28SEP

Libyan International Conference for Health Sciences

The First Libyan International Conference for Health Sciences (Sept. 28 - 29, 2024): Open University, Tripoli, Libya
Mediterranean Journal of Pharmacy and Pharmaceutical Sciences
https://ppj.org.ly/article/doi/10.5281/zenodo.13309953

Mediterranean Journal of Pharmacy and Pharmaceutical Sciences

Original article

Sorghum bicolor-based supplement reduces oxidative stress and pro-inflammatory cytokines to mitigate rotenone-induced Parkinsonian-like motor dysfunctions in rats

Paul A. Adeleke, Olajide S. Annafi, Abayomi M. Ajayi, Benneth Ben-Azu, Olajuwon Okubena, Solomon Umukoro

Downloads: 0
Views: 354

Abstract

Parkinson’s disease is a common movement disorder associated primarily with oxidative stress-mediated degeneration of dopaminergic neurons. Earlier studies showed that Sorghum bicolor-based supplement (SbS) exhibited antioxidant and neuroprotective activities and might likely rescue the death of dopaminergic neurons in Parkinson’s disease. This study examined the effect of SbS on rotenone-induced Parkinsonian-like motor deficits in rats and the involvement of oxidative stress and pro-inflammatory cytokines. Rats were divided into six groups and treated orally with sunflower oil (vehicle-control), rotenone (2.5 mg/kg) alone or in combination with each dose of SbS (50, 100, and 200 mg/kg) and carbidopa (10 mg/kg) on an alternate day for 28 days. The changes in motor functions were evaluated on day 28 and the brain concentrations of oxidative stress biomarkers and pro-inflammatory cytokines (tumor necrosis factor-alpha and interleukin-6) were determined. Rotenone caused motor deficits by impaired locomotor activity in the open field test and induced catalepsy in the bar test, which were attenuated by SbS. Rats pretreated with SbS had reduced brain levels of malondialdehyde, nitrite, and pro-inflammatory cytokines compared to rotenone controls. 100 mg/kg and 200 mg/kg SbS mitigated rotenone-induced depletion of reduced glutathione and antioxidant enzymes in the rat brain. The results suggest that SbS ameliorated rotenone-induced Parkinsonian-like motor dysfunctions by reducing neuronal oxidative stress and pro-inflammatory cytokines in rats.

Keywords

Pro-inflammatory cytokines, motor deficits, oxidative stress, rotenone

References

  1. Pang SY-Y, Ho PW-L, Liu H-F, Leung C-T, Li L, Chang EES, Ramsden DB, Ho S-L (2019) The interplay of aging, genetics and environmental factors in the pathogenesis of Parkinson’s disease. Translational Neuro-degeneration. 8: 23. doi: 10.1186/s40035-019-0165-9
  2. Yin R, Xue J, Tan Y, Fang C, Hu C, Yang Q, Mei X, Qi D (2021) The positive role and mechanism of herbal medicine in Parkinson's disease. Oxidative Medicine and Cellular Longevity. 9923331. ID: 1-23. doi: 10.1155/2021/ 9923331
  3. Alam M, Schmidt WJ (2002) Rotenone destroys dopaminergic neurons and induces Parkinsonian symptoms in rats. Behavioural Brain Research. 136 (1): 317-324. doi: 10.1016/s0166-4328(02)00180-8
  4. Olanow C, Schapira A (2013) Therapeutic prospects for Parkinson's disease. Annals of Neurology. 74 (3):  337-347. doi: 10.1002/ana.24011
  5. Schapira AH, Jenner P (2011) Etiology and pathogenesis of Parkinson's disease. Movement Disorders. 26 (6): 1049-1055. doi: 10.1002/mds.23732
  6. Taylor JM, Main BS, Crack PJ (2013) Neuroinflammation and oxidative stress: co-conspirators in the pathology of Parkinson’s disease. Neurochemistry International. 62 (5): 803-819. doi: 10.1016/j.neuint.2012.12.016
  7. Sutachan JJ, Casas Z, Albarracin SL, Stab BR, Samudio I, Gonzalez J, Barreto G (2012) Cellular and molecular mechanisms of antioxidants in Parkinson's disease. Nutritional Neuroscience. 15 (3): 120-126. doi: 10.1179/ 1476830511Y.0000000033
  8. Trist BG, Hare DJ, Double KL (2019) Oxidative stress in the aging substantia nigra and the etiology of Parkinson's disease. Aging Cell. 18 (6): e13031. doi: 10.1111/acel.13031
  9. Nakabeppu Y, Tsuchimoto D, Yamaguchi H, Sakumi K (2007) Oxidative damage in nucleic acids and Parkinson's disease. Journal of Neuroscience Research. 85 (5): 919-934. doi: 10.1002/jnr.21191
  10. Sarkar S, Raymick J, Imam S (2006) Neuroprotective and therapeutic strategies against Parkinson’s disease: recent perspectives. International Journal of Molecular Sciences. 17 (6): 904. doi: 10.3390/ijms17060904
  11. Puspita L, Chung SY, Shim J-W (2007) Oxidative stress and cellular pathologies in Parkinson’s disease. Molecular Brain. 10 (1): 53. doi: 10.1186/s13041-017-0340-9
  12. Bosco D, Fowler D, Zhang Q, Nieva J, Powers E, Wentworth P, Kelly J (2006) Elevated levels of oxidized cholesterol metabolites in Lewy body disease brains accelerate α-synuclein fibrilization. Nature Chemical Biology. 2 (5): 249-253. doi: 10.1038/nchembio782
  13. Zeevalk GD, Razmpour R, Bernard LP (2008) Glutathione and Parkinson's disease: is this the elephant in the room? Biomedicine & Pharmacotherapy. 62 (4): 236-249. doi: 10.1016/j.biopha.2008.01.017
  14. Hirsch E, Jenner P, Przedborski S (2008) Pathogenesis of Parkinson's disease. Movement Disorders. 28 (1): 24-30. doi: 10.1002/mds.25032
  15. Schonhoff A, Williams G, Wallen Z, Standaert D, Harms A (2020) Innate and adaptive immune responses in Parkinson's disease. Progress in Brain Research. 252: 169-216. doi: 10.1016/bs.pbr.2019.10.006
  16. Sharma V, Bedi O, Gupta M, Deshmukh R (2022) A review: traditional herbs and remedies impacting pathogenesis of Parkinson’s disease. Naunyn Schmiedeberg's Archives of Pharmacology. 395 (5): 495-513.  doi: 10.1007/s00210-022-02223-5
  17. Rai SN, Chaturvedi VK, Singh P, Singh BK, Singh MP (2020) Mucuna pruriens in Parkinson's and in some other diseases: recent advancement and future prospective. 3 Biotech. 10 (12): 522. doi: 10.1007/s13205-020-02532-7
  18. Rai SN, Singh P, Varshney R, Chaturvedi VK, Vamanu E, Singh MP, Singh BK (2021) Promising drug targets and associated therapeutic interventions in Parkinson's disease. Neural Regeneration Research. 16 (9): 1730-1739. doi: 10.4103/1673-5374.306066
  19. Benson KF, Beaman JL, Ou B, Okubena A, Okubena O, Jensen GS (2013) West African Sorghum bicolor leaf sheaths have anti-inflammatory and immune-modulating properties in vitro. Journal of Medicinal Food. 16 (3): 230-238. doi: 10.1089/jmf.2012.0214
  20. Umukoro S, Omogbiya IA, Eduviere AT (2013) Evaluation of the effect of jobelyn® on chemoconvulsants-induced seizure in mice. Basic and Clinical Neuroscience. 4 (2): 125-129. PMID: 25337338.
  21. Oyinbo CA, Dare W, Avwioro O, Igbigbi P (2015) Neuroprotective effect of Jobelyn in the hippocampus of alcoholic rat is mediated in part by alterations in GFAP and NF 789 protein expressions. Advances in Biological Research. 9 (5): 305-317. doi: 10.5829/idosi.abr.2015.9.5.95109
  22. Umukoro S, Oghwere EE, Ben-Azu B, Owoeye O, Ajayi AM, Omorogbe O, Okubena O (2019) Jobelyn® ameliorates neurological deficits in rats with ischemic stroke through inhibition of release of pro-inflammatory cytokines and NF-κB signaling pathway. Pathophysiology. 26 (1): 77-88. doi: 10.1016/j.pathophys.2018.10.00 2
  23. John R, Abolaji AO, Adedara AO, Ajayi AM, Aderibigbe AO, Umukoro S (2022) Jobelyn® extends the life span and improves motor function in Drosophila melanogaster exposed to lipopolysaccharide via augmentation of antioxidant status. Metabolic Brain Disease. 37 (4): 1031-1040. doi: 10.1007/s11011-022-00919-4
  24. Morais LH, Lima MM, Martynhak BJ, Santiago R, Takahashi TT, Ariza D, Barbiero JK, Andreatini R, Vital MA (2012) Characterization of motor, depressive-like and neurochemical alterations induced by a short-term rotenone administration. Pharmacological Reports. 64 (5):1081-1090. doi: 10.1016/s1734-1140(12)70905-2
  25. Arika WM, Kibiti CM, Njagi JM, Ngugi MP (2019) Effects of DCM leaf extract of Gnidia glauca (Fresen) on locomotor activity, anxiety, and exploration‐like behaviors in high‐fat diet‐induced obese rats. Behavioural Neurology. 2019: 7359235. doi: 10.1155/2019/7359235
  26. Costall B, Naylor R (1973) On catalepsy and catatonia and the predictability of the catalepsy test for neuroleptic activity. Psychopharmacologia. 34: 233-241. doi: 10.1007/BF00421964
  27. Ohkawa H, Ohishi N, Yagi K (1979) Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Analytical Biochemistry. 95 (2):  351-358. doi: 10.1016/0003-2697(79)90738-3
  28. Green L, Tannenbaum S, Goldman P (1981) Nitrate synthesis in the germfree and conventional rat. Science. 212 (4490): 56-58. doi: 10.1126/science.6451927
  29. Moron M, Depierre J, Mannervik B (1979) Levels of glutathione, glutathione reductase and glutathione S-transferase activities in rat lung and liver. Biochimica et Biophysica Acta. 582 (1):  67-78. doi: 10.1016/0304-4165(79)90289-7
  30. Goth L (1991) A simple method for determination of serum catalase activity and revision of reference range. Clinica Chimica Acta. 196 (2-3): 143-151. doi: 10.1016/0009-8981(91)90067-m
  31. Misra H, Fridovich I (1972) The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. Journal of Biological Chemistry. 247 (10): 3170-3175. doi: 10.1016/S0021-9258(19) 45228-9
  32. Lowry O, Rosebrough N, Farr A, Randall R (1951) Protein measurement with the folin phenol reagent. The Journal of Biological Chemistry. 193 (1): 265-275. PMID: 14907713.
  33. Dhanalakshmi C, Janakiraman U, Manivasagam T, Justin Thenmozhi A, Essa M, Kalandar A, Guillemin G (2006) Vanillin attenuated behavioural impairments, neurochemical deficits, oxidative stress and apoptosis against rotenone induced rat model of Parkinson’s disease. Neurochemical Research. 41 (8): 1899-1910. doi: 10.1007/s11064-016-1901-5
  34. Saeed A, Shakir L, Khan M, Ali A, Zaidi A (2017) Haloperidol induced Parkinson’s disease mice model and motor-function modulation with Pyridine-3-carboxylic acid. Biomedical Research and Therapy. 4 (5): 1305-1317. doi: 10.15419/BMRAT.V4105.169  
  35. Cooper J, Spielbauer K, Senchuk M, Nadarajan S, Colaiácovo M, Van Raamsdonk J (2018) α-synuclein expression from a single copy transgene increases sensitivity to stress and accelerates neuronal loss in genetic models of Parkinson's disease. Experimental Neurology. 310: 58-69. doi: 10.1016/j.expneurol.2018.09.001
  36. Waku I, Magalhaes M, Alves C, de Oliveira A (2021) Haloperidol‐induced catalepsy as an animal model for parkinsonism: A systematic review of experimental studies. The European Journal of Neuroscience. 53 (11): 3743-3767. doi: 10.1111/ejn.15222
  37. Alabi A, Ajayi A, Ben-Azu B, Bakre A, Umukoro S (2019) Methyl jasmonate abrogates rotenone-induced parkinsonian-like symptoms through inhibition of oxidative stress, release of pro-inflammatory cytokines, and down-regulation of immnopositive cells of NF-κB and α-synuclein expressions in mice. Neurotoxicology. 74: 172-183. doi: 10.1016/j.neuro.2019.07.003
  38. Dawson TB, Dawson VL (2009) Molecular pathways of neurodegeneration in Parkinson's disease. Science. 302 (5646): 819-822. doi: 10.1126/science.1087753
  39. Sherer T, Betarbet R, Testa CM, Seo BB, Richardson JR, Kim JH, Miller GW, Yagi T, Yagi AM, Greenamyre JT (2003) Mechanism of toxicity in rotenone models of Parkinson's disease. Journal of Neuroscience. 23 (34): 10756-10764. doi: 10.1523/JNEUROSCI.23-34-10756.2003
  40. Pan P, Qiao L, Wen X (2016) Safranal prevents rotenone-induced oxidative stress and apoptosis in an in vitro model of Parkinson's disease through regulating Keap1/Nrf2 signaling pathway. Cellular and Molecular Biology. 62 (14): 11-17. PMID: 28145852.
  41. Pearce R, Owen A, Daniel S, Jenner P, Marsden C (1997) Alterations in the distribution of glutathione in the substantia nigra in Parkinson's disease. Journal of Neural Transmission. 104 (6-7): 661-677. doi: 10.1007/ BF01291884
  42. Afe TO, Alabi A, Ajayi AM, Ale AO, Fasesan OA, Ogunsemi OO (2024) Jobelyn® ameliorates anxiety response and oxido-inflammatory markers induced by tramadol use and discontinuation in rats. Mediterranean Journal of Pharmacy and Pharmaceutical Sciences. 4 (1): 93-110. doi: 10.5281/zenodo.10728692
  43. Owumi S, Kazeem A, Wu B, Ishokare L, Arunsi U, Oyelere A (2022) Apigeninidin-rich Sorghum bicolor (L. Moench) extracts suppress A549 cells proliferation and ameliorate toxicity of aflatoxin B1-mediated liver and kidney derangement in rats. Scientific Reports. 12 (1): 7438. doi: 10.1038/s41598-022-10926-1
  44. Wang W, Wang Y, Wagner K, Lee R, Hwang S, Morisseau C, Wulff H, Hammock B (2023) Aflatoxin B1 increases soluble epoxide hydrolase in the brain and induces neuroinflammation and dopaminergic neurotoxicity. International Journal of Molecular Sciences. 24 (12): 9938. doi: 10.3390/ijms24129938

Submitted date:
07/22/2024

Reviewed date:
08/12/2024

Accepted date:
08/15/2024

Publication date:
08/13/2024

66bb35c1a9539572a11fe263 medjpps Articles
Links & Downloads

Mediterr J Pharm Pharm Sci

Share this page
Page Sections