AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
View PDF
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

Molecular mechanisms of glial cells in brain disorders following physical exercise

School of Traditional Chinese Medicine, Jinan University, 510632 Guangzhou, China
Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, 510632 Guangzhou, China
State Key Laboratory of Brain and Cognitive Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077 Hong Kong, China
Show Author Information

Abstract

Physical exercise generally improves health in humans and animals and may enhance cognitive function and neural function in the brain, especially in the prefrontal cortex and hippocampus. It is also a promising intervention for brain disorders such as psychiatric conditions and neurodegenerative diseases. The neuroprotective mechanisms of exercise are related to synaptic plasticity, neurogenesis, and autophagy. Moreover, the therapeutic effects of exercise are associated with glial cell function in the brain. In this review, we examine the relationship between glial cell function and brain disorders. We also consider the role of glial cells in modulating the effects of exercise on the molecular mechanisms and neural circuits involved in central and peripheral brain function. This review demonstrates that glial cells may play an important role in the effects of physical exercise interventions on the brain, particularly in those with neurological disorders.

References

[1]
Prakash RS, Voss MW, Erickson KI, et al. Physical activity and cognitive vitality. Annu Rev Psychol 2015, 66: 769–797.
[2]
Cassilhas RC, Tufik S, de Mello MT. Physical exercise, neuroplasticity, spatial learning and memory. Cell Mol Life Sci 2016, 73(5): 975–983.
[3]
Liang J, Wang HQ, Zeng Y, et al. Physical exercise promotes brain remodeling by regulating epigenetics, neuroplasticity and neurotrophins. Rev Neurosci 2021, 32(6): 615–629.
[4]
De Miguel Z, Khoury N, Betley MJ, et al. Exercise plasma boosts memory and dampens brain inflammation via clusterin. Nature 2021, 600(7889): 494–499.
[5]
De la Rosa A, Olaso-Gonzalez G, Arc-Chagnaud C, et al. Physical exercise in the prevention and treatment of Alzheimer’s disease. J Sport Health Sci 2020, 9(5): 394–404.
[6]
Hoffmann K, Sobol NA, Frederiksen KS, et al. Moderate-to-high intensity physical exercise in patients with Alzheimer’s disease: a randomized controlled trial. J Alzheimers Dis 2016, 50(2): 443–453.
[7]
Zhang SS, Zhu L, Peng Y, et al. Long-term running exercise improves cognitive function and promotes microglial glucose metabolism and morphological plasticity in the hippocampus of APP/PS1 mice. J Neuroinflammation 2022, 19(1): 34.
[8]
Choi SH, Bylykbashi E, Chatila ZK, et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model. Science 2018, 361(6406): eaan8821.
[9]
Shulman LM, Katzel LI, Ivey FM, et al. Randomized clinical trial of 3 types of physical exercise for patients with Parkinson disease. JAMA Neurol 2013, 70(2): 183–190.
[10]
Dutta D, Paidi RK, Raha S, et al. Treadmill exercise reduces α-synuclein spreading via PPARα. Cell Rep 2022, 40(2): 111058.
[11]
Fan TL, Li XT, Zhang X, et al. Influence of aerobic exercise training on mice gut microbiota in Parkinson’s disease. Turk J Biol 2022, 46(4): 288–297.
[12]
Murri MB, Ekkekakis P, Menchetti M, et al. Physical exercise for late-life depression: effects on symptom dimensions and time course. J Affect Disord 2018, 230: 65–70.
[13]
Stonerock GL, Hoffman BM, Smith PJ, et al. Exercise as treatment for anxiety: systematic review and analysis. Ann Behav Med 2015, 49(4): 542–556.
[14]
Wang YJ, Xu YJ, Sheng H, et al. 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.
[15]
Luo J, Tang CF, Chen XB, et al. Impacts of aerobic exercise on depression-like behaviors in chronic unpredictable mild stress mice and related factors in the AMPK/PGC-1α pathway. Int J Environ Res Public Health 2020, 17(6): 2042.
[16]
Moreno-Collazos JM, Orti ES. The effect of physical exercise on neurogenesis factor production in glial cells. Curr Pharm Des 2018, 24(1): 46–55.
[17]
Mee-Inta O, Zhao ZW, Kuo YM. Physical exercise inhibits inflammation and microglial activation. Cells 2019, 8(7): 691.
[18]
Leite MR, Cechella JL, Pinton S, et al. A diphenyl diselenide-supplemented diet and swimming exercise promote neuroprotection, reduced cell apoptosis and glial cell activation in the hypothalamus of old rats. Exp Gerontol 2016, 82: 1–7.
[19]
Zhang J, Guo YL, Wang YX, et al. Long-term treadmill exercise attenuates Aβ burdens and astrocyte activation in APP/PS1 mouse model of Alzheimer’s disease. Neurosci Lett 2018, 666: 70–77.
[20]
Saur L, Baptista PPA, de Senna PN, et al. Physical exercise increases GFAP expression and induces morphological changes in hippocampal astrocytes. Brain Struct Funct 2014, 219(1): 293–302.
[21]
Belaya I, Ivanova M, Sorvari A, et al. Astrocyte remodeling in the beneficial effects of long-term voluntary exercise in Alzheimer’s disease. J Neuroinflammation 2020, 17(1): 271.
[22]
Zhang WL, Chen HQ, Ding LY, et al. Microglial targeted therapy relieves cognitive impairment caused by Cntnap4 deficiency. Exploration 2023, 3(3): 20220160.
[23]
Zhang MR, Chen HQ, Zhang WL, et al. Biomimetic remodeling of microglial riboflavin metabolism ameliorates cognitive impairment by modulating neuroinflammation. Adv Sci 2023, 10(12): e2300180.
[24]
Zhang WL, Ding LY, Zhang MR, et al. Dietary intake of α-ketoglutarate ameliorates α-synuclein pathology in mouse models of Parkinson’s disease. Cell Mol Life Sci 2023, 80(6): 155.
[25]
Zhang WL, Ding LY, Chen HQ, et al. Cntnap4 partial deficiency exacerbates α-synuclein pathology through astrocyte-microglia C3-C3aR pathway. Cell Death Dis 2023, 14(4): 285.
[26]
Ho MS, Verkhratsky A, Duan SM, et al. Editorial: Glia in health and disease. Front Mol Neurosci 2019, 12: 63.
[27]
Bai J, Geng B, Wang XW, et al. Exercise facilitates the M1-to-M2 polarization of microglia by enhancing autophagy via the BDNF/AKT/mTOR pathway in neuropathic pain. Pain Physician 2022, 25(7): E1137–E1151.
[28]
Colonna M, Butovsky O. Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol 2017, 35: 441–468.
[29]
Bradl M, Lassmann H. Oligodendrocytes: biology and pathology. Acta Neuropathol 2010, 119(1): 37–53.
[30]
Guttikonda SR, Sikkema L, Tchieu J, et al. Fully defined human pluripotent stem cell-derived microglia and tri-culture system model C3 production in Alzheimer’s disease. Nat Neurosci 2021, 24(3): 343–354.
[31]
McAlpine CS, Park J, Griciuc A, et al. Astrocytic interleukin-3 programs microglia and limits Alzheimer’s disease. Nature 2021, 595(7869): 701–706.
[32]
Dowlati Y, Herrmann N, Swardfager W, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry 2010, 67(5): 446–457.
[33]
Enache D, Pariante CM, Mondelli V. Markers of central inflammation in major depressive disorder: a systematic review and meta-analysis of studies examining cerebrospinal fluid, positron emission tomography and post-mortem brain tissue. Brain Behav Immun 2019, 81: 24–40.
[34]
Liddelow SA, Guttenplan KA, Clarke LE, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541(7638): 481–487.
[35]
Wang Q, Jie W, Liu JH, et al. An astroglial basis of major depressive disorder? An overview. Glia 2017, 65(8): 1227–1250.
[36]
Zhou BT, Zhu ZQ, Ransom BR, et al. Oligodendrocyte lineage cells and depression. Mol Psychiatry 2021, 26(1): 103–117.
[37]
Seney ML, Huo ZG, Cahill K, et al. Opposite molecular signatures of depression in men and women. Biol Psychiatry 2018, 84(1): 18–27.
[38]
Ferraiuolo L, Meyer K, Sherwood TW, et al. Oligodendrocytes contribute to motor neuron death in ALS via SOD1-dependent mechanism. Proc Natl Acad Sci U S A 2016, 113(42): E6496–E6505.
[39]
Han PP, Zhang W, Kang L, et al. Clinical evidence of exercise benefits for stroke. Adv Exp Med Biol 2017, 1000: 131–151.
[40]
Cattadori G, Segurini C, Picozzi A, et al. Exercise and heart failure: an update. ESC Heart Fail 2018, 5(2): 222–232.
[41]
Ostman C, Jewiss D, King N, et al. Clinical outcomes to exercise training in type 1 diabetes: a systematic review and meta-analysis. Diabetes Res Clin Pract 2018, 139: 380–391.
[42]
Wegner M, Helmich I, Machado S, et al. Effects of exercise on anxiety and depression disorders: review of meta-analyses and neurobiological mechanisms. CNS Neurol Disord Drug Targets 2014, 13(6): 1002–1014.
[43]
Byun K, Hyodo K, Suwabe K, et al. Positive effect of acute mild exercise on executive function via arousal-related prefrontal activations: an fNIRS study. NeuroImage 2014, 98: 336–345.
[44]
Suwabe K, Hyodo K, Byun K, et al. Aerobic fitness associates with mnemonic discrimination as a mediator of physical activity effects: evidence for memory flexibility in young adults. Sci Rep 2017, 7(1): 5140.
[45]
Castells-Sánchez A, Roig-Coll F, Dacosta-Aguayo R, et al. Exercise and fitness neuroprotective effects: molecular, brain volume and psychological correlates and their mediating role in healthy late-middle-aged women and men. Front Aging Neurosci 2021, 13: 615247.
[46]
Lourenco MV, Frozza RL, de Freitas GB, et al. Exercise-linked FNDC5/irisin rescues synaptic plasticity and memory defects in Alzheimer’s models. Nat Med 2019, 25(1): 165–175.
[47]
Klein C, Jonas W, Iggena D, et al. Exercise prevents high-fat diet-induced impairment of flexible memory expression in the water maze and modulates adult hippocampal neurogenesis in mice. Neurobiol Learn Mem 2016, 131: 26–35.
[48]
Morgan JA, Singhal G, Corrigan F, et al. The effects of aerobic exercise on depression-like, anxiety-like, and cognition-like behaviours over the healthy adult lifespan of C57BL/6 mice. Behav Brain Res 2018, 337: 193–203.
[49]
Xiao K, Luo YM, Liang X, et al. Beneficial effects of running exercise on hippocampal microglia and neuroinflammation in chronic unpredictable stress-induced depression model rats. Transl Psychiatry 2021, 11(1): 461.
[50]
Chen K, Zheng YH, Wei JA, et al. Exercise training improves motor skill learning via selective activation of mTOR. Sci Adv 2019, 5(7): eaaw1888.
[51]
Deldicque L, Theisen D, Francaux M. Regulation of mTOR by amino acids and resistance exercise in skeletal muscle. Eur J Appl Physiol 2005, 94(1/2): 1–10.
[52]
Ogasawara R, Fujita S, Hornberger TA, et al. The role of mTOR signalling in the regulation of skeletal muscle mass in a rodent model of resistance exercise. Sci Rep 2016, 6: 31142.
[53]
Lloyd BA, Hake HS, Ishiwata T, et al. Exercise increases mTOR signaling in brain regions involved in cognition and emotional behavior. Behav Brain Res 2017, 323: 56–67.
[54]
Yan L, Wang M, Yang FZ, et al. Physical exercise mediates a cortical FMRP-mTOR pathway to improve resilience against chronic stress in adolescent mice. Transl Psychiatry 2023, 13(1): 16.
[55]
Yau SY, Li A, Hoo RLC, et al. Physical exercise-induced hippocampal neurogenesis and antidepressant effects are mediated by the adipocyte hormone adiponectin. Proc Natl Acad Sci U S A 2014, 111(44): 15810–15815.
[56]
Sleiman SF, Henry J, Al-Haddad R, et al. Exercise promotes the expression of brain derived neurotrophic factor (BDNF) through the action of the ketone body β-hydroxybutyrate. Elife 2016, 5: e15092.
[57]
Bayod S, Del Valle J, Canudas AM, et al. Long-term treadmill exercise induces neuroprotective molecular changes in rat brain. J Appl Physiol 2011, 111(5): 1380–1390.
[58]
Cheng T, Huang XD, Hu XF, et al. Physical exercise rescues cocaine-evoked synaptic deficits in motor cortex. Mol Psychiatry 2021, 26(11): 6187–6197.
[59]
He XF, Liu DX, Zhang Q, et al. Voluntary exercise promotes glymphatic clearance of amyloid beta and reduces the activation of astrocytes and microglia in aged mice. Front Mol Neurosci 2017, 10: 144.
[60]
Vasconcelos-Filho FSL, da Rocha Oliveira LC, de Freitas TBC, et al. Neuroprotective mechanisms of chronic physical exercise via reduction of β-amyloid protein in experimental models of Alzheimer’s disease: a systematic review. Life Sci 2021, 275: 119372.
[61]
Tsai WL, Chen HY, Huang YZ, et al. Long-term voluntary physical exercise exerts neuroprotective effects and motor disturbance alleviation in a rat model of Parkinson’s disease. Behav Neurol 2019, 2019: 4829572.
[62]
Wang J, Tang J, Liang X, et al. Hippocampal PGC-1α-mediated positive effects on parvalbumin interneurons are required for the antidepressant effects of running exercise. Transl Psychiatry 2021, 11(1): 1–11.
[63]
Tang J, Liang X, Dou XY, et al. Exercise rather than fluoxetine promotes oligodendrocyte differentiation and myelination in the hippocampus in a male mouse model of depression. Transl Psychiatry 2021, 11(1): 622.
[64]
Li Y, Luo YM, Tang J, et al. The positive effects of running exercise on hippocampal astrocytes in a rat model of depression. Transl Psychiatry 2021, 11(1): 83.
[65]
Littlefield AM, Setti SE, Priester C, et al. Voluntary exercise attenuates LPS-induced reductions in neurogenesis and increases microglia expression of a proneurogenic phenotype in aged mice. J Neuroinflammation 2015, 12: 138.
[66]
Ke HC, Huang HJ, Liang KC, et al. Selective improvement of cognitive function in adult and aged APP/PS1 transgenic mice by continuous non-shock treadmill exercise. Brain Res 2011, 1403: 1–11.
[67]
Xu ZQ, Zhang LQ, Wang Q, et al. Aerobic Exercise Combined with Antioxidative Treatment does not Counteract Moderate- or Mid-Stage Alzheimer-Like Pathophysiology of APP/PS1 Mice. CNS Neurosci Ther 2013, 19(10): 795–803.
[68]
Rodrigues L, Dutra MF, Ilha J, et al. Treadmill training restores spatial cognitive deficits and neurochemical alterations in the hippocampus of rats submitted to an intracerebroventricular administration of streptozotocin. J Neural Transm 2010, 117(11): 1295–1305.
[69]
Seo DY, Heo JW, Ko JR, et al. Exercise and neuroinflammation in health and disease. Int Neurourol J 2019, 23(Suppl 2): S82–S92.
[70]
Koo JH, Jang YC, Hwang DJ, et al. 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.
[71]
Vitorino LC, Oliveira KF, da Silva WAB, et al. Physical exercise influences astrocytes in the striatum of a Parkinson’s disease male mouse model. Neurosci Lett 2022, 771: 136466.
[72]
Real CC, Garcia PC, Britto LRG. 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(1): 36–49.
[73]
Santin K, da Rocha RF, Cechetti F, et al. Moderate exercise training and chronic caloric restriction modulate redox status in rat hippocampus. Brain Res 2011, 1421: 1–10.
[74]
Kassa RM, Bonafede R, Boschi F, et al. Effect of physical exercise and anabolic steroid treatment on spinal motoneurons and surrounding glia of wild-type and ALS mice. Brain Res 2017, 1657: 269–278.
[75]
da Silva PGC, Domingues DD, de Carvalho LA, et al. Neurotrophic factors in Parkinson’s disease are regulated by exercise: evidence-based practice. J Neurol Sci 2016, 363: 5–15.
[76]
Miranda M, Morici JF, Zanoni MB, et al. Brain-derived neurotrophic factor: a key molecule for memory in the healthy and the pathological brain. Front Cell Neurosci 2019, 13: 363.
[77]
Kim K, Sung YH, Seo JH, et al. Effects of treadmill exercise-intensity on short-term memory in the rats born of the lipopolysaccharide-exposed maternal rats. J Exerc Rehabil 2015, 11(6): 296–302.
[78]
Vaynman S, Ying Z, Gomez-Pinilla F. Hippocampal BDNF mediates the efficacy of exercise on synaptic plasticity and cognition. Eur J Neurosci 2004, 20(10): 2580–2590.
[79]
Lin LF H, Doherty DH, Lile JD, et al. GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science 1993, 260(5111): 1130–1132.
[80]
McCullough MJ, Gyorkos AM, Spitsbergen JM. Short-term exercise increases GDNF protein levels in the spinal cord of young and old rats. Neuroscience 2013, 240: 258–268.
[81]
Cintrón-Colón AF, Spitsbergen JM, Effect of long-term exercise on GDNF expression and innervation in rat skeletal muscle. The FASEB Journal 2019, 33(S1): 700.25–700.25.
[82]
Hosgorler F, Kizildag S, Koc B, et al. Mild-intensity exercise triggers VEGF in the digestive tract via both hypoxic and nonhypoxic mechanisms. Braz Arch Biol Technol 2020, 63: 1–11.
[83]
Lopez-Lopez C, LeRoith D, Torres-Aleman I. Insulin-like growth factor I is required for vessel remodeling in the adult brain. PNAS 2004, 101(26): 9833–9838.
[84]
Majorczyk M, Smoląg D. Effect of physical activity on IGF-1 and IGFBP levels in the context of civilization diseases prevention. Rocz Panstw Zakl Hig 2016, 67(2): 105–111.
[85]
He YM, Wang Q, Wu HD, et al. The role of IGF-1 in exercise to improve obesity-related cognitive dysfunction. Front Neurosci 2023, 17: 1229165.
[86]
Mason JL, Xuan SH, Dragatsis I, et al. Insulin-like growth factor (IGF) signaling through type 1 IGF receptor plays an important role in remyelination. J Neurosci 2003, 23(20): 7710–7718.
[87]
He L, Lu QR. Coordinated control of oligodendrocyte development by extrinsic and intrinsic signaling cues. Neurosci Bull 2013, 29(2): 129–143.
[88]
Janowska J, Gargas J, Ziemka-Nalecz M, et al. Oligodendrocyte response to pathophysiological conditions triggered by episode of perinatal hypoxia-ischemia: role of IGF-1 secretion by glial cells. Mol Neurobiol 2020, 57(10): 4250–4268.
[89]
Munyeshyaka M, Fields RD. Oligodendroglia are emerging players in several forms of learning and memory. Commun Biol 2022, 5(1): 1148.
[90]
Boström P, Wu J, Jedrychowski MP, et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012, 481(7382): 463–468.
[91]
Pedersen L, Idorn M, Olofsson GH, et al. Voluntary running suppresses tumor growth through epinephrine- and IL-6-dependent NK cell mobilization and redistribution. Cell Metab 2016, 23(3): 554–562.
Stress and Brain
Pages 179-190
Cite this article:
Liu Y, Ran S, So K-F, et al. Molecular mechanisms of glial cells in brain disorders following physical exercise. Stress and Brain, 2023, 3(4): 179-190. https://doi.org/10.26599/SAB.2023.9060004

480

Views

62

Downloads

0

Crossref

Altmetrics

Received: 28 November 2023
Revised: 15 December 2023
Accepted: 15 December 2023
Published: 05 December 2023
© The Author(s) 2023

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attributtion-NonCommercial 4.0 License (http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission.

Return