Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

ISSN (Print): 1871-5273
ISSN (Online): 1996-3181

Research Article

Effects of Exercise and Ferulic Acid on Alpha Synuclein and Neuroprotective Heat Shock Protein 70 in An Experimental Model of Parkinsonism Disease

Author(s): Mona H. Askar, Abdelaziz M. Hussein*, Soheir F. Al-Basiony, Refka K. Meseha, Emile F. Metias, Mohamed M. Salama, Ashraf Antar and Aya El-Sayed

Volume 18, Issue 2, 2019

Page: [156 - 169] Pages: 14

DOI: 10.2174/1871527317666180816095707

Price: $65

Abstract

Background & Objective: This study investigated the effects of ferulic acid (FR), muscle exercise (Ex) and combination of them on rotenone (Rot)-induced Parkinson disease (PD) in mice as well as their underlying mechanisms.

Method: 56 male C57BL/6 mice were allocated into 8 equal groups, 1) Normal control (CTL), 2) FR (mice received FR at 20 mg/kg/day), 3) Ex (mice received swimming Ex) and 4) Ex + FR (mice received FR and Ex), 5) Rot (mice received Rot 3 mg/Kg i.p. for 70 days), 6) ROT+ FR (mice received Rot + FR at 20 mg/kg/day), 7) ROT+ Ex (mice received Rot + swimming Ex) and 8) ROT+ Ex + FR (mice received Rot + FR and Ex). ROT group showed significant impairment in motor performance and significant reduction in tyrosine hydroxylase (TH) density and Hsp70 expression (p< 0.05) with Lewy bodies (alpha synuclein) aggregates in corpus striatum. Also, ROT+FR, ROT+EX and ROT + Ex+ FR groups showed significant improvement in behavioral and biochemical changes, however the effect of FR alone was more potent than Ex alone (p< 0.05) and addition of Ex to FR caused no more significant improvement than FR alone.

Conclusion: We concluded that, FR and Ex improved the motor performance in rotenone-induced PD rodent model which might be due to increased Hsp70 expression and TH density in corpus striatum and combination of both did not offer more protection than FR alone.

Keywords: Mice, Hsp70, tyrosine hydroxylase, Parkinson disease, alpha synuclein, ferulic acid, exercise.

Graphical Abstract
[1]
Khedr EM, Al Attar GS, Kandil MR, Kamel NF, Abo Elfetoh NA. Epidemiological study and clinical profile of Parkinson’s disease in the assiut governorate, Egypt: A community-based study neuroepidemiology 2012; 38: 154-63.
[2]
De Lau LM, Breteler MM. Epidemiology of Parkinson’s disease. Lancet Neurol 2006; 5: 525-35.
[3]
Soldner F, Hockemeyer D, Beard C. Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell 2009; 136: 964-77.
[4]
Aridon P, Fabiana G, Giuseppina T, Marco DA, Giovanni S, Gabriella S. Protective role of heat shock proteins in Parkinson’s disease. Neurodegener Dis 2011; 8(4): 155-68.
[5]
Murphy DD, Rueter SM, Trojanowski JQ, Lee VM. Synucleins are developmentally expressed, and alpha-synuclein regulates the size of the presynaptic vesicular pool in primary hippocampal neurons. J Neurosci 2000; 20(9): 3214-20.
[6]
Abeliovich A, Schmitz Y, Choi-Lundberg D. Mice lacking alpha-synuclein display functional deficits in the nigrostriatal dopamine system. Neuron 2000; 25: 239-52.
[7]
Eller M, Williams DR. Alpha-synuclein in parkinson disease and other neurodegenerative disorders. Clin Chem Lab Med 2011; 49(3): 403-8.
[8]
Puschmann A, Wszolek ZK, Farrer M, Gustafson L, Widner H, Nilsson C. Alpha-synuclein multiplications with Parkinsonism, dementia or progressive myoclonus? Parkinsonism Relat Disord 2009; 15(5): 390-2.
[9]
St Martin JL, Klucken J, Outeiro TF. Dopaminergic neuron loss and up-regulation of chaperone protein mRNA induced by targeted over-expression of alpha-synuclein in mouse Substantia nigra. J Neurochem 2007; 100(6): 1449-57.
[10]
Herrera E, Jiménez R, Aruoma OI, Hercberg S, Sánchez-García I, Fraga C. Aspects of antioxidant foods and supplements in health and disease. Nutr Rev 2009; 67(1): 140-4.
[11]
Shahwar D, Raza MA. Shafiq-Ur-Rehman, et a An investigation of phenolic compounds from plant sources as trypsin inhibitors. Nat Prod Res 2012; 26(12): 1087-93.
[12]
Zhang H, Mak S, Cui W, Li W. Tacrine (2)-ferulic acid, a novel multifunctional dimer, attenuates 6-hydroxydopamine-induced apoptosis in PC12 cells by activating Akt pathway. Neurochem Int 2011; 59(7): 981-8.
[13]
Horowitz M, Robinson SD. Heat shock proteins and the heat shock response during hyperthermia and its modulation by altered physiological conditions. Prog Brain Res 2007; 162: 433-46.
[14]
Joshi G, Perluigi M, Sultana R, Agrippino R, Calabrese V, Butterfield DA. In vivo protection of synaptosomes by ferulic acid ethyl ester (FAEE) from oxidative stress mediated by 2,2-azobis(2-amidino-propane) dihydrochloride (AAPH) or 22 Fe(2+)/H(2)O(2): Insight into mechanisms of neuroprotection and relevance to oxidative stress-related neurodegenerative disorders. Neurochem Int 2006; 48: 318-27.
[15]
Perluigi M, Joshi G, Sultana R, et al. In vivo protective effects of ferulic acid ethyl ester against amyloid-beta peptide 1-42-induced oxidative stress. J Neurosci Res 2006; 84(2): 418-26.
[16]
Pothakos K, Kurz MJ, Lau YS. Restorative effect of endurance exercise on behavioral deficits in the chronic mouse model of Parkinson’s disease with severe neurodegeneration. BMC Neurosci 2009; 10: 6.
[17]
Salama M, Ellaithy A, Helmy B. Colchicine protects dopaminergic neurons in a rat model of Parkinson's disease 2012. 11(7): 836-4
[18]
Rukkumani R, Aruna K, Suresh Varma P, Padmanabhan Menon V. Hepatoprotective role of ferulic acid: a dose-dependent study. J Med Food 2004; 7(4): 456-61.
[19]
Haobam R, Sindhu KM, Chandra G, Mohanakumar KP. Swim-test as a function of motor impairment in MPTP model of Parkinson’s disease: A comparative study in two mouse strains. Behav Brain Res 2005; 163: 159-67.
[20]
Carlsson T, Winkler C, Lundblad M, Cenci MA, Bjorklund A, Kirik D. Graft placement and uneven pattern of reinnervation in the striatum is important for development of graft-induced dyskinesia. Neurobiol Dis 2006; 21: 657-68.
[21]
Johnson ME, Bobrovskaya L. An update on the rotenone models of Parkinson’s disease: their ability to reproduce the features of clinical disease and model gene-environment interactions. Neurotoxicology 2014; 46C: 101-16.
[22]
Cannon JR, Tapias V, Na HM. A highly reproducible rotenone model of Parkinson’s disease. Neurobiol Dis 34(2): 279-90.
[23]
Betarbet R, Sherer TB, MacKenzie G. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci 2000; 3(12): 1301-6.
[24]
Sherer TB, Betarbet R, Kim JH, Greenamyre JT. Selective microglial activation in the rat rotenone model of Parkinson’s disease. Neurosci Lett 2003; 341: 87-90.
[25]
Litteljohn D, Mangano E, Clarke M, Bobyn J, Moloney K, Hayley S. Inflammatory mechanisms of neurodegeneration in toxin-based models of Parkinson’s disease. Parkinsons Dis 2011; 2011: 713517.
[26]
Fleming SM, Zhu C, Fernagut PO. Behavioral and immunohistochemical effects of chronic intravenous and subcutaneous infusions of varying doses of rotenone. Exp Neurol 2004; 187: 418-29.
[27]
Salama M, Helmy B, El-Gamal M, Reda A. Role of L-thyroxin in counteracting rotenone induced neurotoxicity in rats. Environ Toxicol Pharmacol 2013; 35(2): 270-7.
[28]
Thakur P, Nehru B. Long-term heat shock proteins (HSPs) induction by carbenoxolone improves hallmark features of Parkinson’s disease in a rotenone-based model. Neuropharmacology 2014; 79: 190-200.
[29]
Larsen KE, Schmitz Y, Troyer MD, et al. Alpha-synuclein overexpression in PC12 and chromaffin cells impairs catecholamine release by interfering with a late step in exocytosis. J Neurosci 2006; 26: 11915-22.
[30]
Chen RH, Wislet-Gendebien S, Samuel F, et al. Alpha-synuclein membrane association is regulated by the Rab3a recycling machinery and presynaptic activity. J Biol Chem 2013; 288: 7438-49.
[31]
DeWitt DC, Rhoades E. Alpha-synuclein can inhibit SNARE-mediated vesicle fusion through direct interactions with lipid bilayers. Biochemistry 2013; 52: 2385-7.
[32]
Young JC. The role of the cytosolic HSP70 chaperone system in diseases caused by misfolding and aberrant trafficking of ion channels. Dis Model Mech 2014; 7: 319-29.
[33]
Jung AE, Fitzsimons HL, Bland RJ, During MJ, Young D. HSP70 and constitutively active HSF1 mediate protection against CDCrel-1-mediated toxicity. Mol Ther J Am Soc Gene Ther 2008; 16: 1048-55.
[34]
Kalia SK, Kalia LV, McLean PJ. Molecular chaperones as rational drug targets for Parkinson’s disease therapeutics. CNS Neurol Disord Drug Targets 2010; 9: 741-53.
[35]
Kumar P, Ambasta RK, Veereshwarayya V, et al. CHIP and HSPs interact with beta-APP in a proteasome-dependent manner and influence Abeta metabolism. Hum Mol Genet 2007; 16: 848-64.
[36]
Nagel F, Falkenburger BH, Tonges L, et al. Tat-Hsp70 protects dopaminergic neurons in midbrain cultures and in the substantia nigra in models of Parkinson’s disease. J Neurochem 2008; 105: 853-64.
[37]
Sherman MY, Goldberg AL. Cellular defenses against unfolded proteins: a cell biologist thinks about neurodegenerative diseases. Neuron 2001; 29: 15-32.
[38]
Klucken J, Shin Y, Masliah E, Hyman BT, McLean PJ. Hsp70 reduces alpha-synuclein aggregation and toxicity. J Biol Chem 2004; 279: 25497-502.
[39]
Luk KC, Mills IP, Trojanowski JQ, Lee VM. Interactions between Hsp70 and the hydrophobic core of alpha-synuclein inhibit fibril assembly. Biochemistry 2008; 47: 12614-25.
[40]
Leverenz JB, Umar I, Wang Q, Montine TJ, et al. Proteomic identification of novel proteins in cortical lewy bodies. Brain Pathol 2007; 17: 139-45.
[41]
Muchowski PJ, Wacker JL. Modulation of neurodegeneration by molecular chaperones. Nat Rev Neurosci 2005; 6: 11-22.
[42]
Wang X, Qin ZH, Leng Y, et al. Prostaglandin A1 inhibits rotenone-induced apoptosis in SH-SY5Y cells. J Neurochem 2002; 83(5): 1094-102.
[43]
Shen HY, He JC, Wang Y, Huang QY, Chen JF. Geldanamycin induces heat shock protein 70 and protects against MPTP-induced dopaminergic neurotoxicity in mice. J Biol Chem 2005; 280: 39962-9.
[44]
Pan T, Li X, Xie W, Jankovic J, Le W. Valproic acid-mediated Hsp70 induction and anti-apoptotic neuroprotection in SH-SY5Y cells. FEBS Lett 2005; 579: 6716-20.
[45]
Picone P, Bondi ML, Montana G, et al. Ferulic acid inhibits oxidative stress and cell death induced by Ab oligomers: Improved delivery by solid lipid nanoparticles. Free Radic Res 2009; 43(11): 1133-45.
[46]
Yabe T, Hirahara H, Harada N, et al. Ferulic acid induces neural progenitor cell proliferation in vitro and in vivo. Neuroscience 2010; 165(2): 515-24.
[47]
Mori T, Koyama N, Guillot-Sestier MV, Tan J, Town T. Ferulic acid is a nutraceutical β-secretase modulator that improves behavioral impairment and alzheimer-like pathology in transgenic mice. PLoS One 2013; 8(2): e55774.
[48]
Ojha S, Javed H, Azimullah S, Abul Khair SB, Haque ME. Neuroprotective potential of ferulic acid in the rotenone model of Parkinson’s disease. Drug Des Devel Ther 2015; 9: 5499-510.
[49]
Kim BW, Koppula S, Park SY, et al. Attenuation of neuroinflammatory responses and behavioral deficits by Ligusticum officinale (Makino) Kitag in stimulated microglia and MPTP-induced mouse model of Parkinson’s disease. J Ethnopharmacol 2015; 164: 388-97.
[50]
Nagarajan S, Chellappan DR, Chinnaswamy P, Thulasingam S. Ferulic acid pretreatment mitigates MPTP-induced motor impairment and histopathological alterations in C57BL/6 mice. Pharm Biol 2015; 10: 1-11.
[51]
Blesa J, Phani S, Jackson-Lewis V, Przedborski S. Classic and new animal models of Parkinson’s disease. J Biomed Biotechnol 2012; 8: 845618.
[52]
Kikugawa M, Tsutsuki H, Ida T. Water-soluble ferulic acid derivatives improve amyloid-β-induced neuronal cell death and dysmnesia through inhibition of amyloid-β aggregation. Biosci Biotechnol Biochem 2016; 80(3): 547-53.
[53]
Al Dakheel A, Kalia LV, Lang AE. Pathogenesis-targeted disease-modifying therapies in Parkinson disease. Neurotherapeutics 2014; 11(1): 6-23.
[54]
Song JX, Sze SC, Ng TB. Anti-parkinsonian drug discovery from herbal medicines: what have we got from neurotoxic models? J Ethnopharmacol 2012; 139(3): 698-711.
[55]
Sutachan JJ, Casas Z, Albarracin SL, Stab BR. Cellular and molecular mechanisms of antioxidants in Parkinson’s disease. Nutr Neurosci 2012; 15(3): 1206.
[56]
Scapagnini G, Butterfield DA, Colombrita C, Sultana R, Pascale A, Calabrese V. Ethyl ferulate, a lipophilic polyphenol, induces HO-1 and protects rat neurons against oxidative stress. Antioxid Redox Signal 2004; 6(5): 811-8.
[57]
Petzinger GM, Walsh JP, Akopian G. Effects of treadmill exercise on dopaminergic transmission in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned mouse model of basal ganglia injury. J Neurosci 2007; 27: 5291-300.
[58]
Lau YS, Patki G, Das-Panja K, Le W, Ahmad SO. Neuroprotective effects and mechanisms of exercise in a chronic mouse model of Parkinson’s disease with moderate neurodegeneration. J Neurosci 2011; 33: 1264-74.
[59]
Wu SY, Wang TF, Yu L, et al. Running exercise protects the substantia nigra dopaminergic neurons against inflammation-induced degeneration via the activation of BDNF singaling pathway. Brain Behav Immun 2011; 25: 135-46.
[60]
Eldar R, Marincek C. Physical activity for elderly persons with neurological impairment: A review. Scand J Rehabil Med 2000; 32: 99-103.
[61]
Cotman CW, Berchtold NC. Exercise: A behavioral intervention to enhance brain health and plasticity. Trends Neurosci 2002; 25: 295-301.
[62]
Faherty CJ, Raviie Shepherd K, Herasimtschuk A, Smeyne RJ. Environmental enrichment in adulthood eliminates neuronal death in experimental Parkinsonism. Brain Res Mol Brain Res 2005; 134: 170-9.
[63]
Smith AD, Zigmond MJ. Can the brain be protected through exercise? Lessons from an animal model of parkinsonism. Exp Neurol 2003; 184: 31-9.
[64]
Shin MS, Kim TW, Lee JM, Ji ES, Lim BV. Treadmill exercise-alleviates nigrostriatal dopaminergic loss of neurons and fibers in rotenone-induced Parkinson rats. J Exerc Rehabil 2017; 13(1): 30-5.
[65]
Lancaster GI, Moller K, Nielsen B, Secher NH, Febbraio MA, Nybo L. Exercise induces the release of heat shock protein 72 from the human brain in vivo. Cell Stress Chaperones 2004; 9: 276-80.
[66]
Shin M-S, Jeong HY, An DI, Lee HY, Sung YH. Treadmill exercise facilitates synaptic plasticity on dopaminergic neurons and fibers in the mouse model with Parkinson’s disease. Neurosci Lett 2016; 621: 28-33.

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