The effect of low dose amphetamine in rotenone-induced toxicity in a mice model of Parkinson’s disease

Document Type : Original Article

Authors

1 Department of Toxicology and Narcotics, National Research Centre, Cairo, Egypt

2 Department of Medical Biochemistry, National Research Centre, Cairo, Egypt

3 Department of Pathology, National Research Centre, Cairo, Egypt

4 Department of Pharmacology, National Research Centre, Cairo, Egypt

Abstract

Objective(s): The effects of low dose amphetamine on oxidative stress and rotenone-induced neurotoxicity and liver injury were examined in vivo in a mice model of Parkinson’s disease.
Materials and Methods: Male mice were treated with rotenone (1.5 mg/kg, every other day for two weeks, subcutaneously). Mice received either the vehicle or amphetamine intraperitoneally at doses of 0.5, 1.0, or 2.0 mg/kg. Oxidative stress was assessed by measurement of the lipid peroxidation product malondialdehyde (MDA), nitric oxide (NO), total anti-oxidant capacity (TAC), and paraoxonase-1 (PON-1) activity in the brain and liver. In addition, brain concentrations of nuclear factor kappa B (NF-κB) and tyrosine hydroxylase were determined and histopathology and Bax/Bcl-2 immunohistochemistry were performed.
Results: The levels of lipid peroxidation and NO were increased and TAC and PON-1 were decreased significantly compared with vehicle-injected control mice. There were also significantly increased NF-κB and decreased tyrosine hydroxylase in the brain following rotenone administration. These changes were significantly attenuated by amphetamine. Rotenone caused neurodegenerative changes in the substantia nigra, cerebral cortex, and hippocampus. The liver showed degenerative changes in hepatocytes and infiltration of Kupffer cells. Bax/Bcl2 ratio was significantly increased in brain and liver tissues. Amphetamine prevented these histopathological changes and the increase in apoptosis evoked by rotenone.
Conclusion: These results suggest that low dose amphetamine exerts anti-oxidant and anti-apoptotic effects, protects against rotenone-induced neurodegeneration, and could prevent neuronal cell degeneration in Parkinson’s disease.

Keywords


1. Van Den Eeden SK, Tanner CM, Bernstein AL, Fross RD, Leimpeter A, Bloch DA, Nelson LM. Incidence of Parkinson’s disease: variation by age, gender, and race/ethnicity. Am J Epidemiol 2003; 157: 1015-1022.
2. Alves G, Forsaa EB, Pedersen KF, Dreetz Gjerstad M, Larsen JP. Epidemiology of Parkinson’s disease. J Neurol 2008; 255:18-32.
3. Rossi A, Berger K, Chen H, Leslie D, Mailman RB, Huang X. Projection of the prevalence of Parkinson’s disease in coming decades: revisited. Mov Disord 2018; 33: 156–159.
4. Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F. Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci 1973; 20: 415-455.
5. Mallet N, Delgado L, Chazalon M, Miguelez C, Baufreton J. Cellular and synaptic dysfunctions in Parkinson’s disease: stepping out of the striatum. Cells 2019; 8: 1005;
6. Jellinger KA. Recent developments in the pathology of Parkinson’s disease. J Neural Transm Suppl 2002; 62:347–376.
7. Santens P, Boon P, Van Roost D, Caemaert J. The pathophysiology of motor symptoms in Parkinson’s disease. Acta Neurol Belg 2003; 103:129–134.
8. Blesa J, Trigo-Damas I, Quiroga-Varela A,  Jackson-Lewis VR. Oxidative stress and parkinson’s disease. Front Neuroanat 2015; 9: 91.
9. De Virgilio A, Greco A, Fabbrini G, Inghilleri M, Rizzo MI, Gallo A, Conte M, Rosato C, Appiani MC, de Vincentiis M. Parkinson’s disease: autoimmunity and neuroinflammation. Autoimmun Rev 2016; 15: 1005-1011.
10. Halliwell B. Oxidative stress and neurodegeneration: where are we now? J Neurochem 2006; 97:1634–1658.
11. Nagatsu T, Sawada M. Biochemistry of postmortem brains in Parkinson’s disease: historical overview and future prospects. J Neural Transm Suppl 2007; 72: 113-120.
12. Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol 2009; 8:382–397.
13. Ritz BR, Paul KC, Bronstein JM. Of pesticides and men: a California story of genes and environment in Parkinson’s disease. Curr Environ Health Rep 2016; 3: 40-52.
14. Sherer TB, Kim JH, Betarbet R, Greenamyre JT. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp Neurol 2003; 179:9-16.
15. Cannon JR, Tapias V, Na HM, Honick AS, Drolet RE, Greenamyre JT. A highly reproducible rotenone model of Parkinson’s disease. Neurobiol Dis 2009; 34:279-290.
16. Liu HQ, Zhu XZ, Weng EQ. Intracellular dopamine oxidation mediates rotenone-induced apoptosis in PC12 cells. Acta Pharmacol Sin 2005; 26:17-26.
17. Li N, Ragheb K, Lawler G, Sturgis J, Rajwa B, Melendez JA, Robinson JP.  Mitochondrial complex I inhibitor rotenone induces apoptosis through enhancing mitochondrial reactive oxygen species production. J Biol Chem 278:8516-8525.
18. Grivennikova VG, Vinogradov AD. Generation of superoxide by the mitochondrial Complex I. Biochim Biophys Acta 2006; 1757:553-561.
19. Schapira AHV, Emre M, Jenner P, Poewe W. Levodopa in the treatment of Parkinson’s disease. Eur J Neurol 2009; 16: 982e989.
20. Connolly BS, Lang AE. Pharmacological treatment of Parkinson disease: a review. JAMA 2014; 311:1670-83.
21. Rascol O, Lozano A, Stern M, Poewe W. Milestones in Parkinson’s disease therapeutics. Mov Disord. 2011;26:1072–1082.
22. Abdel-Salam OME. Drug therapy for Parkinson’s disease: an update. World J Pharmacol 2015; 4: 117-143.
23. Berman S, O’Neill J, Fears S, Bartzokis G, London ED. Abuse of amphetamines and structural abnormalities in brain. Ann N Y Acad Sci 2018; 1141: 195–220.
24. Janssen PA, Leysen JE, Megens AA, Awouters FH. Does phenylethylamine act as an endogenous amphetamine in some patients? Int J Neuropsychopharmcol 1999; 2: 229–240.
25. Borowsky B, Adham N, Jones KA, Raddatz R, Artymyshyn R, Ogozalek KL, Durkin MM, Lakhlani PP, Bonini JA, Pathirana S, et al. Trace amines: identification of a family of mammalian G protein-coupled receptors. Proc Natl Acad Sci 2001; 98:8966–8971.
26. Sitte HH, Freissmuth M. Amphetamines, new psychoactive drugs and the monoamine transporter cycle. Trends Pharmacol Sci 2015 ; 36: 41-50.
27. Koirala S, Shah S, Khanal L. Effect of D‐amphetamine on dopaminergic neurons of substantia nigra and expression of tyrosine hydroxylase in striatum and pre‐frontal cortex of D‐amphetamine treated Wistar rats. Russian OMJ 2014; 3:0401.
28. Halpin LE, Collins SA, Yamamoto BK. Neurotoxicity of methamphetamine and 3,4-methylenedioxymethamphetamine. Life Sci  2014; 97:37-44.
29. Papadopoulos CM, Tsai S-Y, Guillen V, Ortega J, Kartje GL, Wolf WA. Motor recovery and axonal plasticity with short-term amphetamine after stroke. Stroke 2009; 40:294-302.
30. Byard RW, Donkin J, Vink R. The forensic implications of amphetamine intoxication in cases of inflicted blunt craniocerebral trauma. J Forensic Sci 2018; 63:151-152.
31. Richter F, Hamann M, Richter A. Chronic rotenone treatment induces behavioral effects but no pathological signs of Parkinsonism in mice. J Neurosci Res 2007; 85: 681-691.
32. Abdel-Salam OME, Youness ER, Ahmed NA, El-Toumy SA, Souleman AMA, Shaffie N, Abouelfadl DM. Bougainvillea spectabilis flowers extract protects against the rotenone-induced toxicity. Asian Pac J Trop Med 2017; 10:478-490.
33. Nair V, Turner GA. The thiobarbituric acid test for lipid peroxidation: structure of the adduct with malondialdehyde. Lipids 1984; 19: 804-805.
34. Moshage H, Kok B, Huizenga JR. Nitrite and nitrate determination in plasma: a critical evaluation. Clin Chem 1995; 41:892-896.
35. Koracevic D, Koracevic G, Djordjevic V. Method for the measurement of anti-oxidant activity in human fluids. J Clin Pathol 2001; 54: 356-361.
36. Haagen L, Brock A. A new automated method for phenotyping arylesterase (EC 3.1.1.2) based upon inhibition of enzymatic hydrolysis of 4-nitrophenyl acetate by phenyl acetate. Eur J Clin Chem Clin Biochem 1992; 30:391-395.
37. Drury RVA, Walligton EA. Carltons histological techniques, 5th edn., 1980. Oxford University Press, New York, Pronto.
38. Salakou S, Kardamakis D, Tsamandas AC, Zolota V, Apostolakis E, Tzelepi V et al. Increased Bax/Bcl-2 ratio up-regulates caspase-3 and increases apoptosis in the thymus of patients with myasthenia gravis. In Vivo 2007; 21:123-132.
39. Bashkatova V, Alam M, Vanin A, Schmidt WJ. Chronic administration of rotenone increases levels of nitric oxide and lipid peroxidation products in rat brain. Exp Neurol 2004; 186:235-241.
40. Abdel-Salam OME, Omara EA, El-Shamarka ME-S, Hussein JS. Nigrostriatal damage after systemic rotenone and/or lipopolysaccharide and the effect of cannabis. Comp Clin Pathol 2014; 23:1343-1358.
41. Liu C, Wang R, Ji T, Fan Y, Qin Z, Gao X. Effects of ferulic acid in rotenone induced rat model of Parkinson’s disease. J Pharmaceut Med Res 2016; 2: 55-59.
42. Testa CM, Sherer TB, Greenamyre JT. Rotenone induces oxidative stress and dopaminergic neuron damage in organotypic substantia nigra cultures. Brain Res Mol Brain Res 2005; 134: 109-118.
43. Ibrahim NA, Abdel-Salam OM, Khadrawy YA, Hashem AM, Sameer EM. Non-steroidal anti-inflammatory drugs and vitamin C in the rotenone induced nigrostriatal damage in mice. Eur J Clin Biomed Sci 2017; 3: 67-79.
44. Abdel-Salam OME, Sleem AA, Youness ER, Mohammed NA, Omara EA, Shabana ME. Neuroprotective effects of the glutathione precursor N-acetylcysteine against rotenone- induced neurodegeneration. Reactive Oxygen Species 2019; 8:231-244.
45. Kusano C, Ferrari B. Total anti-oxidant capacity: a biomarker in biomedical and nutritional studies. J Cell Mol Biol 2008; 7:1- 15.
46. He Y, Imam SZ, Dong Z, Jankovic J, Ali SF, Appel SH, Le Q. Role of nitric oxide in rotenone-induced nigro-striatal injury. J Neurochem  2003;86:1338-1345.
47. Abdel-Salam OE, Omara EA, Youness ER, Khadrawy YA, Mohammed NA, Sleem AA. Rotenone-induced nigrostriatal toxicity is reduced by methylene blue. J Neurorestoratol 2014;  2:65-80.
48. Xiong N, Xiong J, Jia M, Liu L, Zhang X, Chen Z, et al. The role of autophagy in Parkinson’s disease: Rotenone-based modeling. Behav Brain Funct 2013; 9:13.
49. Wink DA, Feelisch M, Vodovotz Y, Fukuto J, Grisham MB. The chemical biology of nitric oxide. In: Reactive Oxygen Species in Biological Systems (G Colton). Kluwer Academic/Plenum Publishers, New York, NY, USA. 1999, pp. 245-291.
50. Brown GC. Nitric oxide and neuronal death. Nitric Oxide 2010; 23:153-165.
51. Gao B, Chang C, Zhou J, Zhao T, Wang C, Li C, et al. Pycnogenol protects against rotenone-induced neurotoxicity in PC12 cells through regulating NF-κB-iNOS signaling pathway. DNA Cell Biol 2015;  34: 643-649.
52. Abdel-Salam OME, Youness ER, Mohammed NA, Yassen NN, Shaffie N, Sleem AA. Brilliant blue G protects against rotenone-induced neuronal damage in the rat brain. Reactive Oxygen Species 2017; 4:336-350.
53. Moynagh PN. The NF-κB pathway. J Cell Sci 118: 4389-4392.
54. Turner MD, Nedjai B, Hurst T,  Pennington  DJ. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochimica et Biophysica Acta 2014; 1843: 2563-2582.
55. Bowie A, O’Neill LA. Oxidative stress and nuclear factor-kappaB activation: a reassessment of the evidence in the light of recent discoveries. Biochem Pharmacol 2000; 59:13-23.
56. Androutsopoulos VP, Kanavouras K, Tsatsakis AM. Role of paraoxonase 1 (PON1) in organophosphate metabolism: Implications in neurodegenerative diseases. Toxicol Appl Pharmacol 2011; 256: 418-424.
57. Costa LG, Li WF, Richter RJ, Shih DM, Lusis A, Furlong CE. The role of paraoxonase (PON1) in the detoxication of organophosphates and its human polymorphism. Chem Biol Interact 1999; 119-120:429-438.
58. Li WF, Costa LG, Richter RJ, Hagen T, Shih DM, Tward A, et al. Catalytic efficiency determines the in-vivo efficacy of PON1 for detoxifying organophosphorus compounds. Pharmacogenetics 2000; 10:767-779.
59. Narayan S, Liew Z, Paul K, Lee PC, Sinsheimer JS, Bronstein JM, et al. Household organophosphorus pesticide use and Parkinson’s disease. Int J Epidemiol 2013; 42:1476-1485.
60. Furlong CE, Marsillach J, Jarvik GP, Costa LG. Paraoxonases-1, -2 and -3: What are their Functions? Chem Biol Interact 2016;  259: 51-62.
61. Abdel-Salam OME, Youness ER, Mohammed NA, Abu Elhamed WA. Nuclear factor-kappa B and other oxidative stress biomarkers in serum of autistic children. Open J Mol Integ Physiol 2015; 5:18-27.
62. Jamroz-Wisniewska A, Beltowski J, Stelmasiak Z, Bartosik-Psujek H. Paraoxonase 1 activity in different types of multiple sclerosis. Mult Scler 2009;15:399-402.
63. Wehr H, Bednarska-Makaruk M, Graban A, Lipczynska-Lojkowska W, Rodo M, Bochynska A, et al. Paraoxonase activity and dementia. J Neurol Sci 2009; 283:107-108.
64. Zhou Q, Liu C, Liu W, Zhang H, Zhang R, Liu J, et al. Rotenone induction of hydrogen peroxide inhibits mTOR-mediated S6K1 and 4E-BP1/eIF4E pathways, leading to neuronal apoptosis. Toxicol Sci  2015;143:81-96.
65. Brunelle JK , Letai A. Control of mitochondrial apoptosis by the Bcl-2 family. J Cell Sci 2009; 122:437-441.
66. Watabe M, Nakaki T. Rotenone induces apoptosis via activation of Bad in human dopaminergic SH-SY5Y cells. J Pharmacol Exp Ther 2004; 311: 948-953.
67. Kim HJ, Park HJ, Park HK, Chung JH.Tranexamic acid protects against rotenone-induced apoptosis in human neuroblastoma SH-SY5Y cells. Toxicology 2009; 262:171–174.
68. Badiani A, Oates MM, Day HEW, Watson SJ, Akil H, Robinson TE. Amphetamine-induced behavior, dopamine release, and c-fos mRNA expression: modulation by environmental novelty. J Neurosci 1998; 18:10579-10593.
69. Underhill SM, Hullihen PD, Chen J, Fenollar-Ferrer C, Rizzo MA, Ingram SL, Amara SG. Amphetamines signal through intracellular TAAR1 receptors coupled to Gα13 and GαS in discrete subcellular domains. Mol Psychiatry 2019; 10:1038.
70. Sotnikova TD, Beaulieu JM, Barak LS, Wetsel WC, Caron MG, Gainetdinov RR. Dopamine-independent locomotor actions of amphetamines in a novel acute mouse model of parkinson disease. PLoS Biol 2005; 3: e271.
71. Abdel-Salam OME, Sleem AA, Youness ER, Yassen NN, Shaffie N, El-Toumy SA. Capsicum protects against rotenone-induced toxicity in mice brain via reduced oxidative stress and 5-lipoxygenase activation. J Pharm Pharmacol Res 2018; 2:60–77.