Generic placeholder image

当代阿耳茨海默病研究

Editor-in-Chief

ISSN (Print): 1567-2050
ISSN (Online): 1875-5828

Research Article

高强度间歇训练在阿尔茨海默病大鼠模型中预防认知障碍并增加肌肉基因FNDC5和PPARGC1A的表达

卷 19, 期 12, 2022

发表于: 30 December, 2022

页: [830 - 840] 页: 11

弟呕挨: 10.2174/1567205020666221207103109

价格: $65

摘要

背景:阿尔茨海默病是世界上最常见的神经退行性疾病,以神经元结构和功能的进行性丧失为特征,其主要组织病理学标志是脑内β-淀粉样蛋白的积累。 目的:众所周知,运动是一种神经保护因素,肌肉产生和释放肌肉因子,在炎症和代谢功能障碍中发挥内分泌作用。因此,本研究旨在通过长期HIIT训练与注射β淀粉样蛋白1-42引起的阿尔茨海默氏症模型认知损伤建立运动益处之间的关系。 方法:为此,48只雄性Wistar大鼠被分为四组:久坐假组(SS)、训练假组(ST)、久坐阿兹海默症组(AS)和训练阿兹海默症组(AT)。动物接受立体定向手术,并接受a β1-42或生理盐水的海马注射。术后7天,12天的跑步机适应,随后进行5次最大跑步测试(MRT)和55天的HIIT,大鼠进行莫里斯水迷宫测试。然后对动物实施安乐死,提取腓肠肌组织,通过qRT-PCR分析Fibronectin type III domain containing 5 (FNDC5)、PPARG Coactivator 1 Alpha (PPARGC1A)和Integrin subunit beta 5 (ITGB5-R)的表达以及横截面积。 结果:HIIT可防止淀粉样蛋白β1-42输入引起的认知缺陷(p < 0.0001),引起肌纤维的适应(p < 0.0001),调节FNDC5 (p < 0.01)、ITGB5 (p < 0.01)和PPARGC1A (p < 0.01)的基因表达,并诱导FNDC5外周蛋白表达的增加(p < 0.005)。 结论:因此,我们得出结论,HIIT可以预防Aβ1-42输注引起的认知损伤,构成一种调节重要遗传和蛋白质途径的非药物工具。

关键词: 阿尔茨海默病,鸢尾素,运动,HIIT, Aβ1-42, FNDC5, PPARGC1A。

[1]
Calabrò M, Rinaldi C, Santoro G, Crisafulli C. The biological pathways of Alzheimer's disease: A review. AIMS Neurosci 2021; 8.1: 86.
[2]
Tönnies E, Trushina E. Oxidative stress, synaptic dysfunction, and Alzheimer’s disease. J Alzheimers Dis 2017; 57(4): 1105-21.
[http://dx.doi.org/10.3233/JAD-161088] [PMID: 28059794]
[3]
Borumandnia N, Majd HA, Khadembashi N, Olazadeh K, Alaii H. Worldwide patterns in Alzheimer’s disease and other dementias prevalence from 1990 to 2017: A growth mixture models approach. BMC Neurol 2020; 2020: 1-9.
[4]
Valenzuela PL, Castillo-García A, Morales JS, et al. Exercise benefits on Alzheimer’s disease: State-of-the-science. Ageing Res Rev 2020; 62: 101108.
[http://dx.doi.org/10.1016/j.arr.2020.101108] [PMID: 32561386]
[5]
Di Liegro CM, Schiera G, Proia P, Di Liegro I. Physical activity and brain health. Genes (Basel) 2019; 10(9): 720.
[http://dx.doi.org/10.3390/genes10090720] [PMID: 31533339]
[6]
Nay K, Smiles WJ, Kaiser J, et al. Molecular mechanisms underlying the beneficial effects of exercise on brain function and neurological disorders. Int J Mol Sci 2021; 22(8): 4052.
[http://dx.doi.org/10.3390/ijms22084052] [PMID: 33919972]
[7]
Jin Y, Sumsuzzman D, Choi J, Kang H, Lee SR, Hong Y. Molecular and functional interaction of the myokine irisin with physical exercise and Alzheimer’s disease. Molecules 2018; 23(12): 3229.
[http://dx.doi.org/10.3390/molecules23123229] [PMID: 30544500]
[8]
Erickson K, Hillman C, Stillman CM, et al. Physical activity, cognition, and brain outcomes: A review of the 2018 physical activity guidelines. Med Sci Sports Exerc 2019; 51(6): 1242-51.
[http://dx.doi.org/10.1249/MSS.0000000000001936] [PMID: 31095081]
[9]
Cao RY, Zheng H, Redfearn D, Yang J. FNDC5: A novel player in metabolism and metabolic syndrome. Biochimie 2019; 158: 111-6.
[http://dx.doi.org/10.1016/j.biochi.2019.01.001] [PMID: 30611879]
[10]
Ferrer-Martínez A, Ruiz-Lozano P, Chien KR. Mouse PeP: A novel peroxisomal protein linked to myoblast differentiation and development. Dev Dyn 2002; 224(2): 154-67.
[http://dx.doi.org/10.1002/dvdy.10099]
[11]
Tadaishi M, Miura S, Kai Y, Kano Y, Oishi Y, Ezaki O. Skeletal muscle-specific expression of PGC-1α-b, an exercise-responsive isoform, increases exercise capacity and peak oxygen uptake. PLoS One 2011; 6(12): e28290.
[http://dx.doi.org/10.1371/journal.pone.0028290] [PMID: 22174785]
[12]
Boström P, Wu J, Jedrychowski MP, et al. A PGC1α-dependent myokine that drives browning of white fat and thermogenesis. Nature 2012; 481(7382): 463-8.
[13]
Kim H, Wrann CD, Jedrychowski M, et al. The irisin mediates effects on bone and fat through αV integrin receptors. Cell 2018; 175: 1756-68-e1717.
[http://dx.doi.org/10.1016/j.cell.2018.10.025]
[14]
Paxinos G, Watson C. The Rat brain: in stereotaxic coordinates. (6th ed.), Amsterdam: Elsevier 2009.
[15]
Zhang R, Alushin GM, Brown A, Nogales E. Mechanistic origin of microtubule dynamic instability and its modulation by EB proteins. Cell 2015; 162(4): 849-59.
[http://dx.doi.org/10.1016/j.cell.2015.07.012] [PMID: 26234155]
[16]
Zhou W, Lu S, Su Y, et al. Decreasing oxidative stress and neuroinflammation with a multifunctional peptide rescues memory deficits in mice with Alzheimer disease. Free Radic Biol Med 2014; 74: 50-63.
[http://dx.doi.org/10.1016/j.freeradbiomed.2014.06.013] [PMID: 24960578]
[17]
Lee HE, Kim DH, Park SJ, et al. Neuroprotective effect of sinapic acid in a mouse model of amyloid β1-42 protein-induced Alzheimer’s disease. Pharmacol Biochem Behav 2012; 103(2): 260-6.
[http://dx.doi.org/10.1016/j.pbb.2012.08.015] [PMID: 22971592]
[18]
Takeda S, Sato N, Niisato K, et al. Validation of Aβ1-40 administration into mouse cerebroventricles as an animal model for Alzheimer disease. Brain Res 2009; 1280: 137-47.
[http://dx.doi.org/10.1016/j.brainres.2009.05.035] [PMID: 19464276]
[19]
Morris JK, et al. Is Alzheimer’s disease a systemic disease? Biochim Biophys Acta 2014; 1842(9): 1340-9.
[http://dx.doi.org/10.1016/j.bbadis.2014.04.012]
[20]
Morris JK, Vidoni ED, Johnson DK, Van Sciver A, Mahnken JD, Honea RA. Aerobic exercise for Alzheimer’s disease: A randomized controlled pilot trial. PLoS One 2017; 12(2): e0170547.
[http://dx.doi.org/10.1371/journal.pone.0170547]
[21]
Vieira WHB, Goes R, Costa FC, et al. Adaptação enzimática da LDH em ratos submetidos a treinamento aeróbio em esteira e laser de baixa intensidade. Braz J Phys Ther 2006; 10(2): 205-11.
[http://dx.doi.org/10.1590/S1413-35552006000200011]
[22]
Jones JH. Resource Book for the Design of Animal Exercise Protocols. Am J Vet Res 2007; 68(6): 583-3.
[http://dx.doi.org/10.2460/ajvr.68.6.583]
[23]
Segal SK, Cotman CW, Cahill LF. Exercise-induced noradrenergic activation enhances memory consolidation in both normal aging and patients with amnestic mild cognitive impairment. J Alzheimers Dis 2012; 32(4): 1011-8.
[24]
Kobilo T, Liu QR, Gandhi K, Mughal M, Shaham Y, van Praag H. Running is the neurogenic and neurotrophic stimulus in environmental enrichment. Learn Mem 2011; 18(9): 605-9.
[http://dx.doi.org/10.1101/lm.2283011] [PMID: 21878528]
[25]
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): 165-75.
[http://dx.doi.org/10.1038/s41591-018-0275-4] [PMID: 30617325]
[26]
Kuipers SD, Bramham CR. Brain-derived neurotrophic factor mechanisms and function in adult synaptic plasticity: new insights and implications for therapy. Curr Opin Drug Discov Devel 2006; 9(5): 580-6.
[PMID: 17002218]
[27]
Erickson HP. Irisin and FNDC5 in retrospect. Adipocyte 2013; 2(4): 289-93.
[http://dx.doi.org/10.4161/adip.26082] [PMID: 24052909]
[28]
Shan T, Liang X, Bi P, Kuang S. Myostatin knockout drives browning of white adipose tissue through activating the AMPK‐PGC1α‐Fndc5 pathway in muscle. FASEB J 2013; 27(5): 1981-9.
[http://dx.doi.org/10.1096/fj.12-225755] [PMID: 23362117]
[29]
López-Ortiz S, Pinto-Fraga J, Valenzuela PL, et al. Physical exercise and Alzheimer’s disease: Effects on pathophysiological molecular pathways of the disease. Int J Mol Sci 2021; 22(6): 2897.
[http://dx.doi.org/10.3390/ijms22062897] [PMID: 33809300]
[30]
Ramos JS, Dalleck LC, Tjonna AE, Beetham KS, Coombes JS. The impact of high-intensity interval training versus moderate-intensity continuous training on vascular function: a systematic review and meta-analysis. Sports Med 2015; 45(5): 679-92.
[http://dx.doi.org/10.1007/s40279-015-0321-z] [PMID: 25771785]
[31]
Daskalopoulou SS, Cooke AB, Gomez YH, et al. Plasma irisin levels progressively increase in response to increasing exercise workloads in young, healthy, active subjects. Eur J Endocrinol 2014; 171(3): 343-52.
[http://dx.doi.org/10.1530/EJE-14-0204]
[32]
Benedini S, Dozio E, Invernizzi PL, et al. Irisin: A potential link between physical exercise and metabolism-an observational study in differently trained subjects, from elite athletes to sedentary people. J Diabetes Res 2017; 2017: 1039161.
[http://dx.doi.org/10.1155/2017/1039161] [PMID: 28386566]
[33]
Nygaard H, Slettaløkken G, Vegge G, et al. Irisin in blood increases transiently after single sessions of intense endurance exercise and heavy strength training. PLoS One 2015; 10(3): e0121367.
[http://dx.doi.org/10.1371/journal.pone.0121367] [PMID: 25781950]
[34]
Pang M, Yang J, Rao J, et al. Time-dependent changes in increased levels of plasma irisin and muscle PGC-1α and FNDC5 after exercise in mice. Tohoku J Exp Med 2018; 244(2): 93-103.
[http://dx.doi.org/10.1620/tjem.244.93] [PMID: 29415899]
[35]
Cosio PL, Crespo-Posadas M, Velarde-Sotres Á, Pelaez M. Effect of chronic resistance training on circulating irisin: Systematic review and meta-analysis of randomized controlled trials. Int J Environ Res Public Health 2021; 18(5): 2476.
[http://dx.doi.org/10.3390/ijerph18052476] [PMID: 33802329]
[36]
Panati K, Suneetha Y, Narala VR. Irisin/FNDC5-An updated review. Eur Rev Med Pharmacol Sci 2016; 20(4): 689-97.
[PMID: 26957272]
[37]
Pedersen BK. Exercise-induced myokines and their role in chronic diseases. Brain Behav Immun 2011; 25(5): 811-6.
[http://dx.doi.org/10.1016/j.bbi.2011.02.010] [PMID: 21354469]
[38]
Miura S, Kawanaka K, Kai Y, et al. An increase in murine skeletal muscle peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) mRNA in response to exercise is mediated by β-adrenergic receptor activation. Endocrinology 2007; 148(7): 3441-8.
[http://dx.doi.org/10.1210/en.2006-1646] [PMID: 17446185]
[39]
Vasconcelos-Filho FSL, da Rocha-e-Silva RC, Martins JER, et al. Neuroprotector effect of daily 8-minutes of high-intensity interval training in rat Aβ1-42 Alzheimer disease model. Curr Alzheimer Res 2021; 17(14): 1320-33.
[http://dx.doi.org/10.2174/1567205018666210218161856] [PMID: 33602092]
[40]
Rabiee F, Lachinani L, Ghaedi S, Nasr-Esfahani MH, Megraw TL, Ghaedi K. New insights into the cellular activities of Fndc5/Irisin and its signaling pathways. Cell Biosci 2020; 10(1): 51.
[http://dx.doi.org/10.1186/s13578-020-00413-3] [PMID: 32257109]
[41]
Finck BN, Kelly DP. PGC-1 coactivators: Inducible regulators of energy metabolism in health and disease. J Clin Invest 2006; 116(3): 615-22.
[http://dx.doi.org/10.1172/JCI27794] [PMID: 16511594]
[42]
Liang H, Ward WF. PGC-1α: A key regulator of energy metabolism. Adv Physiol Educ 2006; 30(4): 145-51.
[http://dx.doi.org/10.1152/advan.00052.2006] [PMID: 17108241]
[43]
Wang R, Li JJ, Diao S, et al. Metabolic stress modulates Alzheimer’s β-secretase gene transcription via SIRT1-PPARγ-PGC-1 in neurons. Cell Metab 2013; 17(5): 685-94.
[http://dx.doi.org/10.1016/j.cmet.2013.03.016] [PMID: 23663737]
[44]
Corona JC, Duchen MR. PPARγ and PGC-1α as therapeutic targets in Parkinson’s. Neurochem Res 2015; 40(2): 308-16.
[http://dx.doi.org/10.1007/s11064-014-1377-0] [PMID: 25007880]
[45]
Patti ME, Butte AJ, Crunkhorn S, et al. Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: Potential role of PGC1 and NRF1. Proc Natl Acad Sci USA 2003; 100(14): 8466-71.
[http://dx.doi.org/10.1073/pnas.1032913100] [PMID: 12832613]
[46]
Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI. Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med 2004; 350(7): 664-71.
[http://dx.doi.org/10.1056/NEJMoa031314] [PMID: 14960743]
[47]
Mootha VK, Lindgren CM, Eriksson KF, et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 2003; 34(3): 267-73.
[http://dx.doi.org/10.1038/ng1180] [PMID: 12808457]
[48]
Muller YL, Bogardus C, Pedersen O, Baier L. A Gly482Ser missense mutation in the peroxisome proliferator-activated receptor gamma coactivator-1 is associated with altered lipid oxidation and early insulin secretion in Pima Indians. Diabetes 2003; 52(3): 895-8.
[http://dx.doi.org/10.2337/diabetes.52.3.895] [PMID: 12606537]
[49]
Hara K, Tobe K, Okada T, et al. A genetic variation in the PGC-1 gene could confer insulin resistance and susceptibility to Type II diabetes. Diabetologia 2002; 45(5): 740-3.
[http://dx.doi.org/10.1007/s00125-002-0803-z] [PMID: 12107756]
[50]
Baar K, Wende AR, Jones TE, et al. Adaptations of skeletal muscle to exercise: rapid increase in the transcriptional coactivator PGC‐1. FASEB J 2002; 16(14): 1879-86.
[http://dx.doi.org/10.1096/fj.02-0367com] [PMID: 12468452]
[51]
Michael LF, Wu Z, Cheatham RB, et al. Restoration of insulin-sensitive glucose transporter (GLUT4) gene expression in muscle cells by the transcriptional coactivator PGC-1. Proc Natl Acad Sci USA 2001; 98(7): 3820-5.
[http://dx.doi.org/10.1073/pnas.061035098] [PMID: 11274399]
[52]
Lester-Coll N, Rivera EJ, Soscia SJ, Doiron K, Wands JR, de la Monte SM. Intracerebral streptozotocin model of type 3 diabetes: Relevance to sporadic Alzheimer’s disease. J Alzheimers Dis 2006; 9(1): 13-33.
[http://dx.doi.org/10.3233/JAD-2006-9102] [PMID: 16627931]
[53]
Steen E, Terry BM, Rivera EJ, et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease-is this type 3 diabetes? J Alzheimers Dis 2005; 7(1): 63-80.
[http://dx.doi.org/10.3233/JAD-2005-7107] [PMID: 15750215]
[54]
Willette AA, Xu G, Johnson SC, et al. Insulin resistance, brain atrophy, and cognitive performance in late middle-aged adults. Diabetes Care 2013; 36(2): 443-9.
[http://dx.doi.org/10.2337/dc12-0922] [PMID: 23069842]
[55]
Crane PK, Walker R, Hubbard RA, et al. Glucose levels and risk of dementia. N Engl J Med 2013; 369(6): 540-8.
[http://dx.doi.org/10.1056/NEJMoa1215740] [PMID: 23924004]
[56]
Cherbuin N, Sachdev P, Anstey KJ. Higher normal fasting plasma glucose is associated with hippocampal atrophy: The PATH Study. Neurology 2012; 79(10): 1019-26.
[http://dx.doi.org/10.1212/WNL.0b013e31826846de] [PMID: 22946113]
[57]
Chiu SL, Chen CM, Cline HT. Insulin receptor signaling regulates synapse number, dendritic plasticity, and circuit function in vivo. Neuron 2008; 58(5): 708-19.
[http://dx.doi.org/10.1016/j.neuron.2008.04.014] [PMID: 18549783]
[58]
Kim O, Song J. The role of irisin in Alzheimer’s disease. J Clin Med 2018; 7(11): 407.
[http://dx.doi.org/10.3390/jcm7110407] [PMID: 30388754]
[59]
Cardó-Vila M, Arap W, Pasqualini R. α v β 5 integrin-dependent programmed cell death triggered by a peptide mimic of annexin V. Mol Cell 2003; 11(5): 1151-62.
[http://dx.doi.org/10.1016/S1097-2765(03)00138-2] [PMID: 12769841]
[60]
Goldfinger LE. Integrin signaling. Encycl Biol Chem Second Ed 2013; 285: 441-5.
[http://dx.doi.org/10.1016/B978-0-12-378630-2.00447-3]
[61]
Wu M, Fang K, Wang W, Lin W, Guo L, Wang J. Identification of key genes and pathways for Alzheimer’s disease via combined analysis of genome-wide expression profiling in the hippocampus. Biophys Rep 2019; 5(2): 98-109.
[http://dx.doi.org/10.1007/s41048-019-0086-2]
[62]
Reynolds LE, Wyder L, Lively JC, et al. Enhanced pathological angiogenesis in mice lacking β3 integrin or β3 and β5 integrins. Nat Med 2002; 8(1): 27-34.
[http://dx.doi.org/10.1038/nm0102-27] [PMID: 11786903]
[63]
Guillot-Sestier MV, Doty KR, Gate D, et al. Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology. Neuron 2015; 85(3): 534-48.
[http://dx.doi.org/10.1016/j.neuron.2014.12.068] [PMID: 25619654]
[64]
Bi X, Gall CM, Zhou J, Lynch G. Uptake and pathogenic effects of amyloid beta peptide1-42 are enhanced by integrin antagonists and blocked by NMDA receptor antagonists. Neuroscience 2002; 112(4): 827-40.
[http://dx.doi.org/10.1016/S0306-4522(02)00132-X] [PMID: 12088742]
[65]
Wang Z, Wang Z, Zhou Z, Ren Y. Crucial genes associated with diabetic nephropathy explored by microarray analysis. BMC Nephrol 2016; 17(1): 128.
[http://dx.doi.org/10.1186/s12882-016-0343-2] [PMID: 27613243]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy