You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Physical Exercise Enhances Neuroplasticity and Delays Alzheimer’s Disease

Abstract

Accumulating evidence indicates that exercise can improve learning and memory as well as attenuate neurodegeneration, including Alzheimer’s disease (AD). In addition to improving neuroplasticity by altering the synaptic structure and function in various brain regions, exercise also modulates systems like angiogenesis and glial activation that are known to support neuroplasticity. Moreover, exercise helps to maintain a cerebral microenvironment that facilitates synaptic plasticity by enhancing the clearance of Aβ, one of the main culprits of AD pathogenesis. The purpose of this review is to highlight the positive impacts of exercise on promoting neuroplasticity. Possible mechanisms involved in exercise-modulated neuroplasticity are also discussed. Undoubtedly, more studies are needed to design an optimal personalized exercise protocol for enhancing brain function.

INTRODUCTION

In line with population aging, the number of people living with dementia worldwide is set to increase. According to the World Alzheimer Report 2016, there are about 47 million people worldwide living with dementia and this number may reach 74.7 million in 2030 and 131.5 million in 2050 (Alzheimer’s Disease International. World Alzheimer report 2016: Improving healthcare for people living with dementia. https://www.alz.co.uk/research/world-report-2016). Alzheimer’s disease (AD) is the most common form of dementia and possibly contributes to 60–70% of dementia cases (WHO and Alzheimer’s Disease International. Dementia: a public health priority. Geneva, Switzerland: World Health Organization, 2012). The pathological features of AD are characterized by extracellular depositions of amyloid plaques, which are primarily composed of 39–43 amino acids long Aβ peptides, and intracellular accumulations of neurofibrillary tangles, which are mainly composed by hyperphosphorylated tau protein [1]. Accumulations of amyloid plaques and neurofibrillary tangles in the brain have been hypothesized to cause deleterious responses in neuronal function in several brain regions related to cognition, such as the hippocampus and entorhinal cortex [1]. To date, there is no promising pharmacologic treatment (medication) that can halt AD. Therefore, nonpharmacological interventions that could improve or maintain cognitive function have become alternative options for the prevention and treatment of AD.

Epidemiological evidence has suggested that some lifestyle factors, such as regular exercise and cognitive activity, may delay age-related memory impairment and decrease risk of AD [2–4]. A meta-analysis examining the relationship between physical activity and the risk of neurodegenerative disease reported that engaging in physical activity reduces the risk of dementia and AD by 28% and 45%, respectively [5]. Higher levels of total daily physical exercise is associated with a lower risk of developing AD [6, 7]. Exercise can have a positive effect on multiple aspects of the brain, such as an increase in synaptic and cerebrovascular plasticity [8], a decrease in neuropathology [9], and an attenuation in neuroinflammation [10]. The wide variety of beneficial effects induced by exercise enhances overall brain health, which in turn helps to preserve neuronal function and protect against aging-associated loss of cognition. In this review, we summarize the recent advances in the beneficial effects of exercise on brain function and highlight some potential mechanisms.

BENEFICIAL EFFECTS OF EXERCISE ON BRAIN FUNCTION

Behavior level – learning and memory function

A growing number of studies support the idea that physical exercise increases brain function throughout life [11, 12]. In a meta-analysis that included a total of 59 studies (from 1947 to 2009) examining the relationship between physical activity and academic achievement in school-age children, the authors demonstrated significant and positive correlations between physical activity and cognitive outcomes [13]. In another meta-analytic review of 29 randomized controlled trials examining the association between aerobic exercise and neurocognitive performance in a group with a mean age of≥18 years of age, positive association between exercise with attention, processing speed, and executive and memory function was also evident [14]. Particularly, exercise enhances pattern separation in humans, which is defined as a process to remove redundancy from similar input patterns so that events can be separated from each other and interference can be minimized [15]. An acute bout of moderate intensity aerobic exercise improves the pattern separation in young adults [16]. This mnemonic discrimination ability is highly correlated with aerobic fitness. The higher fitness group (i.e., high endurance capacity during exercise) had better performance in the pattern separation test compared with the lower fitness group [17]. The memory-enhancing effect of exercise on young adults has also been demonstrated in a series of hippocampal-associated learning and memory tests designed for rodents. Almost all young animal studies confirmed the memory-facilitating effect (i.e., acquisition and retention) of exercise. These tests included novel object recognition [18, 19], object displacement [19], Morris water maze [18, 20–22], radial arm water maze [23], Y-maze [24], passive avoidance [25, 26] and contextual fear conditioning [27–29].

In addition to benefiting healthy young adults, physical exercise is known to delay age-related cognitive decline. A randomized, controlled trial study that evaluated the association between exercise and cognitive function in 120 healthy participants aged over 65 showed that 6 months of exercise reversed age-related loss in hippocampus volume and improved performance in a computerized spatial memory task [30]. A meta-analysis including 42 studies (from 1966 and 2010) of cognitive interventions of exercise in 3,781 healthy older adults aged 55 and older concluded that aerobic fitness training improves cognitive performance [31]. Similar findings have also been obtained in aged animals. For example, it has been shown that 6-weeks of treadmill exercise ameliorates age-induced losses in short-term (tested by step-down avoidance task) and long-term memory (tested by radial 8-arm maze) functions in 24-month-old rats [32]. Another study demonstrated that one week of mild treadmill exercise was enough to improve spatial learning and memory ability, which was tested by the Morris water maze in 23-month-old rats [33]. These results suggest that exercise not only enhances learning and memory in young adults but also protects aged individuals from age-related cognitive impairment.

Although the beneficial effects of exercise on learning and memory have been well-documented in numerous studies, some studies of rodents failed to find such an association. This discrepancy may be due to the different exercise intensities and durations employed in each study. For example, high levels of running exercise are known to impair hippocampus-dependent spatial memory [34, 35] and other types of memory [36]. It has been speculated that the exercise intensity affects cognitive performance in a reversed U-shaped fashion. In other words, cognition improvement is most effective at moderate-intensity exercise, whereas exercise at low-intensity is less effective and at high-intensity would induce high levels of stress responses, hence impairing cognitive performance [37, 38].

Behavior level – mental function

The beneficial effects of exercise extend beyond cognitive function. A plethora of evidence supports the notion that exercise can prevent or delay the onset of various mental disorders such as anxiety, depression, and posttraumatic stress disorder [39, 40]. Several meta-analysis studies suggest that exercise could reduce depressive and anxiolytic symptoms in adolescents with clinical levels of mental illness [41, 42]. The effects of exercise on mental health are dose-dependent. In adults, a moderate amount of exercise exerts greater benefits in the mental health than low or high doses of exercise [43].

The positive effects of exercise on mood disorders have been examined in various animal models. It has been shown that a 2-week period of treadmill exercise effectively decreased anxiety-like (tested by elevated plus maze) and depressive-like (tested by sucrose preference test) behaviors in a single-prolonged stress-induced post-traumatic stress disorder rat model [44]. Besides stress, mood disorders commonly occur secondary to medical conditions such as obesity and stroke. In high-fat diet induced obesity, and middle cerebral artery occlusion-induced stroke animal models, exercise was also capable of attenuating depression- and anxiety-like behaviors in the modeled animals [45, 46].

Behavior level – motor function

Exercise has a profound effect on motor function. It has been demonstrated that a single session of exercise not only significantly improves performance in a motor learning task during the training session [47] but also leads to longer retention of said motor skills than the control group. This is especially clear when motor memory is assessed one day after practice [48]. In the animal studies, both wheel and treadmill exercise enhanced animals’ motor performance in foot fault-placing and parallel bar-crossing, as well as the staircase reaching task [49], rota-rod test [50], beam walking and cylinder tests [51], and ladder-climbing tests [52], after ischemic or hemorrhagic stroke. Furthermore, wheel-running and treadmill exercise can also rescue motor behaviors (e.g., rotarod performance test, rotational behavior test, ladder rung walking task, and gait parameters) that are impaired by different dopamine-depletion methods such as LPS, 6-OHDA and MPTP [53–55].

Cellular level – neurons

Neuroplasticity is a continuous process that modifies existing neural networks by mediating structural and functional adaptations of synapses in response to changes in behavior. Exercise-induced structural and functional changes in the brain have been reported in both human and animal studies.

Structural changes – macroscopic levels

Using a resting MRI to evaluate human brain structure, it has been shown that aerobic exercise, from several months to a year, increased brain volume in various brain regions, such as the prefrontal and temporal cortex, [56] as well as the hippocampus [30]. Compared to healthy adults with a sedentary lifestyle, higher gray and white matter cluster concentrations (Voxel-based analysis) in the subgyrus, cuneus, and precuneus regions are found in athletes of similar ages [57]. A functional MRI study suggested that aerobic exercise at least enhanced activity in the brain areas which are involved in attentional control tasks [58]. Moreover, twelve months of chronic exercise enhanced functional connectivity in the default-mode network and the frontal executive network [59]. The increase of brain regional volume and activity may reflect an alteration in the number of neurons, synapses, and axonal and dendritic arbors.

Animal studies also support that exercise induces neuronal structural alteration in various brain regions, including the dentate gyrus (DG) and the cornu ammonis (CA) areas of the hippocampus, amygdala, cortex, striatum, and cerebellum [28, 60–63].

Structural changes – microscopic levels

The effect of exercise on neuronal morphology has also been studied in multiple brain regions. These exercise-induced neuroadaptations will be discussed separately.

In the hippocampal DG and CA regions, exercise significantly enhances dendritic length and complexity as well as increases the dendritic spine density of neurons [60, 64–67]. Schaefers et al. has scored the presynaptic turnover rates by quantifying lysosomal accumulations in degrading axon terminals [68]. Their study indicates that four days of wheel-running in adulthood leads to a significant difference in the level of synaptic turnover in the stratum lacunosum-moleculare layer of the CA1 region, suggesting the effects of exercise on pre-synaptic remodeling [68]. At the post-synaptic location, exercise increased the dendritic arborization and spine density in both the CA1 and CA3 regions. The dendritic branches in the apical dendrites and the numbers of dendritic spines on both the apical and basal dendrites of CA1 pyramidal neurons also increased after treadmill and wheel-running exercise [28, 65]. In addition to normal physiological conditions, exercise can also restore impaired neuronal structure in CA regions after brain injury. For example, in the middle cerebral artery occlusion-induced ischemic rats model, the neurons in CA1 and CA3 exhibited the longer dendritic length and higher arborization after two weeks of treadmill exercise as compared to the sedentary group [69]. In the neonatal hypoxia-ischemia rat model, physical activity improved the spine density in the CA1 neurons [70].

The effect of exercise on the structure of amygdalar neurons has also been studied in rats [28] and mice [71]. By using the single neuron labeling technique, the dendritic arborization and spine density of neurons in the basolateral amygdala increased after one month of treadmill exercise [28, 71]. However, it is interesting to note that different forms of exercise induced distinct brain region-dependent neuronal adaptations. For example, 1-month treadmill running exercise, but not running wheel exercise, increased dendrite complexity and spine densities of neurons in the basolateral amygdala [28]. The differential effects between treadmill and wheel exercise may be derived from the difference in exercise intensity as skeletal muscle citrate synthase activity is only elevated in the rats trained by treadmill exercise, but not by running wheel exercise [28]. These results suggest that the influences of exercise on amygdalar neurons may depend on exercise intensity.

The cytoarchitecture of cortical neurons is also affected by exercise. It has been shown that the nitrergic neurons in the cerebral cortex exhibit larger size and extended dendritic arborization after 16-months of moderate exercise, suggesting that nitric oxide may play a role in synaptic plasticity related to exercise [61]. Higher density of dendritic spines in the medial prefrontal cortex pyramidal neurons in rats could be identified after 2-weeks of running wheel exercise when compared to rats that didn’t do the wheel exercise [62]. These results suggest that medial prefrontal cortex pyramidal neurons are much more sensitive to exercise, as such, intensity of wheel-running does not significantly affect amygdalar neurons [28]. By utilizing transcranial two-photon in vivo microscopy, Chen et al. investigated the turnover of dendritic spines in the barrel cortex of mice receiving exercise [72]. They demonstrated that exercise decreases the elimination but does not improve the formation of dendritic spines in the cortical neurons. The net outcome of exercise increased the percentage of mushroom-like spines and decreased the numbers of thin and filopodia spines in the barrel cortex, which represent the facilitation of maturation of dendritic spines by exercise [72].

The cerebellum and striatum are the primary structures of a distributed system for the control of motor functions [73]. Exercise has been shown to increase the density of dendrites and dendritic spines in the cerebellar Purkinje cells of animals [63, 74, 75]. Among the different types of dendritic spines, the stubby, mushroom, and wild spines are more abundant in the cerebellar Purkinje cells in exercise animals than sedentary rats [75]. The changes in morphology of dendritic spines could be related to the regulation of excitability in Purkinje cells due to motor activity [75]. Furthermore, exercise selectively increases the thickness of the molecular layer in the cerebellum [74], implying morphological adaptations in the basket and stellate cells.

Unlike other brain regions, the effects of exercise on neuronal morphology of the striatum is frequently investigated after local neurons undergo pathological challenges. No apparent morphological changes in the striatal neurons are evident in healthy animals after 1-month to 1.5 years of chronic exercise [61, 76–79]. However, in MPTP- and 6-OHDA-treated Parkinson’s disease animal models and in the collagenase-induced intracerebral hemorrhage model, the number of tyrosine hydroxylase-immunostaining positive fibers and dendritic spine density in the striatum are higher in the exercise group than the non-exercise group [77–79]. By using Drd2-eGFP-BAC transgenic (Tg) mice in conjunction with biocytin injection methods, Toy et al. showed that chronic exercise increased dendritic spine densities in the striatal medium spiny neurons in both DA-D1R-direct and DA-D2R-indirect pathways of MPTP-treated animals [77].

Functional changes

The structural changes in neurons (e.g., dendritic complexity, spine density, and newborn neuron maturation in the adult hippocampus) after exercise are highly associated with functional alterations in neurons (e.g., synaptic activity). Long-term potentiation (LTP) is a form of synaptic plasticity characterized by a prolonged increase in synaptic efficiency based on application of a patterned stimulus. Numerous studies have shown that exercise efficiently increases the LTP amplitude in the DG region [18, 21, 80–83]. It has been suggested that exercise alters the expression of LTP in the DG by reducing the threshold of LTP induction. The weak theta burst stimulation, while not reliably inducing a significant amount of LTP in the DG of sedentary animals, can trigger robust short-term potentiation and long-lasting LTP in running animals [80]. Moreover, the effectiveness of exercise on the enhancement of LTP is of a time-dependent manner (number of running days). Patten et al. examined the expression of LTP in animals that received different periods of exercise and found that LTP expression gradually increased in the DG during days 7–28 of the exercise period and reached a significant level after 56 days of running [83].

Exercise-induced improvement of LTP has also been demonstrated in other hippocampal regions. Both treadmill and wheel-running enhance the expression of a single burst of 100-Hz-induced LTP in the Schaffer collateral pathway of CA1 area [84, 85]. In addition, this exercise promotes the occurrence of a tetraethylammonium-induced potassium ion channel blockade, which in turn leads to the expression of LTP in the mossy fiber of the CA3 area [86]. A blockade of NMDA receptors, NR2A or NR2B, inhibit the effects of exercise on LTP enhancement [80, 81], suggesting that the facilitation of LTP by exercise critically relies on the functions of the NMDA and glutamate receptors [87].

Cellular level – glial cells

In parallel with the effects of exercise on neurons, exercise is also known to have multiple effects on glial cells. Astrocytes, the major glial cells in the brain, play a vital role in the regulation of brain energy transmissions from the vasculatures to the neurons [88, 89]. Because proper and efficient astrocyte function is essential for supporting neuronal function [88], it has been suggested that exercise-induced enhancement of neuronal function may occur via the adaptation of astrocyte behavior. Exercise induces widespread plasticity in astrocytes [90]. It has been shown that one month of treadmill exercise increases GFAP-labeled cell numbers and the improvement of astrocyte plasticity in the CA1 area [91]. Furthermore, immunostaining signals of S100β and aquaporin-4, two astrocyte-specific markers, in various brain regions related with cognitive function, such as the hippocampus, medial prefrontal cortex, and orbitofrontal cortex, increased after 12 days of wheel-running in rats [90]. In mice, 4 weeks of physical exercise (a combination of treadmill and running wheel) increased the average length and the area enclosed by each astrocytic projection in the hippocampus [92]. The exercise also changed the orientation of astrocytic projections towards DG in the hippocampus, the region with significant increase in neurogenesis following the exercise [92].

The structure and function of microglia, the major immune cells in the central nervous system, are also affected by exercise. Exercise has been reported to decrease the aging-induced activation and proliferation of microglia in the hippocampus [93, 94]. In parallel to the in vivo findings, microglia isolated from aged rodents that participated in chronic wheel-running exercise have a lower basal level of IL-1β and TNF-α when compared to the microglia isolated from their sedentary littermates [95–97]. Similarly, when stimulating these microglia with LPS, the production of IL-1β was also lower in the chronic wheel-running group. In addition to the inflammatory cytokines, neuron-microglia contact signaling, an important regulator of microglial activation, is affected by exercise. For example, CX3CL1 and CD200 are immunomodulatory factors expressed by neurons to inhibit microglial activation through binding with the CX3CL1 and CD200 receptor on microglia, respectively [98]. Chronic exercise is known to upregulate the expression of the CX3CL1 gene [99] and the CD200/CD200R proteins in the brain [100], supporting that exercise may inhibit microglial activation via the enhancement of neuron-microglia contact signaling.

Molecular level – brain-derived neurotrophic factor (BDNF)

The mechanisms underlying exercise-induced neuroplasticity involve, at least in part, several neurotrophic and growth factors. Among them, BDNF is a well-characterized mediator of neuronal growth [101], plasticity [102, 103], and survival [99]. Human and animal studies collectively suggest that exercise is an active strategy to upregulate the expression of BDNF, which plays an essential role in exercise-induced neuroplasticity [104, 105]. Different types of exercise (i.e., voluntary wheel-running and mandatory treadmill running) all seem to be capable of enhancing the production of BDNF in the hippocampus of young and aged animals [22, 28, 106–108]. It is believed that BDNF binds to its receptor, tropomyosin receptor kinase B (TrkB), which increases the phosphorylation levels of cAMP response-element-binding (CREB) and the translation of synaptic-plasticity related proteins [22, 28, 103, 109, 110], and finally enhances neuroplasticity. It has been demonstrated that animals with higher levels of BDNF and TrkB receptor in the hippocampus exhibit better performance in radial water mazes [23] and passive avoidance tasks [26]. On the contrary, blocking the TrkB receptor with a TrkB receptor-IgG chimera or antagonist inhibits the efficacy of exercise-induced upregulation of synaptic plasticity-related proteins significantly [103, 109], resulting in a disruption of benefits on cognitive function. Behavior tests in the Morris water maze, passive avoidance, and contextual fear condition also suggest that the learning and memory abilities enhanced by exercise are reduced to sedentary levels after treating the animals with TrkB receptor blockers [26, 28, 111].

Molecular level – insulin-like growth factor 1 (IGF-1)

IGF-1 is also a key modulator of neuronal functions in the central nervous system, including synaptic plasticity, synapse density, neurotransmission, neurogenesis and neuron differentiation [112–114]. Chronic exercise-enhanced adult hippocampal neurogenesis, learning, and memory performance have been attributed to IGF-1 signaling in the hippocampus [115–117]. The levels of IGF-1 in the blood positively correlated with the time animals spent in the target quadrant during the probe trial of the Morris water maze [115]. The levels of IGF-1 in the brain are also increased after exercise, which is a result of uptake in circulating IGF-1 [118]. Infusion of anti-IGF-I antiserum or neutralization of hippocampal IGF-1 receptor inhibits the exercise-induced brain uptake of circulating IGF-I, disrupts neurogenesis, and lessens the effects of exercise on memory retention [116, 117, 119]. Exercise-induced upregulation of BDNF may be partially affected by the IGF-1 pathway. Blockade of the IGF-I receptor during exercise inhibits the ability of exercise to enhance the expressions of pro-BDNF and BDNF in the hippocampus [119].

Molecular level – vascular endothelial growth factor (VEGF)

VEGF, an angiogenic protein, is known to have neuroprotective and neurotrophic functions [120]. VEGF can be synthesized and released by peripheral vascular endothelial cells and brain cells, including astrocytes, ependymal cells, and neuronal stem cells [121]. Intracerebroventricular injection of VEGF or gene transfer of VEGF in the hippocampus increases the number of BrdU labeling cells in the subventricular zone and the subgranular zone of the DG, suggesting an intimate association between angiogenesis and neurogenesis [122, 123]. The changes in the vascular niche promotes local signaling that directly regulates or indirectly activates other regulatory factors to stimulate the proliferation, differentiation, and survival of newborn neurons [124]. Voluntary exercise is known to increase the expressions of VEGF receptors, fms-like tyrosine kinase (Flt-1/VEGF1), and fetal liver kinase (Flk-1 or VEGFR2) in the hippocampus [125]. The activation of the intracellular tyrosine kinase domains of the VEGF receptors induces the activation of several downstream signaling pathways which in turn enhances the proliferation of neuron precursors [126, 127]. Peripheral VEGF seems to play an essential role in exercise-induced adult hippocampal neurogenesis [128, 129]. Blockade of VEGF signaling by intravenous injection of adenoviruses carrying the chimeric VEGFR1 receptor or conditionally knocking down skeletal myofiber-specific VEGF gene nullifies running-induced adult hippocampal neurogenesis [128, 129].

Molecular level – nerve growth factor (NGF)

NGF also plays a role in promoting neuronal function, especially the survival of neural progenitors [130]. Results gathered from microarray and nuclease protection assays indicate that the expression of hippocampal NGF is increased after wheel- running, with a peak at 2 to 3 days after exercise [87, 131]. The expressions of hippocampal NGF and one of its receptors, tropomyosin receptor kinase A (TrkA), are increased after 8-weeks of treadmill running in rodents. Similar to BDNF/TrkB signaling, the binding of NGF to TrkA stimulates the downstream transcription factor, CREB, and induces various gene transcriptions related to cell survival and neuroplasticity [132].

PHYSICAL ACTIVITY, AGING, AND AD

Physical activity and aging

Aging is an irreversible and inescapable process. Age alone increases the risk of dementia and AD [133]. Even in the absence of overt disease symptom, increasing age is associated with decreasing cognitive function of varying severity in human beings. The hippocampus is considerably vulnerable to aging and is involved in the development of aging-related deficits of neuroplasticity and memory functions [134]. Aging-associated memory deficits are accompanied by abnormal structural and functional changes in the hippocampus. Aging also reduces hippocampus size, induces hippocampal neuron loss [135, 136], suppresses dendritic arbors and spine density of the hippocampal neurons [137–139], decreases the degrees of DG neurogenesis [140, 141], and represses the field excitatory postsynaptic potentials [142, 143] and LTP induction [144] in the hippocampus.

Several lines of evidence indicate that exercise can ameliorate the structural and functional changes in the brain during aging. In the pioneer study, Samorajski et al. showed that exercise (spontaneously wheel-running) significantly increased recent memory in the passive avoidance test in adults (10–14 month), middle-aged (20–24 month), and old (28–30 month) C57BL/6J male mice [145]. Later studies also demonstrated that physical activity increases the cognitive reserve and prevents aging-related memory decline [108, 146, 147]. Regular aerobic exercise increases the hippocampal volume by 2%, effectively countering age-related loss in volume in older adults (55–80 years old) without dementia [30]. Six-weeks of moderate treadmill training reverses the age-related declines in the complexity of dendritic arbors and the density of the dendritic spines of hippocampal CA1 pyramidal neurons in mice [139]. Furthermore, six-weeks of moderate-intensity running enhanced the LTP induced by high-frequency stimulation in the CA1 regions and hippocampal memory in mice of different ages, even when the memory impairment had progressed to an advanced stage [139]. It is widely accepted that exercise enhances the adult hippocampal neurogenesis that is critical for hippocampal memory functions [21, 148–150]. The decline in neurogenesis in aged (19 months of age) mice is reversed to 50% of young (3 months of age) control levels by 45-days of voluntary wheel-running [141]. Yang et al. demonstrated that the hippocampal neurogenesis dramatically decreased by the time mice reached nine months of age and 5-weeks of treadmill running attenuated the decrease of the number of neural stem/progenitor cells during aging and enhanced the maturation of newborn neurons [66].

Physical activity and AD – neuroprotection

It is well known that neurotrophic and growth factors (BDNF, IGF-1, VEGF, NGF) are attenuated in AD [151–154]. Therefore, one of the potential mechanisms for exercise to protect the brain against AD may be via the upregulations of neurotrophic and growth factors. In the following section, we will discuss exercise-induced neuroprotection against AD with focuses on the four trophic factors.

Both clinical and basic research documents that BDNF mRNA and proteins, including proBDNF, are severely decreased in AD, especially in the hippocampus [155–157]. Several lines of evidence have suggested that decreases in brain BDNF and NGF levels contribute to AD pathology [158, 159]. On the contrary, administration of lentiviral vectors to constitutively express BDNF in the hippocampus can prevent cell death, reverse neuronal atrophy, and ameliorate behavioral deficits, hence delaying the development of AD [160]. As a physiological approach to increase the production of neurotrophic factors, exercise has been reported to alleviate hippocampal functional impairment in AD through the induction of BDNF expression in the hippocampus [71, 105]. Exercise-induced BDNF signaling activation has been applied to a variety of AD models, including the Aβ injection-induced non-Tg AD model [161], APP/PS1 [71, 162], APOE4 [163], NSE/APPsw [164] and NSE/PS2 m [165, 166] Tg animal models. Increased BDNF signaling due to exercise is suggested to protect against AD-induced learning and memory deficits through enhancing neurogenesis [167] and LTP expression [161, 162], modulating dendritic morphology [71], and reversing Aβ-induced neurotoxicity [168].

Neurotrophic signaling not only directly enhances neuronal function but also decreases AD pathological burden. An in-vitro study suggests that APP processing shifts towards the non-amyloidogenic α-secretase pathway in response to BDNF treatment [169], which is known to reduce the production of Aβ1 - 40 and Aβ1 - 42. Beyond amyloid pathology, BDNF also affects the tau pathology through mediating tau phosphorylation. In P19 neurons, BDNF stimulation induces a rapid decrease in tau phosphorylation [170]. The effects of BDNF on tau dephosphorylation is known to be TrkB dependent. TrkB activation induces AKT-dependent phosphorylation of GSK-3β at the Ser9 site, resulting in the inhibition of tau phosphorylation [171]. The K252a treatment, a Trk receptor inhibitor, attenuates the effect of BDNF stimulation on tau dephosphorylation [170].

The reduction of the cerebrospinal fluid/plasma IGF-I ratio has been found in AD patients and AD model mice [172, 173], reflecting an impaired uptake of IGF-I from the serum to the CSF. Exercise is known to increase the uptake of IGF-1 in the central nervous system [118]. Elevated IGF-1 not only benefits neuronal functions but also influences the production of Aβ and the formation of neurofibrillary tangles [174]. IGF-1 also affects brain amyloidosis [175, 176]. Crossing APPswe/PS1dE9 Tg mice with serum IGF-I deficient LID mice accelerates brain amyloidosis significantly [175]. Conversely, increases of serum IGF-1 levels decrease the Aβ load in the brain of aged APP/PS2 Tg mice [176]. IGF-1-induced inhibition of amyloidosis could be due to, at least in part, an increase of Aβ clearance. This is because IGF-1 increases the levels of Aβ carrying proteins, such as apolipoprotein J, transthyretin, and albumin, which are all known to facilitate Aβ clearance via blood-brain barrier (BBB) transportation [176, 177]. Antagonization of IGF-1 by TNF-α reduces IGF-I signaling at the BBB, resulting in a decrease of the expression of Aβ-carrying proteins and an increase of Aβ levels in the brain [177]. Furthermore, in vitro cell culture studies (i.e., NT2N cells and rat primary cortical neurons) reveal that IGF-1 induces the phosphorylation of GSK3α (ser21)/β (ser9) and attenuates tau phosphorylation through activating IRS1/PI3K/AKT/mTOR pathway [178, 179]. It has been reported that exercise increases the activity in the IRS/PI3/AKT pathway to dephosphorylate GSK-3α/β [180] and inhibit tau phosphorylation in APP/PS1 [181] and NSE/htau23 Tg mice [182] as well as in the ICV-STZ non-Tg AD model [183]. This supports the assertion that exercise-induced IGF-1 signaling plays an important role in preventing pathologies associated with AD.

Exercise elevates VEGF transcription, mRNA, and protein in the brain [184]. In addition to supporting neurogenesis, VEGF could protect neurons against AD via the following potential mechanisms. Firstly, VEGF increases angiogenesis to help the maintenance of BBB integrity [185, 186], which is essential in preventing the entry of systemic Aβ [187]. Secondly, VEGF regulates the expression of Aβ transporters on the BBB [188]. It has been shown that an implant of VEGF-secreting fibroblast microcapsules into the brain of APP/PS1 mice not only enhances brain vessel density but also increases the level of LRP-1 expression, which is an Aβ efflux transporter on the BBB, accompanied by a reduction of Aβ deposition in the brain [189]. Thirdly, VEGF directly affects APP processing by decreasing β-secretase activity [190, 191]. Applying VEGF to Tg2576 mouse brain slices decreases β-secretase activity and production of both soluble Aβ1 - 40 and Aβ1 - 42 [190]. Finally, VEGF directly binds to Aβ and eliminates Aβ toxicity. VEGF contains a heparin-binding domain that can be recognized by Aβ [192]; therefore, it is capable of binding to Aβ (at around the 26–35 amino acid region), hence inhibiting Aβ aggregation and Aβ neurotoxicity [192].

Decreases in mature NGF expression as well as increases in Pro-NGF expression have been found in postmortem AD brains [152, 193, 194], suggesting that diminished conversion of pro-NGF to mature NGF and increased degradation in the activity of mature NGF in AD brains [152]. It is known that proNGF preferentially binds to p75NTR and initiates the activation of apoptotic pathways, which may contribute to AD neurodegeneration [195, 196]. Increased mature NGF due to exercise could protect neurons against AD pathologies through acting on the trkA receptor to promote neurite generation, neuronal survival, and synapse formation [197, 198]. It has been reported that exercise-induced increases of mature NGF expression and decreases of Bax expression, the downstream molecule of p75NTR, are involved in the inhibition of neuronal death during AD development [165]. An in-vivo study of Tg mice expressing a neutralizing antibody directed against NGF showed a progressive development of amyloid deposition and neurofibrillary pathology in the brain [199, 200]. An in-vitro study using PC12 differentiated cells revealed that withdrawal of NGF caused the overproduction of Aβ peptides, increasing tau phosphorylation and neurite degeneration [201, 202]. Similar to BDNF, mature NGF increases the metabolism of APP from amyloidogenic towards non-amyloidogenic processing through binding to the TrkA receptor [203, 204]. Mature NGF is also able to decrease the hyperphosphorylation of tau via increasing the phosphorylation of GSK3 β at the Ser 9 site [204].

Physical activity and AD – glia cells regulations

Accumulated evidence suggests a strong association between risk factors of cerebrovascular disorders, such as hypertension, diabetes, hypercholesterolaemia, hyperhomocysteinaemia, and APOE4, with AD [205]. Astrocytes play an important role in maintaining neurovascular function [205]. The prodromal stage of AD has been linked to dysregulated astrocytes in the neurovascular unit, which may then cause disruptions of neuronal metabolic support, impairment of synaptic functions, onset of neuronal death, and decreases of degradation and clearance of Aβ [206–208]. One of the exercise-induced neuroprotective pathways is mediated by astrocytes. For example, S100B, a calcium-binding protein, is predominantly secreted by astrocytes and acts as a neurotrophic factor to support neuronal survival. In the sporadic AD animal model, five weeks of treadmill exercise could reverse the reduced extracellular levels of S100B [209], suggesting that exercise can alleviate neuronal dysfunction via controlling astrocyte signaling. Furthermore, exercise increases the Aβ clearance abilities of astrocytes in the AD brain. The recently identified glymphatic pathway, which critically relies on AQP4 in astrocytes, contributes to Aβ clearance. Mice lacking the AQP4 in astrocytes exhibit slower paravascular cerebrospinal fluid-interstitial fluid exchange and lower Aβ1-40 clearance rate than those of intact mice [210, 211]. Chronic exercise can increase Aβ clearance through the glymphatic pathway [212]. Two-photon imaging reveals that six weeks of wheel-running increases AQP4 expression in the perivascular region and increases the rate of paravascular cerebrospinal fluid-interstitial fluid exchange in the brain of aged mice; this is accompanied by a decrease of Aβ accumulation in the cortex and hippocampus [212].

In addition to amyloid plaque and neurofibrillary tangle deposition, neuroinflammation is considered a key feature of AD pathology. AD inflammation is characterized by the presence of reactive astrocytes and activated microglia surrounding amyloid plaques. Chronic exercise has been reported to decrease neuroinflammation in AD. In the Tg animal models, wheel-running and treadmill exercise decreased the levels of hippocampal pro-inflammatory cytokines (i.e., IL-1β and TNF-α) of Tg2576 AD mice and NSE/htau23 Tg mice to levels indistinguishable from wild-type mice [213, 214].

Physical activity and AD – Aβ transportation and degradation

LRP-1 and RAGE are major Aβ carrier proteins that regulate Aβ efflux and influx, respectively, in transportation across the BBB [215, 216]. The expression of LRP-1 and RAGE can be detected in both neurons and glial cells [215, 216]. It has been demonstrated that exercise increases LRP-1 and decreases RAGE expression in the hippocampus of Tg2576 [217] and APP/PS1 [71, 181] mice, suggesting enhanced brain-to-blood Aβ transportation after exercise. Also, exercise can increase the activity of Aβ degradation enzymes that prevent the aggregation of Aβ. Tg2576 mice given a 12-week period of exercise significantly increased hippocampal levels of neprilysin, insulin-degrading enzyme, and matrix metallopeptidase 9; all of which are Aβ proteolytic enzymes [217]. Interestingly, exercise-induced changes in the expression levels of Aβ transport proteins and proteolytic degrading enzymes depend on the intensity of exercise training [217].

CONCLUSION

Although much progress has been made in AD studies, effective pharmacological treatments remain evasive. Alternatively, non-pharmacological strategies to delay AD have become a major health issue. There is no doubt that brain health can be improved through physical exercise. Exercise benefits neuroplasticity in health and disease stages by targeting different aspects of brain function. Firstly, increases of trophic factors exert net effects on enhancing neuroplasticity, and cognitive and behavioral function. Secondly, exercise synchronously changes cerebrovascular function and glial cells to support enhanced neuroplasticity. Finally, lowering of toxic Aβ and tau by exercise decreases neuronal vulnerability, which may help the maintenance of synaptic function. Overall, exercise enhances neuronal plasticity (brain reservoir) and could be a strategy to delay the onset of AD.

CONFLICT OF INTEREST

The authors declare that there is no conflict of interest.

ACKNOWLEDGMENTS

This study was supported by grants 107-2320-B-006 -054 -MY3 and 107-2514-S-006-006 from the Taiwan Ministry of Science and Technology.

REFERENCES

[1] 

Spires-Jones TL , Hyman BT . The intersection of amyloid beta and tau at synapses in Alzheimer’s disease. Neuron. (2014) ;82: (4):756–71.

[2] 

Lindsay J , Laurin D , Verreault R , Hebert R , Helliwell B , Hill GB , et al. Risk factors for Alzheimer’s disease: A prospective analysis from the Canadian Study of Health and Aging. Am J Epidemiol. (2002) ;156: (5):445–53.

[3] 

Barnes DE , Yaffe K , Satariano WA , Tager IB . A longitudinal study of cardiorespiratory fitness and cognitive function in healthy older adults. J Am Geriatr Soc. (2003) ;51: (4):459–65.

[4] 

Larson EB , Wang L , Bowen JD , McCormick WC , Teri L , Crane P , et al. Exercise is associated with reduced risk for incident dementia among persons 65 years of age and older. Ann Intern Med. (2006) ;144: (2):73–81.

[5] 

Hamer M , Chida Y . Physical activity and risk of neurodegenerative disease: A systematic review of prospective evidence. Psychol Med. (2009) ;39: (1):3–11.

[6] 

Buchman AS , Boyle PA , Yu L , Shah RC , Wilson RS , Bennett DA . Total daily physical activity and the risk of AD and cognitive decline in older adults. Neurology. (2012) ;78: (17):1323–9.

[7] 

Lautenschlager NT , Cox KL , Flicker L , Foster JK , van Bockxmeer FM , Xiao J , et al. Effect of physical activity on cognitive function in older adults at risk for Alzheimer disease: A randomized trial. JAMA. (2008) ;300: (9):1027–37.

[8] 

Lange-Asschenfeldt C , Kojda G . Alzheimer’s disease, cerebrovascular dysfunction and the benefits of exercise: From vessels to neurons. Exp Gerontol. (2008) ;43: (6):499–504.

[9] 

Liang KY , Mintun MA , Fagan AM , Goate AM , Bugg JM , Holtzman DM , et al. Exercise and Alzheimer’s disease biomarkers in cognitively normal older adults. Ann Neurol. (2010) ;68: (3):311–8.

[10] 

Spielman LJ , Little JP , Klegeris A . Physical activity and exercise attenuate neuroinflammation in neurological diseases. Brain Res Bull. (2016) ;125: :19–29.

[11] 

Hillman CH , Erickson KI , Kramer AF . Be smart, exercise your heart: Exercise effects on brain and cognition. Nat Rev Neurosci. (2008) ;9: (1):58–65.

[12] 

Verburgh L , Konigs M , Scherder EJ , Oosterlaan J . Physical exercise and executive functions in preadolescent children, adolescents and young adults: A meta-analysis. Br J Sports Med. (2014) ;48: (12):973–9.

[13] 

Fedewa AL , Ahn S . The effects of physical activity and physical fitness on children’s achievement and cognitive outcomes: A meta-analysis. Res Q Exerc Sport. (2011) ;82: (3):521–35.

[14] 

Smith PJ , Blumenthal JA , Hoffman BM , Cooper H , Strauman TA , Welsh-Bohmer K , et al. Aerobic exercise and neurocognitive performance: A meta-analytic review of randomized controlled trials. Psychosom Med. (2010) ;72: (3):239–52.

[15] 

Kesner RP . Role of the hippocampus in mediating interference as measured by pattern separation processes. Behav Processes. (2013) ;93: : 148–54.

[16] 

Suwabe K , Hyodo K , Byun K , Ochi G , Yassa MA , Soya H . Acute moderate exercise improves mnemonic discrimination in young adults. Hippocampus. (2017) ;27: (3):229–34.

[17] 

Suwabe K , Hyodo K , Byun K , Ochi G , Fukuie T , Shimizu T , et al. Aerobic fitness associates with mnemonic discrimination as a mediator of physical activity effects: Evidence for memory flexibility in young adults. Sci Rep. (2017) ;7: (1):5140.

[18] 

O’Callaghan RM , Ohle R , Kelly AM . The effects of forced exercise on hippocampal plasticity in the rat: A comparison of LTP, spatial- and non-spatial learning. Behav Brain Res. (2007) ;176: (2):362–6.

[19] 

Griffin EW , Bechara RG , Birch AM , Kelly AM . Exercise enhances hippocampal-dependent learning in the rat: Evidence for a BDNF-related mechanism. Hippocampus. (2009) ;19: (10):973–80.

[20] 

Ang ET , Dawe GS , Wong PT , Moochhala S , Ng YK . Alterations in spatial learning and memory after forced exercise. Brain Res. (2006) ;1113: (1):186–93.

[21] 

van Praag H , Christie BR , Sejnowski TJ , Gage FH . Running enhances neurogenesis, learning, and long-term potentiation in mice. Proc Natl Acad Sci U S A. (1999) ;96: (23):13427–31.

[22] 

Liu YF , Chen HI , Wu CL , Kuo YM , Yu L , Huang AM , et al. Differential effects of treadmill running and wheel running on spatial or aversive learning and memory: Roles of amygdalar brain-derived neurotrophic factor and synaptotagmin I. The Journal of physiology. (2009) ;587: (Pt 13):3221–31.

[23] 

Berchtold NC , Castello N , Cotman CW . Exercise and time-dependent benefits to learning and memory. Neuroscience. (2010) ;167: (3):588–97.

[24] 

Van der Borght K , Havekes R , Bos T , Eggen BJ , Van der Zee EA . Exercise improves memory acquisition and retrieval in the Y-maze task: Relationship with hippocampal neurogenesis. Behav Neurosci. (2007) ;121: (2):324–34.

[25] 

Chen HI , Lin LC , Yu L , Liu YF , Kuo YM , Huang AM , et al. Treadmill exercise enhances passive avoidance learning in rats: The role of down-regulated serotonin system in the limbic system. Neurobiol Learn Mem. (2008) ;89: (4):489–96.

[26] 

Liu YF , Chen HI , Yu L , Kuo YM , Wu FS , Chuang JI , et al. Upregulation of hippocampal TrkB and synaptotagmin is involved in treadmill exercise-enhanced aversive memory in mice. Neurobiol Learn Mem. (2008) ;90: (1):81–9.

[27] 

Baruch DE , Swain RA , Helmstetter FJ . Effects of exercise on Pavlovian fear conditioning. Behav Neurosci. (2004) ;118: (5):1123–7.

[28] 

Lin TW , Chen SJ , Huang TY , Chang CY , Chuang JI , Wu FS , et al. Different types of exercise induce differential effects on neuronal adaptations and memory performance. Neurobiol Learn Mem. (2012) ;97: (1):140–7.

[29] 

Greenwood BN , Strong PV , Foley TE , Fleshner M . A behavioral analysis of the impact of voluntary physical activity on hippocampus-dependent contextual conditioning. Hippocampus. (2009) ;19: (10):988–1001.

[30] 

Erickson KI , Voss MW , Prakash RS , Basak C , Szabo A , Chaddock L , et al. Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci U S A. (2011) ;108: (7):3017–22.

[31] 

Hindin SB , Zelinski EM . Extended practice and aerobic exercise interventions benefit untrained cognitive outcomes in older adults: A meta-analysis. J Am Geriatr Soc. (2012) ;60: (1):136–41.

[32] 

Kim SE , Ko IG , Kim BK , Shin MS , Cho S , Kim CJ , et al. Treadmill exercise prevents aging-induced failure of memory through an increase in neurogenesis and suppression of apoptosis in rat hippocampus. Exp Gerontol. (2010) ;45: (5):357–65.

[33] 

Albeck DS , Sano K , Prewitt GE , Dalton L . Mild forced treadmill exercise enhances spatial learning in the aged rat. Behav Brain Res. (2006) ;168: (2):345–8.

[34] 

Aguiar AS Jr , Boemer G , Rial D , Cordova FM , Mancini G , Walz R , et al. High-intensity physical exercise disrupts implicit memory in mice: Involvement of the striatal glutathione antioxidant system and intracellular signaling. Neuroscience. (2010) ;171: (4):1216–27.

[35] 

Blustein JE , McLaughlin M , Hoffman JR . Exercise effects stress-induced analgesia and spatial learning in rats. Physiol Behav. (2006) ;89: (4):582–6.

[36] 

Garcia-Capdevila S , Portell-Cortes I , Torras-Garcia M , Coll-Andreu M , Costa-Miserachs D . Effects of long-term voluntary exercise on learning and memory processes: Dependency of the task and level of exercise. Behav Brain Res. (2009) ;202: (2):162–70.

[37] 

Brisswalter J , Collardeau M , Rene A . Effects of acute physical exercise characteristics on cognitive performance. Sports Med. (2002) ;32: (9):555–66.

[38] 

Kashihara K , Maruyama T , Murota M , Nakahara Y . Positive effects of acute and moderate physical exercise on cognitive function. J Physiol Anthropol. (2009) ;28: (4):155–64.

[39] 

Zschucke E , Gaudlitz K , Strohle A . Exercise and physical activity in mental disorders: Clinical and experimental evidence. J Prev Med Public Health.. (2013) ;46: (Suppl 1):S12–21.

[40] 

Asmundson GJ , Fetzner MG , Deboer LB , Powers MB , Otto MW , Smits JA . Let’s get physical: A contemporary review of the anxiolytic effects of exercise for anxiety and its disorders. Depress Anxiety. (2013) ;30: (4):362–73.

[41] 

Radovic S , Gordon MS , Melvin GA . Should we recommend exercise to adolescents with depressive symptoms? A meta-analysis. J Paediatr Child Health. (2017) ;53: (3):214–20.

[42] 

Gordon BR , McDowell CP , Lyons M , Herring MP . The Effects of Resistance Exercise Training on Anxiety: A Meta-Analysis and Meta-Regression Analysis of Randomized Controlled Trials. Sports Med. (2017) ;47: (12):2521–32.

[43] 

Kim YS , Park YS , Allegrante JP , Marks R , Ok H , Ok Cho K , et al. Relationship between physical activity and general mental health. Prev Med. (2012) ;55: (5):458–63.

[44] 

Patki G , Li L , Allam F , Solanki N , Dao AT , Alkadhi K , et al. Moderate treadmill exercise rescues anxiety and depression-like behavior as well as memory impairment in a rat model of posttraumatic stress disorder. Physiol Behav. (2014) ;130: :47–53.

[45] 

Park HS , Lee JM , Cho HS , Park SS , Kim TW . Physical exercise ameliorates mood disorder-like behavior on high-fat-diet-induced obesity in mice. Psychiatry Res. (2017) ;250: :71–7.

[46] 

Zhang Q , Zhang J , Yan Y , Zhang P , Zhang W , Xia R . Proinflammatory cytokines correlate with early exercise attenuating anxiety-like behavior after cerebral ischemia. Brain Behav. (2017) ;7: (11):e00854.

[47] 

Statton MA , Encarnacion M , Celnik P , Bastian AJ . A Single Bout of Moderate Aerobic Exercise Improves Motor Skill Acquisition. PloS one. (2015) ;10: (10):e0141393.

[48] 

Roig M , Skriver K , Lundbye-Jensen J , Kiens B , Nielsen JB . A single bout of exercise improves motor memory. PloS one. (2012) ;7: (9):e44594.

[49] 

Ploughman M , Attwood Z , White N , Dore JJ , Corbett D . Endurance exercise facilitates relearning of forelimb motor skill after focal ischemia. Eur J Neurosci. (2007) ;25: (11):3453–60.

[50] 

Park JW , Bang MS , Kwon BS , Park YK , Kim DW , Shon SM , et al. Early treadmill training promotes motor function after hemorrhagic stroke in rats. Neurosci Lett. (2010) ;471: (2):104–8.

[51] 

Takamatsu Y , Tamakoshi K , Waseda Y , Ishida K . Running exercise enhances motor functional recovery with inhibition of dendritic regression in the motor cortex after collagenase-induced intracerebral hemorrhage in rats. Behav Brain Res. (2016) ;300: :56–64.

[52] 

Chang HC , Yang YR , Wang PS , Wang RY . Quercetin enhances exercise-mediated neuroprotective effects in brain ischemic rats. Med Sci Sports Exerc. (2014) ;46: (10):1908–16.

[53] 

Wu SY , Wang TF , Yu L , Jen CJ , Chuang JI , Wu FS , et al. Running exercise protects the substantia nigra dopaminergic neurons against inflammation-induced degeneration via the activation of BDNF signaling pathway. Brain Behav Immun. (2011) ;25: (1):135–46.

[54] 

Jadavji NM , Kolb B , Metz GA . Enriched environment improves motor function in intact and unilateral dopamine-depleted rats. Neuroscience. (2006) ;140: (4):1127–38.

[55] 

Chuang CS , Chang JC , Cheng FC , Liu KH , Su HL , Liu CS . Modulation of mitochondrial dynamics by treadmill training to improve gait and mitochondrial deficiency in a rat model of Parkinson’s disease. Life Sci. (2017) ;191: :236–44.

[56] 

Colcombe SJ , Erickson KI , Scalf PE , Kim JS , Prakash R , McAuley E , et al. Aerobic exercise training increases brain volume in aging humans. J Gerontol A Biol Sci Med Sci. (2006) ;61: (11):1166–70.

[57] 

Tseng BY , Uh J , Rossetti HC , Cullum CM , Diaz-Arrastia RF , Levine BD , et al. Masters athletes exhibit larger regional brain volume and better cognitive performance than sedentary older adults. J Magn Reson Imaging. (2013) ;38: (5):1169–76.

[58] 

Colcombe SJ , Kramer AF , Erickson KI , Scalf P , McAuley E , Cohen NJ , et al. Cardiovascular fitness, cortical plasticity, and aging. Proc Natl Acad Sci U S A. (2004) ;101: (9):3316–21.

[59] 

Voss MW , Prakash RS , Erickson KI , Basak C , Chaddock L , Kim JS , et al. Plasticity of brain networks in a randomized intervention trial of exercise training in older adults. Front Aging Neurosci. (2010) :2.

[60] 

Redila VA , Christie BR . Exercise-induced changes in dendritic structure and complexity in the adult hippocampal dentate gyrus. Neuroscience. (2006) ;137: (4):1299–307.

[61] 

Pietrelli A , Lopez-Costa JJ , Goni R , Lopez EM , Brusco A , Basso N . Effects of moderate and chronic exercise on the nitrergic system and behavioral parameters in rats. Brain Res. (2011) ;1389: : 71–82.

[62] 

Eddy MC , Green JT . Running wheel exercise reduces renewal of extinguished instrumental behavior and alters medial prefrontal cortex neurons in adolescent, but not adult, rats. Behav Neurosci. (2017) ;131: (6):460–9.

[63] 

Huang TY , Lin LS , Cho KC , Chen SJ , Kuo YM , Yu L , et al. Chronic treadmill exercise in rats delicately alters the Purkinje cell structure to improve motor performance and toxin resistance in the cerebellum. Journal of applied physiology. (2012) ;113: (6):889–95.

[64] 

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. (2005) ;486: (1):39–47.

[65] 

Stranahan AM , Khalil D , Gould E . Running induces widespread structural alterations in the hippocampus and entorhinal cortex. Hippocampus. (2007) ;17: (11):1017–22.

[66] 

Yang TT , Lo CP , Tsai PS , Wu SY , Wang TF , Chen YW , et al. Aging and Exercise Affect Hippocampal Neurogenesis via Different Mechanisms. PLoS One. (2015) ;10: (7):e0132152.

[67] 

Wu CW , Chang YT , Yu L , Chen HI , Jen CJ , Wu SY , et al. Exercise enhances the proliferation of neural stem cells and neurite growth and survival of neuronal progenitor cells in dentate gyrus of middle-aged mice. Journal of Applied Physiology. (2008) ;105: (5):1585–94.

[68] 

Schaefers AT , Grafen K , Teuchert-Noodt G , Winter Y . Synaptic remodeling in the dentate gyrus, CA3, CA1, subiculum, and entorhinal cortex of mice: Effects of deprived rearing and voluntary running. Neural Plast. (2010) ;2010: :870573.

[69] 

Shih PC , Yang YR , Wang RY . Effects of exercise intensity on spatial memory performance and hippocampal synaptic plasticity in transient brain ischemic rats. PloS One. (2013) ;8: (10):e78163.

[70] 

Rojas JJ , Deniz BF , Miguel PM , Diaz R , Hermel Edo E , Achaval M , et al. Effects of daily environmental enrichment on behavior and dendritic spine density in hippocampus following neonatal hypoxia-ischemia in the rat. Exp Neurol. (2013) ;241: :25–33.

[71] 

Lin TW , Shih YH , Chen SJ , Lien CH , Chang CY , Huang TY , et al. Running exercise delays neurodegeneration in amygdala and hippocampus of Alzheimer’s disease (APP/PS1) transgenic mice. Neurobiol Learn Mem. (2015) ;118: : 189–97.

[72] 

Chen K , Zhang L , Tan M , Lai CS , Li A , Ren C , et al. Treadmill exercise suppressed stress-induced dendritic spine elimination in mouse barrel cortex and improved working memory via BDNF/TrkB pathway. Transl Psychiatry. (2017) ;7: (3):e1069.

[73] 

Laforce R Jr , Doyon J . Distinct contribution of the striatum and cerebellum to motor learning. Brain Cogn. (2001) ;45: (2):189–211.

[74] 

Pysh JJ , Weiss GM . Exercise during development induces an increase in Purkinje cell dendritic tree size. Science. (1979) ; 206: (4415):230–2.

[75] 

Gonzalez-Burgos I , Gonzalez-Tapia D , Zamora DA , Feria-Velasco A , Beas-Zarate C . Guided motor training induces dendritic spine plastic changes in adult rat cerebellar purkinje cells. Neurosci Lett. (2011) ;491: (3):216–20.

[76] 

Shin MS , Jeong HY , An DI , Lee HY , Sung YH . Treadmill exercise facilitates synaptic plasticity on dopaminergic neurons and fibers in the mouse model with Parkinson’s disease. Neurosci Lett. (2016) ;621: : 28–33.

[77] 

Toy WA , Petzinger GM , Leyshon BJ , Akopian GK , Walsh JP , Hoffman MV , et al. Treadmill exercise reverses dendritic spine loss in direct and indirect striatal medium spiny neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson’s disease. Neurobiol Dis. (2014) ;63: : 201–9.

[78] 

Takamatsu Y , Ishida A , Hamakawa M , Tamakoshi K , Jung CG , Ishida K . Treadmill running improves motor function and alters dendritic morphology in the striatum after collagenase-induced intracerebral hemorrhage in rats. Brain Res. (2010) ;1355: 165–73.

[79] 

Yoon MC , Shin MS , Kim TS , Kim BK , Ko IG , Sung YH , et al. Treadmill exercise suppresses nigrostriatal dopaminergic neuronal loss in 6-hydroxydopamine-induced Parkinson’s rats. Neurosci Lett. (2007) ;423: (1):12–7.

[80] 

Farmer J , Zhao X , van Praag H , Wodtke K , Gage FH , Christie BR . Effects of voluntary exercise on synaptic plasticity and gene expression in the dentate gyrus of adult male Sprague-Dawley rats in vivo. Neuroscience. (2004) ;124: (1):71–9.

[81] 

Vasuta C , Caunt C , James R , Samadi S , Schibuk E , Kannangara T , et al. Effects of exercise on NMDA receptor subunit contributions to bidirectional synaptic plasticity in the mouse dentate gyrus. Hippocampus. (2007) ;17: (12):1201–8.

[82] 

Radahmadi M , Hosseini N , Alaei H . Effect of exercise, exercise withdrawal, and continued regular exercise on excitability and long-term potentiation in the dentate gyrus of hippocampus. Brain Res. (2016) ;1653: : 8–13.

[83] 

Patten AR , Sickmann H , Hryciw BN , Kucharsky T , Parton R , Kernick A , et al. Long-term exercise is needed to enhance synaptic plasticity in the hippocampus. Learn Mem. (2013) ;20: (11):642–7.

[84] 

D’Arcangelo G , Triossi T , Buglione A , Melchiorri G , Tancredi V . Modulation of synaptic plasticity by short-term aerobic exercise in adult mice. Behav Brain Res. (2017) ;332: : 59–63.

[85] 

Duffy SN , Craddock KJ , Abel T , Nguyen PV . Environmental enrichment modifies the PKA-dependence of hippocampal LTP and improves hippocampus-dependent memory. Learn Mem. (2001) ;8: (1):26–34.

[86] 

Kim SE , Ko IG , Shin MS , Kim CJ , Jin BK , Hong HP , et al. Treadmill exercise and wheel exercise enhance expressions of neutrophic factors in the hippocampus of lipopolysaccharide-injected rats. Neurosci Lett. (2013) ;538: : 54–9.

[87] 

Molteni R , Ying Z , Gomez-Pinilla F . Differential effects of acute and chronic exercise on plasticity-related genes in the rat hippocampus revealed by microarray. Eur J Neurosci. (2002) ;16: (6):1107–16.

[88] 

Tsai SF , Chen PC , Calkins MJ , Wu SY , Kuo YM . Exercise Counteracts Aging-Related Memory Impairment: A Potential Role for the Astrocytic Metabolic Shuttle. Front Aging Neurosci. (2016) ;8: : 57.

[89] 

Barres BA . The mystery and magic of glia: A perspective on their roles in health and disease. Neuron. (2008) ;60: (3):430–40.

[90] 

Brockett AT , LaMarca EA , Gould E . Physical exercise enhances cognitive flexibility as well as astrocytic and synaptic markers in the medial prefrontal cortex. PloS One. (2015) ;10: (5):e0124859.

[91] 

Saur L , Baptista PP , de Senna PN , Paim MF , do Nascimento P , Ilha J , et al. Physical exercise increases GFAP expression and induces morphological changes in hippocampal astrocytes. Brain Struct Funct. (2014) ;219: (1):293–302.

[92] 

Fahimi A , Baktir MA , Moghadam S , Mojabi FS , Sumanth K , McNerney MW , et al. Physical exercise induces structural alterations in the hippocampal astrocytes: Exploring the role of BDNF-TrkB signaling. Brain Struct Funct. (2017) ;222: (4):1797–808.

[93] 

Kohman RA , DeYoung EK , Bhattacharya TK , Peterson LN , Rhodes JS . Wheel running attenuates microglia proliferation and increases expression of a proneurogenic phenotype in the hippocampus of aged mice. Brain Behav Immun. (2012) ;26: (5):803–10.

[94] 

Kohman RA , Bhattacharya TK , Wojcik E , Rhodes JS . Exercise reduces activation of microglia isolated from hippocampus and brain of aged mice. J Neuroinflammation. (2013) ;10: : 114.

[95] 

Barrientos RM , Frank MG , Crysdale NY , Chapman TR , Ahrendsen JT , Day HE , et al. Little exercise, big effects: Reversing aging and infection-induced memory deficits, and underlying processes. J Neurosci. (2011) ;31: (32):11578–86.

[96] 

Speisman RB , Kumar A , Rani A , Foster TC , Ormerod BK . Daily exercise improves memory, stimulates hippocampal neurogenesis and modulates immune and neuroimmune cytokines in aging rats. Brain Behav Immun. (2013) ;28: : 25–43.

[97] 

Martin SA , Pence BD , Greene RM , Johnson SJ , Dantzer R , Kelley KW , et al. Effects of voluntary wheel running on LPS-induced sickness behavior in aged mice. Brain Behav Immun. (2013) ;29: ,113–23.

[98] 

Biber K , Neumann H , Inoue K , Boddeke HW . Neuronal ‘On’ and ‘Off’ signals control microglia. Trends Neurosci. (2007) ;30: (11):596–602.

[99] 

Tong L , Shen H , Perreau VM , Balazs R , Cotman CW . Effects of exercise on gene-expression profile in the rat hippocampus. Neurobiol Dis. (2001) ;8: (6):1046–56.

[100] 

Sung YH , Kim SC , Hong HP , Park CY , Shin MS , Kim CJ , et al. Treadmill exercise ameliorates dopaminergic neuronal loss through suppressing microglial activation in Parkinson’s disease mice. Life Sci. (2012) ;91: (25-26):1309–16.

[101] 

Duman RS , Monteggia LM . A neurotrophic model for stress-related mood disorders. Biol Psychiatry. (2006) ;59: (12):1116–27.

[102] 

McAllister AK , Katz LC , Lo DC . Neurotrophins and synaptic plasticity. Annu Rev Neurosci. (1999) ;22: : 295–318.

[103] 

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. (2003) ;122: (3):647–57.

[104] 

Szuhany KL , Bugatti M , Otto MW . A meta-analytic review of the effects of exercise on brain-derived neurotrophic factor. J Psychiatr Res. (2015) ;60: : 56–64.

[105] 

Cotman CW , Berchtold NC . Exercise: A behavioral intervention to enhance brain health and plasticity. Trends Neurosci. (2002) ;25: (6):295–301.

[106] 

Garza AA , Ha TG , Garcia C , Chen MJ , Russo-Neustadt AA . Exercise, antidepressant treatment, and BDNF mRNA expression in the aging brain. Pharmacol Biochem Behav. (2004) ;77: (2):209–20.

[107] 

Soya H , Nakamura T , Deocaris CC , Kimpara A , Iimura M , Fujikawa T , et al. BDNF induction with mild exercise in the rat hippocampus. Biochem Biophys Res Commun. (2007) ;358: (4):961–7.

[108] 

Aguiar AS Jr , Castro AA , Moreira EL , Glaser V , Santos AR , Tasca CI , et al. 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. (2011) ;132: (11-12):560–7.

[109] 

Gomez-Pinilla F , So V , Kesslak JP . Spatial learning induces neurotrophin receptor and synapsin I in the hippocampus. Brain Res. (2001) ;904: (1):13–9.

[110] 

Ferreira AF , Real CC , Rodrigues AC , Alves AS , Britto LR . Short-term, moderate exercise is capable of inducing structural, BDNF-independent hippocampal plasticity. Brain Res. (2011) ;1425:111–22.

[111] 

Vaynman S , Ying Z , Gomez-Pinilla F . Hippocampal BDNF mediates the efficacy of exercise on synaptic plasticity and cognition. Eur J Neurosci. (2004) ;20: (10):2580–90.

[112] 

Fernandez AM , Torres-Aleman I . The many faces of insulin-like peptide signalling in the brain. Nat Rev Neurosci. (2012) ;13: (4):225–39.

[113] 

Brooker GJ , Kalloniatis M , Russo VC , Murphy M , Werther GA , Bartlett PF . Endogenous IGF-1 regulates the neuronal differentiation of adult stem cells. J Neurosci Res. (2000) ;59: (3):332–41.

[114] 

Aleman A , Torres-Aleman I . Circulating insulin-like growth factor I and cognitive function: Neuromodulation throughout the lifespan. Prog Neurobiol. (2009) ;89: (3):256–65.

[115] 

Cetinkaya C , Sisman AR , Kiray M , Camsari UM , Gencoglu C , Baykara B , et al. Positive effects of aerobic exercise on learning and memory functioning, which correlate with hippocampal IGF-1 increase in adolescent rats. Neurosci Lett. (2013) ;549: : 177–81.

[116] 

Llorens-Martin M , Torres-Aleman I , Trejo JL . Exercise modulates insulin-like growth factor 1-dependent and -independent effects on adult hippocampal neurogenesis and behaviour. Mol Cell Neurosci. (2010) ;44: (2):109–17.

[117] 

Trejo JL , Carro E , Torres-Aleman I . Circulating insulin-like growth factor I mediates exercise-induced increases in the number of new neurons in the adult hippocampus. J Neurosci. (2001) ;21: (5):1628–34.

[118] 

Carro E , Trejo JL , Busiguina S , Torres-Aleman I . Circulating insulin-like growth factor I mediates the protective effects of physical exercise against brain insults of different etiology and anatomy. J Neurosci. (2001) ;21: (15):5678–84.

[119] 

Ding Q , Vaynman S , Akhavan M , Ying Z , Gomez-Pinilla F . Insulin-like growth factor I interfaces with brain-derived neurotrophic factor-mediated synaptic plasticity to modulate aspects of exercise-induced cognitive function. Neuroscience. (2006) ;140: (3):823–33.

[120] 

Gora-Kupilas K , Josko J . The neuroprotective function of vascular endothelial growth factor (VEGF). Folia Neuropathol. (2005) ;43: (1):31–9.

[121] 

Nowacka MM , Obuchowicz E . Vascular endothelial growth factor (VEGF) and its role in the central nervous system: A new element in the neurotrophic hypothesis of antidepressant drug action. Neuropeptides. (2012) ;46: (1):1–10.

[122] 

Cao L , Jiao X , Zuzga DS , Liu Y , Fong DM , Young D , et al. VEGF links hippocampal activity with neurogenesis, learning and memory. Nat Genet. (2004) ;36: (8):827–35.

[123] 

Jin K , Zhu Y , Sun Y , Mao XO , Xie L , Greenberg DA . Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc Natl Acad Sci U S A. (2002) ;99: (18):11946–50.

[124] 

Ballard HJ . Exercise makes your brain bigger: Skeletal muscle VEGF and hippocampal neurogenesis. The Journal of Physiology. (2017) ;595: (17):5721–2.

[125] 

Stevenson ME , Behnke VK , Swain RA . Exercise pattern and distance differentially affect hippocampal and cerebellar expression of FLK-1 and FLT-1 receptors in astrocytes and blood vessels. Behav Brain Res. (2018) ;337: : 8–16.

[126] 

Petrova TV , Makinen T , Alitalo K . Signaling via vascular endothelial growth factor receptors. Exp Cell Res. (1999) ;253: (1):117–30.

[127] 

Palmer TD , Willhoite AR , Gage FH . Vascular niche for adult hippocampal neurogenesis. J Comp Neurol. (2000) ;425: (4):479–94.

[128] 

Fabel K , Fabel K , Tam B , Kaufer D , Baiker A , Simmons N , et al. VEGF is necessary for exercise-induced adult hippocampal neurogenesis. Eur J Neurosci. (2003) ;18: (10):2803–12.

[129] 

Rich B , Scadeng M , Yamaguchi M , Wagner PD , Breen EC . Skeletal myofiber vascular endothelial growth factor is required for the exercise training-induced increase in dentate gyrus neuronal precursor cells. The Journal of Physiology. (2017) ;595: (17):5931–43.

[130] 

Frielingsdorf H , Simpson DR , Thal LJ , Pizzo DP . Nerve growth factor promotes survival of new neurons in the adult hippocampus. Neurobiol Dis. (2007) ;26: (1):47–55.

[131] 

Neeper SA , Gomez-Pinilla F , Choi J , Cotman CW . Physical activity increases mRNA for brain-derived neurotrophic factor and nerve growth factor in rat brain. Brain Res. (1996) ;726: (1-2):49–56.

[132] 

Shaywitz AJ , Greenberg ME . CREB: A stimulus-induced transcription factor activated by a diverse array of extracellular signals. Annu Rev Biochem. (1999) ;68: : 821–61.

[133] 

Bishop NA , Lu T , Yankner BA . Neural mechanisms of ageing and cognitive decline. Nature. (2010) ;464: (7288):529–35.

[134] 

Petersen RC , Smith GE , Waring SC , Ivnik RJ , Kokmen E , Tangelos EG . Aging, memory, and mild cognitive impairment. Int Psychogeriatr. (1997) ;9: (Suppl 1):65–9.

[135] 

Golomb J , Kluger A , de Leon MJ , Ferris SH , Convit A , Mittelman MS , et al. Hippocampal formation size in normal human aging: A correlate of delayed secondary memory performance. Learn Mem. (1994) ;1: (1):45–54.

[136] 

Enzinger C , Fazekas F , Matthews PM , Ropele S , Schmidt H , Smith S , et al. Risk factors for progression of brain atrophy in aging: Six-year follow-up of normal subjects. Neurology. (2005) ;64: (10):1704–11.

[137] 

Bondareff W . Synaptic atrophy in the senescent hippocampus. Mech Ageing Dev. (1979) ;9: (1-2):163–71.

[138] 

Meyer G , Ferres-Torres R . [Quantitative age-dependent variations in dendritic spines in the hippocampus (CA1, CA3 and fascia dentata) of the albino mouse]. J Hirnforsch. (1978) ;19: (4):371–8.

[139] 

Tsai SF , Ku NW , Wang TF , Yang YH , Shih YH , Wu SY , et al. Long-Term Moderate Exercise Rescues Age-Related Decline in Hippocampal Neuronal Complexity and Memory. Gerontology. (2018) : 1–11.

[140] 

Kuhn HG , Dickinson-Anson H , Gage FH . Neurogenesis in the dentate gyrus of the adult rat: Age-related decrease of neuronal progenitor proliferation. J Neurosci. (1996) ;16: (6):2027–33.

[141] 

van Praag H , Shubert T , Zhao C , Gage FH . Exercise enhances learning and hippocampal neurogenesis in aged mice. J Neurosci. (2005) ;25: (38):8680–5.

[142] 

Deupree DL , Bradley J , Turner DA . Age-related alterations in potentiation in the CA1 region in F344 rats. Neurobiol Aging. (1993) ;14: (3):249–58.

[143] 

Barnes CA . Memory deficits associated with senescence: A neurophysiological and behavioral study in the rat. J Comp Physiol Psychol. (1979) ;93: (1):74–104.

[144] 

Tombaugh GC , Rowe WB , Chow AR , Michael TH , Rose GM . Theta-frequency synaptic potentiation in CA1 in vitro distinguishes cognitively impaired from unimpaired aged Fischer 344 rats. J Neurosci. (2002) ;22: (22):9932–40.

[145] 

Samorajski T , Delaney C , Durham L , Ordy JM , Johnson JA , Dunlap WP . Effect of exercise on longevity, body weight, locomotor performance, and passive-avoidance memory of C57BL/6J mice. Neurobiol Aging. (1985) ;6: (1):17–24.

[146] 

Gomes da Silva S , Unsain N , Masco DH , Toscano-Silva M , de Amorim HA , Silva Araujo BH , et al. Early exercise promotes positive hippocampal plasticity and improves spatial memory in the adult life of rats. Hippocampus. (2012) ;22: (2):347–58.

[147] 

Cotman CW , Berchtold NC , Christie LA . Exercise builds brain health: Key roles of growth factor cascades and inflammation. Trends Neurosci. (2007) ;30: (9):464–72.

[148] 

Cooper C , Moon HY , van Praag H On the Run for Hippocampal Plasticity. Cold Spring Harb Perspect Med. (2018) ;8: (4).

[149] 

Lazarov O , Mattson MP , Peterson DA , Pimplikar SW , van Praag H . When neurogenesis encounters aging and disease. Trends Neurosci. (2010) ;33: (12):569–79.

[150] 

van Praag H , Kempermann G , Gage FH . Running increases cell proliferation and neurogenesis in the adult mouse dentate gyrus. Nat Neurosci. (1999) ;2: (3):266–70.

[151] 

Laske C , Stransky E , Leyhe T , Eschweiler GW , Maetzler W , Wittorf A , et al. BDNF serum and CSF concentrations in Alzheimer’s disease, normal pressure hydrocephalus and healthy controls. J Psychiatr Res. (2007) ;41: (5):387–94.

[152] 

Bruno MA , Leon WC , Fragoso G , Mushynski WE , Almazan G , Cuello AC . Amyloid beta-induced nerve growth factor dysmetabolism in Alzheimer disease. J Neuropathol Exp Neurol. (2009) ;68: (8):857–69.

[153] 

Solerte SB , Cerutti N , Mirani M , Ceresini G , Giusti A , Ferrari E , et al. Impairment of secretory pattern of IGF-I from lymphomononuclear cells in aging and dementia of the Alzheimer’s and vascular type. J Endocrinol Invest. (2002) ;25: (10 Suppl):47–50.

[154] 

Solerte SB , Ferrari E , Cuzzoni G , Locatelli E , Giustina A , Zamboni M , et al. Decreased release of the angiogenic peptide vascular endothelial growth factor in Alzheimer’s disease: Recovering effect with insulin and DHEA sulfate. Dement Geriatr Cogn Disord. (2005) ;19: (1):1–10.

[155] 

Phillips HS , Hains JM , Armanini M , Laramee GR , Johnson SA , Winslow JW . BDNF mRNA is decreased in the hippocampus of individuals with Alzheimer’s disease. Neuron. (1991) ;7: (5):695–702.

[156] 

Hock C , Heese K , Hulette C , Rosenberg C , Otten U . Region-specific neurotrophin imbalances in Alzheimer disease: Decreased levels of brain-derived neurotrophic factor and increased levels of nerve growth factor in hippocampus and cortical areas. Arch Neurol. (2000) ;57: (6):846–51.

[157] 

Peng S , Wuu J , Mufson EJ , Fahnestock M . Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer’s disease. J Neurochem. (2005) ;93: (6):1412–21.

[158] 

Fumagalli F , Racagni G , Riva MA . The expanding role of BDNF: A therapeutic target for Alzheimer’s disease? Pharmacogenomics J. (2006) ;6: (1):8–15.

[159] 

Budni J , Bellettini-Santos T , Mina F , Garcez ML , Zugno AI . The involvement of BDNF, NGF and GDNF in aging and Alzheimer’s disease. Aging Dis. (2015) ;6: (5):331–41.

[160] 

Nagahara AH , Merrill DA , Coppola G , Tsukada S , Schroeder BE , Shaked GM , et al. Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat Med. (2009) ;15: (3):331–7.

[161] 

Dao AT , Zagaar MA , Levine AT , Alkadhi KA . Comparison of the Effect of Exercise on Late-Phase LTP of the Dentate Gyrus and CA1 of Alzheimer’s Disease Model. Mol Neurobiol. (2016) ;53: (10):6859–68.

[162] 

Liu HL , Zhao G , Cai K , Zhao HH , Shi LD . Treadmill exercise prevents decline in spatial learning and memory in APP/PS1 transgenic mice through improvement of hippocampal long-term potentiation. Behav Brain Res. (2011) ;218: (2):308–14.

[163] 

Nichol K , Deeny SP , Seif J , Camaclang K , Cotman CW . Exercise improves cognition and hippocampal plasticity in APOE epsilon4 mice. Alzheimers Dement. (2009) ;5: (4):287–94.

[164] 

Um HS , Kang EB , Leem YH , Cho IH , Yang CH , Chae KR , et al. 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. (2008) ;22: (4):529–39.

[165] 

Um HS , Kang EB , Koo JH , Kim HT , Jin L , Kim EJ , et al. Treadmill exercise represses neuronal cell death in an aged transgenic mouse model of Alzheimer’s disease. Neurosci Res. (2011) ;69: (2):161–73.

[166] 

Koo JH , Kwon IS , Kang EB , Lee CK , Lee NH , Kwon MG , et al. Neuroprotective effects of treadmill exercise on BDNF and PI3-K/AKT signaling pathway in the cortex of transgenic mice model of Alzheimer’s disease. J Exerc Nutrition Biochem. (2013) ;17: (4):151–60.

[167] 

Kim BK , Shin MS , Kim CJ , Baek SB , Ko YC , Kim YP . Treadmill exercise improves short-term memory by enhancing neurogenesis in amyloid beta-induced Alzheimer disease rats. J Exerc Rehabil. (2014) ;10: (1):2–8.

[168] 

Arancibia S , Silhol M , Mouliere F , Meffre J , Hollinger I , Maurice T , et al. Protective effect of BDNF against beta-amyloid induced neurotoxicity in vitro and in vivo in rats. Neurobiol Dis. (2008) ;31: (3):316–26.

[169] 

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. (2005) ;95: (4):1059–68.

[170] 

Elliott E , Atlas R , Lange A , Ginzburg I . Brain-derived neurotrophic factor induces a rapid dephosphorylation of tau protein through a PI-3 Kinase signalling mechanism. Eur J Neurosci. (2005) ;22: (5):1081–9.

[171] 

Li Z , Tan F , Thiele CJ . Inactivation of glycogen synthase kinase-3beta contributes to brain-derived neutrophic factor/TrkB-induced resistance to chemotherapy in neuroblastoma cells. Mol Cancer Ther. (2007) ;6: (12 Pt 1):3113–21.

[172] 

Talbot K , Wang HY , Kazi H , Han LY , Bakshi KP , Stucky A , et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Invest. (2012) ;122: (4):1316–38.

[173] 

Trueba-Saiz A , Cavada C , Fernandez AM , Leon T , Gonzalez DA , Fortea Ormaechea J , et al. Loss of serum IGF-I input to the brain as an early biomarker of disease onset in Alzheimer mice. Transl Psychiatry. (2013) ;3: : e330.

[174] 

Gasparini L , Xu H . Potential roles of insulin and IGF-1 in Alzheimer’s disease. Trends Neurosci. (2003) ;26: (8):404–6.

[175] 

Poirier R , Fernandez AM , Torres-Aleman I , Metzger F . Early brain amyloidosis in APP/PS1 mice with serum insulin-like growth factor-I deficiency. Neurosci Lett. (2012) ;509: (2):101–4.

[176] 

Carro E , Trejo JL , Gerber A , Loetscher H , Torrado J , Metzger F , et al. Therapeutic actions of insulin-like growth factor I on APP/PS2 mice with severe brain amyloidosis. Neurobiol Aging. (2006) ;27: (9):1250–7.

[177] 

Carro E , Trejo JL , Gomez-Isla T , LeRoith D , Torres-Aleman I . Serum insulin-like growth factor I regulates brain amyloid-beta levels. Nat Med. (2002) ;8: (12):1390–7.

[178] 

Hong M , Lee VM . Insulin and insulin-like growth factor-1 regulate tau phosphorylation in cultured human neurons. J Biol Chem. (1997) ;272: (31):19547–53.

[179] 

Lesort M , Johnson GV . Insulin-like growth factor-1 and insulin mediate transient site-selective increases in tau phosphorylation in primary cortical neurons. Neuroscience. (2000) ;99: (2):305–16.

[180] 

Howlett KF , Sakamoto K , Yu H , Goodyear LJ , Hargreaves M . Insulin-stimulated insulin receptor substrate-2-associated phosphatidylinositol 3-kinase activity is enhanced in human skeletal muscle after exercise. Metabolism. (2006) ;55: (8):1046–52.

[181] 

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. (2013) ;256: : 261–72.

[182] 

Kang EB , Cho JY . Effect of treadmill exercise on PI3K/AKT/mTOR, autophagy, and Tau hyperphosphorylation in the cerebral cortex of NSE/htau23 transgenic mice. J Exerc Nutrition Biochem. (2015) ;19: (3):199–209.

[183] 

Kang EB , Cho JY . Effects of treadmill exercise on brain insulin signaling and beta-amyloid in intracerebroventricular streptozotocin induced-memory impairment in rats. J Exerc Nutrition Biochem. (2014) ;18: (1):89–96.

[184] 

Tang K , Xia FC , Wagner PD , Breen EC . Exercise-induced VEGF transcriptional activation in brain, lung and skeletal muscle. Respir Physiol Neurobiol. (2010) ;170: (1):16–22.

[185] 

Ross MD , Wekesa AL , Phelan JP , Harrison M . Resistance exercise increases endothelial progenitor cells and angiogenic factors. Med Sci Sports Exerc. (2014) ;46: (1):16–23.

[186] 

Zlokovic BV . Neurovascular mechanisms of Alzheimer’s neurodegeneration. Trends Neurosci. (2005) ;28: (4):202–8.

[187] 

Poetsch V , Neuhaus W , Noe CR . Serum-derived immunoglobulins neutralize adverse effects of amyloid-beta peptide on the integrity of a blood-brain barrier in vitro model. J Alzheimers Dis. (2010) ;21: (1):303–14.

[188] 

Jeynes B , Provias J . Evidence for altered LRP/RAGE expression in Alzheimer lesion pathogenesis. Curr Alzheimer Res. (2008) ;5: (5):432–7.

[189] 

Spuch C , Antequera D , Portero A , Orive G , Hernandez RM , Molina JA , et al. The effect of encapsulated VEGF-secreting cells on brain amyloid load and behavioral impairment in a mouse model of Alzheimer’s disease. Biomaterials. (2010) ;31: (21):5608–18.

[190] 

Burger S , Noack M , Kirazov LP , Kirazov EP , Naydenov CL , Kouznetsova E , et al. Vascular endothelial growth factor (VEGF) affects processing of amyloid precursor protein and beta-amyloidogenesis in brain slice cultures derived from transgenic Tgmouse brain. Int J Dev Neurosci. (2009) ;27: (6):517–23.

[191] 

Burger S , Yafai Y , Bigl M , Wiedemann P , Schliebs R . Effect of VEGF and its receptor antagonist SU-an inhibitor of angiogenesis, on processing of the beta-amyloid precursor protein in primary neuronal cells derived from brain tissue of Tgmice. Int J Dev Neurosci. (2010) ;28: (7):597–604.

[192] 

Yang SP , Kwon BO , Gho YS , Chae CB . Specific interaction of VEGF165 with beta-amyloid, and its protective effect on beta-amyloid-induced neurotoxicity. J Neurochem. (2005) ;93: (1):118–27.

[193] 

Fahnestock M , Michalski B , Xu B , Coughlin MD . The precursor pro-nerve growth factor is the predominant form of nerve growth factor in brain and is increased in Alzheimer’s disease. Mol Cell Neurosci. (2001) ;18: (2):210–20.

[194] 

Counts SE , He B , Prout JG , Michalski B , Farotti L , Fahnestock M , et al. Cerebrospinal Fluid proNGF: A Putative Biomarker for Early Alzheimer’s Disease. Curr Alzheimer Res. (2016) ;13: (7):800–8.

[195] 

Nykjaer A , Lee R , Teng KK , Jansen P , Madsen P , Nielsen MS , et al. Sortilin is essential for proNGF-induced neuronal cell death. Nature. (2004) ; 427: (6977): 843–8.

[196] 

Mufson EJ , He B , Nadeem M , Perez SE , Counts SE , Leurgans S , et al. Hippocampal proNGF signaling pathways and beta-amyloid levels in mild cognitive impairment and Alzheimer disease. J Neuropathol Exp Neurol. (2012) ;71: (11):1018–29.

[197] 

Chao MV Retrograde transport redux. Neuron. (2003) ;39: (1):1–2.

[198] 

Gavazzi I , Cowen T . Can the neurotrophic hypothesis explain degeneration and loss of plasticity in mature and ageing autonomic nerves? J Auton Nerv Syst. (1996) ;58: (1-2):1–10.

[199] 

Capsoni S , Ugolini G , Comparini A , Ruberti F , Berardi N , Cattaneo A . Alzheimer-like neurodegeneration in aged antinerve growth factor transgenic mice. Proc Natl Acad Sci U S A. (2000) ;97: (12):6826–31.

[200] 

Capsoni S , Giannotta S , Cattaneo A . Beta-amyloid plaques in a model for sporadic Alzheimer’s disease based on transgenic anti-nerve growth factor antibodies. Mol Cell Neurosci. (2002) ;21: (1):15–28.

[201] 

Matrone C , Di Luzio A , Meli G , D’Aguanno S , Severini C , Ciotti MT , et al. Activation of the amyloidogenic route by NGF deprivation induces apoptotic death in PC12 cells. J Alzheimers Dis. (2008) ;13: (1):81–96.

[202] 

Nuydens R , Dispersyn G , de Jong M , van den Kieboom G , Borgers M , Geerts H . Aberrant tau phosphorylation and neurite retraction during NGF deprivation in PC12 cells. Biochem Biophys Res Commun. (1997) ;240: (3):687–91.

[203] 

Canu N , Amadoro G , Triaca V , Latina V , Sposato V , Corsetti V , et al. The Intersection of NGF/TrkA Signaling and Amyloid Precursor Protein Processing in Alzheimer’s Disease Neuropathology. Int J Mol Sci. (2017) ;18: (6).

[204] 

Lv Q , Lan W , Sun W , Ye R , Fan X , Ma M , et al. Intranasal nerve growth factor attenuates tau phosphorylation in brain after traumatic brain injury in rats. J Neurol Sci. (2014) ;345: (1-2):48–55.

[205] 

Iadecola C Neurovascular regulation in the normal brain and in Alzheimer’s disease. Nat Rev Neurosci. (2004) ;5: (5):347–60.

[206] 

Pekny M , Pekna M , Messing A , Steinhauser C , Lee JM , Parpura V , et al. Astrocytes: A central element in neurological diseases. Acta Neuropathol. (2016) ;131: (3):323–45.

[207] 

Verkhratsky A , Olabarria M , Noristani HN , Yeh CY , Rodriguez JJ . Astrocytes in Alzheimer’s disease. Neurotherapeutics. (2010) ;7: (4):399–412.

[208] 

Wyss-Coray T , Loike JD , Brionne TC , Lu E , Anankov R , Yan F , et al. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat Med. (2003) ;9: (4):453–7.

[209] 

Rodrigues L , Dutra MF , Ilha J , Biasibetti R , Quincozes-Santos A , Leite MC , et al. Treadmill training restores spatial cognitive deficits and neurochemical alterations in the hippocampus of rats submitted to an intracerebroventricular administration of streptozotocin. J Neural Transm (Vienna). (2010) ;117: (11):1295–305.

[210] 

Kress BT , Iliff JJ , Xia M , Wang M , Wei HS , Zeppenfeld D , et al. Impairment of paravascular clearance pathways in the aging brain. Ann Neurol. (2014) ;76: (6):845–61.

[211] 

Iliff JJ , Wang M , Liao Y , Plogg BA , Peng W , Gundersen GA , et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med.. (2012) ;4: (147):147ra11.

[212] 

He XF , Liu DX , Zhang Q , Liang FY , Dai GY , Zeng JS , et al. Voluntary Exercise Promotes Glymphatic Clearance of Amyloid Beta and Reduces the Activation of Astrocytes and Microglia in Aged Mice. Front Mol Neurosci. (2017) ;10: : 144.

[213] 

Nichol KE , Poon WW , Parachikova AI , Cribbs DH , Glabe CG , Cotman CW . Exercise alters the immune profile in TgAlzheimer mice toward a response coincident with improved cognitive performance and decreased amyloid. J Neuroinflammation. (2008) ;5: : 13.

[214] 

Leem YH , Lee YI , Son HJ , Lee SH . Chronic exercise ameliorates the neuroinflammation in mice carrying NSE/htau23. Biochem Biophys Res Commun. (2011) ;406: (3):359–65.

[215] 

Kanekiyo T , Bu G . The low-density lipoprotein receptor-related protein 1 and amyloid-beta clearance in Alzheimer’s disease. Front Aging Neurosci. (2014) ;6: : 93.

[216] 

Sasaki N , Toki S , Chowei H , Saito T , Nakano N , Hayashi Y , et al. Immunohistochemical distribution of the receptor for advanced glycation end products in neurons and astrocytes in Alzheimer’s disease. Brain Res. (2001) ;888: (2):256–62.

[217] 

Moore KM , Girens RE , Larson SK , Jones MR , Restivo JL , Holtzman DM , et al. A spectrum of exercise training reduces soluble Abeta in a dose-dependent manner in a mouse model of Alzheimer’s disease. Neurobiol Dis. (2016) ;85: : 218–24.