Next Article in Journal
Arrestin Domain Containing 3 Reverses Epithelial to Mesenchymal Transition and Chemo-Resistance of TNBC Cells by Up-Regulating Expression of miR-200b
Next Article in Special Issue
Microglial Phenotyping in Neurodegenerative Disease Brains: Identification of Reactive Microglia with an Antibody to Variant of CD105/Endoglin
Previous Article in Journal
Hepatocellular Toxicity of Paris Saponins I, II, VI and VII on Two Kinds of Hepatocytes-HL-7702 and HepaRG Cells, and the Underlying Mechanisms
Previous Article in Special Issue
Synthesis and Evaluation of Novel Pyrazole Ethandiamide Compounds as Inhibitors of Human THP-1 Monocytic Cell Neurotoxicity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Physical Exercise Inhibits Inflammation and Microglial Activation

1
Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan
2
Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan 70101, Taiwan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 5 June 2019 / Revised: 8 July 2019 / Accepted: 9 July 2019 / Published: 9 July 2019
(This article belongs to the Special Issue Microglia in Neurodegenerative Diseases)

Abstract

:
Accumulating evidence indicates that exercise can enhance brain function and attenuate neurodegeneration. Besides improving neuroplasticity by altering the synaptic structure and function in various brain regions, exercise also modulates multiple systems that are known to regulate neuroinflammation and glial activation. Activated microglia and several pro-inflammatory cytokines play active roles in the pathogenesis of neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease. The purpose of this review is to highlight the impacts of exercise on microglial activation. Possible mechanisms involved in exercise-modulated microglial activation are also discussed. Undoubtedly, more studies are needed in order to disclose the detailed mechanisms, but this approach offers therapeutic potential for improving the brain health of millions of aging people where pharmacological intervention has failed.

1. Introduction

Exercise impacts our body at multiple levels, including the central nervous system (CNS). In responding to exercise-related stress (e.g., hypoxia, heat, free radicals, etc.) and injuries, the body launches multiple endogenous protective and repair systems by altering gene expression and releasing a range of factors that prepare the body for the next challenge. These factors, amongst others, involve trophic effects, anti-oxidation, energy metabolism, and anti-inflammation. Some of these factors enhance brain function and ameliorate brain disorders by inducing neuroplasticity, increasing metabolic efficiency, and improving anti-oxidative capacity [1,2,3,4]. Others maintain brain homeostasis and protect brain from pathological insults by regulating glial activation and neuroinflammation. Activated microglia and several pro-inflammatory cytokines play active roles in the pathogenesis of neurodegenerative diseases, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). It has been well-documented that although acute, high-intensity exercise may cause muscle injury and induce inflammation, long-term exercise at low-to-moderate intensity negatively regulates the inflammatory response. Here, we review literature that provides empirical evidence showing potential mechanisms for the inflammation-modulating effect of physical exercise, with focus on the effect of anti-microglial activation.

2. Microglia Are the Central Modulators of Neuroinflammation

Microglia are the primary immune cells in the CNS. One of their major functions is to maintain the homeostasis of the CNS [5]. In addition to controlling innate and adaptive immune responses in the CNS, microglia also play a significant role in remodeling the dendritic spine formations and modulating neuronal activity [6,7]. In the healthy brain, microglia exist in a resting state characterized by ramified morphology. However, resting microglia are not inactive. Their processes continually survey a defined microenvironment [8,9]. Thus, it has been suggested that “surveying microglia” is a more appropriate name to describe their active surveillance mode [9]. When exposed to pathological insult, microglia transform from a resting stage into a spectrum of activated stages. Activated microglia are characterized by changes in morphology and expression of cytoplasmic and surface proteins [10,11]. Some of the morphological changes (e.g., retraction of ramified processes) and altered protein expressions (e.g., MHC II, CD11b, CD68) have been adopted as markers of microglia activation. The two types of activation states (i.e., the classical activation M1-proinflammatory type and the alternative activation M2-anti-inflammatory type) in macrophages have been introduced into the field of microglia [12,13,14]. Nonetheless, it has been argued whether this M1/M2 polarization occurs in microglia in tissue, especially in the human brains [15].
The activity of microglia can be induced by a plethora of pro-inflammatory factors such as neutrophil-related cytokines, oxidative stress, nucleotide-binding domain, leucine rich containing protein (NLRP-inflammasome), and lipid-mediated proteins derived from metabolic disorders [16,17,18]. Different neurotransmitters are also known to activate microglia via their respective receptors, such as purinergic receptors, glutamate receptors, dopaminergic receptors and cholinergic receptors [9]. The immune cells and molecules (e.g., complement, inflammasome associated protein, gut microbiota, and the bacterial secreted metabolisms [19,20,21]) known to induce peripheral inflammation can also induce microglial activation.
The activity of microglia can be inhibited by anti-inflammatory factors, such as cluster of differentiation-200 (CD200)/CD200 receptor (CD200R), interleukin-10 (IL-10), triggering receptor expressed on myeloid cell-2 (TREM2), and vitamin D3 [22,23,24,25]. These proteins downregulate inflammatory cytokines released by microglia and are involved in the essential signaling pathways in these cells. For example, CD200R can inhibit the activation of the extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (MAPK) pathways, which cause cytokine production [26]. IL-10 prevents the lipopolysaccharide (LPS)-induced production and secretion of pro-inflammatory cytokines through the toll-like receptor-4 (TLR4) and nuclear factor kappa B (NF-κB) signaling cascade [27,28]. Moreover, TREM2 downregulates the PI3K/Akt and NF-κB signaling and attenuates the LPS-induced inflammatory responses [29]. Remarkably, expression levels of several anti-microglial activation factors are significantly decreased in AD and PD [30,31]. The deficiency of anti-inflammatory factors results in enhanced microglial activation and subsequently leads to the neuroinflammation, which has been shown to impair in adult hippocampal neurogenesis, neuronal synaptic morphology, and synaptic plasticity [32,33]. Furthermore, it has been hypothesized that inflammation is important for the death of dopaminergic neurons [34]. The levels of the proinflammatory cytokines TNF-α, IFN-γ, IL-1β, and IL-6 are elevated in the cerebrospinal fluid, serum, striatum, and substantia nigra of PD patients [35]. Using positron emission tomography scans with radiotracers for activated microglia and dopamine transporter, a negative correlation between these two markers in the dopaminergic nigrostriatal system has been reported in early PD patients [36], which suggests that microglia are activated early in the disease.
In Table 1, we list studies that have described how different forms of exercise can modulate some of these factors to affect microglia activation.

3. Exercise Regulates Microglial Activation by Increasing Anti-Inflammatory Factors

3.1. Anti-Inflammatory Cytokines

In response to muscular contractions, myocytes produce and release numerous molecules, termed myokines [56]. Among them, IL-6 was the first identified myokine [56,57,58]. IL-6 has both pro-inflammatory and anti-inflammatory effects. These effects are mediated by distinct signaling pathways: classic-signaling and trans-signaling. The pro-inflammatory effects of IL-6 depend on trans-signaling, in which IL-6 binds to a soluble form of IL-6 receptor; whereas the anti-inflammatory effects of IL-6 depended on classic-signaling, in which IL-6 binds to the membrane-bound non-signaling α-receptor [59,60]. Unlike pro-inflammatory cytokines (e.g., IL-1 and TNFα), IL-6, which can be produced in contracting skeletal muscles and secreted to the vascular system, does not promote major inflammatory mediators [61]. In terms of the anti-inflammatory effects, IL-6 myokine upregulates the expressions of anti-inflammatory cytokine IL-10 and the levels of IL-1 receptor antagonist (IL-1Ra) [39]. Interestingly, the levels of IL-6 are reduced in several neurodegenerative diseases, including AD and PD [60].
In addition to IL-6, other myokines may also have different kinds of anti-inflammatory cytokine properties. It has been shown that long-term exercise can increase the production and secretion of IL-10 in the skeletal muscles [38]. In a peripheral nerve injury mouse model, two weeks of low-intensity exercise was found to inhibit peripheral and central neuroinflammatory responses via upregulation of IL-4 [37]. These anti-inflammatory myokines can be readily transported into the CNS from the peripheral circulation [62]. When IL-10 interacts with its receptor on microglia, it enhances the suppressor of cytokine signaling 3, an inhibitor of cytokine-induced signaling responses resulting in inhibition of inhibits microglial activation [63].
Moreover, exercise can also stimulate the expression of IL-1Ra in the CNS [37]. IL-1Ra has a higher affinity for the IL-1R than IL-1α or IL-1β. Blocking the binding of IL-1 to its receptor interrupts the pro-inflammatory IL-1 signaling cascade and related microglial activity [64]. Therefore, exercise can upregulate the expression of anti-inflammatory cytokines and inhibit microglial activation.

3.2. CD200-CD200R

CD200 is an immunomodulatory factor expressed by neurons in the CNS [65]. It inhibits microglial activation through binding with CD200R on microglial plasma membranes [65]. It has been demonstrated that long-term treadmill running increases expression levels of both CD200 and CD200R in mouse brains [40]. When CD200-CD200R interaction occurs, it recruits downstream tyrosine kinases 1 and 2, phosphotyrosine-binding domain proteins. After phosphorylation, these proteins bind to RasGAP and Src homology 2-containing inositol phosphatase, leading to inhibition of downstream Ras/ERK-MAPK inflammatory signaling pathways. Furthermore, CD200/CD200R can also inhibit JNK and p38 MAPK. The inactivation of MAPK signaling results in the suppression of pro-inflammatory cytokine secretion [26,66,67]. Exercise may increase the expression of CD200/CD200R to suppress MAPK signaling pathway and inhibit microglial activation.

3.3. TREM2

TREM2, an immunoglobulin superfamily receptor, is mainly expressed by microglia in the CNS [68]. It plays essential roles in microglial phagocytosis and cytoskeleton rearrangement [69]. A soluble form of TREM2, derived from the cell surface receptor, can promote the survival of microglia and their phagocytic activity [70,71]. It has also been suggested that increasing TREM2 levels is an effective therapeutic approach for delaying the pathogenesis of AD [70]. Mutations in TREM2 resulting in loss of function can lead to increased risk of developing AD. Long-term physical exercise increases levels of soluble TREM2 in the CSF of AD patients [41]. When TREM2 forms a complex with DNAX activation proteins 12 (DAP12), a transmembrane adapter protein on the microglial membrane, TREM2 activation causes DAP12 phosphorylation via Src family kinases. The TREM2-DAP12 interaction stimulates PI3K/Akt downstream effect and subsequently blocks MAPK cascade at the RAF level. These signaling pathways lead to the inhibition of inflammatory responses driven by TLR4 in microglia [72,73]. Thus, exercise may regulate the activation status of microglia via upregulation of TREM2.

3.4. Heat-Shock Proteins (HSP)

During physical exercise period, neuroprotective HSPs are synthesized and released from skeletal muscle into blood circulation. Hence, HSPs can be considered as myokines. HSPs are categorized into several sub-types (e.g., HSP60, HSP70, and HSP72) [50,51,52,74] and their functions are multifaceted, but mainly in maintaining cellular homeostasis and protein stability [75]. HSPs are associated with endotoxin tolerance response [76]. Noticeably, HSP60 and HSP70 decrease the cellular responses to LPS as well as TLR4 expression, resulting in downregulation of secretion of pro-inflammatory molecules, including TNF-α and IL-1β [77,78,79,80]. Therefore, exercise-induced upregulation of HSPs may negatively regulate the productions of pro-inflammatory cytokines from glial cells.

3.5. Metabolic Factors

The efficacy of both acute resistance and chronic endurance exercises enhances the production of Sirtuin-1 (SIRT1), an NAD+-dependent protein deacetylase [44,45,46]. SIRT1 plays critical roles in regulating inflammation and cell survival by deacetylation of transcription factors [81]. Overexpression of SIRT1 deacetylates the NF-κB subunit p65, hence suppresses its activity, resulting in decreased production of proinflammatory cytokines [82,83,84]. Moreover, SIRT1 can activate the expression of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) to stimulate the antioxidant response [85]. Nrf2-activated gene expression leads to antioxidant enzymes being released, including glutathione peroxidase and heme oxygenase-1 [86]. Another function of Nrf2 is an antagonist of the NF-κB cascade, which causes reduced production of proinflammatory mediators [87]. More importantly, SIRT1 directly stimulates the transcriptional co-activator peroxisome proliferator-activated receptor gamma coactivator-1-alpha (PGC-1α), which controls inflammation. PGC-1α restrains the activity of NF-κB-mediated microglial activation, but at the same time enhances the phagocytic activity of microglia through the STAT3 and STAT6 pathways [88,89]. Thus, the SIRT1-related Nrf2 and NF-κB pathways may contribute to exercise-induced inhibition of microglial activation and neuroinflammation.
In a diet-induced obesity and leptin resistance mouse model, increased leptin levels are found in the hippocampus [47]. Accumulated leptin induces local astroglial activation and secretion of IL-1β and TNF-α, which subsequently induces microglial activation [47]. Long-term voluntary exercise decreases the degree of microglial activation in the hippocampus by increasing leptin sensitivity and decreasing the levels of IL-1β and TNF-α [47].

3.6. Brain-Derived Neurotrophic Factor (BDNF)

BDNF is one of the most studied neurotrophic factors whose production in the brain is increased by exercise [48,49,90]. BDNF is primarily produced by astrocytes and microglia in brain and has multiple effects on neurogenesis and inflammation modulation. Using the 5xFAD AD model mouse which develops severe AD-like pathology at an early age and cognitive dysfunction, it was found that inducing adult hippocampal neurogenesis (AHN) alone did not reverse cognitive dysfunction or pathology, but inducing AHN and BDNF levels through exercise resulted in reduced Aβ load, increased expression of synaptic proteins and improved cognition [90]. These effects were only observed in 5xFAD mice with pathology and not age-matched controls. The exercise induced secretions of BDNF, IL- 6 and fibronectin type III domain–containing protein–5 (FNDC5). It has been demonstrated that muscle contraction-induced activation of PGC-1α stimulates the release of irisin from its membrane-bound precursor FNDC5 [91]. The circulating irisin then enters the brain and upregulates BDNF expression in the brain [91]. Alternatively, ketone body β-hydroxybutyrate released from the liver during exercise has also been shown to induce BDNF expression in the brain [92]. BDNF can regulate brain functions in multiple aspects, including neuronal cell survival, adult hippocampal neurogenesis, and neuroplasticity [93]. Furthermore, BDNF can alleviate microglial activation in several brain disease models [94,95,96,97]. However, the decreased microglial activation is generally considered an indirect consequence of reduced neuronal injury elicited by the BDNF neurotrophic effect, not a direct effect of BDNF.
Nonetheless, we propose that BDNF may directly influence microglial activation through the following pathways. By binding to its receptor TrkB, BDNF can induce ERK activation and CREB phosphorylation, which can block the activity of NF-κB and transcription of certain anti-inflammatory genes [98,99]. Furthermore, BDNF can activate Akt signaling, which suppresses the activity of glycogen synthase kinase 3, resulting in a decrease of NF-κB activation and an increase of CREB activation [100]. BDNF is also known to modulate mitogen-activated protein kinase phosphatase 1, resulting in the reduction of p38 and JNK phosphorylation [101,102,103]. The effect of BDNF on microglial activation requires further in-depth investigation to determine the interactions of the effects of exercise-induced BDNF on neurogenesis and microglia/astrocyte responses.

3.7. Antioxidants

It is known that free radical substances are produced during exercise. However, endurance and long-term exercises essentially induce adaptive responses by upregulation of endogenous antioxidants, including glutathione peroxidase (GSH) and superoxide dismutase [53,104,105]. Noticeably, GSH plays vital roles in maintaining redox balance and anti-inflammatory mediators found in astrocytes and microglia [106,107]. The increased GSH synthesis can attenuate the releasing pro-inflammatory factors TNF-α and IL-6. Moreover, it negatively regulates the p38 MAPK, JNK, and NF-κB inflammatory pathways in glial cells [108,109,110], which may also inhibit the production of free radicals. In a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse study, long-term treadmill running decreased the MPTP-induced upregulation of NADPH oxidase and microglial activation [111]. Therefore, long-term exercise-induced upregulation of antioxidant levels play a crucial role in the protection against neuroinflammation by microglia. The induced upregulation of antioxidants is involved in protection not only to oxidative stress but also to neurodegenerative diseases, including AD and PD [112].

3.8. Glymphatic System

The glymphatic system is known as the brain waste clearance system, involving glial cells and lymphatic vessels [113,114]. This system drains the fluid and solutes from the parenchyma (i.e., interstitial fluid) into the cerebrospinal fluid and eventually to the deep cervical lymph nodes [115]. Aquaporin-4 is a key protein component, which is found along the astrocytic endfeet contacting to the cellular membranes of perivascular cells. It functions in brain-water homeostasis and importantly facilitates the pathway of perivascular interstitial fluid-cerebrospinal fluid exchange [116,117]. Impaired glymphatic function is evident in aging and neurodegenerative diseases [118,119]. On the other hand, both voluntary wheel and mandatory treadmill exercise enhanced the efficiency of glymphatic system in normal aging and AD mice [42,120]. Furthermore, the degrees of astrocytic and microglial activations in aged mice were also decreased, whereas the amount of aquaporin-4 increased through exercise [43]. It has been suggested that the anti-microglial activation effect of exercise is due to the enhanced glymphatic system, which facilitates the clearance of metabolic wastes and toxins, such as amyloid-β and α-synuclein, from the brain [121,122].

4. Effect of Exercise on Proinflammatory Cytokines and Chemokines

In this section, studies showing that exercise inhibits microglial activation by suppressing the production of pro-inflammatory factors will be discussed. The key studies are summarized in Table 2.

4.1. Proinflammatory Cytokines and Chemokines

Exercise can inhibit microglial activation and alleviate pathogenesis of AD and PD in both patients and animal models by downregulating the expression of proinflammatory cytokines [123,124,125,126]. For example, in the APPswe/PS1De9 double-transgenic mouse model of AD, long-term treadmill exercise has been shown to suppress oxidative stress and microglia-induced neuroinflammation by decreasing the level of IL-1β and TNF-α [127]. IL-1β, IL-18, and TNF-α are considered the major proinflammatory cytokines in the CNS [128]. IL-1β and IL-18 can promote inflammasome production to enhance the inflammatory response [116]. In the APP/PS1 model of AD, IL-1β secretion has been shown to enhance NLRP3 inflammasome in microglia, while deficiency of NLRP3 inflammasome decreases deposition of amyloid-β [129]. Furthermore, in several neurological disease models, including multiple sclerosis, PD, and AD, NLRP3 and NLRC4 inflammasomes have been demonstrated to play critical roles in the activation of microglia and astrocytes [18,129,130]. In ovariectomized mice, long-term treadmill running has been shown to decrease the release of IL-1β and IL-18 and inhibit microglial activation from NLRP3 inflammasome production in the hippocampus [123].
Chemokines are responsible for the recruitment of cells of monocytic origin to sites of inflammation, which may aggravate the degrees of local microglial activation [131]. In obese mice induced by a high-fat western diet, expression of a number of proinflammatory molecules, including chemokines CCL2 and CXCL10, are increased in the prefrontal cortex [132]. Long-term wheel running is known to decrease the expression levels of CXCL10 and CCL2, which may lessen the degrees of subsequent microglial activation [132].

4.2. Toll-Like Receptor (TLR) Signaling Pathway

TLRs are a class of pattern recognition receptors that play a key role in the innate immune response, including the activation of microglia. Several studies have implied that exercise can inhibit microglial activation by regulating TLR signaling pathways. In the mouse model of PD, soluble α-synuclein indirectly lead to oxidative stress when binding to surface receptors TLR2, TLR4, and CD11b in microglia, resulting in activation of neuroinflammation [133,134,135]. Interestingly, long-term treadmill running suppressed α-synuclein (α-Syn)/ TLR2 mediated neuroinflammation and the associated microglia and NADPH oxidase activation [111]. In another study, long-term treadmill running was found to decrease the high-fat-diet induced hippocampal neuroinflammation, including the degree of microglial activation, levels of proinflammatory cytokines (TNF-α and IL-1β) and cyclooxygenase-2, as well as the expression levels of TLR-4 and its downstream proteins [136]. In these two studies, the authors attributed the exercise-induced anti-microglial activation to downregulation of TLR signaling pathways. However, the actual causal relationship between TLRs and microglial activation remains to be confirmed.

4.3. Gut Microbiota

Bidirectional communication between the brain and the gut has been suggested [138]. The brain can alter gut function by influencing motility, secretion, blood flow, and gut-associated immune function; whereas, the microbiome and products or metabolites secreted from the microbiome can modulate neuronal, immune, metabolic and endocrine pathways [21,139]. It has been suggested that information exchange along the brain-gut-microbe axis can be achieved by various routes, including the vagus nerve, the hypothalamic-pituitary-adrenal axis, and neurotransmitters and hormones, some of which can be produced by the microbiota [140,141]. For example, the gastrointestinal inflammatory tone could cross into brain via vagal afferent neurons [142]. It has been shown that pathogenic gut bacteria, such as Campylobacter jejuni or Salmonella typhimurium can, via the vagal afferents, induce neuronal activation in the nucleus of the solitary tract, suggesting vagal innervation of the GI tract is capable of detecting host-pathogen interactions in the intestine prior to the challenge producing a systemic response [142].
Intriguingly, gastrointestinal abnormalities such as constipation are highly associated with PD [143]. Recently, studies indicated that α-synuclein accumulation appears early in the gut and propagates via the vagus nerve to the brain in a prion-like fashion [144,145]. In an α-synuclein overexpressed mouse model of PD, α-synuclein pathology and microglial activation are regulated by gut microbiota, whereas antibiotic treatment ameliorates these pathologies [145]. Likewise, the dysregulation of gut microbiota is also known to regulate microglia activation and contribute to AD pathogenesis [146,147,148]. These findings strongly indicate that intestinal dysbiosis can induce microglial activation and other inflammatory responses in the brain.
It has been shown that physical exercise can modify the microbiome and influence the activation status of microglia, as well as performance of learning and memory [137]. However, exactly how exercise can modulate gut microbiota remains unclear, although several potential mechanisms have been suggested. These include indirect effects of exercise on the brain-gut-microbe axis, diet-microbe-host metabolic interactions, neuro-endocrine, and neuro-immune interactions, most likely via the regulation of vagus efferents with respect to the immune system (e.g., spleen and lymph nodes) and gut [142,149,150]

5. Conclusions

Undoubtedly, physical exercise can modulate microglial activation in the CNS. Current literature states that low-intensity exercise is sufficient to induce an anti-microglial activation effect by regulating the expressions of various factors. Some of these factors (e.g., myokines) can directly inhibit microglial activation via assorted mechanisms that subsequently prevent neuroinflammation in the CNS. Considerable evidence also suggests that exercise may inhibit microglial activation by downregulation of the levels of pro-inflammatory factors. However, the mechanism for the exercise-related downregulation of pro-inflammatory factors is less clear, as pro-inflammatory cytokines can be secreted from various sources (e.g., injured neurons, astrocytes, and microglia). Thus, the anti-microglial activation effect of exercise can be interpreted indirectly by upregulating the levels of trophic factors, which then lead to reduced neuronal injury and degrees of microglial activation. Furthermore, there are a few reports suggesting that physical exercise can shift the composition of the gut microbiome, which then affects both peripheral and central inflammation, including microglial activation in the CNS. Although some mechanisms are still waiting to be determined, it should be emphasized that physical activity represents a natural strong anti-inflammatory strategy to improve brain function [43].

Author Contributions

O.M.I., Z.W.Z., and Y.M.K. contributed to writing the manuscript. All authors read and approved the final manuscript for publication.

Funding

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

Conflicts of Interest

The authors have declared that no conflict of interest exists.

Abbreviations

ADAlzheimer’s disease
BDNFbrain-derived neurotrophic factor
CD200cluster of differentiation-200
CNScentral nervous system
DAP122-DNAX activation protein 12
ERKextracellular signal-regulated kinase
FNDC5fibronectin type III domain–containing protein–5
GSHglutathione peroxidase
HSPheat-shock protein
ILinterleukin
IL-1RaIL-1 receptor antagonist
JNKc-Jun N-terminal kinase
LPSlipopolysaccharide
MAPKmitogen-activated protein kinase
MPTP1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
NF-κBnuclear factor kappa B
NLRPnucleotide-binding domain, leucine rich containing protein
Nrf2nuclear factor erythroid 2-related factor 2
PDParkinson’s disease
PGC-1αproliferator-activated receptor gamma coactivator-1-alpha
PI3K/AKTphosphoinositide-3-kinase/AKT
SIRT1sirtuin-1
TLRtoll-like receptor
TNF-αtumor necrosis factor-alpha
TREM2triggering receptor expressed on myeloid cell-2

References

  1. Nagamatsu, L.S.; Flicker, L.; Kramer, A.F.; Voss, M.W.; Erickson, K.I.; Hsu, C.L.; Liu-Ambrose, T. Exercise is medicine, for the body and the brain. Br. J. Sports Med. 2014, 48, 943–944. [Google Scholar] [CrossRef] [PubMed]
  2. Lin, T.W.; Chen, S.J.; Huang, T.Y.; Chang, C.Y.; Chuang, J.I.; Wu, F.S.; Kuo, Y.M.; Jen, C.J. Different types of exercise induce differential effects on neuronal adaptations and memory performance. Neurobiol. Learn. Mem. 2012, 97, 140–147. [Google Scholar] [CrossRef] [PubMed]
  3. Tsai, S.F.; Ku, N.W.; Wang, T.F.; Yang, Y.H.; Shih, Y.H.; Wu, S.Y.; Lee, C.W.; Yu, M.; Yang, T.T.; Kuo, Y.M. Long-Term Moderate Exercise Rescues Age-Related Decline in Hippocampal Neuronal Complexity and Memory. Gerontology 2018, 64, 551–561. [Google Scholar] [CrossRef] [PubMed]
  4. Gomez-Cabrera, M.C.; Domenech, E.; Vina, J. Moderate exercise is an antioxidant: Upregulation of antioxidant genes by training. Free Radic. Biol. Med. 2008, 44, 126–131. [Google Scholar] [CrossRef] [PubMed]
  5. Perry, V.H.; Teeling, J. Microglia and macrophages of the central nervous system: The contribution of microglia priming and systemic inflammation to chronic neurodegeneration. Semin. Immunopathol. 2013, 35, 601–612. [Google Scholar] [CrossRef] [PubMed]
  6. Ferro, A.; Qu, W.; Lukowicz, A.; Svedberg, D.; Johnson, A.; Cvetanovic, M. Inhibition of NF-κB signaling in IKKβF/F; LysM Cre mice causes motor deficits but does not alter pathogenesis of Spinocerebellar ataxia type 1. PLoS ONE 2018, 13, e0200013. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, X.; Zhao, L.; Zhang, J.; Fariss, R.N.; Ma, W.; Kretschmer, F.; Wang, M.; Qian, H.H.; Badea, T.C.; Diamond, J.S. Requirement for microglia for the maintenance of synaptic function and integrity in the mature retina. J. Neurosci. 2016, 36, 2827–2842. [Google Scholar] [CrossRef] [PubMed]
  8. Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma In Vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef]
  9. Wolf, S.A.; Boddeke, H.; Kettenmann, H. Microglia in physiology and disease. Annu. Rev. Physiol. 2017, 79, 619–643. [Google Scholar] [CrossRef]
  10. Hickman, S.E.; Kingery, N.D.; Ohsumi, T.K.; Borowsky, M.L.; Wang, L.-C.; Means, T.K.; El Khoury, J. The microglial sensome revealed by direct RNA sequencing. Nat. Neurosci. 2013, 16, 1896. [Google Scholar] [CrossRef]
  11. Butovsky, O.; Weiner, H.L. Microglial signatures and their role in health and disease. Nat. Rev. Neurosci. 2018, 19, 622–635. [Google Scholar] [CrossRef] [PubMed]
  12. Saijo, K.; Glass, C.K. Microglial cell origin and phenotypes in health and disease. Nat. Rev. Immunol. 2011, 11, 775–787. [Google Scholar] [CrossRef] [PubMed]
  13. Orihuela, R.; McPherson, C.A.; Harry, G.J. Microglial M1/M2 polarization and metabolic states. Br. J. Pharmacol. 2016, 173, 649–665. [Google Scholar] [CrossRef] [PubMed]
  14. Ortega-Gomez, A.; Perretti, M.; Soehnlein, O. Resolution of inflammation: An integrated view. EMBO Mol. Med. 2013, 5, 661–674. [Google Scholar] [CrossRef] [PubMed]
  15. Hanisch, U.-K.; Kettenmann, H. Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 2007, 10, 1387. [Google Scholar] [CrossRef] [PubMed]
  16. Kim, E.J.; Kwon, K.J.; Park, J.Y.; Lee, S.H.; Moon, C.H.; Baik, E.J. Neuroprotective effects of prostaglandin E2 or cAMP against microglial and neuronal free radical mediated toxicity associated with inflammation. J. Neurosci. Res. 2002, 70, 97–107. [Google Scholar] [CrossRef] [PubMed]
  17. Bozoyan, L.; Dumas, A.; Patenaude, A.; Vallieres, L. Interleukin-36gamma is expressed by neutrophils and can activate microglia, but has no role in experimental autoimmune encephalomyelitis. J. Neuroinflamm. 2015, 12, 173. [Google Scholar] [CrossRef]
  18. Freeman, L.; Guo, H.; David, C.N.; Brickey, W.J.; Jha, S.; Ting, J.P. NLR members NLRC4 and NLRP3 mediate sterile inflammasome activation in microglia and astrocytes. J. Exp. Med. 2017, 214, 1351–1370. [Google Scholar] [CrossRef] [Green Version]
  19. Maneu, V.; Noailles, A.; Gomez-Vicente, V.; Carpena, N.; Cuenca, N.; Gil, M.L.; Gozalbo, D. Immunosuppression, peripheral inflammation and invasive infection from endogenous gut microbiota activate retinal microglia in mouse models. Microbiol. Immunol. 2016, 60, 617–625. [Google Scholar] [CrossRef] [Green Version]
  20. Heneka, M.T. Inflammasome activation and innate immunity in Alzheimer’s disease. Brain Pathol. 2017, 27, 220–222. [Google Scholar] [CrossRef]
  21. Erny, D.; de Angelis, A.L.H.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
  22. Perry, V.H.; Nicoll, J.A.; Holmes, C. Microglia in neurodegenerative disease. Nat. Rev. Neurol. 2010, 6, 193–201. [Google Scholar] [CrossRef] [PubMed]
  23. Shrikant, P.; Weber, E.; Jilling, T.; Benveniste, E.N. Intercellular adhesion molecule-1 gene expression by glial cells. Differential mechanisms of inhibition by IL-10 and IL-6. J. Immunol. 1995, 155, 1489–1501. [Google Scholar] [PubMed]
  24. Shrivastava, K.; Gonzalez, P.; Acarin, L. The immune inhibitory complex CD200/CD200R is developmentally regulated in the mouse brain. J. Comp. Neurol. 2012, 520, 2657–2675. [Google Scholar] [CrossRef] [PubMed]
  25. Boontanrart, M.; Hall, S.D.; Spanier, J.A.; Hayes, C.E.; Olson, J.K. Vitamin D3 alters microglia immune activation by an IL-10 dependent SOCS3 mechanism. J. Neuroimmunol. 2016, 292, 126–136. [Google Scholar] [CrossRef]
  26. Zhang, S.; Cherwinski, H.; Sedgwick, J.D.; Phillips, J.H. Molecular mechanisms of CD200 inhibition of mast cell activation. J. Immunol. 2004, 173, 6786–6793. [Google Scholar] [CrossRef] [PubMed]
  27. Meng, J.; Ni, J.; Wu, Z.; Jiang, M.; Zhu, A.; Qing, H.; Nakanishi, H. The Critical Role of IL-10 in the Antineuroinflammatory and Antioxidative Effects of Rheum tanguticum on Activated Microglia. Oxidative Med. Cell. Longev. 2018, 2018. [Google Scholar] [CrossRef]
  28. Chen, Z.; Jalabi, W.; Shpargel, K.B.; Farabaugh, K.T.; Dutta, R.; Yin, X.; Kidd, G.J.; Bergmann, C.C.; Stohlman, S.A.; Trapp, B.D. Lipopolysaccharide-induced microglial activation and neuroprotection against experimental brain injury is independent of hematogenous TLR4. J. Neurosci. 2012, 32, 11706–11715. [Google Scholar] [CrossRef]
  29. Li, C.; Zhao, B.; Lin, C.; Gong, Z.; An, X. TREM2 inhibits inflammatory responses in mouse microglia by suppressing the PI3K/NF-κB signaling. Cell Biol. Int. 2019, 43, 360–372. [Google Scholar] [CrossRef]
  30. Zhang, S.; Wang, X.J.; Tian, L.P.; Pan, J.; Lu, G.Q.; Zhang, Y.J.; Ding, J.Q.; Chen, S.D. CD200-CD200R dysfunction exacerbates microglial activation and dopaminergic neurodegeneration in a rat model of Parkinson’s disease. J. Neuroinflamm. 2011, 8, 154. [Google Scholar] [CrossRef]
  31. Liu, D.; Cao, B.; Zhao, Y.; Huang, H.; McIntyre, R.S.; Rosenblat, J.D.; Zhou, H. Soluble TREM2 changes during the clinical course of Alzheimer’s disease: A meta-analysis. Neurosci. Lett. 2018, 686, 10–16. [Google Scholar] [CrossRef] [PubMed]
  32. Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s Dement. 2018, 4, 575–590. [Google Scholar] [CrossRef] [PubMed]
  33. Ekdahl, C.T.; Claasen, J.H.; Bonde, S.; Kokaia, Z.; Lindvall, O. Inflammation is detrimental for neurogenesis in adult brain. Proc. Natl. Acad. Sci. USA 2003, 100, 13632–13637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Block, M.L.; Zecca, L.; Hong, J.S. Microglia-mediated neurotoxicity: Uncovering the molecular mechanisms. Nat. Rev. Neurosci. 2007, 8, 57–69. [Google Scholar] [CrossRef] [PubMed]
  35. Hirsch, E.C.; Hunot, S. Neuroinflammation in Parkinson’s disease: A target for neuroprotection? Lancet Neurol. 2009, 8, 382–397. [Google Scholar] [CrossRef]
  36. Ouchi, Y.; Yoshikawa, E.; Sekine, Y.; Futatsubashi, M.; Kanno, T.; Ogusu, T.; Torizuka, T. Microglial activation and dopamine terminal loss in early Parkinson’s disease. Ann. Neurol. 2005, 57, 168–175. [Google Scholar] [CrossRef] [PubMed]
  37. Bobinski, F.; Teixeira, J.M.; Sluka, K.A.; Santos, A.R.S. Interleukin-4 mediates the analgesia produced by low-intensity exercise in mice with neuropathic pain. Pain 2018, 159, 437–450. [Google Scholar] [CrossRef]
  38. Calegari, L.; Nunes, R.B.; Mozzaquattro, B.B.; Rossato, D.D.; Dal Lago, P. Exercise training improves the IL-10/TNF-α cytokine balance in the gastrocnemius of rats with heart failure. Braz. J. Phys. Ther. 2018, 22, 154–160. [Google Scholar] [CrossRef]
  39. Steensberg, A.; Fischer, C.P.; Keller, C.; Møller, K.; Pedersen, B.K. IL-6 enhances plasma IL-1ra, IL-10, and cortisol in humans. Am. J. Physiol. Endocrinol. Metab. 2003, 285, E433–E437. [Google Scholar] [CrossRef]
  40. Sung, Y.H.; Kim, S.C.; Hong, H.P.; Park, C.Y.; Shin, M.S.; Kim, C.J.; Seo, J.H.; Kim, D.Y.; Kim, D.J.; Cho, H.J. Treadmill exercise ameliorates dopaminergic neuronal loss through suppressing microglial activation in Parkinson’s disease mice. Life Sci. 2012, 91, 1309–1316. [Google Scholar] [CrossRef]
  41. Jensen, C.S.; Bahl, J.M.; Ostergaard, L.B.; Hogh, P.; Wermuth, L.; Heslegrave, A.; Zetterberg, H.; Heegaard, N.H.H.; Hasselbalch, S.G.; Simonsen, A.H. Exercise as a potential modulator of inflammation in patients with Alzheimer’s disease measured in cerebrospinal fluid and plasma. Exp. Gerontol. 2019, 121, 91–98. [Google Scholar] [CrossRef] [PubMed]
  42. He, X.F.; Liu, D.X.; Zhang, Q.; Liang, F.Y.; Dai, G.Y.; Zeng, J.S.; Pei, Z.; Xu, G.Q.; Lan, Y. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Basco, D.; Blaauw, B.; Pisani, F.; Sparaneo, A.; Nicchia, G.P.; Mola, M.G.; Reggiani, C.; Svelto, M.; Frigeri, A. AQP4-dependent water transport plays a functional role in exercise-induced skeletal muscle adaptations. PLoS ONE 2013, 8, e58712. [Google Scholar] [CrossRef]
  44. Suwa, M.; Nakano, H.; Radak, Z.; Kumagai, S. Endurance exercise increases the SIRT1 and peroxisome proliferator-activated receptor gamma coactivator-1alpha protein expressions in rat skeletal muscle. Metabolism 2008, 57, 986–998. [Google Scholar] [CrossRef] [PubMed]
  45. Lai, C.H.; Ho, T.J.; Kuo, W.W.; Day, C.H.; Pai, P.Y.; Chung, L.C.; Liao, P.H.; Lin, F.H.; Wu, E.T.; Huang, C.Y. Exercise training enhanced SIRT1 longevity signaling replaces the IGF1 survival pathway to attenuate aging-induced rat heart apoptosis. Age 2014, 36, 9706. [Google Scholar] [CrossRef]
  46. Guerra, B.; Guadalupe-Grau, A.; Fuentes, T.; Ponce-Gonzalez, J.G.; Morales-Alamo, D.; Olmedillas, H.; Guillen-Salgado, J.; Santana, A.; Calbet, J.A. SIRT1, AMP-activated protein kinase phosphorylation and downstream kinases in response to a single bout of sprint exercise: Influence of glucose ingestion. Eur. J. Appl. Physiol. 2010, 109, 731–743. [Google Scholar] [CrossRef] [PubMed]
  47. Koga, S.; Kojima, A.; Ishikawa, C.; Kuwabara, S.; Arai, K.; Yoshiyama, Y. Effects of diet-induced obesity and voluntary exercise in a tauopathy mouse model: Implications of persistent hyperleptinemia and enhanced astrocytic leptin receptor expression. Neurobiol. Dis. 2014, 71, 180–192. [Google Scholar] [CrossRef] [Green Version]
  48. Fang, Z.H.; Lee, C.H.; Seo, M.K.; Cho, H.; Lee, J.G.; Lee, B.J.; Park, S.W.; Kim, Y.H. Effect of treadmill exercise on the BDNF-mediated pathway in the hippocampus of stressed rats. Neurosci. Res. 2013, 76, 187–194. [Google Scholar] [CrossRef]
  49. Saucedo Marquez, C.M.; Vanaudenaerde, B.; Troosters, T.; Wenderoth, N. High-intensity interval training evokes larger serum BDNF levels compared with intense continuous exercise. J. Appl. Physiol. 2015, 119, 1363–1373. [Google Scholar] [CrossRef] [Green Version]
  50. Khadir, A.; Kavalakatt, S.; Cherian, P.; Warsame, S.; Abubaker, J.A.; Dehbi, M.; Tiss, A. Physical Exercise Enhanced Heat Shock Protein 60 Expression and Attenuated Inflammation in the Adipose Tissue of Human Diabetic Obese. Front. Endocrinol. 2018, 9, 16. [Google Scholar] [CrossRef]
  51. Milne, K.J.; Noble, E.G. Exercise-induced elevation of HSP70 is intensity dependent. J. Appl. Physiol. 2002, 93, 561–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Walsh, R.C.; Koukoulas, I.; Garnham, A.; Moseley, P.L.; Hargreaves, M.; Febbraio, M.A. Exercise increases serum Hsp72 in humans. Cell Stress Chaperones 2001, 6, 386. [Google Scholar] [CrossRef]
  53. Sen, C.K.; Marin, E.; Kretzschmar, M.; Hanninen, O. Skeletal muscle and liver glutathione homeostasis in response to training, exercise, and immobilization. J. Appl. Physiol. 1992, 73, 1265–1272. [Google Scholar] [CrossRef] [PubMed]
  54. Piercy, K.L.; Troiano, R.P.; Ballard, R.M.; Carlson, S.A.; Fulton, J.E.; Galuska, D.A.; George, S.M.; Olson, R.D. The physical activity guidelines for Americans. JAMA 2018, 320, 2020–2028. [Google Scholar] [CrossRef] [PubMed]
  55. Inoue, K.; Okamoto, M.; Shibato, J.; Lee, M.C.; Matsui, T.; Rakwal, R.; Soya, H. Long-Term Mild, rather than Intense, Exercise Enhances Adult Hippocampal Neurogenesis and Greatly Changes the Transcriptomic Profile of the Hippocampus. PLoS ONE 2015, 10, e0128720. [Google Scholar] [CrossRef] [PubMed]
  56. Pedersen, B.K.; Akerstrom, T.C.A. Role of myokines in exercise and metabolism. J. Appl. Physiol. 2007, 103, 1093–1098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Pedersen, B.K. Muscular interleukin-6 and its role as an energy sensor. Med. Sci. Sports Exerc. 2012, 44, 392–396. [Google Scholar] [CrossRef] [PubMed]
  58. Febbraio, M.A.; Pedersen, B.K. Contraction-induced myokine production and release: Is skeletal muscle an endocrine organ? Exerc. Sport Sci. Rev. 2005, 33, 114–119. [Google Scholar] [CrossRef] [PubMed]
  59. Taga, T.; Hibi, M.; Hirata, Y.; Yamasaki, K.; Yasukawa, K.; Matsuda, T.; Hirano, T.; Kishimoto, T. Interleukin-6 triggers the association of its receptor with a possible signal transducer, gp130. Cell 1989, 58, 573–581. [Google Scholar] [CrossRef]
  60. Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Biophys. Acta 2011, 1813, 878–888. [Google Scholar] [CrossRef] [Green Version]
  61. Pedersen, B.K.; Steensberg, A.; Schjerling, P. Muscle-derived interleukin-6: Possible biological effects. J. Physiol. 2001, 536, 329–337. [Google Scholar] [CrossRef] [PubMed]
  62. Kelly, A.M. Exercise-Induced Modulation of Neuroinflammation in Models of Alzheimer’s Disease. Brain Plast. 2018, 4, 81–94. [Google Scholar] [CrossRef] [PubMed]
  63. Cianciulli, A.; Dragone, T.; Calvello, R.; Porro, C.; Trotta, T.; Lofrumento, D.D.; Panaro, M.A. IL-10 plays a pivotal role in anti-inflammatory effects of resveratrol in activated microglia cells. Int. Immunopharmacol. 2015, 24, 369–376. [Google Scholar] [CrossRef] [PubMed]
  64. Rosenzweig, J.M.; Lei, J.; Burd, I. Interleukin-1 receptor blockade in perinatal brain injury. Front. Pediatr. 2014, 2, 108. [Google Scholar] [CrossRef] [PubMed]
  65. Biber, K.; Neumann, H.; Inoue, K.; Boddeke, H.W. Neuronal ‘On’ and ‘Off’ signals control microglia. Trends Neurosci. 2007, 30, 596–602. [Google Scholar] [CrossRef] [PubMed]
  66. Huber, M.; Helgason, C.D.; Damen, J.E.; Liu, L.; Humphries, R.K.; Krystal, G. The src homology 2-containing inositol phosphatase (SHIP) is the gatekeeper of mast cell degranulation. Proc. Natl. Acad. Sci. USA 1998, 95, 11330–11335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Lemay, S.; Davidson, D.; Latour, S.; Veillette, A. Dok-3, a novel adapter molecule involved in the negative regulation of immunoreceptor signaling. Mol. Cell. Biol. 2000, 20, 2743–2754. [Google Scholar] [CrossRef]
  68. Colonna, M.; Wang, Y. TREM2 variants: New keys to decipher Alzheimer disease pathogenesis. Nat. Rev. Neurosci. 2016, 17, 201–207. [Google Scholar] [CrossRef] [PubMed]
  69. Filipello, F.; Morini, R.; Corradini, I.; Zerbi, V.; Canzi, A.; Michalski, B.; Erreni, M.; Markicevic, M.; Starvaggi-Cucuzza, C.; Otero, K.; et al. The Microglial Innate Immune Receptor TREM2 Is Required for Synapse Elimination and Normal Brain Connectivity. Immunity 2018, 48, 979–991. [Google Scholar] [CrossRef]
  70. Zhong, L.; Chen, X.F.; Wang, T.; Wang, Z.; Liao, C.; Wang, Z.; Huang, R.; Wang, D.; Li, X.; Wu, L.; et al. Soluble TREM2 induces inflammatory responses and enhances microglial survival. J. Exp. Med. 2017, 214, 597–607. [Google Scholar] [CrossRef] [Green Version]
  71. Zhong, L.; Xu, Y.; Zhuo, R.; Wang, T.; Wang, K.; Huang, R.; Wang, D.; Gao, Y.; Zhu, Y.; Sheng, X.; et al. Soluble TREM2 ameliorates pathological phenotypes by modulating microglial functions in an Alzheimer’s disease model. Nat. Commun. 2019, 10, 1365. [Google Scholar] [CrossRef] [PubMed]
  72. Mecca, C.; Giambanco, I.; Donato, R.; Arcuri, C. Microglia and Aging: The Role of the TREM2-DAP12 and CX3CL1-CX3CR1 Axes. Int. J. Mol. Sci. 2018, 19, 318. [Google Scholar] [CrossRef] [PubMed]
  73. Turnbull, I.R.; Colonna, M. Activating and inhibitory functions of DAP12. Nat. Rev. Immunol. 2007, 7, 155. [Google Scholar] [CrossRef] [PubMed]
  74. Flynn, M.G.; McFarlin, B.K.; Markofski, M.M. The Anti-Inflammatory Actions of Exercise Training. Am. J. Lifestyle Med. 2007, 1, 220–235. [Google Scholar] [CrossRef] [PubMed]
  75. Fehrenbach, E.; Niess, A.M. Role of heat shock proteins in the exercise response. Exerc. Immunol. Rev. 1999, 5, 57–77. [Google Scholar] [PubMed]
  76. Fan, H.; Cook, J.A. Molecular mechanisms of endotoxin tolerance. J. Endotoxin Res. 2004, 10, 71–84. [Google Scholar] [CrossRef] [PubMed]
  77. Kilmartin, B.; Reen, D.J. HSP60 induces self-tolerance to repeated HSP60 stimulation and cross-tolerance to other pro-inflammatory stimuli. Eur. J. Immunol. 2004, 34, 2041–2051. [Google Scholar] [CrossRef] [PubMed]
  78. Aneja, R.; Odoms, K.; Dunsmore, K.; Shanley, T.P.; Wong, H.R. Extracellular heat shock protein-70 induces endotoxin tolerance in THP-1 cells. J. Immunol. 2006, 177, 7184–7192. [Google Scholar] [CrossRef] [PubMed]
  79. Yu, W.W.; Cao, S.N.; Zang, C.X.; Wang, L.; Yang, H.Y.; Bao, X.Q.; Zhang, D. Heat shock protein 70 suppresses neuroinflammation induced by alpha-synuclein in astrocytes. Mol. Cell. Neurosci. 2018, 86, 58–64. [Google Scholar] [CrossRef] [PubMed]
  80. Ferat-Osorio, E.; Sánchez-Anaya, A.; Gutiérrez-Mendoza, M.; Boscó-Gárate, I.; Wong-Baeza, I.; Pastelin-Palacios, R.; Pedraza-Alva, G.; Bonifaz, L.C.; Cortés-Reynosa, P.; Pérez-Salazar, E.; et al. Heat shock protein 70 down-regulates the production of toll-like receptor-induced pro-inflammatory cytokines by a heat shock factor-1/constitutive heat shock element-binding factor-dependent mechanism. J. Inflamm. 2014, 11, 19. [Google Scholar] [CrossRef] [PubMed]
  81. Haigis, M.C.; Sinclair, D.A. Mammalian sirtuins: Biological insights and disease relevance. Annu. Rev. Pathol. 2010, 5, 253–295. [Google Scholar] [CrossRef] [PubMed]
  82. Yang, H.; Zhang, W.; Pan, H.; Feldser, H.G.; Lainez, E.; Miller, C.; Leung, S.; Zhong, Z.; Zhao, H.; Sweitzer, S.; et al. SIRT1 activators suppress inflammatory responses through promotion of p65 deacetylation and inhibition of NF-κB activity. PLoS ONE 2012, 7, e46364. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, J.; Zhou, Y.; Mueller-Steiner, S.; Chen, L.F.; Kwon, H.; Yi, S.; Mucke, L.; Gan, L. SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-κB signaling. J. Biol. Chem. 2005, 280, 40364–40374. [Google Scholar] [CrossRef] [PubMed]
  84. Mendes, K.L.; Lelis, D.F.; Santos, S.H.S. Nuclear sirtuins and inflammatory signaling pathways. Cytokine Growth Factor Rev. 2017, 38, 98–105. [Google Scholar] [CrossRef] [PubMed]
  85. Pedruzzi, L.M.; Stockler-Pinto, M.B.; Leite, M., Jr.; Mafra, D. Nrf2-keap1 system versus NF-κB: The good and the evil in chronic kidney disease? Biochimie 2012, 94, 2461–2466. [Google Scholar] [CrossRef] [PubMed]
  86. Jung, K.A.; Kwak, M.K. The Nrf2 system as a potential target for the development of indirect antioxidants. Molecules 2010, 15, 7266–7291. [Google Scholar] [CrossRef] [PubMed]
  87. Saldanha, J.F.; Leal Vde, O.; Stenvinkel, P.; Carraro-Eduardo, J.C.; Mafra, D. Resveratrol: Why is it a promising therapy for chronic kidney disease patients? Oxidative Med. Cell. Longev. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  88. Navas-Enamorado, I.; Bernier, M.; Brea-Calvo, G.; de Cabo, R. Influence of anaerobic and aerobic exercise on age-related pathways in skeletal muscle. Ageing Res. Rev. 2017, 37, 39–52. [Google Scholar] [CrossRef]
  89. Yang, X.; Xu, S.; Qian, Y.; Xiao, Q. Resveratrol regulates microglia M1/M2 polarization via PGC-1alpha in conditions of neuroinflammatory injury. Brain Behav. Immun. 2017, 64, 162–172. [Google Scholar] [CrossRef]
  90. Choi, S.H.; Bylykbashi, E.; Chatila, Z.K.; Lee, S.W.; Pulli, B.; Clemenson, G.D.; Kim, E.; Rompala, A.; Oram, M.K.; Asselin, C.; et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model. Science 2018, 361. [Google Scholar] [CrossRef]
  91. Wrann, C.D.; White, J.P.; Salogiannnis, J.; Laznik-Bogoslavski, D.; Wu, J.; Ma, D.; Lin, J.D.; Greenberg, M.E.; Spiegelman, B.M. Exercise induces hippocampal BDNF through a PGC-1alpha/FNDC5 pathway. Cell Metab. 2013, 18, 649–659. [Google Scholar] [CrossRef] [PubMed]
  92. Sleiman, S.F.; Henry, J.; Al-Haddad, R.; El Hayek, L.; Abou Haidar, E.; Stringer, T.; Ulja, D.; Karuppagounder, S.S.; Holson, E.B.; Ratan, R.R.; et al. Exercise promotes the expression of brain derived neurotrophic factor (BDNF) through the action of the ketone body beta-hydroxybutyrate. eLife 2016, 5. [Google Scholar] [CrossRef] [PubMed]
  93. Greenberg, M.E.; Xu, B.; Lu, B.; Hempstead, B.L. New insights in the biology of BDNF synthesis and release: Implications in CNS function. J. Neurosci. 2009, 29, 12764–12767. [Google Scholar] [CrossRef] [PubMed]
  94. Jiang, Y.; Wei, N.; Lu, T.; Zhu, J.; Xu, G.; Liu, X. Intranasal brain-derived neurotrophic factor protects brain from ischemic insult via modulating local inflammation in rats. Neuroscience 2011, 172, 398–405. [Google Scholar] [CrossRef] [PubMed]
  95. Jiang, Y.; Wei, N.; Zhu, J.; Lu, T.; Chen, Z.; Xu, G.; Liu, X. Effects of brain-derived neurotrophic factor on local inflammation in experimental stroke of rat. Mediat. Inflamm. 2010, 2010. [Google Scholar] [CrossRef] [PubMed]
  96. Makar, T.K.; Trisler, D.; Sura, K.T.; Sultana, S.; Patel, N.; Bever, C.T. Brain derived neurotrophic factor treatment reduces inflammation and apoptosis in experimental allergic encephalomyelitis. J. Neurol. Sci. 2008, 270, 70–76. [Google Scholar] [CrossRef] [PubMed]
  97. Bovolenta, R.; Zucchini, S.; Paradiso, B.; Rodi, D.; Merigo, F.; Navarro Mora, G.; Osculati, F.; Berto, E.; Marconi, P.; Marzola, A.; et al. Hippocampal FGF-2 and BDNF overexpression attenuates epileptogenesis-associated neuroinflammation and reduces spontaneous recurrent seizures. J. Neuroinflamm. 2010, 7, 81. [Google Scholar] [CrossRef]
  98. Dong, Y.; Pu, K.; Duan, W.; Chen, H.; Chen, L.; Wang, Y. Involvement of Akt/CREB signaling pathways in the protective effect of EPA against interleukin-1beta-induced cytotoxicity and BDNF down-regulation in cultured rat hippocampal neurons. BMC Neurosci. 2018, 19, 52. [Google Scholar] [CrossRef]
  99. Qin, L.; Bouchard, R.; Pugazhenthi, S. Regulation of cyclic AMP response element-binding protein during neuroglial interactions. J. Neurochem. 2016, 136, 918–930. [Google Scholar] [CrossRef] [Green Version]
  100. Li, X.; Jope, R.S. Is glycogen synthase kinase-3 a central modulator in mood regulation? Neuropsychopharmacology 2010, 35, 2143–2154. [Google Scholar] [CrossRef]
  101. Suri, D.; Vaidya, V.A. Glucocorticoid regulation of brain-derived neurotrophic factor: Relevance to hippocampal structural and functional plasticity. Neuroscience 2013, 239, 196–213. [Google Scholar] [CrossRef]
  102. Jeanneteau, F.; Deinhardt, K.; Miyoshi, G.; Bennett, A.M.; Chao, M.V. The MAP kinase phosphatase MKP-1 regulates BDNF-induced axon branching. Nat. Neurosci. 2010, 13, 1373–1379. [Google Scholar] [CrossRef] [PubMed]
  103. Littlefield, A.M.; Setti, S.E.; Priester, C.; Kohman, R.A. Voluntary exercise attenuates LPS-induced reductions in neurogenesis and increases microglia expression of a proneurogenic phenotype in aged mice. J. Neuroinflamm. 2015, 12, 138. [Google Scholar] [CrossRef]
  104. Powers, S.K.; Jackson, M.J. Exercise-induced oxidative stress: Cellular mechanisms and impact on muscle force production. Physiol. Rev. 2008, 88, 1243–1276. [Google Scholar] [CrossRef] [PubMed]
  105. Radak, Z.; Taylor, A.W.; Ohno, H.; Goto, S. Adaptation to exercise-induced oxidative stress from muscle to brain. Exerc. Immunol. Rev. 2001, 7, 90–107. [Google Scholar] [PubMed]
  106. Chatterjee, S.; Noack, H.; Possel, H.; Keilhoff, G.; Wolf, G. Glutathione levels in primary glial cultures: Monochlorobimane provides evidence of cell type-specific distribution. Glia 1999, 27, 152–161. [Google Scholar] [CrossRef]
  107. Haddad, J.J.; Harb, H.L. l-gamma-Glutamyl-l-cysteinyl-glycine (glutathione; GSH) and GSH-related enzymes in the regulation of pro- and anti-inflammatory cytokines: A signaling transcriptional scenario for redox(y) immunologic sensor(s)? Mol. Immunol. 2005, 42, 987–1014. [Google Scholar] [CrossRef] [PubMed]
  108. Pocernich, C.B.; Butterfield, D.A. Elevation of glutathione as a therapeutic strategy in Alzheimer disease. Biochim. Biophys. Acta 2012, 1822, 625–630. [Google Scholar] [CrossRef] [Green Version]
  109. Lee, M.; Tazzari, V.; Giustarini, D.; Rossi, R.; Sparatore, A.; Del Soldato, P.; McGeer, E.; McGeer, P.L. Effects of hydrogen sulfide-releasing l-DOPA derivatives on glial activation: Potential for treating Parkinson disease. J. Biol. Chem. 2010, 285, 17318–17328. [Google Scholar] [CrossRef]
  110. Rojo, A.I.; McBean, G.; Cindric, M.; Egea, J.; Lopez, M.G.; Rada, P.; Zarkovic, N.; Cuadrado, A. Redox control of microglial function: Molecular mechanisms and functional significance. Antioxid. Redox Signal. 2014, 21, 1766–1801. [Google Scholar] [CrossRef]
  111. Koo, J.H.; Jang, Y.C.; Hwang, D.J.; Um, H.S.; Lee, N.H.; Jung, J.H.; Cho, J.Y. Treadmill exercise produces neuroprotective effects in a murine model of Parkinson’s disease by regulating the TLR2/MyD88/NF-κB signaling pathway. Neuroscience 2017, 356, 102–113. [Google Scholar] [CrossRef] [PubMed]
  112. Simioni, C.; Zauli, G.; Martelli, A.M.; Vitale, M.; Sacchetti, G.; Gonelli, A.; Neri, L.M. Oxidative stress: Role of physical exercise and antioxidant nutraceuticals in adulthood and aging. Oncotarget 2018, 9, 17181. [Google Scholar] [CrossRef] [PubMed]
  113. Iliff, J.J.; Wang, M.; Liao, Y.; Plogg, B.A.; Peng, W.; Gundersen, G.A.; Benveniste, H.; Vates, G.E.; Deane, R.; Goldman, S.A.; et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci. Transl. Med. 2012, 4, 147ra111. [Google Scholar] [CrossRef] [PubMed]
  114. Iliff, J.J.; Nedergaard, M. Is there a cerebral lymphatic system? Stroke 2013, 44, S93–S95. [Google Scholar] [CrossRef] [PubMed]
  115. Ma, Q.; Ineichen, B.V.; Detmar, M.; Proulx, S.T. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat. Commun. 2017, 8, 1434. [Google Scholar] [CrossRef] [PubMed]
  116. Mariathasan, S.; Newton, K.; Monack, D.M.; Vucic, D.; French, D.M.; Lee, W.P.; Roose-Girma, M.; Erickson, S.; Dixit, V.M. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature 2004, 430, 213. [Google Scholar] [CrossRef]
  117. Nedergaard, M. Garbage truck of the brain. Science 2013, 340, 1529–1530. [Google Scholar] [CrossRef]
  118. Kress, B.T.; Iliff, J.J.; Xia, M.; Wang, M.; Wei, H.S.; Zeppenfeld, D.; Xie, L.; Kang, H.; Xu, Q.; Liew, J.A.; et al. Impairment of paravascular clearance pathways in the aging brain. Ann. Neurol. 2014, 76, 845–861. [Google Scholar] [CrossRef]
  119. Peng, W.; Achariyar, T.M.; Li, B.; Liao, Y.; Mestre, H.; Hitomi, E.; Regan, S.; Kasper, T.; Peng, S.; Ding, F.; et al. Suppression of glymphatic fluid transport in a mouse model of Alzheimer’s disease. Neurobiol. Dis. 2016, 93, 215–225. [Google Scholar] [CrossRef]
  120. Von Holstein-Rathlou, S.; Petersen, N.C.; Nedergaard, M. Voluntary running enhances glymphatic influx in awake behaving, young mice. Neurosci. Lett. 2018, 662, 253–258. [Google Scholar] [CrossRef]
  121. Tarasoff-Conway, J.M.; Carare, R.O.; Osorio, R.S.; Glodzik, L.; Butler, T.; Fieremans, E.; Axel, L.; Rusinek, H.; Nicholson, C.; Zlokovic, B.V.; et al. Clearance systems in the brain-implications for Alzheimer disease. Nat. Rev. Neurol. 2015, 11, 457–470. [Google Scholar] [CrossRef] [PubMed]
  122. Zou, W.; Pu, T.; Feng, W.; Lu, M.; Zheng, Y.; Du, R.; Xiao, M.; Hu, G. Blocking meningeal lymphatic drainage aggravates Parkinson’s disease-like pathology in mice overexpressing mutated alpha-synuclein. Transl. Neurodegener. 2019, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, Y.; Xu, Y.; Sheng, H.; Ni, X.; Lu, J. Exercise amelioration of depression-like behavior in OVX mice is associated with suppression of NLRP3 inflammasome activation in hippocampus. Behav. Brain Res. 2016, 307, 18–24. [Google Scholar] [CrossRef] [PubMed]
  124. Real, C.C.; Garcia, P.C.; Britto, L.R.G. Treadmill Exercise Prevents Increase of Neuroinflammation Markers Involved in the Dopaminergic Damage of the 6-OHDA Parkinson’s Disease Model. J. Mol. Neurosci. 2017, 63, 36–49. [Google Scholar] [CrossRef] [PubMed]
  125. Real, C.C.; Doorduin, J.; Kopschina Feltes, P.; Vallez Garcia, D.; de Paula Faria, D.; Britto, L.R.; de Vries, E.F. Evaluation of exercise-induced modulation of glial activation and dopaminergic damage in a rat model of Parkinson’s disease using [(11)C]PBR28 and [(18)F]FDOPA PET. J. Cereb. Blood Flow Metab. 2019, 39, 989–1004. [Google Scholar] [CrossRef]
  126. Xu, Q.; Park, Y.; Huang, X.; Hollenbeck, A.; Blair, A.; Schatzkin, A.; Chen, H. Physical activities and future risk of Parkinson disease. Neurology 2010, 75, 341–348. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, X.; He, Q.; Huang, T.; Zhao, N.; Chen, X.; Li, T.; Bi, J. Treadmill exercise decreases Aβ deposition and counteracts cognitive decline in APP/PS1 mice, possibly via hippocampal microglia modifications. Front. Aging Neurosci. 2019, 11, 78. [Google Scholar] [CrossRef]
  128. Wang, W.-Y.; Tan, M.-S.; Yu, J.-T.; Tan, L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann. Transl. Med. 2015, 3. [Google Scholar] [CrossRef]
  129. Heneka, M.T.; Kummer, M.P.; Stutz, A.; Delekate, A.; Schwartz, S.; Vieira-Saecker, A.; Griep, A.; Axt, D.; Remus, A.; Tzeng, T.C.; et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 2013, 493, 674–678. [Google Scholar] [CrossRef]
  130. Jamilloux, Y.; Pierini, R.; Querenet, M.; Juruj, C.; Fauchais, A.L.; Jauberteau, M.O.; Jarraud, S.; Lina, G.; Etienne, J.; Roy, C.R. Inflammasome activation restricts Legionella pneumophila replication in primary microglial cells through flagellin detection. Glia 2013, 61, 539–549. [Google Scholar] [CrossRef]
  131. Gerard, C.; Rollins, B.J. Chemokines and disease. Nat. Immunol. 2001, 2, 108. [Google Scholar] [CrossRef] [PubMed]
  132. Carlin, J.L.; Grissom, N.; Ying, Z.; Gomez-Pinilla, F.; Reyes, T.M. Voluntary exercise blocks Western diet-induced gene expression of the chemokines CXCL10 and CCL2 in the prefrontal cortex. Brain Behav. Immun. 2016, 58, 82–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Rocha, E.M.; De Miranda, B.; Sanders, L.H. Alpha-synuclein: Pathology, mitochondrial dysfunction and neuroinflammation in Parkinson’s disease. Neurobiol. Dis. 2018, 109, 249–257. [Google Scholar] [CrossRef] [PubMed]
  134. Zhang, Q.-S.; Heng, Y.; Yuan, Y.-H.; Chen, N.-H. Pathological α-synuclein exacerbates the progression of Parkinson’s disease through microglial activation. Toxicol. Lett. 2017, 265, 30–37. [Google Scholar] [CrossRef] [PubMed]
  135. Kim, C.; Lee, H.-J.; Masliah, E.; Lee, S.-J. Non-Cell-Autonomous Neurotoxicity of α-Synuclein through Microglial Toll-Like Receptor 2. Exp. Neurobiol. 2016, 25, 113–119. [Google Scholar] [CrossRef] [PubMed]
  136. Kang, E.B.; Koo, J.H.; Jang, Y.C.; Yang, C.H.; Lee, Y.; Cosio-Lima, L.M.; Cho, J.Y. Neuroprotective Effects of Endurance Exercise against High-Fat Diet-Induced Hippocampal Neuroinflammation. J. Neuroendocrinol. 2016, 28. [Google Scholar] [CrossRef] [PubMed]
  137. Abraham, D.; Feher, J.; Scuderi, G.L.; Szabo, D.; Dobolyi, A.; Cservenak, M.; Juhasz, J.; Ligeti, B.; Pongor, S.; Gomez-Cabrera, M.C.; et al. Exercise and probiotics attenuate the development of Alzheimer’s disease in transgenic mice: Role of microbiome. Exp. Gerontol. 2019, 115, 122–131. [Google Scholar] [CrossRef]
  138. Dinan, T.G.; Cryan, J.F. Gut instincts: Microbiota as a key regulator of brain development, ageing and neurodegeneration. J. Physiol. 2017, 595, 489–503. [Google Scholar] [CrossRef]
  139. Lynch, S.V.; Pedersen, O. The Human Intestinal Microbiome in Health and Disease. N. Engl. J. Med. 2016, 375, 2369–2379. [Google Scholar] [CrossRef]
  140. Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbe communication in health and disease. Front. Physiol. 2011, 2, 94. [Google Scholar] [CrossRef]
  141. Stilling, R.M.; Bordenstein, S.R.; Dinan, T.G.; Cryan, J.F. Friends with social benefits: Host-microbe interactions as a driver of brain evolution and development? Front. Cell. Infect. Microbiol. 2014, 4, 147. [Google Scholar] [CrossRef] [PubMed]
  142. Riley, T.P.; Neal-McKinney, J.M.; Buelow, D.R.; Konkel, M.E.; Simasko, S.M. Capsaicin-sensitive vagal afferent neurons contribute to the detection of pathogenic bacterial colonization in the gut. J. Neuroimmunol. 2013, 257, 36–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Verbaan, D.; Marinus, J.; Visser, M.; van Rooden, S.M.; Stiggelbout, A.M.; van Hilten, J.J. Patient-reported autonomic symptoms in Parkinson disease. Neurology 2007, 69, 333–341. [Google Scholar] [CrossRef] [PubMed]
  144. Shannon, K.M.; Keshavarzian, A.; Dodiya, H.B.; Jakate, S.; Kordower, J.H. Is alpha-synuclein in the colon a biomarker for premotor Parkinson’s disease? Evidence from 3 cases. Mov. Disord. 2012, 27, 716–719. [Google Scholar] [CrossRef] [PubMed]
  145. Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480. [Google Scholar] [CrossRef]
  146. Tse, J.K. Gut microbiota, nitric oxide, and microglia as prerequisites for neurodegenerative disorders. ACS Chem. Neurosci. 2017, 8, 1438–1447. [Google Scholar] [CrossRef] [PubMed]
  147. Jiang, C.; Li, G.; Huang, P.; Liu, Z.; Zhao, B. The Gut Microbiota and Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 58, 1–15. [Google Scholar] [CrossRef]
  148. Kong, Y.; Jiang, B.; Luo, X. Gut microbiota influences Alzheimer’s disease pathogenesis by regulating acetate in Drosophila model. Fut. Microbiol. 2018, 13, 1117–1128. [Google Scholar] [CrossRef]
  149. O’Sullivan, O.; Cronin, O.; Clarke, S.F.; Murphy, E.F.; Molloy, M.G.; Shanahan, F.; Cotter, P.D. Exercise and the microbiota. Gut Microbes 2015, 6, 131–136. [Google Scholar] [CrossRef] [Green Version]
  150. Bonaz, B.; Sinniger, V.; Pellissier, S. The Vagus Nerve in the Neuro-Immune Axis: Implications in the Pathology of the Gastrointestinal Tract. Front. Immunol. 2017, 8, 1452. [Google Scholar] [CrossRef]
Table 1. Exercise regulates microglial activation by increasing anti-inflammatory factors.
Table 1. Exercise regulates microglial activation by increasing anti-inflammatory factors.
EffectsModelsTime, Frequency& Duration #Intensity $TypeReference
Cytokines↑ [IL-4]spinal cordMouse30 min/day, 5 days/week, Long-termLowTreadmill[37]
↑ [IL-10]skeletal muscleRat60 min/day, 5 days/week, Long-termLowTreadmill[38]
↑ [IL-6, IL-10]plasmaHuman3 h 26 minN/AMarathon[39]
Membrane proteins↑ [CD200, CD200R]midbrainMouse30 min/day, 5 days/week, Long-termLow & ModerateTreadmill[40]
↑ [TREM2]CSFHumanLong-termN/APhysical exercise[41]
↑ [Aquaporin-4]astrocyteMouseLong-termN/AWheel[42]
↑ [Aquaporin-4]astrocyteRat MouseShort- & long-termHighTreadmill &Wheel[43]
Metabolism↑ [SIRT-1]skeletal muscleRat60 & 90 min/day, 7 days/week, Long-termLow & HighTreadmill[44]
↑ [SIRT-1]cardiac muscleRat20–60 min/day, 5 days/week, Long-termN/ASwimming[45]
↑ [SIRT-1]skeletal muscleHuman30 sN/ASprint exercise[46]
↑ [leptin sensitivity]hippocampusMouseLong-term~5.6 ± 1.2 km/dayWheel[47]
Neurotrophic factors↑ [BDNF]serumRat30 min/day, Short-termLowTreadmill[48]
↑ [BDNF]serumHuman20 minHighCycling[49]
Heat shock proteins↑ [HSP60]subcutaneous adipose tissueHuman60 min/day, Long-termModerateAerobic, Treadmill, & Cycling[50]
↑ [HSP70]skeletal muscleRat60 min/day, Short-termMod/HighTreadmill[51]
↑ [HSP70]serum, skeletal muscleHuman60 min/day, Short-termModerateTreadmill[52]
Antioxidant↑ [Glutathione]skeletal muscleRat2 h/day, 5 days/week, Long-termHighTreadmill[53]
↑ [Glutathione]skeletal muscleDog40 km/day, 5 days/week, Long-termHighTreadmill[53]
# Exercise duration: acute: single bout; short-term: ≤2 weeks; long-term: >2 weeks. $ For human studies, we followed the criteria of Physical Activity Guidelines for Americans [54] to grade the intensity of exercise. For animal studies, we followed the method described in Inoue et al. [55] by using lactate threshold as a criterion to grade the intensity of exercise.
Table 2. Exercise regulates microglial activation by decreasing pro-inflammatory factors.
Table 2. Exercise regulates microglial activation by decreasing pro-inflammatory factors.
EffectsModelsTime, Frequency & Duration #Intensity $TypeReference
Toll-like receptors↓ [TLR2]substantia nigra Mouse60 min/day, 5 days/week, Long-termLowTreadmill [111]
↓ [TLR4]hippocampusRat30 min/day, 5 days/week, Long-termLowTreadmill [136]
Cytokines↓ [IL-1β, TNF-α]substantia nigra Mouse60 min/day, 5 days/week, Long-termLowTreadmill [111]
↓ [IL-1β, TNF-α] hippocampusMouse45 min/day, 5 days/week, Long-termLowTreadmill[127]
↓ [IL-1β, TNF-α]hippocampus Rat30 min/day, 5 days/week, Long-termLowTreadmill [136]
↓ [IL-1β, IL-18]hippocampusMouse60 min/day, 7 days/week, Long-termLowTreadmill [123]
Chemokines↓ [CCL2, CXCL10]prefrontal cortexMouse30 min/day, Long-termN/AWheel[132]
Inflammasome↓ [NLRP3]hippocampus Mouse60 min/day, 7 days/week, Long-termLowTreadmill [123]
Free radicals ↓ [NADPH oxidase]substantia nigra Mouse60 min/day, 5 days/week, Long-termLowTreadmill [111]
MicrobiomeShifting gut microbiotaMouse60 min/day, 4 days/week, Long-termLow/HighTreadmill[137]
# Exercise duration: acute: single bout; short-term: ≤2 weeks; long-term: >2 weeks. $ For human studies, we followed the criteria of Physical Activity Guidelines for Americans [54] to grade the intensity of exercise. For animal studies, we followed the method described in Inoue et al. [55] by using lactate threshold as a criterion to grade the intensity of exercise.

Share and Cite

MDPI and ACS Style

Mee-inta, O.; Zhao, Z.-W.; Kuo, Y.-M. Physical Exercise Inhibits Inflammation and Microglial Activation. Cells 2019, 8, 691. https://0-doi-org.brum.beds.ac.uk/10.3390/cells8070691

AMA Style

Mee-inta O, Zhao Z-W, Kuo Y-M. Physical Exercise Inhibits Inflammation and Microglial Activation. Cells. 2019; 8(7):691. https://0-doi-org.brum.beds.ac.uk/10.3390/cells8070691

Chicago/Turabian Style

Mee-inta, Onanong, Zi-Wei Zhao, and Yu-Min Kuo. 2019. "Physical Exercise Inhibits Inflammation and Microglial Activation" Cells 8, no. 7: 691. https://0-doi-org.brum.beds.ac.uk/10.3390/cells8070691

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop