Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Rev. Neurosci.

2021; aop

Juan Liang, Huiqing Wang, Yan Zeng, Yi Qu, Qian Liu, Fengyan Zhao, Jianan Duan, Yin Jiang,
Shiping Li, Junjie Ying, Jinhui Li* and Dezhi Mu*

Physical exercise promotes brain remodeling by


regulating epigenetics, neuroplasticity and
neurotrophins
https://doi.org/10.1515/revneuro-2020-0099 analysis. It is composed of billions of nerve cells. They
Received September 1, 2020; accepted October 26, 2020; connect through synapses to form a complex neural
published online February 15, 2021
network, finely regulating a multitude of body functions.
Although it has been reported that brain damage is usually
Abstract: Exercise has been shown to have beneficial effects
irreversible, increasing evidence shows that physical ex-
on brain functions in humans and animals. Exercise can
ercise plays a protective role in various diseases, such as
improve memory and learning in age-related neurodegener-
Alzheimer’s disease (AD), Parkinson’s disease (PD),
ative diseases. In animal models, physical exercise regulates
depression, and stroke (Cass 2017; Ignacio et al. 2019; Pin-
epigenetics, promotes synaptic plasticity and hippocampal
Barre and Laurin 2015; Xu et al. 2019). The positive effects
neurogenesis, regulates the expression levels of neurotrophic
are reflected in improved memory and cognition, enhanced
factors, and improves cognitive function. Therefore, exercise
motor function, better-regulated mood, and strengthened
is very important for brain rehabilitation and remodeling. The
motor rehabilitation (Cass 2017; Kvam et al. 2016; Pin-Barre
purpose of this review is to explore the mechanisms by which
and Laurin 2015; Sharp and Hewitt 2014). Over the life span
exercise exerts positive effects on brain function. This
of a human, brain atrophy can be demonstrated by using
knowledge implies that physical exercise can be used as a
neuroimaging techniques (Nosheny et al. 2019), and the
non-drug therapy for neurological diseases.
plasma levels of some neurotrophic factors, such as brain-
Keywords: brain; epigenetics; improvement; neurotrophic derived neurotrophic factor (BDNF), insulin-like growth
factors; physical exercise; plasticity. factor 1 (IGF-1), and vascular endothelial growth factor
(VEGF), are decreased in the aged population (Ashpole
et al. 2015; Muñoz et al. 2018; Siuda et al. 2017). Researchers
Introduction have found that exercise can upregulate the levels of
neurotrophic factors, enhance angiogenesis, and promote
The central nervous system plays important roles in ho- hippocampal neurogenesis from animal studies (Cotman
meostasis, learning, memory function, and information et al. 2007; Ding et al. 2006a,b; Trejo et al. 2001). This
review summarizes various mechanisms such as epige-
netic, neuroplasticity, and neurotrophic factors by which
Juan Liang and Huiqing Wang contributed equally to this review. exercise may improve brain function. We think that these
mechanisms will help brain rehabilitation and remodeling
*Corresponding authors: Jinhui Li and Dezhi Mu, Department of in patients with brain disorders. Furthermore, these find-
Pediatrics, West China Second University Hospital, Sichuan ings can be used as the basic theory to support exercise-
University, Chengdu 610041, China; and Key Laboratory of Birth
induced improvements and enhance the quality of life in
Defects and Related Diseases of Women and Children (Ministry of
Education), West China Second University Hospital, Sichuan patients with various neurological diseases.
University, Chengdu 610041, China, E-mail: yoyolee824@163.com
(J. Li), mudz@scu.edu.cn (D. Mu).
Juan Liang, Huiqing Wang, Yan Zeng, Yi Qu, Qian Liu, Fengyan Zhao, Exercise benefits neuroprotection
Yin Jiang, Shiping Li and Junjie Ying, Department of Pediatrics, West
China Second University Hospital, Sichuan University, Chengdu by regulating epigenetics
610041, China; and Key Laboratory of Birth Defects and Related
Diseases of Women and Children (Ministry of Education), West China
Epigenetic inheritance refers to the change of gene
Second University Hospital, Sichuan University, Chengdu 610041,
China
expression and regulation without changing the DNA
Jianan Duan, West China Hospital, Sichuan University, Chengdu sequence. Epigenetic information differs from classical
610041, China genetic information that involves changes in gene quality
2 J. Liang et al.: Exercise promotes brain remodeling

and gene expression levels caused by alterations of the pathway (Bilang-Bleuel et al. 2005; Chandramohan et al. 2008;
gene sequence. Although epigenetic information does not Reul and Nutt 2008). The adaptive response of epigenetics and
change genes, the quantity of the encoded protein can be gene expression is a part of hippocampal neuroplasticity,
changed. Epigenetics was originally defined as the heri- which may be involved in memory formation for events to
table but reversible effect on gene expression without avoid anxiety and improve cognition in depression and stress-
altering DNA base pairs, primarily including histone related psychiatric disorders (Collins et al. 2009).
modification, DNA methylation, chromatin remodeling,
and microRNAs (miRNAs) (Burggren 2016; Tammen et al.
2013). Figure 1A shows exercise-induced epigenetic factors DNA methylation and chromatin remodeling
regulation on brain remodeling.
Besides the effect on HAT and HDAC, exercise has an in-
fluence on DNA methylation (Kashimoto et al. 2016). DNA
Histone modification methylation, which is related to gene expression, refers to
the process in which the enzyme DNA methyltransferase
Histone modification is one of the key mechanisms of gene adds a methyl group to a cytosine moiety of a genomic
transcription regulation, achieved by both histone acetyl- CpG dinucleotide at its 5′ carbon position (Jung and
transferase (HAT) and histone deacetylase (HDAC). The Pfeifer 2015). Both acute and chronic exercise can signif-
HAT can activate the transcription process of specific icantly affect DNA methylation in a highly gene-specific
genes, whereas HDAC can inhibit this process (Ali et al. manner (Voisin et al. 2015). Exercise can improve the
2018). Exercise can activate both HAT and HDAC, thereby cognitive processes in mouse models of aging, upregu-
influencing histone modification and DNA replication late BDNF levels, and downregulate the hippocampal
(Maejima et al. 2018a,b). Recently, evidence has shown expression of the neurotrophic factor receptor p75, which
that exercise can upregulate HAT activity, increase histone is related to necrosis and apoptosis of neurons (Maejima
acetylation level, and downregulate HDAC activity in the et al. 2018a,b). It has been reported that exercise can
hippocampus (Elsner et al. 2011). The decrease in HDAC reduce CpG methylation in the Bdnf promoter IV, leading
activity may be related to neuronal apoptosis (Elsner et al. to DNA demethylation of this promoter (Gomez-Pinilla
2011; Ryu et al. 2003; Saha and Pahan 2006). In et al. 2011).
senescence-accelerated mice prone 8 (SAMP8) model, ex- Chromatin remodeling, an epigenetic mechanism, is a
ercise was found to increase HAT activity and decrease the biological process characterized by a series of changes in
activity of HDAC thereby improving cognitive function. The chromatin nucleosomes mediated by the chromatin
results of this study suggest that exercise-induced alter- remodeling complex (Tammen et al. 2013). Exercise can
ations of HAT and HDAC in SAMP8 mice have an impact on increase the transcription and translation of BDNF and
chromatin remodeling, autophagy, and memory formation promote the chromatin remodeling of the Bdnf gene, which
(Cosin-Tomas et al. 2014). In rat models of stress (novelty may be associated with the exercise-induced hypo-
exposure, forced swimming, etc.), exercise induced his- methylation of the Bdnf promoter IV (Gomez-Pinilla et al.
tone H3 phospho-acetylation and increased the number of 2011; Vaynman et al. 2004). Exercise also up-regulates the
cFos-positive neurons in the dentate gyrus, which may be phosphorylation level of methyl-CpG binding protein
associated with an increased ability to cope with stress. The (MeCP2), which can increase the release of MeCP2 and
researchers also found that the intervention group showed activate BDNF transcription (Gomez-Pinilla et al. 2011).
more immobility behavior than the control group (Collins Without stimulus, MeCP2 suppresses BDNF transcription,
et al. 2009). In addition, previous research has shown that but this inhibitory MeCP2 effect disappears when neurons
histone H3 phospho-acetylation and cFos induction in are depolarized (Chen et al. 2003; Martinowich et al. 2003).
dentate granule cells are related to the immobility response In addition, exercise can improve the activated stage of
after re-test forced swimming (Bilang-Bleuel et al. 2005; both Ca2+/calmodulin-dependent protein kinase II (CaM-
Chandramohan et al. 2008), which may be involved in the KII) and cyclic adenosine monophosphate (cAMP)-
memory formation of post-traumatic stress disorder (Reul response element-binding protein (CREB) in the hippo-
and Chandramohan 2007). This behavior after stressful campus (Gomez-Pinilla et al. 2011), which is vital for
events may be associated with the activation of glucocor- exercise-induced effects on synaptic plasticity and cogni-
ticoid receptor-dependent histone modifications and the tion (Vaynman et al. 2003). Therefore, exercise can regu-
N-methyl-D-aspartate (NMDA)/extracellular signal-regulated late brain function and plasticity to play a neuroprotective
kinase/mitogen- and stress-activated kinase signaling role via chromatin remodeling of the Bdnf gene.
J. Liang et al.: Exercise promotes brain remodeling 3

Figure 1: The mechanisms of physical exercise on neuroplasticity promotion.


(A) The influence of epigenetics, neurogenesis, and neurotrophins on brain remodeling. (B) Peripheral metabolic and hormonal substances
involved in brain remodeling. HAT, histone acetyltransferase; HDAC, histone deacetylase; Bdnf/BDNF, brain-derived neurotrophic factor;
MiRNA, microRNA; IGF-1, insulin-like growth factor 1; Gpld1, glycosylphosphatidylinositol phospholipase D1; CTSB, cathepsin B; VEGF,
vascular endothelial growth factor; FNDC5, fibronectin type III domain-containing protein 5; AMPK, AMP-activated protein kinase; mTOR,
mammalian target of rapamycin; Nrf2, Nuclear Factor E2-Related Factor 2; DCX, doublecortin; MCT, monocarboxylate transporter; Flk-1, fetal
liver kinase 1; TrkB, tyrosine kinase receptor B; LTP, long-term potentiation.

miRNAs 22 nucleotides encoded by endogenous genes (Tammen


et al. 2013). After physical exercise, the levels of miR-129-1-
miRNAs have been reported to play an important role in 3p, miR-144-5p, and miR-10b-5p are transiently increased,
posttranscriptional gene regulation, which are a type of whereas that of miR-708-5p is decreased (Fernandes et al.
noncoding single-stranded RNA molecule with a length of 2018). Studies have shown that miR-129-1-3p is related to
4 J. Liang et al.: Exercise promotes brain remodeling

cell proliferation, whereas miR-144-5p, miR-10b-5p, and brain (Ismail et al. 2017). However, some studies show that
miR-708-5p are involved in neurological diseases such as neuroplasticity can be enhanced after adult brain injury
depression, AD, and PD (Forstner et al. 2015; Hoss et al. and may be stimulated by physical exercise or functional
2016; Mo et al. 2017; Satoh et al. 2015; Wang et al. 2015). electrical stimulation (Figure 1A) (Cassilhas et al. 2016;
Some miRNAs such as miRNA-7, miRNA-9, and miRNA-34a Cramer et al. 2011; Hara 2015).
are significantly increased in patients with AD, whereas
miRNA-132 is increased in the SAMP8 mouse mode (Dong
et al. 2018; Zhao et al. 2015). Furthermore, abnormal Synaptic plasticity
miRNA gene expression is involved in the pathogenesis of
SAMP8 mice (Cheng et al. 2013). Interestingly, exercise can Synaptic plasticity includes short-term synaptic plasticity
inhibit the high expression levels of miRNA-132 and, and long-term synaptic plasticity (Citri and Malenka 2008).
thereby, improve the cognition in the SAMP8 model (Dong Exercise increases the synaptic strength of the perforant
et al. 2018). pathway connecting the entorhinal cortex with the dentate
It has been reported that physical exercise can promote gyrus, which can enhance long-term potentiation (LTP)
adult hippocampal neurogenesis, and this effect may be (van Praag et al. 1999a,b). In age-related neurodegenera-
used as a preventive measure for age-related diseases tive diseases, the decline of cognitive function is accom-
(Farioli-Vecchioli et al. 2014; Kronenberg et al. 2006; panied by a decrease in hippocampal neurogenesis (Ma
Overall et al. 2013). The study by Pons-Espinal et al. (2019) et al. 2017). The main pathological features of AD are the
discovered that exercise can promote the proliferation of formation of neurofibrillary tangles and amyloid-β (Aβ)
neural precursor cells through the downregulation of plaques (DeTure and Dickson 2019). Aβ1–42 can suppress
miRNA 135a-5p, indicating that this miRNA may be a novel LTP in the dentate gyrus through NMDA receptor signal
target for therapeutic strategies in pathological brain ag- transduction (Babri et al. 2012). BDNF-mediated excitatory
ing. In addition, Mojtahedi et al. (2013) showed that the synaptic regulation is reported to be involved in LTP. At the
level of miRNA-124 is significantly increased in both low- level of post-synaptic regulation, BDNF-tyrosine kinase
and high-intensity exercise. miRNA-124, a specific miRNA receptor B (TrkB) signal induces NMDA phosphorylation
that is highly conserved, has a higher content in adult and increases the opening of ion channels; the influx of
mammalian brain tissue (Lagos-Quintana et al. 2002; Lewis calcium ions can activate CaMKII to maintain enhanced
et al. 2005), and is associated with the transformation of synaptic efficiency (Leal et al. 2017). In a rat model of AD,
neurons from proliferation to differentiation (Krichevsky exercise can protect synaptic transmission and LTP in the
et al. 2003; Krichevsky et al. 2006; Smirnova et al. 2005). dentate gyrus and normalize the levels of synaptic
However, another study has indicated that exercise may plasticity-related molecules like CaMKII, calcineurin, and
downregulate miRNA-124 levels and upregulate hippo- BDNF (Dao et al. 2015). Thus, exercise can prevent
campal glucocorticoid receptor expression, leading to a AD-induced negative effects on dentate gyrus LTP and
positive effect on stress resilience in single-housed mice AD-related pathways. Farmer et al. (2004) showed that
(Pan-Vazquez et al. 2015). These findings suggest that exercise can significantly increase the transcription levels
exercise-induced regulation of miRNAs may be involved in of the NR2B subtype of the NMDA receptor, as well as BDNF
neurological diseases. and glutamate receptor-5 in the dentate gyrus. This
observation may be related to the phenomenon that exer-
cise can reduce the LTP threshold and promote neuro-
genesis (Farmer et al. 2004). Exercise can also reduce the
Exercise benefits neuroprotection load of argentophilic senile plaques, inflammatory re-
by enhancing neuroplasticity sponses, and mitochondrial dysfunction, leading to a
reduction in cognitive impairment and enhanced hippo-
Neuroplasticity refers to the ability of the nervous system to campal neurogenesis in AD model mice (Kim et al. 2019). In
initiate adaptive responses by improving its structure, a vascular dementia rat model, exercise was found to in-
function, and connection following stimulation (von crease the level of synapsin I, postsynaptic density-95,
Bernhardi et al. 2017). Neuroplasticity involves various microtubule-associated proteins, and tau protein in the
mechanisms such as synaptic plasticity, dendritic remod- hippocampus (Lin et al. 2015). In the same study, exercise
eling, and neurogenesis (Amtul and Atta Ur 2015; Ismail could also prevent the loss of neuronal dendrites and
et al. 2017). It has been reported that the developing brain neurons in the hippocampal CA1 and CA2 zones, enhance
has more patterns of neuroplasticity compared to the adult the growth of dendrites, and improve cognition (Lin et al.
J. Liang et al.: Exercise promotes brain remodeling 5

2015). Furthermore, researchers also illustrate that invol- (Ghalandari-Shamami et al. 2019) found that exposure to
untary exercise induced by functional electrical stimula- exercise can improve behavioral and morphological im-
tions has more protective effects on cognition compared to pairments induced by stress. They demonstrated that ex-
forced and voluntary exercise. ercise can reverse the deficits regarding dendritic length
and branching in the prefrontal cortex. Another study
showed that the treadmill exercise in a rat model of stroke
Dendritic remodeling can improve the neurological deficits induced by ischemia-
reperfusion injury through enhancement of dendritic and
Dendritic morphology is a critical factor for the normal synaptic plasticity, which may be due to the upregulation
functioning of the nervous system and shows great di- of the caveolin-1/VEGF signal transduction pathway (Xie
versity among different types of neurons (Ascoli 2006; et al. 2019). These exercise-induced benefits indicate that
Bozelos et al. 2016). The complexity of the dendritic the promotion of dendritic remodeling can be used as a
morphology directly affects the function of neural tissues therapeutic target in some neurological diseases to
(Chklovskii 2004; Spruston 2008). Dendritic remodeling improve the outcome in affected patients and support
can be observed under physiological conditions and medication effects.
following neuropathological stimuli, indicating that den-
drites are dynamic structures (Bozelos et al. 2016).
The degeneration of dendrites and loss of synapses Adult hippocampal neurogenesis
may be one of the mechanisms underlying brain impair-
ment after neurological disorders, such as PD, AD, Not long ago, it was generally believed that neurons cannot
depression, and other brain injuries (Qiao et al. 2016; Reza- regenerate following the physiological or pathological
Zaldivar et al. 2020; Xiong et al. 2019). Recent studies have death of a neuron. However, the discovery of neural stem
shown that exercise can enhance the dendritic complexity cells raised the possibility of plasticity and post-injury
in various subregions of the hippocampus including the repair of central nervous tissue (Altman 1962; Altman and
dentate gyrus, cornu ammonis, and entorhinal cortex Das 1965; Bond et al. 2015; Martínez-Cerdeño and Noctor
(Eadie et al. 2005; Redila and Christie 2006; Stranahan 2018). Neural stem cells, having the characteristics of self-
et al. 2007). In patients with PD, the loss of dopaminergic renewal and pluripotency, exist in the hippocampal sub-
neurons in the substantia nigra and the degeneration of the granular zone and the subventricular zone of the adult
substantia nigra-striatum pathway causes a dysfunction of mammalian central nervous system (Bond et al. 2015; Li
the cortical-basal ganglia loop. PD patients present et al. 2019).
symptoms related to a hypodopaminergic and relative Adult hippocampal neurogenesis takes place in the
hypercholinergic state (Kalia and Lang 2015). In a mouse hippocampal subgranular zone, which is the process of
model of PD, the density of dendritic spines containing neural progenitor cells renewing and regenerating neurons
dopamine receptors is reduced but exercise can prevent the in the hippocampus (Abbott and Nigussie 2020; Eriksson
density loss of dendritic spines in striatal medium spiny et al. 1998). Exercise can enhance cell proliferation, sur-
neurons and upregulate the expression of the postsynaptic vival, and neuronal differentiation in the hippocampus of
density-95 protein, leading to symptom improvements in adult mice, leading to an increased hippocampal storage
PD mice (Toy et al. 2014). capacity (van Praag et al. 1999a,b). Conversely, aging-
Similarly, evidence indicates that stress-induced related cognitive decline may be due to decreased neuro-
reduction in neurogenesis, dendritic atrophy, and loss of genesis in the dentate gyrus (Bizon et al. 2004; Kemper-
spines in hippocampal and prefrontal cortex neurons mann et al. 1998). van Praag et al. (2005) found that the
are involved in the pathophysiology of depression numbers of bromodeoxyuridine (BrdU)-positive cells were
(Qiao et al. 2016; Yau et al. 2011). Using an animal model of increased in both young and old groups after wheel
corticosterone-induced stress, Yau et al. (2011) demon- running, and more cells tended to become neurons in the
strated that the inhibited hippocampal neurogenesis and exercise group compared with age-matched controls (van
diminished spine density were reversed by physical exer- Praag et al. 1999a,b). Additionally, enhanced neurogenesis
cise. Wheel-running can enhance hippocampal neuro- stimulated by increased exercise levels or enriched envi-
genesis, increase dendritic length, and recover spine ronment did not affect the percentage of glial cells in either
density, leading to improvements in spatial memory and age group (Kempermann et al. 1998; van Praag et al. 2005).
depressive behavior in a rat stress model (Yau et al. 2011). Exercise can also promote hippocampal neurogenesis in
In line with these results, Ghalandari-Shamami et al. an animal model of corticosterone-induced stress, leading
6 J. Liang et al.: Exercise promotes brain remodeling

to improvements in spatial memory and depression-like found differences were dependent on the brain region.
symptoms (Yau et al. 2011). Moreover, exercise can reverse After 10 min of exercise, the CBF of the gray matter was
the suppression of cell proliferation in the subventricular decreased, but this change was not significant after 40 min,
zone (Lee et al. 2016). Therefore, physical exercise can be whereas an increase in CBF was detected in the white
regarded as a therapy for stress-related disorders. matter (MacIntosh et al. 2014). This difference may reflect
Exercise can promote the proliferation and differenti- that physical exercise has selective acute effects on CBF
ation of neural progenitor cells in the hippocampus by due to region-specific reactions of blood vessels to
activating extracellular signal-regulated kinases pathways exercise-induced alterations. Besides, neurogenesis is
in a rat model of stroke (Liu et al. 2018). It has been reported associated with angiogenesis, and angiogenesis is associ-
that voluntary exercise can reverse the reduction of cell ated with the microvascular blood volume (Aronen et al.
proliferation and neurogenesis in adults following prena- 2000; Louissaint et al. 2002; Palmer et al. 2000). Thus, the
tal ethanol exposure (Redila et al. 2006). However, the increase of CBF may indicate there is a relationship on
underlying mechanism by which exercise reverses cell exercise-induced angiogenesis and neurogenesis. Howev-
proliferation and neurogenesis after injury remains to be er, a multicenter single-blind randomized controlled trial
elucidated. Farioli-Vecchioli et al. (2014) demonstrated did not confirm that the CBF is changed by physical exer-
that running can shorten the cell cycle length, especially in cise over 16 weeks. The exercise-induced cardiorespiratory
proliferating neural progenitor cells of the dentate gyrus. fitness was not reflected in CBF alterations. This may be
They found that in mice with a silenced antiproliferative due to the 16-week exercise for this relatively small sample
gene Btg1, running reversed the excessive proliferation of is too short for MRI to detect the effect (van der Kleij et al.
the dentate gyrus stem cell pool due to weakened inhibi- 2018).
tion and increased the cell proliferation and neurogenesis In summary, exercise can promote hippocampal neu-
in the adult dentate gyrus. Exercise selectively affected the rogenesis by protecting the newly generated cells, and
cell cycle of specific neurogenic differentiation 1 (Neu- regulating CBF. Study shows that hippocampal neuro-
roD1)-positive progenitor cells, shortened the cycle of cells genesis may persist during the whole life span (Eriksson
positive for glial fibrillary acidic protein and sex- et al. 1998). Boldrini et al. (Boldrini et al. 2018) assessed
determining region Y related HMG box 2(Sox2), as well as autoptic hippocampal specimens from individuals without
NeuroD1-positive progenitor cells, and reactivated the cognition impairment aged 14–79 years. They found that
plasticity of neural stem cells (Farioli-Vecchioli et al. 2014). neural progenitor cells were present in the dentate gyrus
Their study illustrates that running-induced proliferation even of elderly people. Cells with the ability to differentiate
reversal is associated with cell cycle dynamics and pro- and proliferate existed in the dentate gyrus of the elderly,
vides the first evidence that a shortening of the S phase is suggesting that aging may not lead to a functional decline
the main target for the running-induced enhancement of of neurons. However, in the dentate gyrus, the number of
neurogenesis (Farioli-Vecchioli et al. 2014). It seems that cells with proliferative potential declines dramatically in
regulation of cell cycle dynamics maybe the fundamental the first year after birth. The average density of prolifer-
effect of running-induced adult neurogenesis in the hip- ating neurons is about 1618/mm2 after birth and reduced to
pocampus (Farioli-Vecchioli et al. 2014; Farioli-Vecchioli 0.14% of this value at the age of 13 years. By adulthood,
and Tirone 2015). those cells have completely disappeared (Sorrells et al.
Furthermore, exercise-induced cardiorespiratory 2018). Therefore, the existence of hippocampal neuro-
fitness may reflect in cerebral blood flow (CBF) alterations, genesis in humans and other species demands more
leading to neurogenesis. Using arterial spin-labeling research to confirm it.
magnetic resonance imaging, Binnewijzend et al. (Binne-
wijzend et al. 2013) showed that the CBF is reduced in pa-
tients with mild cognitive impairment or AD. In mice,
exercise mainly increases the CBF in the dentate gyrus,
Circulating factors on brain
which may be related to the increase in neurogenesis remodeling
(Pereira et al. 2007). Meanwhile, MacIntosh et al. (MacIn-
tosh et al. 2014) showed that cerebral perfusion is robustly BDNF
affected after a single 1-h aerobic exercise for healthy
adults. Using a voxel-wise analysis, they demonstrated Physical exercise can improve temporal lobe function, in-
that the left medial postcentral gyrus presented an elevated crease serum BDNF levels, and participate in exercise-
perfusion level after 40 min of aerobic exercise, and they induced changes in human cognitive function. This
J. Liang et al.: Exercise promotes brain remodeling 7

suggests that the exercise-induced upregulation of this (Wu et al. 2011). Nam at al (Nam et al. 2014). showed in an
neurotrophin may be important for neuroprotection animal model of D-galactose-induced aging that the num-
(Griffin et al. 2011). Depressive disorder is a common ber of Ki67-/doublecortin-positive cells is decreased after D-
chronic psychiatric disease characterized by mood galactose treatment, but treadmill exercises can signifi-
depression. According to recent statistics, 264 million cantly increase their number. They also found that the
people are affected by this disorder worldwide (Collabo- upregulation of BDNF and pCREB expression can reverse
rators 2018). Serotonin (5-HT) is involved in mood regula- the reduction in neural stem cell numbers, cell prolifera-
tion, and a decrease in its secretion level and activity is tion, and neural differentiation induced by D-galactose
related to the occurrence of depression (Dell’Osso et al. administration. In an animal model of ischemia, long-time
2016). Decreased 5-HT levels can cause depression (Kurdi treadmill exercise enhances cell proliferation, promotes
and Flora 2019), and the 5-HT transporter (SERT) is the neuroblast differentiation, and significantly increases
main target of various antidepressant drugs. Physical ex- BDNF levels in the hippocampus (Ahn et al. 2016). Recent
ercise can increase 5-HT levels of raphe and up-regulate the evidence revealed that BDNF is associated with the regu-
expression of 5-HT transporter and 5-HT receptor 1A in the lation of synaptic plasticity in pyramidal neurons and
cortex and hippocampus (Pietrelli et al. 2018), leading to granule cells and that BDNF can enhance dendritic spine
cognition improvements. BDNF-dependent hippocampal density and LTP in hippocampal neurons (De Vincenti et al.
neurogenesis following exercise also requires serotonin 2019; von Bohlen und Halbach and von Bohlen und Hal-
regulation (Pietrelli et al. 2018). BDNF can promote the bach 2018). Furthermore, exercise can reverse the loss of
maturation and sprouting of 5-HT neurons, and increased dopaminergic neuron and improve the motor deficits,
activity of 5-HT neurons may regulate neuroplasticity which were induced by peripheral lipopolysaccharide
(Klempin et al. 2013). Physical exercise can significantly (LPS) injection. The researchers found that the LPS com-
increase hippocampus BDNF level (Uysal et al. 2015). The bined with infusion of recombinant-BDNF group showed
relationship of BDNF and 5-HT system is bidirectional and that substantia nigra pars compacta had more tyrosine
interacted through an auto/paracrine feedback loop hydroxylase-positive cells, which indicated BDNF can
(Martinowich and Lu 2008). Serotonergic neurons in the protect the loss of dopaminergic neuron induced by LPS
dorsal and median raphe co-express BDNF and its receptor (Wu et al. 2011). Therefore, BDNF plays a pivotal role in
TrkB (Madhav et al. 2001). In addition, BDNF can be exercise-induced neuroplasticity.
transported retrogradely from 5-HT terminals of the stria-
tum and hippocampus to neuronal somata of the raphe
nucleus to exert physiological roles (Anderson et al. 1995). IGF-1
Evidences showed that BDNF can promote the maturation
and sprouting of 5-HT neuron, which may lead to the IGF is a versatile hormone, of which 95% is produced by the
enhancement of 5-HT neurons functions to regulate the liver. It can also be synthesized in the cartilage, muscle,
neuroplasticity (Mamounas et al. 1995; Martinowich and spinal cord, and other tissues. IGF-1 can promote cell
Lu 2008; Mattson et al. 2004; Pietrelli et al. 2018). BDNF mitosis, differentiation, proliferation, and inhibit cell
also can enhance the expression of serotonergic markers in apoptosis (Halmos and Suba 2019). Plasma IGF levels
raphe neurons (Martinowich and Lu 2008). Djalali et al. change with age, they peak at adolescence and then
(Djalali et al. 2005) demonstrated that BDNF may indirectly decrease gradually with age. The decrease in intracranial
affect the serotonergic system via the regulation of S100β vascular density in old mice is accompanied by the
and myelin basic protein in glial cells. Conversely, 5-HT decrease in growth hormone and IGF-1 in the plasma
can also be involved in neuroplasticity mediated by BDNF (Sonntag et al. 1997).
(Pietrelli et al. 2018). The activation of serotonin receptors Exercise induces the upregulation of IGF-1 (Trejo et al.
combined with cAMP production and CREB activation can 2001), whereas blocking of the IGF-1 receptor (IGF-1R) can
induce Bdnf gene transcription (Mattson et al. 2004). eliminate exercise-induced enhancements of Bdnf gene
Besides of the effects on serotonergic neurons, it has expression, as well as synapsin I, phosphorylated-CaMKII,
been reported that BNDF contributes to the differentiation and phosphorylated-mitogen activated protein kinase
and survival of neuronal progenitor cells and the prolifer- (MAPK) II pathways (Ding et al. 2006a,b). And a previous
ation of neural stem cells, and can also promote the sur- study showed that the activation of CaMKII can maintain
vival of progenitor cells (Lee et al. 2002; Sairanen et al. the BDNF-mediated excitatory synaptic regulation during
2005). And exercise can also protect the substantia nigra LTP (Leal et al. 2017). The CREB was an important molec-
dopaminergic neurons through BDNF signaling pathway ular on the transcription of essential prosurvival genes. The
8 J. Liang et al.: Exercise promotes brain remodeling

Ras–MAPK signal pathway, which can be activated by the VEGF


phosphorylated-TrkB, was combined with the activation
CREB (Reichardt 2006). In vivo, BDNF-induced LTP of VEGF, a homodimeric glycoprotein, can be induced under
dentate gyrus was depended on the activation ERK coupled hypoxia conditions. As an important factor to induce
to CREB (Ying et al. 2002). BDNF has powerful effects on angiogenesis, VEGF promotes vascular growth and
synapsin I during exercise, and the interactions among remodeling (Carmeliet 2005). Exercise increases VEGF
CAMKII, MAPK, NMDA receptors, and BDNF can signifi- levels and angiogenesis in aged rats (Ding et al. 2006a,b).
cantly regulate exercise-induced neuroplasticity (Vayn- VEGF expression is induced by exercise and may be
man et al. 2004; Vaynman et al. 2003). Therefore, the IGF-1/ involved in the regulation of hippocampal blood flow and
IGF-1R pathway might be involved in the regulation of neurogenesis. In humans, the inhibition of the VEGF signal
CaMKII, MAPKII, synapsin I, and BDNF, thereby modu- transduction pathway is related to anxiety and depression
lating neuroplasticity (Ding et al. 2006a,b; Dyer et al. 2016; (Gormanns et al. 2011).
Vaynman et al. 2003). Circulating VEGF was increased after exercise (Cotman
The IGF-1R can be expressed by the neural stem cells et al. 2007), which can cross the blood-brain barrier,
and all neural cells over the entire life span (Baron-Van selectively act on specific areas of the brain, and promote
Evercooren et al. 1991; Popken et al. 2005). The IGF-1/ hippocampal neurogenesis (Fabel et al. 2003; Trejo et al.
IGF-1R signal can be activated by PI3K-Akt-mTOR kinase 2001). In a murine middle cerebral artery occlusion (MCAO)
and Ras-Raf-MAP pathways (Sun and D’Ercole 2006; Ye model, the numbers of BDNF- and BrdU-/NeuN-, BrdU-/
and D’Ercole 2006), which regulates neuronal precursor synapsin I- and CYFIP1-positive cells were significantly
proliferation, and neuronal differentiation (O’Kusky et al. increased in the MCAO exercise group, whereas the
2000; Zhang et al. 2014). In treadmill mouse exercise administration of a caveolin-1 inhibitor to this MCAO ex-
model (Chen et al. 2019a,b), the mTOR signaling pathway ercise group significantly reduced the numbers of these
can be activated to enhance the formation of cortical cells that are related to neurogenesis and neuroplasticity
synapses after exercise, and both in vivo and in vitro ex- (Chen et al. 2019a,b). Moreover, the caveolin-1 inhibitor
periments of exercise can enhance the postsynaptic group showed more severe morphological alterations of
excitability and neuronal activity of layer five pyramidal cortical neurons (Chen et al. 2019a,b), and the upregulation
neurons in the motor cortex. These authors also found of the caveolin-1/VEGF signal transduction pathway can
that exercise promotes axonal myelination and dendritic enhance dendritic and synaptic plasticity (Xie et al. 2019).
spine formation. Conversely, those positive enhance- Depression-like behavior, reduction of hippocampal
ments can be eliminated by rapamycin inhibition (Chen vascular density and neurogenesis under chronic stress
et al. 2019a,b). Therefore, the mTOR/IGF-1/IGF-1R stimulations can be improved through regular exercise and
pathway plays an important role in exercise-induced the newly generated neurons survival can also increase,
brain remodeling. The Ras-related GTPase RIT1 may be whereas the VEGF receptor fetal liver kinase one inhibitor
the key downstream component of the IGF-1 signaling exercise group eliminated those exercise-induced im-
pathway to enhance exercise-induced hippocampal provements (Kiuchi et al. 2012). Therefore, exercise-
neurogenesis, because IGF-1 can regulate Sox2 activation induced neurogenesis and angiogenesis, and anti-
and neuronal differentiation via RIT1. This IGF- depression may be partly depended on VEGF signal.
1-mediated response promotes pro-neuronal Ascl1 and
Neurod1 gene expression and regulates Sox2 stability and
transcriptional activity in hippocampal neural progenitor Metabolic and hormonal substances
cells (Mir et al. 2017). Besides, the level of exercise-
induced hippocampal neurogenesis is downregulated in Physical exercise can increase the plasma level of adipo-
RIT1 knockdown mice (Mir et al. 2017). Thus, exercise- nectin (de Carvalho et al. 2018). Adiponectin, which is an
induced hippocampal neurogenesis, mediated by IGF-1, adipocyte-secreted hormone, is known as an important
is associated with the RIT1-Sox2 signaling cascade. protein related to glucose and fat metabolism. Adiponectin
In another study, researchers found that exercise upre- can relieve metabolic disorders, regulate inflammation and
gulates the expression of proteasomes through the oxidative stress, and promote regeneration (Abou-Samra
IGF-1/nuclear factor erythroid 2-related factor pathway, et al. 2020). Furthermore, adiponectin has neuroprotective
suggesting that exercise may promote hippocampal effects. Voluntary running rescues impaired hippocam-
neurogenesis through effects involving hippocampal pal neurogenesis in a diabetic mouse model, but voluntary
proteasomes (Niu et al. 2019). exercise cannot restore neurogenesis in diabetic
J. Liang et al.: Exercise promotes brain remodeling 9

adiponectin knockdown mice (Yau et al. 2018). Yau et al. NeuN+/BrdU+ cells in the dorsal gyrus are higher compared
(2014) revealed that the enhanced hippocampal neuro- to phosphate buffered saline injection controls. Interest-
genesis and antidepressant effects of adiponectin may be ingly, lactate-induced neurogenesis increases the survival
associated with adiponectin receptor 1-mediated activation of newly generated neurons (Lev-Vachnish et al. 2019),
of the AMP-activated protein kinase (Yau et al. 2014). whereas exercise is not only associated with neuronal
Therefore, adiponectin is a vital mediator of exercise- proliferation, but also neuronal maturation (Ma et al. 2017).
induced neuroprotection. Lactate can enter neurons through the monocarboxylate
In mice and humans, exercise induces in stimulated transporter (MCT)-1 and MCT-2 or activate them via the
muscles the gene expression of fibronectin type III domain- hydroxycarboxylic acid receptor 1. Inhibition of MCT-2 can
containing protein 5 (FNDC5), leading to irisin increases suppress lactate-induced neurogenesis, and the expres-
(Bostrom et al. 2012). Irisin, a circulating hormone pro- sion of MCT-2 on mature neurons is a potential target for
duced after exercise, regulates energy metabolism. Irisin is lactate-mediated metabolic and signal transduction (Lev-
involved in neuroplasticity and memory improvement Vachnish et al. 2019). It has been suggested that lactate
(Lourenco et al. 2019). The levels of FNDC5/irisin are may regulate NMDA receptor activity via MCT-2 to enhance
decreased in the hippocampus and cerebrospinal fluid of neuroplasticity (Yang et al. 2014). Therefore, the lactate
mouse models of AD and AD patients. Blocking FNDC5/ released after exercise can promote the function of neu-
irisin can cause LTP and memory impairments, whereas rotrophic factors and enhance neurogenesis. However, it
increasing FNDC5/irisin can protect synaptic plasticity and has been reported that high lactate levels can be detected
memory function. Upregulation of FNDC5/irisin levels in in the cerebrospinal fluid of AD patients, suggesting that
the hippocampus via peripheral adenovirus delivery of lactate may be a risk factor for cerebral amyloidosis (Red-
FNDC5 prevented Aβ-induced memory impairments jems-Bennani et al. 1998). In another study, the β-hy-
(Lourenco et al. 2019). Therefore, exercise can increase the droxybutyric acid level was found to be upregulated after
level of FNDC5/irisin, which in turn can improve neuro- long-term exercise. β-Hydroxybutyric acid can activate the
plasticity and cognition. Bdnf promoter I, leading to BDNF transcription, and in-
It has been reported that exercise can enhance the crease the levels of TrkB receptor-dependent neurotrans-
expression of plasma cathepsin B (CTSB), leading to in- mitters (Sleiman et al. 2016). Therefore, exercise may
creases in doublecortin and BDNF expression in mice, modulate the metabolism, thereby regulating the expres-
rhesus monkeys, and humans. Plasma CTSB levels are sion of lactate and β-hydroxybutyric acid, which is bene-
correlated with memory, and spatial memory and hippo- ficial to brain health (El Hayek et al. 2019; Sleiman et al.
campal neurogenesis are inhibited in CTSB knockout mice. 2016).
Therefore, increased CTSB levels induced by exercise may These findings implicate that physical exercise can
serve as a mediator of brain remodeling (Moon et al. 2016). enhance hippocampal neurogenesis by regulating the
Glycosylphosphatidylinositol phospholipase D1 (Gpld1), levels of adiponectin, FNDC5/irisin, CTSB, Gpld1, lactate
which is related to high-density lipoprotein, is abundant in and β-hydroxybutyric acid (Figure 1B). But how exercise-
mammalian serum (Deeg et al. 2001). High levels of Gpld1 induced peripheral effects affect brain remodeling requires
are synthesized by the liver of mice following exercise, and more research. Additionally, Table 1 briefly summarizes
these elevated Gpld1 levels promote hippocampal neuro- the independent variables on exercise-induced effects
genesis and improve cognition in aging mice. Besides, regarding brain remodeling.
Gpld1 levels are increased in the blood of healthy elderly
people who regularly exercise (Horowitz et al. 2020). Hor-
owitz et al. (2020) suggested that Gpld1 can stimulate Conclusions
hippocampal neurogenesis and improves cognition by
cleaving the GPI-anchored substrate and triggering a In summary, physical exercise can delay or even reverse
downstream signaling cascade. the effects of aging on the brain, improve cognitive decline,
Furthermore, it is well-known that lactate is produced and reduce the vulnerability to some neurological dis-
after exercise (Wahl et al. 2018). It can cross the blood-brain eases. Exercise can reduce the pathological damage on AD,
barrier to upregulate BDNF expression in the brain and PD, depression and stroke, leading to brain rehabilitation.
activate the TrkB signal transduction pathway (El Hayek The regulation of neurotrophic factors, such as BDNF,
et al. 2019). Lev-Vachnish et al. (Lev-Vachnish et al. 2019) IGF-1, and VEGF, plays a pivotal role in exercise-induced
injected mice with 1.75 g/kg L-lactate to mimic exercise- brain remodeling. Exercise also can promote BDNF
induced lactate levels. They found that the numbers of expression, facilitate chromatin remodeling of the Bdnf
10 J. Liang et al.: Exercise promotes brain remodeling

Table : Independent variables on exercise-induced effects on Future work should clarify the relationships between
brain remodeling. changes in the brain and peripheral levels of these sub-
stances following exercise. Therefore, physical exercise
Independent Mechanisms Effects
can not only promote fitness but also enhance brain
variables
remodeling by regulating epigenetic information, pro-
Epigenetics Histone modification ↑ acetylation
moting neuroplasticity, and modulating the expression of
factors ↑ phospho-acetylation
neurotrophic factors.
DNA methylation and ↓ bdnf promoter IV CpG
Chromatin remodeling methylation
miRNAs ↓ miRNA- ↑ Author contributions: All the authors have accepted
miRNA- responsibility for the entire content of this submitted
Neuroplasticity Synaptic plasticity ↑ SYN ↑ PSD- ↑ MAP- manuscript and approved submission.
↑ tau protein
Research funding: Our work was supported by the National
↑ synaptic transmission
↑ LTP Science Foundation of China (81630038, 81971433,
Dendritic remodeling ↑ dendritic length ↑ 81971428, 81771634, 81842011, 81330016, 81801629), the
spine density National Key R&D Program of China (2017YFA 0104200),
↑ neurons density the grants from the Ministry of Education of China
Hippocampal ↑ NPCs proliferation and
(IRT0935), the grants from the Science and Technology
neurogenesis differentiation
Bureau of Sichuan Province (2016TD0002), the grant from
↑ CBF
Trophic factors Neurotrophic factors ↑ BDNF ↓ p receptor the clinical discipline program (Neonatology) from the
↑ IGF- ↑ BrdU-labeled Ministry of Health of China (1311200003303), and the
cells ↑ HN Fundamental Research Funds for the Central Universities.
↑ VEGF ↑ angiogenesis Conflict of interest statement: The authors declare that the
Circulating Hormonal substances ↑ Adiponectin ↑FNDC/
research was conducted in the absence of any commercial
factors irisin
↑ CTSB ↑ doublecortin or financial relationships that could be construed as a
Metabolic substances ↑ Gpld ↑ lactate potential conflict of interest.
↑ β-hydroxybutyric acid

↑, increased. ↓, decreased. BDNF, brain-derived neurotrophic factor;


SYN, synapsin-I; PSD-, postsynaptic density-; MAP-, References
microtubule associated protein ; AD, Alzheimer’s disease; LTP, long-
term potentiation; NPCs, neural progenitor cells; CBF, cerebral blood Abbott, L.C. and Nigussie, F. (2020). Adult neurogenesis in the
flow; IGF-, insulin-like growth factor ; BrdU, bromodeoxyuridine; mammalian dentate gyrus. Anat. Histol. Embryol. 49: 3–16.
HN, hippocampal neurogenesis; VEGF, vascular endothelial growth Abou-Samra, M., Selvais, C.M., Dubuisson, N., and Brichard, S.M.
factor; FNDC, fibronectin type III domain-containing protein ; CTSB, (2020). Adiponectin and its mimics on skeletal muscle: insulin
cathepsin B; Gpld, glycosylphosphatidylinositol phospholipase D. sensitizers, fat burners, exercise mimickers, muscling pills … or
everything together? Int. J. Mol. Sci. 21: 2620.
Ahn, J.H., Choi, J.H., Park, J.H., Kim, I.H., Cho, J.H., Lee, J.C., Koo, H.M.,
gene, and alter the methylation status of the Bdnf promoter Hwangbo, G., Yoo, K.Y., Lee, C.H., et al. (2016). Long-term
exercise improves memory deficits via restoration of myelin and
IV. Altered BDNF expression is associated with synapsin
microvessel damage, and enhancement of neurogenesis in the
I, 5-HT, CAMKII, MAPK, and NMDA receptors, which all aged gerbil hippocampus after ischemic stroke. Neurorehabil.
may be involved in exercise-induced neuroplasticity. Neural Repair. 30: 894–905.
IGF-1/IGF-1R upregulation after exercise modulates Ali, I., Conrad, R.J., Verdin, E., and Ott, M. (2018). Lysine acetylation
neuronal precursor proliferation, and neuronal differenti- goes global: from epigenetics to metabolism and therapeutics.
Chem. Rev. 118: 1216–1252.
ation. Exercise can regulate the dendritic morphology via
Altman, J. (1962). Are new neurons formed in the brains of adult
the caveolin-1/VEGF signaling pathway. Exercise-induced
mammals? Science 135: 1127–1128.
effects are involved in the regulation of cell cycle dy- Altman, J. and Das, G.D. (1965). Autoradiographic and histological
namics, leading to the proliferation of neural progenitor evidence of postnatal hippocampal neurogenesis in rats.
cells in the dentate gyrus. Additionally, the regulation of J. Comp. Neurol. 124: 319–335.
histone modifications and miRNAs play a critical role in Amtul, Z. and Atta Ur, R. (2015). Neural plasticity and memory:
molecular mechanism. Rev. Neurosci. 26: 253–268.
brain remodeling after exercises. Furthermore, exercise-
Anderson, K.D., Alderson, R.F., Altar, C.A., DiStefano, P.S., Corcoran,
induced changes in peripheral levels of molecules such as T.L., Lindsay, R.M., and Wiegand, S.J. (1995). Differential
adiponectin, FNDC5/irisin, CTSB, Gpld1, lactate and β-hy- distribution of exogenous BDNF, NGF, and NT-3 in the brain
droxybutyric acid are also related to brain remodeling. corresponds to the relative abundance and distribution of high-
J. Liang et al.: Exercise promotes brain remodeling 11

affinity and low-affinity neurotrophin receptors. J. Comp. Neurol. Chandramohan, Y., Droste, S.K., Arthur, J.S., and Reul, J.M. (2008).
357: 296–317. The forced swimming-induced behavioural immobility response
Aronen, H.J., Pardo, F.S., Kennedy, D.N., Belliveau, J.W., Packard, S.D., involves histone H3 phospho-acetylation and c-Fos induction in
Hsu, D.W., Hochberg, F.H., Fischman, A.J., and Rosen, B.R. dentate gyrus granule neurons via activation of the N-methyl-
(2000). High microvascular blood volume is associated with high D-aspartate/extracellular signal-regulated kinase/mitogen- and
glucose uptake and tumor angiogenesis in human gliomas. Clin. stress-activated kinase signalling pathway. Eur. J. Neurosci. 27:
Cancer Res. 6: 2189–2200. 2701–2713.
Ascoli, G.A. (2006). Mobilizing the base of neuroscience data: the case Chen, W.G., Chang, Q., Lin, Y., Meissner, A., West, A.E., Griffith, E.C.,
of neuronal morphologies. Nat. Rev. Neurosci. 7: 318–324. Jaenisch, R., and Greenberg, M.E. (2003). Derepression of BDNF
Ashpole, N.M., Sanders, J.E., Hodges, E.L., Yan, H., and Sonntag, W.E. transcription involves calcium-dependent phosphorylation of
(2015). Growth hormone, insulin-like growth factor-1 and the MeCP2. Science 302: 885–889.
aging brain. Exp. Gerontol. 68: 76–81. Cheng, X.R., Cui, X.L., Zheng, Y., Zhang, G.R., Li, P., Huang, H., Zhao,
Babri, S., Amani, M., Mohaddes, G., Alihemmati, A., and Ebrahimi, H. Y.Y., Bo, X.C., Wang, S.Q., Zhou, W.X., et al. (2013). Nodes and
(2012). Effect of aggregated β-amyloid (1-42) on synaptic biological processes identified on the basis of network analysis
plasticity of hippocampal dentate gyrus granule cells in vivo. in the brain of the senescence accelerated mice as an
BioImpacts BI 2: 189–194. Alzheimer’s disease animal model. Front. Aging Neurosci. 5: 65.
Baron-Van Evercooren, A., Olichon-Berthe, C., Kowalski, A., Visciano, Chen, Z., Hu, Q., Xie, Q., Wu, S., Pang, Q., Liu, M., Zhao, Y., Tu, F., Liu,
G., and Van Obberghen, E. (1991). Expression of IGF-I and insulin C., and Chen, X. (2019a). Effects of treadmill exercise on motor
receptor genes in the rat central nervous system: a and cognitive function recovery of MCAO mice through the
developmental, regional, and cellular analysis. J. Neurosci. Res. caveolin-1/VEGF signaling pathway in ischemic penumbra.
28: 244–253. Neurochem. Res. 44: 930–946.
Bilang-Bleuel, A., Ulbricht, S., Chandramohan, Y., De Carli, S., Droste, Chen, K., Zheng, Y., Wei, J.-A., Ouyang, H., Huang, X., Zhang, F., Lai,
S.K., and Reul, J.M. (2005). Psychological stress increases C.S.W., Ren, C., So, K.-F., and Zhang, L. (2019b). Exercise training
histone H3 phosphorylation in adult dentate gyrus granule improves motor skill learning via selective activation of mTOR.
neurons: involvement in a glucocorticoid receptor-dependent Sci. Adv. 5: eaaw1888.
behavioural response. Eur. J. Neurosci. 22: 1691–1700. Chklovskii, D.B. (2004). Synaptic connectivity and neuronal
Binnewijzend, M.A., Kuijer, J.P., Benedictus, M.R., van der Flier, W.M., morphology: two sides of the same coin. Neuron 43: 609–617.
Wink, A.M., Wattjes, M.P., van Berckel, B.N., Scheltens, P., and Citri, A. and Malenka, R.C. (2008). Synaptic plasticity: multiple forms,
Barkhof, F. (2013). Cerebral blood flow measured with 3D functions, and mechanisms. Neuropsychopharmacology 33:
pseudocontinuous arterial spin-labeling MR imaging in 18–41.
Alzheimer disease and mild cognitive impairment: a marker for Collins, A., Hill, L.E., Chandramohan, Y., Whitcomb, D., Droste, S.K.,
disease severity. Radiology 267: 221–230. and Reul, J.M. (2009). Exercise improves cognitive responses to
Bizon, J.L., Lee, H.J., and Gallagher, M. (2004). Neurogenesis in a rat psychological stress through enhancement of epigenetic
model of age-related cognitive decline. Aging Cell 3: 227–234. mechanisms and gene expression in the dentate gyrus. PLoS One
Boldrini, M., Fulmore, C.A., Tartt, A.N., Simeon, L.R., Pavlova, I., 4: e4330.
Poposka, V., Rosoklija, G.B., Stankov, A., Arango, V., Dwork, A.J., Cosin-Tomas, M., Alvarez-Lopez, M.J., Sanchez-Roige, S., Lalanza, J.F.,
et al. (2018). Human hippocampal neurogenesis persists Bayod, S., Sanfeliu, C., Pallas, M., Escorihuela, R.M., and
throughout aging. Cell Stem Cell 22: 589–599, e585. Kaliman, P. (2014). Epigenetic alterations in hippocampus of
Bond, A.M., Ming, G.-L., and Song, H. (2015). Adult mammalian neural SAMP8 senescent mice and modulation by voluntary physical
stem cells and neurogenesis: five decades later. Cell Stem Cell exercise. Front. Aging Neurosci. 6: 51.
17: 385–395. Cotman, C.W., Berchtold, N.C., and Christie, L.A. (2007). Exercise
Bostrom, P., Wu, J., Jedrychowski, M.P., Korde, A., Ye, L., Lo, J.C., builds brain health: key roles of growth factor cascades and
Rasbach, K.A., Bostrom, E.A., Choi, J.H., Long, J.Z., et al. (2012). A inflammation. Trends Neurosci. 30: 464–472.
PGC1-α-dependent myokine that drives brown-fat-like Cramer, S.C., Sur, M., Dobkin, B.H., O’Brien, C., Sanger, T.D.,
development of white fat and thermogenesis. Nature 481: Trojanowski, J.Q., Rumsey, J.M., Hicks, R., Cameron, J., Chen, D.,
463–468. et al. (2011). Harnessing neuroplasticity for clinical applications.
Bozelos, P., Stefanou, S.S., Bouloukakis, G., Melachrinos, C., and Brain 134: 1591–1609.
Poirazi, P. (2016). REMOD: a tool for analyzing and remodeling Dao, A.T., Zagaar, M.A., and Alkadhi, K.A. (2015). Moderate treadmill
the dendritic architecture of neural cells. Front. Neuroanat. 9: exercise protects synaptic plasticity of the dentate gyrus and
156. related signaling cascade in a rat model of Alzheimer’s disease.
Burggren, W. (2016). Epigenetic evaluation and new perspectives Mol. Neurobiol. 52: 1067–1076.
inheritance and its role in evolutionary biology: re-evaluation de Carvalho, F.P., Moretto, T.L., Benfato, I.D., Barthichoto, M.,
and new perspectives. Biology 5: 24. Ferreira, S.M., Costa-Júnior, J.M., and de Oliveira, C.A.M. (2018).
Carmeliet, P. (2005). VEGF as a key mediator of angiogenesis in Central and peripheral effects of physical exercise without
cancer. Oncology 69(Suppl. 3): 4–10. weight reduction in obese and lean mice. Biosci. Rep. 38:
Cass, S.P. (2017). Alzheimer’s disease and exercise: a literature BSR20171033.
review. Curr. Sports Med. Rep. 16: 19–22. De Vincenti, A.P., Ríos, A.S., Paratcha, G., and Ledda, F. (2019).
Cassilhas, R.C., Tufik, S., and de Mello, M.T. (2016). Physical exercise, Mechanisms that modulate and diversify BDNF Functions:
neuroplasticity, spatial learning and memory. Cell. Mol. Life Sci. implications for hippocampal synaptic plasticity. Front. Cell.
73: 975–983. Neurosci. 13: 135.
12 J. Liang et al.: Exercise promotes brain remodeling

Deeg, M.A., Bowen, R.F., Williams, M.D., Olson, L.K., Kirk, E.A., and and gene expression in the dentate gyrus of adult male Sprague-
LeBoeuf, R.C. (2001). Increased expression of GPI-specific Dawley rats in vivo. Neuroscience 124: 71–79.
phospholipase D in mouse models of type 1 diabetes. Am. Fernandes, J., Vieira, A.S., Kramer-Soares, J.C., Da Silva, E.A., Lee,
J. Physiol. Endocrinol. Metab. 281: E147–154. K.S., Lopes-Cendes, I., and Arida, R.M. (2018). Hippocampal
Dell’Osso, L., Carmassi, C., Mucci, F., and Marazziti, D. (2016). microRNA-mRNA regulatory network is affected by physical
Depression, serotonin and tryptophan. Curr. Pharm. Des. 22: exercise. Biochim. Biophys. Acta Gen. Subj. 1862: 1711–1720.
949–954. Forstner, A.J., Hofmann, A., Maaser, A., Sumer, S., Khudayberdiev, S.,
DeTure, M.A. and Dickson, D.W. (2019). The neuropathological Mühleisen, T.W., Leber, M., Schulze, T.G., Strohmaier, J.,
diagnosis of Alzheimer’s disease. Mol. Neurodegener. 14: 32. Degenhardt, F., et al. (2015). Genome-wide analysis implicates
Ding, Y.H., Li, J., Zhou, Y., Rafols, J.A., Clark, J.C., and Ding, Y. (2006a). microRNAs and their target genes in the development of bipolar
Cerebral angiogenesis and expression of angiogenic factors in disorder. Transl. Psychiatry 5: e678.
aging rats after exercise. Curr. Neurovasc. Res. 3: 15–23. GBD 2017 Disease and Injury Incidence and Prevalence Collaborators
Ding, Q., Vaynman, S., Akhavan, M., Ying, Z., and Gomez-Pinilla, F. (2018). Global, regional, and national incidence, prevalence, and
(2006b). Insulin-like growth factor I interfaces with brain-derived years lived with disability for 354 diseases and injuries for 195
neurotrophic factor-mediated synaptic plasticity to modulate countries and territories, 1990-2017: a systematic analysis for
aspects of exercise-induced cognitive function. Neuroscience the Global Burden of Disease Study 2017. Lancet 392:
140: 823–833. 1789–1858.
Djalali, S., Höltje, M., Grosse, G., Rothe, T., Stroh, T., Grosse, J., Deng, Ghalandari-Shamami, M., Nourizade, S., Yousefi, B., Vafaei, A.A.,
D.R., Hellweg, R., Grantyn, R., Hörtnagl, H., et al. (2005). Effects of Pakdel, R., and Rashidy-Pour, A. (2019). Beneficial effects of
brain-derived neurotrophic factor (BDNF) on glial cells and physical activity and crocin against adolescent stress induced
serotonergic neurones during development. J. Neurochem. 92: anxiety or depressive-like symptoms and dendritic morphology
616–627. remodeling in prefrontal cortex in adult male rats. Neurochem.
Dong, J., Liu, Y., Zhan, Z., and Wang, X. (2018). MicroRNA-132 is Res. 44: 917–929.
associated with the cognition improvement following voluntary Gomez-Pinilla, F., Zhuang, Y., Feng, J., Ying, Z., and Fan, G. (2011).
exercise in SAMP8 mice. Brain Res. Bull. 140: 80–87. Exercise impacts brain-derived neurotrophic factor plasticity by
Dyer, A.H., Vahdatpour, C., Sanfeliu, A., and Tropea, D. (2016). The role engaging mechanisms of epigenetic regulation. Eur. J. Neurosci.
of insulin-like growth factor 1 (IGF-1) in brain development, 33: 383–390.
maturation and neuroplasticity. Neuroscience 325: 89–99. Gormanns, P., Mueller, N.S., Ditzen, C., Wolf, S., Holsboer, F., and
Eadie, B.D., Redila, V.A., and Christie, B.R. (2005). Voluntary exercise Turck, C.W. (2011). Phenome-transcriptome correlation unravels
alters the cytoarchitecture of the adult dentate gyrus by anxiety and depression related pathways. J. Psychiatr. Res. 45:
increasing cellular proliferation, dendritic complexity, and spine 973–979.
density. J. Comp. Neurol. 486: 39–47. Griffin, E.W., Mullally, S., Foley, C., Warmington, S.A., O’Mara, S.M.,
El Hayek, L., Khalifeh, M., Zibara, V., and Abi Assaad, R. (2019). and Kelly, A.M. (2011). Aerobic exercise improves hippocampal
Lactate mediates the effects of exercise on learning and function and increases BDNF in the serum of young adult males.
memory through SIRT1-dependent activation of hippocampal Physiol. Behav. 104: 934–941.
brain-derived neurotrophic factor (BDNF). J. Neurosci. 39: Halmos, T. and Suba, I. (2019). The physiological role of growth
2369–2382. hormone and insulin-like growth factors. Orv. Hetil. 160:
Elsner, V.R., Lovatel, G.A., Bertoldi, K., Vanzella, C., Santos, F.M., 1774–1783.
Spindler, C., de Almeida, E.F., Nardin, P., and Siqueira, I.R. Hara, Y. (2015). Brain plasticity and rehabilitation in stroke patients.
(2011). Effect of different exercise protocols on histone J. Nippon Med. Sch. 82: 4–13.
acetyltransferases and histone deacetylases activities in rat Horowitz, A.M., Fan, X., Bieri, G., Smith, L.K., Sanchez-Diaz, C.I.,
hippocampus. Neuroscience 192: 580–587. Schroer, A.B., Gontier, G., Casaletto, K.B., Kramer, J.H., Williams,
Eriksson, P.S., Perfilieva, E., Björk-Eriksson, T., Alborn, A.-M., K.E., et al. (2020). Blood factors transfer beneficial effects of
Nordborg, C., Peterson, D.A., and Gage, F.H. (1998). exercise on neurogenesis and cognition to the aged brain.
Neurogenesis in the adult human hippocampus. Nat. Med. 4: Science 369: 167–173.
1313–1317. Hoss, A.G., Labadorf, A., Beach, T.G., Latourelle, J.C., and Myers, R.H.
Fabel, K., Fabel, K., Tam, B., Kaufer, D., Baiker, A., Simmons, N., Kuo, (2016). microRNA Profiles in Parkinson’s disease prefrontal
C.J., and Palmer, T.D. (2003). VEGF is necessary for exercise- cortex. Front. Aging Neurosci. 8: 36.
induced adult hippocampal neurogenesis. Eur. J. Neurosci. 18: Ignacio, Z.M., da Silva, R.S., Plissari, M.E., Quevedo, J., and Reus, G.Z.
2803–2812. (2019). Physical exercise and neuroinflammation in major
Farioli-Vecchioli, S., Mattera, A., Micheli, L., Ceccarelli, M., Leonardi, depressive disorder. Mol. Neurobiol. 56: 8323–8335.
L., Saraulli, D., Costanzi, M., Cestari, V., Rouault, J.P., and Tirone, Ismail, F.Y., Fatemi, A., and Johnston, M.V. (2017). Cerebral plasticity:
F. (2014). Running rescues defective adult neurogenesis by windows of opportunity in the developing brain. Eur. J. Paediatr.
shortening the length of the cell cycle of neural stem and Neurol. 21: 23–48.
progenitor cells. Stem Cells 32: 1968–1982. Jung, M. and Pfeifer, G.P. (2015). Aging and DNA methylation. BMC
Farioli-Vecchioli, S. and Tirone, F. (2015). Control of the cell cycle in Biol. 13: 7.
adult neurogenesis and its relation with physical exercise. Brain Kalia, L.V. and Lang, A.E. (2015). Parkinson’s disease. Lancet 386:
Plast. 1: 41–54. 896–912.
Farmer, J., Zhao, X., van Praag, H., Wodtke, K., Gage, F.H., and Christie, Kashimoto, R.K., Toffoli, L.V., Manfredo, M.H.F., Volpini, V.L., Martins-
B.R. (2004). Effects of voluntary exercise on synaptic plasticity Pinge, M.C., Pelosi, G.G., and Gomes, M.V. (2016). Physical
J. Liang et al.: Exercise promotes brain remodeling 13

exercise affects the epigenetic programming of rat brain and Liu, W., Wu, W., Lin, G., Cheng, J., Zeng, Y., and Shi, Y. (2018). Physical
modulates the adaptive response evoked by repeated restraint exercise promotes proliferation and differentiation of
stress. Behav. Brain Res. 296: 286–289. endogenous neural stem cells via ERK in rats with cerebral
Kempermann, G., Kuhn, H.G., and Gage, F.H. (1998). Experience- infarction. Mol. Med. Rep. 18: 1455–1464.
induced neurogenesis in the senescent dentate gyrus. Louissaint, A., Jr., Rao, S., Leventhal, C., and Goldman, S.A. (2002).
J. Neurosci. 18: 3206–3212. Coordinated interaction of neurogenesis and angiogenesis in the
Kim, D., Cho, J., and Kang, H. (2019). Protective effect of exercise adult songbird brain. Neuron 34: 945–960.
training against the progression of Alzheimer’s disease in 3xTg- Lourenco, M.V., Frozza, R.L., de Freitas, G.B., Zhang, H., Kincheski,
AD mice. Behav. Brain Res. 374: 112105. G.C., Ribeiro, F.C., Goncalves, R.A., Clarke, J.R., Beckman, D.,
Kiuchi, T., Lee, H., and Mikami, T. (2012). Regular exercise cures Staniszewski, A., et al. (2019). Exercise-linked FNDC5/irisin
depression-like behavior via VEGF-Flk-1 signaling in chronically rescues synaptic plasticity and memory defects in Alzheimer’s
stressed mice. Neuroscience 207: 208–217. models. Nat. Med. 25: 165–175.
Klempin, F., Beis, D., Mosienko, V., Kempermann, G., Bader, M., and Ma, C.L., Ma, X.T., Wang, J.J., Liu, H., Chen, Y.F., and Yang, Y.
Alenina, N. (2013). Serotonin is required for exercise-induced (2017). Physical exercise induces hippocampal neurogenesis
adult hippocampal neurogenesis. J. Neurosci. 33: 8270–8275. and prevents cognitive decline. Behav. Brain Res. 317:
Krichevsky, A.M., King, K.S., Donahue, C.P., Khrapko, K., and Kosik, 332–339.
K.S. (2003). A microRNA array reveals extensive regulation of MacIntosh, B.J., Crane, D.E., Sage, M.D., Rajab, A.S., Donahue, M.J.,
microRNAs during brain development. RNA 9: 1274–1281. McIlroy, W.E., and Middleton, L.E. (2014). Impact of a single bout
Krichevsky, A.M., Sonntag, K.-C., Isacson, O., and Kosik, K.S. (2006). of aerobic exercise on regional brain perfusion and activation
Specific microRNAs modulate embryonic stem cell-derived responses in healthy young adults. PLoS One 9: e85163.
neurogenesis. Stem Cells 24: 857–864. Madhav, T.R., Pei, Q., and Zetterström, T.S. (2001). Serotonergic cells
Kronenberg, G., Bick-Sander, A., Bunk, E., Wolf, C., Ehninger, D., and of the rat raphe nuclei express mRNA of tyrosine kinase B (trkB),
Kempermann, G. (2006). Physical exercise prevents age-related the high-affinity receptor for brain derived neurotrophic factor
decline in precursor cell activity in the mouse dentate gyrus. (BDNF). Brain Res. Mol. Brain Res. 93: 56–63.
Neurobiol. Aging 27: 1505–1513. Maejima, H., Kanemura, N., Kokubun, T., Murata, K., and Takayanagi,
Kurdi, F.N. and Flora, R. (2019). Physical exercise increased brain- K. (2018a). Exercise enhances cognitive function and
derived neurotrophic factor in elderly population with neurotrophin expression in the hippocampus accompanied by
depression. Open Access Maced. J. Med. Sci. 7: 2057–2061. changes in epigenetic programming in senescence-accelerated
Kvam, S., Kleppe, C.L., Nordhus, I.H., and Hovland, A. (2016). Exercise mice. Neurosci. Lett. 665: 67–73.
as a treatment for depression: a meta-analysis. J. Affect. Disord. Maejima, H., Ninuma, S., Okuda, A., Inoue, T., and Hayashi, M.
202: 67–86. (2018b). Exercise and low-level GABAA receptor inhibition
Lagos-Quintana, M., Rauhut, R., Yalcin, A., Meyer, J., Lendeckel, W., modulate locomotor activity and the expression of BDNF
and Tuschl, T. (2002). Identification of tissue-specific microRNAs accompanied by changes in epigenetic regulation in the
from mouse. Curr. Biol. 12: 735–739. hippocampus. Neurosci. Lett. 685: 18–23.
Leal, G., Bramham, C.R., and Duarte, C.B. (2017). BDNF and Mamounas, L.A., Blue, M.E., Siuciak, J.A., and Altar, C.A. (1995). Brain-
hippocampal synaptic plasticity. Vitam. Horm. 104: 153–195. derived neurotrophic factor promotes the survival and sprouting
Lee, J., Duan, W., and Mattson, M.P. (2002). Evidence that brain- of serotonergic axons in rat brain. J. Neurosci. 15: 7929–7939.
derived neurotrophic factor is required for basal neurogenesis Martínez-Cerdeño, V. and Noctor, S.C. (2018). Neural progenitor cell
and mediates, in part, the enhancement of neurogenesis by terminology. Front. Neuroanat. 12: 104.
dietary restriction in the hippocampus of adult mice. Martinowich, K., Hattori, D., Wu, H., Fouse, S., He, F., Hu, Y., Fan, G.,
J. Neurochem. 82: 1367–1375. and Sun, Y.E. (2003). DNA methylation-related chromatin
Lee, J.C., Yau, S.Y., Lee, T.M.C., Lau, B.W., and So, K.F. (2016). remodeling in activity-dependent BDNF gene regulation. Science
Voluntary wheel running reverses the decrease in subventricular 302: 890–893.
zone neurogenesis caused by corticosterone. Cell Transplant. 25: Martinowich, K. and Lu, B. (2008). Interaction between BDNF and
1979–1986. serotonin: role in mood disorders. Neuropsychopharmacology
Lev-Vachnish, Y., Cadury, S., Rotter-Maskowitz, A., Feldman, N., 33: 73–83.
Roichman, A., Illouz, T., Varvak, A., Nicola, R., Madar, R., and Mattson, M.P., Maudsley, S., and Martin, B. (2004). BDNF and 5-HT: a
Okun, E. (2019). L-Lactate promotes adult hippocampal dynamic duo in age-related neuronal plasticity and
neurogenesis. Front. Neurosci. 13: 403. neurodegenerative disorders. Trends Neurosci. 27: 589–594.
Lewis, B.P., Burge, C.B., and Bartel, D.P. (2005). Conserved seed Mir, S., Cai, W., Carlson, S.W., Saatman, K.E., and Andres, D.A. (2017).
pairing, often flanked by adenosines, indicates that thousands IGF-1 mediated neurogenesis involves a novel RIT1/Akt/Sox2
of human genes are microRNA targets. Cell 120: 15–20. cascade. Sci. Rep. 7: 3283.
Li, A., Yau, S.Y., Machado, S., Wang, P., Yuan, T.F., and So, K.F. (2019). Mo, M., Xiao, Y., Huang, S., Cen, L., Chen, X., Zhang, L., Luo, Q., Li, S.,
Enhancement of hippocampal plasticity by physical exercise as a Yang, X., Lin, X., and Xu, P. (2017). MicroRNA expressing profiles
polypill for stress and depression: a review. CNS Neurol. Disord. in A53T mutant alpha-synuclein transgenic mice and
Drug Targets 18: 294–306. Parkinsonian. Oncotarget 8: 15–28.
Lin, Y., Dong, J., Yan, T., He, X., Zheng, X., Liang, H., and Sui, M. (2015). Mojtahedi, S., Kordi, M.R., Hosseini, S.E., Omran, S.F., and Soleimani,
Involuntary, forced and voluntary exercises are equally capable M. (2013). Effect of treadmill running on the expression of genes
of inducing hippocampal plasticity and the recovery of cognitive that are involved in neuronal differentiation in the hippocampus
function after stroke. Neurol. Res. 37: 893–901. of adult male rats. Cell Biol. Int. 37: 276–283.
14 J. Liang et al.: Exercise promotes brain remodeling

Moon, H.Y., Becke, A., Berron, D., Becker, B., Sah, N., Benoni, G., Qiao, H., Li, M.-X., Xu, C., Chen, H.-B., An, S.-C., and Ma, X.-M. (2016).
Janke, E., Lubejko, S.T., Greig, N.H., Mattison, J.A., et al. (2016). Dendritic spines in depression: what we learned from animal
Running-induced systemic cathepsin B secretion is associated models. Neural Plast. 2016: 8056370.
with memory function. Cell Metab. 24: 332–340. Redila, V.A. and Christie, B.R. (2006). Exercise-induced changes in
Muñoz, A., Corrêa, C.L., Lopez-Lopez, A., Costa-Besada, M.A., Diaz- dendritic structure and complexity in the adult hippocampal
Ruiz, C., and Labandeira-Garcia, J.L. (2018). Physical exercise dentate gyrus. Neuroscience 137: 1299–1307.
improves aging-related changes in angiotensin, IGF-1, SIRT1, Redila, V.A., Olson, A.K., Swann, S.E., Mohades, G., Webber, A.J.,
SIRT3, and VEGF in the substantia nigra. J. Gerontol. 73: Weinberg, J., and Christie, B.R. (2006). Hippocampal cell
1594–1601. proliferation is reduced following prenatal ethanol exposure but
Nam, S.M., Kim, J.W., Yoo, D.Y., Yim, H.S., Kim, D.W., Choi, J.H., Kim, can be rescued with voluntary exercise. Hippocampus 16:
W., Jung, H.Y., Won, M.H., Hwang, I.K., et al. (2014). Physical 305–311.
exercise ameliorates the reduction of neural stem cell, cell Redjems-Bennani, N., Jeandel, C., Lefebvre, E., Blain, H., Vidailhet, M.,
proliferation and neuroblast differentiation in senescent mice and Guéant, J.L. (1998). Abnormal substrate levels that depend
induced by D-galactose. BMC Neurosci. 15: 116. upon mitochondrial function in cerebrospinal fluid from
Niu, X., Zhao, Y., Yang, N., Zhao, X., Zhang, W., Bai, X., Li, A., Yang, W., Alzheimer patients. Gerontology 44: 300–304.
and Lu, L. (2019). Proteasome activation by insulin-like growth Reichardt, L.F. (2006). Neurotrophin-regulated signalling pathways.
factor-1/nuclear factor erythroid 2-related factor 2 signaling Philos. Trans. R. Soc. Lond. B Biol. Sci. 361: 1545–1564.
promotes exercise-induced neurogenesis. Stem Cells, https:// Reul, J.M. and Chandramohan, Y. (2007). Epigenetic mechanisms in
doi.org/10.1002/stem.3102. stress-related memory formation. Psychoneuroendocrinology
Nosheny, R.L., Insel, P.S., Mattsson, N., Tosun, D., Buckley, S., Truran, 32(Suppl. 1): S21–25.
D., Schuff, N., Aisen, P.S., and Weiner, M.W. (2019). Associations Reul, J.M. and Nutt, D.J. (2008). Glutamate and cortisol – a critical
among amyloid status, age, and longitudinal regional brain confluence in PTSD? J. Psychopharmacol. 22: 469–472.
atrophy in cognitively unimpaired older adults. Neurobiol. Aging Reza-Zaldivar, E.E., Hernández-Sápiens, M.A., Minjarez, B., Gómez-
82: 110–119. Pinedo, U., Sánchez-González, V.J., Márquez-Aguirre, A.L., and
O’Kusky, J.R., Ye, P., and D’Ercole, A.J. (2000). Insulin-like growth Canales-Aguirre, A.A. (2020). Dendritic spine and synaptic
factor-I promotes neurogenesis and synaptogenesis in the plasticity in Alzheimer’s disease: a focus on microRNA. Front. Cell
hippocampal dentate gyrus during postnatal development. Dev. Biol. 8: 255.
J. Neurosci. 20: 8435–8442. Ryu, H., Lee, J., Olofsson, B.A., Mwidau, A., Dedeoglu, A., Escudero,
Overall, R.W., Walker, T.L., Leiter, O., Lenke, S., Ruhwald, S., and M., Flemington, E., Azizkhan-Clifford, J., Ferrante, R.J., and Ratan,
Kempermann, G. (2013). Delayed and transient increase of adult R.R. (2003). Histone deacetylase inhibitors prevent oxidative
hippocampal neurogenesis by physical exercise in DBA/2 mice. neuronal death independent of expanded polyglutamine repeats
PLoS One 8: e83797. via an Sp1-dependent pathway. Proc. Natl. Acad. Sci. U.S.A. 100:
Palmer, T.D., Willhoite, A.R., and Gage, F.H. (2000). Vascular niche for 4281–4286.
adult hippocampal neurogenesis. J. Comp. Neurol. 425: Saha, R.N. and Pahan, K. (2006). HATs and HDACs in
479–494. neurodegeneration: a tale of disconcerted acetylation
Pan-Vazquez, A., Rye, N., Ameri, M., McSparron, B., Smallwood, G., homeostasis. Cell Death Differ. 13: 539–550.
Bickerdyke, J., Rathbone, A., Dajas-Bailador, F., and Toledo- Sairanen, M., Lucas, G., Ernfors, P., Castrén, M., and Castrén, E.
Rodriguez, M. (2015). Impact of voluntary exercise and housing (2005). Brain-derived neurotrophic factor and antidepressant
conditions on hippocampal glucocorticoid receptor, miR-124 and drugs have different but coordinated effects on neuronal
anxiety. Mol. Brain 8: 40. turnover, proliferation, and survival in the adult dentate gyrus.
Pereira, A.C., Huddleston, D.E., Brickman, A.M., Sosunov, A.A., Hen, J. Neurosci. 25: 1089–1094.
R., McKhann, G.M., Sloan, R., Gage, F.H., Brown, T.R., and Small, Satoh, J., Kino, Y., and Niida, S. (2015). MicroRNA-Seq data analysis
S.A. (2007). An in vivo correlate of exercise-induced pipeline to identify blood biomarkers for Alzheimer’s disease
neurogenesis in the adult dentate gyrus. Proc. Natl. Acad. Sci. from public data. Biomark. Insights 10: 21–31.
U.S.A. 104: 5638–5643. Sharp, K. and Hewitt, J. (2014). Dance as an intervention for people
Pietrelli, A., Matkovic, L., Vacotto, M., Lopez-Costa, J.J., Basso, N., and with Parkinson’s disease: a systematic review and meta-
Brusco, A. (2018). Aerobic exercise upregulates the analysis. Neurosci. Biobehav. Rev. 47: 445–456.
BDNF-serotonin systems and improves the cognitive function in Siuda, J., Patalong-Ogiewa, M., Żmuda, W., Targosz-Gajniak, M.,
rats. Neurobiol. Learn. Mem. 155: 528–542. Niewiadomska, E., Matuszek, I., Jędrzejowska-Szypułka, H., Lewin-
Pin-Barre, C. and Laurin, J. (2015). Physical exercise as a Kowalik, J., and Rudzińska-Bar, M. (2017). Cognitive impairment
diagnostic, rehabilitation, and preventive tool: influence on and BDNF serum levels. Neurol. Neurochir. Pol. 51: 24–32.
neuroplasticity and motor recovery after stroke. Neural Plast. Sleiman, S.F., Henry, J., Al-Haddad, R., El Hayek, L., Abou Haidar, E.,
2015: 608581. Stringer, T., Ulja, D., Karuppagounder, S.S., Holson, E.B., Ratan,
Pons-Espinal, M., Gasperini, C., Marzi, M.J., Braccia, C., Armirotti, A., R.R., et al. (2016). Exercise promotes the expression of brain
Pötzsch, A., Walker, T.L., Fabel, K., Nicassio, F., Kempermann, G., derived neurotrophic factor (BDNF) through the action of the
et al. (2019). MiR-135a-5p is critical for exercise-induced adult ketone body beta-hydroxybutyrate. eLife 5: e15092.
neurogenesis. Stem Cell Rep. 12: 1298–1312. Smirnova, L., Gräfe, A., Seiler, A., Schumacher, S., Nitsch, R., and
Popken, G.J., Dechert-Zeger, M., Ye, P., and D’Ercole, A.J. (2005). Brain Wulczyn, F.G. (2005). Regulation of miRNA expression during
development. Adv. Exp. Med. Biol. 567: 187–220. neural cell specification. Eur. J. Neurosci. 21: 1469–1477.
J. Liang et al.: Exercise promotes brain remodeling 15

Sonntag, W.E., Lynch, C.D., Cooney, P.T., and Hutchins, P.M. (1997). von Bohlen und Halbach, O., and von Bohlen und Halbach, V. (2018).
Decreases in cerebral microvasculature with age are associated BDNF effects on dendritic spine morphology and hippocampal
with the decline in growth hormone and insulin-like growth factor function. Cell Tissue Res. 373: 729–741.
1. Endocrinology 138: 3515–3520. Wahl, P., Zwingmann, L., Manunzio, C., Wolf, J., and Bloch, W. (2018).
Sorrells, S.F., Paredes, M.F., Cebrian-Silla, A., Sandoval, K., Qi, D., Higher accuracy of the lactate minimum test compared to
Kelley, K.W., James, D., Mayer, S., Chang, J., Auguste, K.I., et al. established threshold concepts to determine maximal lactate
(2018). Human hippocampal neurogenesis drops sharply steady state in running. Int. J. Sports Med. 39: 541–548.
in children to undetectable levels in adults. Nature 555: Wang, X., Sundquist, K., Hedelius, A., Palmér, K., Memon, A.A., and
377–381. Sundquist, J. (2015). Circulating microRNA-144-5p is associated
Spruston, N. (2008). Pyramidal neurons: dendritic structure and with depressive disorders. Clin. Epigenet. 7: 69.
synaptic integration. Nat. Rev. Neurosci. 9: 206–221. Wu, S.Y., Wang, T.F., Yu, L., Jen, C.J., Chuang, J.I., Wu, F.S., Wu, C.W.,
Stranahan, A.M., Khalil, D., and Gould, E. (2007). Running induces and Kuo, Y.M. (2011). Running exercise protects the substantia
widespread structural alterations in the hippocampus and nigra dopaminergic neurons against inflammation-induced
entorhinal cortex. Hippocampus 17: 1017–1022. degeneration via the activation of BDNF signaling pathway. Brain
Sun, L.Y. and D’Ercole, A.J. (2006). Insulin-like growth factor-I Behav. Immun. 25: 135–146.
stimulates histone H3 and H4 acetylation in the brain in vivo. Xie, Q., Cheng, J., Pan, G., Wu, S., Hu, Q., Jiang, H., Wang, Y., Xiong, J.,
Endocrinology 147: 5480–5490. Pang, Q., and Chen, X. (2019). Treadmill exercise ameliorates
Tammen, S.A., Friso, S., and Choi, S.W. (2013). Epigenetics: the link focal cerebral ischemia/reperfusion-induced neurological deficit
between nature and nurture. Mol. Aspects Med. 34: 753–764. by promoting dendritic modification and synaptic plasticity via
Toy, W.A., Petzinger, G.M., Leyshon, B.J., Akopian, G.K., Walsh, J.P., upregulating caveolin-1/VEGF signaling pathways. Exp. Neurol.
Hoffman, M.V., Vuckovic, M.G., and Jakowec, M.W. (2014). 313: 60–78.
Treadmill exercise reverses dendritic spine loss in direct and Xiong, Y., Mahmood, A., and Chopp, M. (2019). Remodeling dendritic
indirect striatal medium spiny neurons in the 1-methyl-4-phenyl- spines for treatment of traumatic brain injury. Neural Regen. Res.
1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson’s 14: 1477–1480.
disease. Neurobiol. Dis. 63: 201–209. Xu, X., Fu, Z., and Le, W. (2019). Exercise and Parkinson’s disease. Int.
Trejo, J.L., Carro, E., and Torres-Aleman, I. (2001). Circulating insulin- Rev. Neurobiol. 147: 45–74.
like growth factor I mediates exercise-induced increases in the Yang, J., Ruchti, E., Petit, J.M., Jourdain, P., Grenningloh, G., and
number of new neurons in the adult hippocampus. J. Neurosci. Allaman, I. (2014). Lactate promotes plasticity gene expression
21: 1628–1634. by potentiating NMDA signaling in neurons. Proc. Natl. Acad. Sci.
Uysal, N., Kiray, M., Sisman, A.R., Camsari, U.M., Gencoglu, C., U.S.A. 111: 12228–12233.
Baykara, B., Cetinkaya, C., and Aksu, I. (2015). Effects of Yau, S.-Y., Lau, B.W.-M., Tong, J.-B., Wong, R., Ching, Y.-P., Qiu, G.,
voluntary and involuntary exercise on cognitive functions, and Tang, S.-W., Lee, T.M.C., and So, K.-F. (2011). Hippocampal
VEGF and BDNF levels in adolescent rats. Biotech. Histochem. 90: neurogenesis and dendritic plasticity support running-improved
55–68. spatial learning and depression-like behaviour in stressed rats.
van der Kleij, L.A., Petersen, E.T., Siebner, H.R., Hendrikse, J., PLoS One 6: e24263.
Frederiksen, K.S., Sobol, N.A., Hasselbalch, S.G., and Garde, E. Yau, S.-Y., Lee, T.H.-Y., Li, A., Xu, A., and So, K.-F. (2018). Adiponectin
(2018). The effect of physical exercise on cerebral blood flow in mediates running-restored hippocampal neurogenesis in
Alzheimer’s disease. NeuroImage 20: 650–654. streptozotocin-induced type 1 diabetes in mice. Front. Neurosci.
van Praag, H., Christie, B.R., Sejnowski, T.J., and Gage, F.H. (1999a). 12: 679.
Running enhances neurogenesis, learning, and long-term Yau, S.Y., Li, A., Hoo, R.L.C., Ching, Y.P., Christie, B.R., Lee, T.M.C., Xu,
potentiation in mice. Proc. Natl. Acad. Sci. U.S.A. 96: A., and So, K.-F. (2014). Physical exercise-induced hippocampal
13427–13431. neurogenesis and antidepressant effects are mediated by the
van Praag, H., Kempermann, G., and Gage, F.H. (1999b). Running adipocyte hormone adiponectin. Proc. Natl. Acad. Sci. U.S.A. 111:
increases cell proliferation and neurogenesis in the adult mouse 15810–15815.
dentate gyrus. Nat. Neurosci. 2: 266–270. Ye, P. and D’Ercole, A.J. (2006). Insulin-like growth factor actions
van Praag, H., Shubert, T., Zhao, C., and Gage, F.H. (2005). Exercise during development of neural stem cells and progenitors in the
enhances learning and hippocampal neurogenesis in aged mice. central nervous system. J. Neurosci. Res. 83: 1–6.
J. Neurosci. 25: 8680–8685. Ying, S.-W., Futter, M., Rosenblum, K., Webber, M.J., Hunt, S.P., Bliss,
Vaynman, S., Ying, Z., and Gomez-Pinilla, F. (2003). Interplay between T.V.P., and Bramham, C.R. (2002). Brain-derived neurotrophic
brain-derived neurotrophic factor and signal transduction factor induces long-term potentiation in intact adult
modulators in the regulation of the effects of exercise on hippocampus: requirement for ERK activation coupled to CREB
synaptic-plasticity. Neuroscience 122: 647–657. and upregulation of Arc synthesis. J. Neurosci. 22: 1532–1540.
Vaynman, S., Ying, Z., and Gomez-Pinilla, F. (2004). Hippocampal Zhang, X., Zhang, L., Cheng, X., Guo, Y., Sun, X., Chen, G., Li, H., Li, P.,
BDNF mediates the efficacy of exercise on synaptic plasticity and Lu, X., Tian, M., et al. (2014). IGF-1 promotes Brn-4 expression
cognition. Eur. J. Neurosci. 20: 2580–2590. and neuronal differentiation of neural stem cells via the PI3K/Akt
Voisin, S., Eynon, N., Yan, X., and Bishop, D.J. (2015). Exercise pathway. PLoS One 9: e113801.
training and DNA methylation in humans. Acta Physiol. 213: Zhao, Y., Pogue, A.I., and Lukiw, W.J. (2015). MicroRNA (miRNA)
39–59. signaling in the human CNS in sporadic Alzheimer’s disease
von Bernhardi, R., Bernhardi, L.E., and Eugenin, J. (2017). What is (AD)-novel and unique pathological features. Int. J. Mol. Sci. 16:
neural plasticity?. Adv. Exp. Med. Biol. 1015: 1–15. 30105–30116.

You might also like