Chae KR, Hwang DY, Cho JY. Exercise training acts as a therapeutic strategy for reduction of the pathogenic phenotypes for Alzheimer’s disease in an NSE/APPsw-transgenic model. Int J Mol Med 22: 529–539, 2008. [PubMed] [Google Scholar]
- 107.van Praag H, Shubert T, Zhao C, Gage FH. Exercise enhances learning and hippocampal neurogenesis in aged mice. J Neurosci 25: 8680–8685, 2005. doi: 10.1523/JNEUROSCI.1731-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Vassar R. BACE1 inhibitor drugs in clinical trials for Alzheimer’s disease. Alzheimers Res Ther 6: 89, 2014. doi: 10.1186/s13195-014-0089-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, Teplow DB, Ross S, Amarante P, Loeloff R, Luo Y, Fisher S, Fuller J, Edenson S, Lile J, Jarosinski MA, Biere AL, Curran E, Burgess T, Louis JC, Collins F, Treanor J, Rogers G, Citron M. Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science 286: 735–741, 1999. doi: 10.1126/science.286.5440.735. [DOI] [PubMed] [Google Scholar]
- 110.Vassar R, Kandalepas PC. The β-secretase enzyme BACE1 as a therapeutic target for Alzheimer’s disease. Alzheimers Res Ther 3: 20, 2011. doi: 10.1186/alzrt82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Vaynman S, Ying Z, Gomez-Pinilla F. Hippocampal BDNF mediates the efficacy of exercise on synaptic plasticity and cognition. Eur J Neurosci 20: 2580–2590, 2004. doi: 10.1111/j.1460-9568.2004.03720.x. [DOI] [PubMed] [Google Scholar]
- 112.Vaynman S, Ying Z, Gomez-Pinilla F. Interplay between brain-derived neurotrophic factor and signal transduction modulators in the regulation of the effects of exercise on synaptic-plasticity. Neuroscience 122: 647–657, 2003. doi: 10.1016/j.neuroscience.2003.08.001. [DOI] [PubMed] [Google Scholar]
- 113.Vaynman S, Ying Z, Wu A, Gomez-Pinilla F. Coupling energy metabolism with a mechanism to support brain-derived neurotrophic factor-mediated synaptic plasticity. Neuroscience 139: 1221–1234, 2006. doi: 10.1016/j.neuroscience.2006.01.062. [DOI] [PubMed] [Google Scholar]
- 114.Voss MW, Vivar C, Kramer AF, van Praag H. Bridging animal and human models of exercise-induced brain plasticity. Trends Cogn Sci 17: 525–544, 2013. doi: 10.1016/j.tics.2013.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Wang R, Li JJ, Diao S, Kwak YD, Liu L, Zhi L, Büeler H, Bhat NR, Williams RW, Park EA, Liao FF. Metabolic stress modulates Alzheimer’s β-secretase gene transcription via SIRT1-PPARγ-PGC-1 in neurons. Cell Metab 17: 685–694, 2013. doi: 10.1016/j.cmet.2013.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Whitmer RA. The epidemiology of adiposity and dementia. Curr Alzheimer Res 4: 117–122, 2007. doi: 10.2174/156720507780362065. [DOI] [PubMed] [Google Scholar]
- 117.Winter B, Breitenstein C, Mooren FC, Voelker K, Fobker M, Lechtermann A, Krueger K, Fromme A, Korsukewitz C, Floel A, Knecht S. High impact running improves learning. Neurobiol Learn Mem 87: 597–609, 2007. doi: 10.1016/j.nlm.2006.11.003. [DOI] [PubMed] [Google Scholar]
- 118.Wolk DA, Grachev ID, Buckley C, Kazi H, Grady MS, Trojanowski JQ, Hamilton RH, Sherwin P, McLain R, Arnold SE. Association between in vivo fluorine 18-labeled flutemetamol amyloid positron emission tomography imaging and in vivo cerebral cortical histopathology. Arch Neurol 68: 1398–1403, 2011. doi: 10.1001/archneurol.2011.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Xu WL, Atti AR, Gatz M, Pedersen NL, Johansson B, Fratiglioni L. Midlife overweight and obesity increase late-life dementia risk: a population-based twin study. Neurology 76: 1568–1574, 2011. doi: 10.1212/WNL.0b013e3182190d09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Yan R. Stepping closer to treating Alzheimer’s disease patients with BACE1 inhibitor drugs. Transl Neurodegener 5: 13, 2016. doi: 10.1186/s40035-016-0061-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Yan R, Fan Q, Zhou J, Vassar R. Inhibiting BACE1 to reverse synaptic dysfunctions in Alzheimer’s disease. Neurosci Biobehav Rev 65: 326–340, 2016. doi: 10.1016/j.neubiorev.2016.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Yang LB, Lindholm K, Yan R, Citron M, Xia W, Yang XL, Beach T, Sue L, Wong P, Price D, Li R, Shen Y. Elevated beta-secretase expression and enzymatic activity detected in sporadic Alzheimer disease. Nat Med 9: 3–4, 2003. doi: 10.1038/nm0103-3. [DOI] [PubMed] [Google Scholar]
- 123.Yasojima K, McGeer EG, McGeer PL. Relationship between beta amyloid peptide generating molecules and neprilysin in Alzheimer disease and normal brain. Brain Res 919: 115–121, 2001. doi: 10.1016/S0006-8993(01)03008-6. [DOI] [PubMed] [Google Scholar]
- 124.Zhang T, Pan BS, Zhao B, Zhang LM, Huang YL, Sun FY. Exacerbation of poststroke dementia by type 2 diabetes is associated with synergistic increases of beta-secretase activation and beta-amyloid generation in rat brains. Neuroscience 161: 1045–1056, 2009. doi: 10.1016/j.neuroscience.2009.04.032. [DOI] [PubMed] [Google Scholar]
- 125.Zhao G, Liu HL, Zhang H, Tong XJ. Treadmill exercise enhances synaptic plasticity, but does not alter β-amyloid deposition in hippocampi of aged APP/PS1 transgenic mice. Neuroscience 298: 357–366, 2015. doi: 10.1016/j.neuroscience.2015.04.038. [DOI] [PubMed] [Google Scholar]