Assessment of the neuroprotective effect of green synthesized iron oxide nanoparticles capped with curcumin against a rat model of Parkinson’s disease

Document Type : Original Article

Authors

1 Medical Physiology Department, Medical Division, National Research Centre, Giza, Egypt

2 Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt

Abstract

Objective(s): The current study aims to investigate the protective effect of iron oxide nanoparticles capped with curcumin (FeONPs-Cur) against motor impairment and neurochemical changes in a rat model of Parkinson’s disease (PD) induced by reserpine.
Materials and Methods: Rats were grouped into control, PD model induced by reserpine, and PD model treated with FeONPs-Cur (8 rats/group). The open field test was used to assess motor activity. The concentration of dopamine (DA), norepinephrine (NE), serotonin (5-HT), lipid peroxidation (MDA), reduced glutathione (GSH), and nitric oxide (NO), and the activities of Na+,K+,ATPase, acetylcholinesterase (AchE), and monoamine oxidase (MAO) were determined in the midbrain and striatum. Data were analyzed by ANOVA at P-value<0.05. 
Results: The PD model exhibited a decrease in motor activity. In the midbrain and striatum of the PD model, DA, NE, and 5-HT levels decreased significantly (P-value<0.05). However, an increase in MAO, NO, and MDA was observed. GSH, AchE and Na+,K+,ATPase decreased significantly in the two brain areas. FeONPs-Cur prevented the decline of dopamine and norepinephrine and reduced oxidative stress in both areas.  It also prevented the increased MAO activity in the two areas and the inhibited activity of AchE and Na+,K+,ATPase in the midbrain. These changes were associated with improvements in motor activity.
Conclusion: The present data indicate that FeONPs-Cur could prevent the motor deficits induced in the PD rat model by restoring dopamine and norepinephrine in the midbrain and striatum. The antioxidant activity of FeONPs-Cur contributed to its protective effect. These effects nominate FeONPs-Cur as an antiparkinsonian candidate. 

Keywords

Main Subjects


1. Balestrino R, Schapira AHV. Parkinson disease. Eur J Neurol 2020; 27: 27-42.
2. Tysnes OB, Storstein A. Epidemiology of Parkinson’s disease. J Neural Transm 2017; 124: 901–905.
3. Ikenaka K, Suzuki M, Mochizuki H, Nagai Y. Lipids as trans-acting effectors for α-synuclein in the pathogenesis of Parkinson’s disease. Front Neurosci 2019; 13:693. 
4. DeMaagd G, Philip A. Parkinson’s disease and its management: Part 1: disease entity, risk factors, pathophysiology, clinical presentation, and diagnosis. P T 2015; 40: 504–532.
5. Kim GH, Kim JE, Rhie SJ, Yoon S. The role of oxidative stress in neurodegenerative diseases. Exp Neurobiol 2015; 24: 325–340.
6. Callio J, Oury TD, Chu CT. Manganese superoxide dismutase protects against 6-hydroxydopamine injury in mouse brains. J Biol Chem 2005; 280:18536–18542. 
7. Perier C, Bové J, Vila M, Przedborski S. The rotenone model of Parkinson’s disease. Trends Neurosci 2003; 26: 345–346.
8. Liu C. Targeting the cholinergic system in Parkinson’s disease. Acta Pharmacol Sin 2020; 41: 453–463. 
9. Sharma S, Rabbani SA, Agarwal T, Baboota S, Pottoo FH, Kadian R. Nanotechnology driven approaches for the management of Parkinson’s disease: current status and future perspectives. Curr Drug Metab 2021; 22: 287–298.
10. Braak H, Del Tredici K, Rüb U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging 2003; 24:197–211.
11. Lee TK, Yankee EL. A review on Parkinson’s disease treatment Neuroimmunol Neuroinflammation 2021; 8: 222
12. Cheng K, Peng S, Xu C, Sun S. Porous hollow Fe3O4 nanoparticles for targeted delivery and controlled release of cisplatin. J Am Chem Soc 2009; 131: 10637-10644. 
13. Maeng JH, Lee DH, Jung KH, Bae YH, Park IS, Jeong S, et al. Multifunctional doxorubicin loaded superparamagnetic iron oxide nanoparticles for chemotherapy and magnetic resonance imaging in liver cancer. Biomaterials 2010; 31: 4995-5006.
14. Zhang Y, Wang Z, Li X, Wang L, Yin M, Wang L, et al. Dietary iron oxide nanoparticles delay aging and ameliorate neurodegeneration in drosophila. Adv Mater 2016; 28: 1387-93. 
15. Yarjanli Z, Ghaedi K, Esmaeili A, Rahgozar S, Zarrabi A. Iron oxide nanoparticles may damage to the neural tissue through iron accumulation, oxidative stress, and protein aggregation. BMC Neurosci 2017; 18:51-62. 
16. Ahmed EM. Hydrogel: preparation, characterization, and applications: A review. J Adv Res 2015; 6: 105–121.
17. Kumar B, Smita K, Cumbal L, Debut A. Biogenic synthesis of iron oxide nanoparticles for 2-arylbenzimidazole fabrication. J Saudi Chem Soc 2014; 18: 364-369.
18. Mirza AU, Kareem A, Nami SAA, Khan MS, Rehman S, Bhat SA, et al. Biogenic synthesis of iron oxide nanoparticles using Agrewia optiva and Prunus persica phyto species: Characterization, antibacterial and antioxidant activity. J Photochem Photobiol B 2018; 185: 262–274.
19. Bibi I, Nazar N, Ata S, Sultan M, Ali A, Abbas A, et al. Green synthesis of iron oxide nanoparticles using pomegranate seeds extract and photocatalytic activity evaluation for the degradation of textile dye. J Mater Res Technol 2019; 8: 6115–6124.
20. Fatemi M, Mollania N, Momeni-Moghaddam M, Sadeghifar F. Extracellular biosynthesis of magnetic iron oxide nanoparticles by Bacillus cereus strain HMH1: Characterization and in vitro cytotoxicity analysis on MCF-7 and 3T3 cell lines. J Biotechnol 2018; 270: 1–11.
21. Peralta-Videa JR, Huang Y, Parsons JG, Zhao L, Lopez-Moreno L, Hernandez-Viezcas JA, et al. Plant-based green synthesis of metallic nanoparticles: scientific curiosity or a realistic alternative to chemical synthesis? Nanotechnol. Environ Eng 2016; 1: 1–29.
22. Khadrawy YA, Hosny EN, Magdy M, Mohammed HS. Antidepressant effects of curcumin-coated iron oxide nanoparticles in a rat model of depression. Eur J Pharmacol 2021; 908:174384. 
23. Naserzadeh P, Hafez AA, Abdorahim M, Abdollahifar MA, Shabani R, Peirovi H, et al. Curcumin loading potentiates the neuroprotective efficacy of Fe3O4 magnetic nanoparticles in cerebellum cells of schizophrenic rats. Biomed Pharmacother 2018; 108: 1244-1252.
24. Elbialy NS, Aboushoushah SF, Alshammari WW. Long-term biodistribution and toxicity of curcumin capped iron oxide nanoparticles after single-dose administration in mice. Life Sci 2019; 230:76-83.
25. Fernandes VS, Santos JR, Leão AH, Medeiros AM, Melo TG, Izídio GS, et al. Repeated treatment with a low dose of reserpine as a progressive model of Parkinson’s disease. Behav Brain Res 2012; 231: 154-163.
26. Brown RE, Corey SC, Moore AK. Differences in measures of exploration and fear in MHC-congenic C57BL/6J and B6-H-2K mice. Behavior Genetics 1999; 29: 263-271.
27. Ruiz-Larrea MB, Leal AM, Liza M, Lacort M, de Groot H. Anti-oxidant effects of estradiol and 2-hydroxyestradiol on iron-induced lipid peroxidation of rat liver microsomes. Steroids 1994; 59: 383–388.
28. Montgomery HAC, Dymock JF. The determination of nitrite in water. Analyst 1961; 6: 414–416.
29. Beutler E, Duron O, Kelly OM. Improved method for the determination of blood glutathione. J Lab Clin Med 1963: 61: 882–888.
30. Gorun V, Proinov I, Baltescu V, Balaban G, Barzu O. Modified Ellman procedure for assay of cholinesterase in crude-enzymatic preparations. Anal Biochem 1997; 86: 324–326.
31. Tsakiris S, Angelogianni P, Schulpis KH, Stavridis JC. Protective effect of L-phenylalanine on rat brain acetylcholinesterase inhibition induced by free radicals. Clin Biochem 2000; 33: 103-106.
32. Holt A, Sharman DS, Baker GB, Palcic MM. A continuous spectrophotometric assay for monoamine oxidase and related enzymes in tissue homogenates. Anal Biochem 1997; 244: 384-392.
33. Ciarlone AE. Further modification of a fluoromertric method for analyzing brain amines. Microchem J 1978; 23: 9–12.
34. Colpaert FC. Pharmacological characteristics of tremor, rigidity and hypokinesia induced by reserpine in rat. Neuropharmacology 1987: 26:1431–1440.
35. Oe T, Tsukamoto M, Nagakura Y. Reserpine causes biphasic nociceptive sensitivity alteration in conjunction with brain biogenic amine tones in rats. Neuroscience 2021; 169: 1860–1871.
36. Vergo S, Johansen JL, Leist M, Lotharius J. Vesicular monoamine transporter 2 regulates the sensitivity of rat dopaminergic neurons to disturbed cytosolic dopamine levels. Brain Res 2007; 1185: 18–32.
37. Spina MB, Cohen G. Dopamine turnover and glutathione oxidation: implications for Parkinson disease. Proc Natl Acad Sci U S A 1989; 86:1398–1400.
38. Angelova PR, Horrocks MH, Klenerman D, Gandhi S, Abramov AY, Shchepinov MS. Lipid peroxidation is essential for α-synuclein-induced cell death. J Neurochem 2015; 133: 582-589.
39. Eve DJ, Nisbet AP, Kingsbury AE, Hewson EL, Daniel SE, Lees AJ, et al. Basal ganglia neuronal nitric oxide synthase mRNA expression in Parkinson’s disease. Mol Brain Res 1998; 63: 62–71. 
40. Joniec I, Ciesielska A, Kurkowska-Jastrzebska I, Przybylkowski A, Czlonkowska A, Czlonkowski A. Age- and sex-differences in the nitric oxide synthase expression and dopamine concentration in the murine model of Parkinson’s disease induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Brain Res 2009; 1261: 7–19.
41. Cavalcanti-Kwiatkoski R, Raisman-Vozari R, Ginestet L, Del Bel E. Altered expression of neuronal nitric oxide synthase in weaver mutant mice. Brain Res 2010; 1326: 40–50.
42. Tzschentke TM. Pharmacology and behavioral pharmacology of the mesocortical dopamine system. Prog Neurobiol 2001; 6: 241–320.
43. Michelotti GA, Price DT, Schwinn DA. Alpha 1-adrenergic receptor regulation: basic science and clinical implications. Pharmacol Ther 2000; 88: 281–309.
44. Kish J, Tong J, Hornykiewicz O, Rajput A, Chang LJ, Guttman M, et al. Preferential loss of serotonin markers in caudate versus putamen in Parkinson’s disease. Brain 2008; 131: 120–131.
45. Kerenyi L, Ricaurte GA, Schretlen DJ, McCann U, Varga J, Mathews WB, et al. Positron emission tomography of striatal serotonin transporters in Parkinson disease. Arch neurol 2003; 60: 1223–1229.
46. Chaudhuri KR, Martinez-Martin P, Schapir AH, Stocch F, Seth K, Odin P, et al. International multicenter pilot study of the first comprehensive self-completed nonmotor symptoms questionnaire for Parkinson’s disease: The NMSQuest study. Mov Disord 2006; 21: 916–923.
47. Perry EK, Curtis M, Dick DJ, Candy JM, Atack JR, Bloxham CA, et al. Cholinergic correlates of cognitive impairment in Parkinson’s disease: comparisons with Alzheimer’s disease. J Neurol Neurosurg Psychiatry 1985; 48: 413–421.
48. Dautan D, Souza AS, Huerta-Ocampo I, Valencia M, Assous M, Witten IB, Deisseroth K, Tepper JM, Bolam JP, Gerdjikov TV, Mena-Segovia J. Segregated cholinergic transmission modulates dopamine neurons integrated in distinct functional circuits. Nat Neurosci 2016; 19: 1025–1033. 
49. Jellinger K. The pedunculopontine nucleus in Parkinson’s disease, progressive supranuclear palsy and Alzheimer’s disease. J Neurol Neurosurg Psychiatry 1988; 51: 540–543.
50. Bohnen NI, Müller MLTM, Koeppe RA, Studenski SA, Kilbourn MA, Frey KA, et al. History of falls in Parkinson disease is associated with reduced cholinergic activity. Neurology 2009; 73: 1670–1676.
51. Kondabolu K, Roberts EA, Bucklin M, McCarthy MM, Kopell N, Han X. Striatal cholinergic interneurons generate beta and gamma oscillations in the corticostriatal circuit and produce motor deficits PNAS 2016; 113: E3159–E3168.
52. Tanimura A, Pancani T, Lim SAO, Tubert C, Melendez AE, Shen W, et al. Striatal cholinergic interneurons and Parkinson’s disease. Eur J Neurosci 2018; 47: 1148-1158.
53. Antunes MS, Ladd FVL, Ladd A, Moreira AL, Boeira SP, Cattelan Souza L. Hesperidin protects against behavioral alterations and loss of dopaminergic neurons in 6-OHDA-lesioned mice: The role of mitochondrial dysfunction and apoptosis. Metab Brain Dis 2021; 36: 153–167.
54. Scheiner-Bobis G. The sodium pump. Its molecular properties and mechanics of ion transport. Eur J Biochem 2002; 269: 2424–2433.
55. Sibarov DA, Bolshakov AE, Abushik PA, Krivoi II, Antonov SM. Na+,K+-ATPase functionally interacts with the plasma membrane Na+, Ca2+ exchanger to prevent Ca2+ overload and neuronal apoptosis in excitotoxic stress. J Pharmacol Exp Ther 2012; 343: 596–607.
56. Sindhu K, Rajaram A, Sreeram KJ, Rajaram R. Curcumin conjugated gold nanoparticle synthesis and its biocompatibility. RSC Adv 2014; 4:1808–1818.
57. Sundaram PA, Augustine R, Kannan M. Extracellular biosynthesis of iron oxide nanoparticles by Bacillus subtilis strains isolated from rhizosphere soil. Biotechnol Bioproc E 2012; 17: 835–840.
58. Batra J, Sood A. Iron deficiency anaemia: Effect on congnitive development in children: a review. Ind J Clin Biochem 2005; 20: 119–125.
59. Bezem MT, Baumann A, Skjærven L, Meyer R, Kursula P, Martinez A, et al. Stable preparations of tyrosine hydroxylase provide the solution structure of the full-length enzyme. Sci Rep 2016; 6: e30390.
60. Chen Z, Yin JJ, Zhou YT, Zhang Y, Song L, Song M, et al. Dual enzyme-like activities of iron oxide nanoparticles and their implication for diminishing cytotoxicity. ACS Nano 2012; 6: 4001- 4012. 
61. Lakshmi PP, Krishna MG, Venkateswara RK, Shanker K. Neuroprotective effect of green synthesized iron oxide nanoparticles using aqueous extract of convolvulus pluricaulis plant in the management of Alzheimer’s disease. IJPPR  2017; 9: 703-709.