Exercise Regulates Microglial Activation by Increasing Anti-Inflammatory Factors: Comparison
Please note this is a comparison between Version 3 by Jessie Wu and Version 2 by Jessie Wu.

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. 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. 

  • anti-inflammatory
  • excercise

1. Anti-Inflammatory Cytokines

In response to muscular contractions, myocytes produce and release numerous molecules, termed myokines [1]. Among them, IL-6 was the first identified myokine [1][2][3]. 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 [4][5]. 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 [6]. 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) [7]. Interestingly, the levels of IL-6 are reduced in several neurodegenerative diseases, including AD and Parkinson’s disease (PD) [5].
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 [8]. 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 [9]. These anti-inflammatory myokines can be readily transported into the central nervous system (CNS) from the peripheral circulation [10]. 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 [11].
Moreover, exercise can also stimulate the expression of IL-1Ra in the CNS [9]. 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 [12]. Therefore, exercise can upregulate the expression of anti-inflammatory cytokines and inhibit microglial activation.

2. Cluster of D200-CD200ifferentiation-200-Cluster of Differentiation-200 Receptor 

Cluster of differentiation-200 (CD200) is an immunomodulatory factor expressed by neurons in the CNS [13]. It inhibits microglial activation through binding with CD200R on microglial plasma membranes [13]. It has been demonstrated that long-term treadmill running increases expression levels of both CD200 and CD200R in mouse brains [14]. 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 [15][16][17]. Exercise may increase the expression of CD200/CD200R to suppress MAPK signaling pathway and inhibit microglial activation.

3. Triggering REMeceptor Expressed on Myeloid Cell-2

Triggering receptor expressed on myeloid cell-2 (TREM2), an immunoglobulin superfamily receptor, is mainly expressed by microglia in the CNS [18]. It plays essential roles in microglial phagocytosis and cytoskeleton rearrangement [19]. A soluble form of TREM2, derived from the cell surface receptor, can promote the survival of microglia and their phagocytic activity [20][21]. It has also been suggested that increasing TREM2 levels is an effective therapeutic approach for delaying the pathogenesis of AD [20]. 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 [22]. 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 [23][24]. Thus, exercise may regulate the activation status of microglia via upregulation of TREM2.

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) [25][26][27][28] and their functions are multifaceted, but mainly in maintaining cellular homeostasis and protein stability [29]. HSPs are associated with endotoxin tolerance response [30]. 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 [31][32][33][34]. Therefore, exercise-induced upregulation of HSPs may negatively regulate the productions of pro-inflammatory cytokines from glial cells.

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 [35][36][37]. SIRT1 plays critical roles in regulating inflammation and cell survival by deacetylation of transcription factors [38]. Overexpression of SIRT1 deacetylates the NF-κB subunit p65, hence suppresses its activity, resulting in decreased production of proinflammatory cytokines [39][40][41]. Moreover, SIRT1 can activate the expression of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) to stimulate the antioxidant response [42]. Nrf2-activated gene expression leads to antioxidant enzymes being released, including glutathione peroxidase and heme oxygenase-1 [43]. Another function of Nrf2 is an antagonist of the NF-κB cascade, which causes reduced production of proinflammatory mediators [44]. 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 [45][46]. 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].

6. Brain-Derived Neurotrophic Factor (BDNF)

Brain-derived neurotrophic factor (BDNF) is one of the most studied neurotrophic factors whose production in the brain is increased by exercise [48][49][50]. 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 [50]. 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 [51]. The circulating irisin then enters the brain and upregulates BDNF expression in the brain [51]. Alternatively, ketone body β-hydroxybutyrate released from the liver during exercise has also been shown to induce BDNF expression in the brain [52]. BDNF can regulate brain functions in multiple aspects, including neuronal cell survival, adult hippocampal neurogenesis, and neuroplasticity [53]. Furthermore, BDNF can alleviate microglial activation in several brain disease models [54][55][56][57]. 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, wresearchers 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 [58][59]. 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 [60]. BDNF is also known to modulate mitogen-activated protein kinase phosphatase 1, resulting in the reduction of p38 and JNK phosphorylation [61][62][63]. 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.

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 [64][65][66]. Noticeably, GSH plays vital roles in maintaining redox balance and anti-inflammatory mediators found in astrocytes and microglia [67][68]. 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 [69][70][71], 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 [72]. 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 [73].

8. Glymphatic System

The glymphatic system is known as the brain waste clearance system, involving glial cells and lymphatic vessels [74][75]. 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 [76]. 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 [77][78]. Impaired glymphatic function is evident in aging and neurodegenerative diseases [79][80]. On the other hand, both voluntary wheel and mandatory treadmill exercise enhanced the efficiency of glymphatic system in normal aging and AD mice [81][82]. Furthermore, the degrees of astrocytic and microglial activations in aged mice were also decreased, whereas the amount of aquaporin-4 increased through exercise [83]. 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 [84][85].

9. 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 [86][87][88][89]. 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 [90]. IL-1β, IL-18, and TNF-α are considered the major proinflammatory cytokines in the CNS [91]. IL-1β and IL-18 can promote inflammasome production to enhance the inflammatory response [77]. 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 [92]. 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 [92][93][94]. 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 [86].
Chemokines are responsible for the recruitment of cells of monocytic origin to sites of inflammation, which may aggravate the degrees of local microglial activation [95]. 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 [96]. Long-term wheel running is known to decrease the expression levels of CXCL10 and CCL2, which may lessen the degrees of subsequent microglial activation [96].

References

  1. Pedersen, B.K.; Akerstrom, T.C.A. Role of myokines in exercise and metabolism. J. Appl. Physiol. 2007, 103, 1093–1098.
  2. Pedersen, B.K. Muscular interleukin-6 and its role as an energy sensor. Med. Sci. Sports Exerc. 2012, 44, 392–396.
  3. 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.
  4. 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.
  5. 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.
  6. Pedersen, B.K.; Steensberg, A.; Schjerling, P. Muscle-derived interleukin-6: Possible biological effects. J. Physiol. 2001, 536, 329–337.
  7. 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.
  8. 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.
  9. 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.
  10. Kelly, A.M. Exercise-Induced Modulation of Neuroinflammation in Models of Alzheimer’s Disease. Brain Plast. 2018, 4, 81–94.
  11. 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.
  12. Rosenzweig, J.M.; Lei, J.; Burd, I. Interleukin-1 receptor blockade in perinatal brain injury. Front. Pediatr. 2014, 2, 108.
  13. Biber, K.; Neumann, H.; Inoue, K.; Boddeke, H.W. Neuronal ‘On’ and ‘Off’ signals control microglia. Trends Neurosci. 2007, 30, 596–602.
  14. 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.
  15. 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.
  16. 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.
  17. 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.
  18. Colonna, M.; Wang, Y. TREM2 variants: New keys to decipher Alzheimer disease pathogenesis. Nat. Rev. Neurosci. 2016, 17, 201–207.
  19. 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.
  20. 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.
  21. 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.
  22. 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.
  23. 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.
  24. Turnbull, I.R.; Colonna, M. Activating and inhibitory functions of DAP12. Nat. Rev. Immunol. 2007, 7, 155.
  25. 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.
  26. Milne, K.J.; Noble, E.G. Exercise-induced elevation of HSP70 is intensity dependent. J. Appl. Physiol. 2002, 93, 561–568.
  27. 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.
  28. Flynn, M.G.; McFarlin, B.K.; Markofski, M.M. The Anti-Inflammatory Actions of Exercise Training. Am. J. Lifestyle Med. 2007, 1, 220–235.
  29. Fehrenbach, E.; Niess, A.M. Role of heat shock proteins in the exercise response. Exerc. Immunol. Rev. 1999, 5, 57–77.
  30. Fan, H.; Cook, J.A. Molecular mechanisms of endotoxin tolerance. J. Endotoxin Res. 2004, 10, 71–84.
  31. 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.
  32. 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.
  33. 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.
  34. 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.
  35. 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.
  36. 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.
  37. 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.
  38. Haigis, M.C.; Sinclair, D.A. Mammalian sirtuins: Biological insights and disease relevance. Annu. Rev. Pathol. 2010, 5, 253–295.
  39. 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.
  40. 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.
  41. Mendes, K.L.; Lelis, D.F.; Santos, S.H.S. Nuclear sirtuins and inflammatory signaling pathways. Cytokine Growth Factor Rev. 2017, 38, 98–105.
  42. 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.
  43. Jung, K.A.; Kwak, M.K. The Nrf2 system as a potential target for the development of indirect antioxidants. Molecules 2010, 15, 7266–7291.
  44. 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.
  45. 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.
  46. 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.
  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.
  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.
  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.
  50. 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.
  51. 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.
  52. 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.
  53. 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.
  54. 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.
  55. 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.
  56. 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.
  57. 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.
  58. 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.
  59. Qin, L.; Bouchard, R.; Pugazhenthi, S. Regulation of cyclic AMP response element-binding protein during neuroglial interactions. J. Neurochem. 2016, 136, 918–930.
  60. Li, X.; Jope, R.S. Is glycogen synthase kinase-3 a central modulator in mood regulation? Neuropsychopharmacology 2010, 35, 2143–2154.
  61. Suri, D.; Vaidya, V.A. Glucocorticoid regulation of brain-derived neurotrophic factor: Relevance to hippocampal structural and functional plasticity. Neuroscience 2013, 239, 196–213.
  62. 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.
  63. 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.
  64. 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.
  65. Powers, S.K.; Jackson, M.J. Exercise-induced oxidative stress: Cellular mechanisms and impact on muscle force production. Physiol. Rev. 2008, 88, 1243–1276.
  66. 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.
  67. 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.
  68. 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.
  69. Pocernich, C.B.; Butterfield, D.A. Elevation of glutathione as a therapeutic strategy in Alzheimer disease. Biochim. Biophys. Acta 2012, 1822, 625–630.
  70. 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.
  71. 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.
  72. 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.
  73. 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.
  74. 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.
  75. Iliff, J.J.; Nedergaard, M. Is there a cerebral lymphatic system? Stroke 2013, 44, S93–S95.
  76. 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.
  77. 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.
  78. Nedergaard, M. Garbage truck of the brain. Science 2013, 340, 1529–1530.
  79. 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.
  80. 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.
  81. 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.
  82. 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.
  83. 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.
  84. 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.
  85. 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.
  86. 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.
  87. 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.
  88. 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 PBR28 and FDOPA PET. J. Cereb. Blood Flow Metab. 2019, 39, 989–1004.
  89. 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.
  90. 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.
  91. 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.
  92. 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.
  93. 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.
  94. 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.
  95. Gerard, C.; Rollins, B.J. Chemokines and disease. Nat. Immunol. 2001, 2, 108.
  96. 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.
More
Video Production Service