Bee venom attenuates neurodegeneration and motor impairment and modulates the response to L-dopa or rasagiline in a mice model of Parkinson’s disease

Document Type : Original Article

Authors

1 Holding Company for Biological Products, Vaccines and Drugs (VACSERA), Cairo, Egypt

2 Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt

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

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

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

Abstract

Objective(s): This study aimed to investigate the effect of bee venom, a form of alternative therapy, on rotenone-induced Parkinson’s disease (PD) in mice. Moreover, the possible modulation by bee venom of the effect of L-dopa/carbidopa or rasagiline was examined.
Materials and Methods: Rotenone (1.5 mg/kg, subcutaneously; SC) was administered every other day for two weeks and at the same time mice received the vehicle (DMSO, SC), bee venom (0.065, 0.13, and 0.26 mg/kg; intradermal; ID), L-dopa/carbidopa (25 mg/kg, intraperitoneal; IP), L-dopa/carbidopa+bee venom (0.13 mg/kg, ID), rasagiline (1 mg/kg, IP) or rasagiline+bee venom (0.13 mg/kg, ID). Then, wire hanging and staircase tests were performed and mice were euthanized and brains’ striata separated. Oxidative stress biomarkers namely, malondialdehyde (MDA), nitric oxide (NO), reduced glutathione (GSH), paraoxonase-1 (PON-1), and total antioxidant capacity (TAC) were measured. Additionally, butyrylcholinesterase (BuChE), monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), and dopamine (DA) were evaluated. Brain histopathological changes and caspase-3- expression were done.
Results: Bee venom significantly enhanced motor performance and inhibited rotenone-induced oxidative/nitrosative stress, observed as a reduction in both MDA and NO along with increasing GSH, PON-1, and TAC. Besides, bee venom decreased MCP-1, TNF-α, and caspase-3 expression together with an increase in BuChE activity and DA content.
Conclusion: Bee venom alone or in combination with L-dopa/carbidopa or rasagiline alleviated neuronal degeneration compared with L-dopa/carbidopa or rasagiline treatment only. Bee venom via its antioxidant and cytokine reducing potentials might be of value either alone or as adjunctive therapy in the management of PD.

Keywords


1. 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.
2. Wirdefeldt K, Adami HO, Cole P, Trichopoulos D, Mandel J. Epidemiology and etiology of Parkinson’s disease: A review of the evidence. Eur J Epidemiol 2011; 26:S1-S58.
3. Le W, Sayana P, Jankovic J. Animal Models of Parkinson’s disease: A gateway to therapeutics. Neuro Therapeutics 2014; 11:92-110.
4. Cai M, Choi SM, Yang EJ. The effects of bee venom acupuncture on the central nervous system and muscle in an animal hSOD1G93A mutant. Toxins 2015, 7, 846-858.
5. Awad K, Abushouk AI, Abdel Karim AH, Mohammed M, Negida A, Shalash AS. (2017). Bee venom for the treatment of Parkinson’s disease: How far is it possible? Biomed Pharmacother 2017;91:295-302.
6. Hossen MS, Shapla UM, Gan SH, Khalil MI. Impact of bee venom enzymes on diseases and immune responses. Molecules 2016;22:25.
7. Eiseman JL, von Bredow J, Alvares AP. Effect of honeybee (Apis mellifera) venom on the course of adjuvant-induced arthritis and depression of drug metabolism in the rat. Biochem Pharmacol 1982; 31:1139–1146.
8. Yang EJ, Kim SH, Yang SC, Lee SM, Choi SM. Melittin restores proteasome function in an animal model of ALS. J Neuroinflammation 2011; 8: 69.
9. Alvarez-Fischer D, Noelker C, Vulinović F. et al. Bee venom and its component apamin as neuroprotective agents in a Parkinson disease mouse model. PLoS ONE 2013; 8: e61700.
10. Koburova KL, Michailova SG, Shkenderov SV. Further investigation on the anti-inflammatory properties of adolapin-Bee venom polypeptide. Acta Physiol Pharmacol Bulg 1985; 11: 50-55.
11. Ostrovsky DA, Ehrlich A. Bee venom acupuncture in addition to anti-Parkinsonian medications may improve activities of daily living and motor symptoms more than medication alone in idiopathic Parkinson’s disease. J Altern Complement Med 2019;15:71-73.
12. Lewitt PA. Levodopa for the treatment of Parkinson’s disease. N Engl J Med 2008; 359: 2468-2476.
13. Anelyssa D’Abreu. Parkinson’s disease: A quick update.  R I Med J 2018; 101:34-36.
14. Ondo WG. Motor complications in Parkinson’s disease. Int J Neurosci 2011; 121: 37-44.
15. Abdel-Salam OME. Drug therapy for Parkinson’s disease: An update. World J Pharmacol 2015; 4: 117-143.
16. Doo AR1, Kim ST, Kim SN, Moon W, Yin CS, Chae Y, et al. Neuroprotective effects of bee venom pharmaceutical acupuncture in acute 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced mouse model of Parkinson’s disease. Neurol Res 2010; 32:88-91.
17. Chung ES1, Kim H, Lee G, Park S, Kim H, Bae H. Neuro-protective effects of bee venom by suppression of neuroinflammatory responses in a mouse model of Parkinson’s disease: role of regulatory T cells. Brain Behav Immun 2012;26:1322-1330.
18. Nicolas Maurice, Thierry Deltheil, Christophe Melon, Bertrand Degos, Christiane Mourre, Marianne Amalric, et al. Bee venom alleviates motor deficits and modulates the transfer of cortical information through the basal ganglia in rat models of parkinson’s disease. PloS One 2015; 10:e0142838.
19. Jung SY, Lee KW, Choi SM, Yang EJ. Bee venom protects against rotenone-induced cell death in NSC34 motor neuron cells. Toxins 2015; 7: 3715-3726.
20. Cho SY, Shim SR, Rhee HY, Park HJ, Jung WS, Moon SK, et al. Effectiveness of acupuncture and bee venom acupuncture in idiopathic Parkinson’s disease. Parkinsonism Relat Disord. 2012; 18:948-952.
21. Paget GE, Barnes JM. In: Laurence, D.R. and Bacharach, A.L., Eds., Evaluation of Drug Activities, Academic Press, Massachusetts, 135-166.
22. Thiffault C, Langston JW, Di Monte DA. Increased striatal dopamine turnover following acute administration of rotenone to mice. Brain Res 2000; 885:283-288.
23. Uchiyama M, Mihara M. Determination of malondialdehyde precursor in tissue by thiobarbituritic acid test. Anal Biochem 1978; 86: 271-278.
24. Ruiz-Larrea MB, Leal AM, Liza M, Lacort M, de Groot H. Antioxidant effects of estradiol and 2-hydroxyestradiol on iron-induced lipid peroxidation of rat liver microsomes. Steroids. 1994; 59:383–388.
25. Moshage H, Kok B, Huizenga JR, Jansen PL. Nitrite and nitrate determination in plasma: a critical evaluation. Clin Chem 1995; 41:892–896.
26. Ellman GL. Tissue sulfhydryl groups. Arch Biochem Biophys 1959;82:70–77
27. E Beutler, O Duron, BM Kelly. Improved Method for the Determination of Blood Glutathione. J Lab Clin Med 1963; 61:882-888.
28. Bulaj G, Kortemme T, Goldenberg DP. Ionization-reactivity relationships for cysteine thiols in polypeptides. Biochem 1998; 37: 8965-8972.
29. Koracevic D, Koracevic G, Djordjevic V, Andrejevic S, Cosic V. Method for the measurements of antioxidants activity in human fluids. J Clin Pathol 2001; 54: 356-361.
30. Higashino K, Takahashi Y, Yamamura Y. Release of phenyl acetate esterase from liver microsomes by carbon tetrachloride. Clin Chim Acta 1972; 41:313-320.
31. Watson AD, Berliner JA, Hama SY. et al. Protective effect of high density lipoprotein associated paraoxonase. Inhibition of the biological activity of minimally oxidized low density lipoprotein. J Clin Invest 1995; 96:2882-2891.
32. Sanberg PR, Martinez R, Douglas Shytle, David W Cahill. The catalepsy test: Is a standardized method possible? Motor Activity and Movement Disorders 1996. pp. 197–211.
33. Baird AL, Meldrum A, Dunnett SB. The staircase tests of skilled reaching in mice. Brain Res Bull 2001; 54:243-250.
34. Li N, Ragheb K, Lawler G, Sturgis J, Rajwa B, Melendez JA, et al. Robinson Mitochondrial complex I inhibitor rotenone induces apoptosis through enhancing mitochondrial reactive oxygen species production. J Biol Chem 2003; 278: 8516–8525.
35. Samantaray S, Knaryan VH, Guyton MK, Matzelle DD, Ray SK, Banik NL. The parkinsonian neurotoxin rotenone activates calpain and caspase-3 leading to motoneuron degeneration in spinal cord of Lewis rats. Neuroscience 2007; 146: 741-755.
36. Im AR, Kim YH, Uddin MR. et al. Betaine protects against rotenone-induced neurotoxicity in PC12 cells. Cell Mol  Neurobiol 2013; 33: 625–635.
37. Tamilselvam K, Braidy N, Manivasagam T, Essa MM, Prasad NR, Karthikeyan S, et al. Neuroprotective effects of hesperidin, a plant flavanone, on rotenone-induced oxidative stress and apoptosis in a cellular model for Parkinson’s disease. Oxid Med Cell Longev 2013; 2013:102741.
38. Valentina Bashkatova , Mesbah Alam, Anatoly Vanin, Werner J Schmidt. Chronic administration of rotenone increases levels of nitric oxide and lipid peroxidation products in rat brain. Exp Neurol 2004;186:235-41.
39. 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.
40. Liu L, Arun A, Ellis L, Peritore C, Donmez G. SIRT2 enhances 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigrostriatal damage via apoptotic pathway. Front Aging Neurosci 2014; 6:184.
41. Draganov DI, La Du BN. Pharmacogenetics of paraoxonases: a brief review. Naunyn Schmiedeberg’s Arch Pharmacol 2004; 369:78–88
42. Lee PC, Rhodes SL, Sinsheimer JS, Bronstein J, Ritz B. Functional paraoxonase 1 variants modify the risk of Parkinson’s disease due to organophosphate exposure. Environ Int 2013; 56:42-47
43. Abdel-Salam OME, Youness ER, Khadrawy YA, Mohammed NA, Abdel-Rahman RF, Omara EA, et al. The effect of cannabis on oxidative stress and neurodegeneration induced by intrastriatal rotenone injection in rats. Comp Clin Pathol 2015; 24:359-378.
44. Son DJ, Lee JW, Lee YH, Song HS, Lee CK, Hong JT.Therapeutic application of anti-arthritis,pain-releasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacol Ther 2007;115: 246-270.
45. Gajski G, Garaj-Vrhovac V. Radioprotective effects of honeybee venom (Apismellifera) against 915-MHZ microwave radiation-induced DNA damage in wister rat lymphocytes: In vitro study. Int J Toxicol 2009; 28: 88–98.
46. Varanda EA, Monti R, Tavares DC. Inhibitory effect of propolis and bee venom on the mutagenicity of some direct- and indirect-acting mutagens. Teratog Carcinog Mutagen 1999; 19: 403-413.
47. Park YC, Koh PS, Seo BK, Lee JW, Cho NS, Park HS, et al. Long-term effectiveness of bee venom acupuncture and physiotherapy in the treatment of adhesive capsulitis: A one-year follow-up analysis of a previous randomized controlled trial. J Altern Complement Med 2014; 20: 919–924.
48. Yoon H, Kim MJ, Yoon I, Li DX, Bae H, Kim SK. Nicotinic acetylcholine receptors mediate the suppressive effect of an injection of diluted bee venom into the GV3 acupoint on oxaliplatin-induced neuropathic cold allodynia in rats. Biol Pharm Bull 2015; 38: 710–714.
49. Huh JE, Baek YH, Lee MH, Choi DY, Park DS, Lee JD. Bee venom inhibits tumor angiogenesis and metastasis by inhibiting tyrosine phosphorylation of VEGFR-2 in LLC-tumor-bearing mice. Cancer Lett 2010; 292: 98–110.
50. Chung ES, Lee G, Lee C, Ye M, Chung HS, Kim H, et al. Bee venom phospholipase A2, a novel foxp3+ regulatory t cell inducer, protects dopaminergic neurons by modulating neuroinflammatory responses in a mouse model of Parkinson’s disease. J Immunol 2015; 195: 4853–4860.
51. Park S, Baek H, Jung KH, Lee G, Lee H, Kang GH, et al. Bee venom phospholipase A2 suppresses allergic airway inflammation in an ovalbumin-induced asthma model through the induction of regulatory t cells. Immun Inflamm Dis 2015; 3: 386–397.
52. MK Rakha, RA Tawfiq, MM Sadek, MA Anwer, SM Salama, AF Mohamed, et al. Neurotherapeutic effects of bee venom in a rotenone-induced mouse model of Parkinson’s disease. Neurophysiol 2019; 50:445-455.
53. Chalimoniuk M, Stolecka A, Ziemińska E, Stepień A, Langfort J, Strosznajder JB. Involvement of multiple protein kinases in cPLA2 phosphorylation, arachidonic acid release, and cell death in in vivo and in vitro models of 1-methyl-4-phenylpyridinium-induced Parkinsonism–the possible key role of PKG. J Neurochem 2009; 110: 307–317.
54. Oliveras-Salvá M, Van der Perren A, Casadei N, et al. rAAV2/7 vector-mediated overexpression of alphasynuclein in mouse substantianigra induces protein aggregation and progressive dose-dependent neurodegeneration. Mol Neurodegener 2013; 8: 44-33.
55. Sacino AN, Thomas MA, Ceballos-Diaz C, Cruz PE, Rosario AM, Lewis J, et al. Conformational templating of alpha-synuclein aggregates in neuronal-glial cultures. Mol Neurodegener 2013; 8: 17.
56. Ye M, Chung HS, Lee C, Song JH, Shim I, Kim YS, et al. Bee venom phospholipase A2 ameliorates motor dysfunction and modulates microglia activation in Parkinson’s disease alpha-synuclein transgenic mice. Exp Mol Med 2016;48:e244.
57. Hwang DS, Kim SK, Bae H.Therapeutic effects of bee venom on immunological and neurological diseases review. Toxins (Basel) 2015;7:2413-2421.
58. Mervat Y. Hanafi, Eman LM Zaher, Soha EM ElAdely, Ahmed Sakr, Ahmed HM Ghobashi, Madiha H. Hemly, et al. The therapeutic effects of bee venom on some metabolic and anti-oxidant parameters associated with HFDinduced nonalcoholic fatty liver in rats. Exp Ther Med. 2018; 15:5091-5099.
59. Stojkovska I, Wagner BM, Morrison BE. Parkinson’s disease and enhanced inflammatory response. Exp Biol Med 2015; 240: 1387–1395.
60. Lee G, Bae H. Anti-inflammatory applications of melittin, a major component of bee venom: Detailed mechanism of action and adverse effects. Molecules 2016; 21:616.
61. Samar F. Darwish, Wesam M. El-Bakly, Hossam M. Arafa, Ebtehal El-Demerdash. Targeting TNF-α and NF-κB activation by Bee venom: Role in suppressing adjuvant induced arthritis and methotrexate hepatotoxicity in rats. PLoS One 2013; 8:e79284.
62. Andreas Aufschnaiter, Verena Kohler, Shaden Khalifa, Aida Abd El-Wahed, Ming Du, Hesham El-Seedi, et al. Apitoxin and its components against cancer, neurodegeneration and rheumatoid arthritis: Limitations and possibilities. Toxins 2020; 12: 66.
63. Chen L, Deltheil T, Turle-Lorenzo N, Liberge  M, Rosier C, Watabe  I, et al. SK channel blockade reverses cognitive and motor deficits induced by nigrostriatal dopamine lesions in rats. Int J Neuropsycho pharmacol. 2014; 17:1295–1306.
64. Hartmann A, Müllner J, Meier N, Hesekamp H, Meerbeeck  PV, Habert MO, et al. Bee venom for the treatment of Parkinson disease. PLoS One 2016;11:e0158235.  
65. Rizzi G, Tan KR. Dopamine and Acetylcholine, a Circuit Point of View in Parkinson’s disease. Front Neural Circuits 2017;11:110.
66. Han S, Lee K, Yeo J, Kweon H, Woo S, Lee ML, et al. Effect of honey bee venom on microglial cells nitric oxide and tumor necrosis factor-alpha production stimulated by LPS. J Ethnopharmacol 2007;111:176-81.
67. Zhang S, Liu Y, Ye Y, Wang XR, Lin LT, Xiao LY, et al. Bee venom therapy: Potential mechanisms and therapeutic applications. Toxicon 2018;148:64-73.
68. de Souza JM, Goncalves BDC, Gomez MV, Vieira LB, Ribeiro FM. Animal Toxins as Therapeutic Tools to Treat Neurodegenerative Diseases. Front Pharmacol 2018;9:145.