Neuroprotective effects of ferulic acid and thymoquinone against deltamethrin-induced neurotoxicity in Drosophila melanogaster

Hala Abdulbaki, Mohammad Ali Al-Deeb

Abstract


Background: Recent research indicates a link between exposure to pyrethroid pesticides and negative neurodevelopmental outcomes such as neurodegenerative disorders, low intelligence quotient (IQ), and attention disorders. Thus this study aims (1) to investigate the effect of exposing Drosophila melanogaster adult male flies to 0.59 µM deltamethrin (DLM) for 24 h on their survival, climbing behavior, and acetylcholinesterase (AChE) activity, as well as the gene expression levels of pleddcdataanat1dop1r1dop2r, and dopecr genes, and (2) to assess the protective effect of 250 µM Ferulic acid (FA), 25 µM Thymoquinone (TQ), and their combinations on the survival, climbing behavior, and AChE activity of D. melanogaster adult male flies exposed to 0.59 µM DLM for 72 h.

Methods: In the first experiment, adult male wild-type flies were exposed to DLM incorporated into a 10% sucrose solution for 24 h, whereas, in the second experiment, they were exposed to DLM and the individual neuroprotective agents, or their mixture for 72 h.

Results: Flies exposed to DLM exhibited higher mortality rates and shorter climbing distances in the negative geotaxis assay compared to control flies. Additionally, exposure to DLM upregulated the genes ddcdatdop1r1dop2r, and dopecr significantly. Moreover, exposure to DLM for 72 h resulted in higher mortality rates and severe locomotor defects. However, the neuroprotective agents were effective in mitigating these toxic effects. While DLM inhibited AChE activity, concurrent exposure to FA ameliorated the inhibition.

Conclusion: This study demonstrates the efficacy of natural compounds in reducing DLM-induced toxicity in D. melanogaster and highlights the potential use of this model in studying and developing therapeutic strategies for movement disorders.

Keywords: Drosophila melanogaster; Neurotoxicity; Ferulic acid; Thymoquinone; Deltamethrin; Negative geotaxis    

 


Full Text:

PDF

References


El Golli-Bennour E, Timoumi R, Annaibi E, Mokni M, Omezzine A, Bacha H, Abid-Essefi S. Protective effects of kefir against deltamethrin-induced hepatotoxicity in rats. Environmental Science and Pollution Research, (2019); 26(18): 18856–18865.

Han B, Lv Z, Zhang X, Lv Y, Li S, Wu P, Yang Q, Li J, Qu B, Zhang Z. Deltamethrin induces liver fibrosis in quails via activation of the TGF-β1/Smad signaling pathway. Environmental Pollution, (2020); 259: 113870.

Moser VC. Animal models of chronic pesticide neurotoxicity. Human & Experimental Toxicology, (2007); 26(4): 321–332.

Tewari A, Banga HS, Gill JPS. Sublethal chronic effects of oral dietary exposure to deltamethrin in Swiss albino mice. Toxicology and Industrial Health, (2018); 34(6): 423–432.

Abdel-Daim MM, Abuzead SMM, Halawa SM. Protective role of Spirulina platensis against acute deltamethrin-induced toxicity in rats. PloS one, (2013); 8(9): e72991–e72991.

Romero A, Ares I, Ramos E, Castellano V, Martínez M, Martínez-Larrañaga MR, Anadón A, Martínez MA. Evidence for dose-additive effects of a type II pyrethroid mixture. In vitro assessment. Environmental Research, (2015); 138: 58–66.

Li HY, Wu SY, Ma Q, Shi N. The pesticide deltamethrin increases free radical production and promotes nuclear translocation of the stress response transcription factor Nrf2 in rat brain. Toxicology and industrial health, (2011); 27(7): 579–590.

Mani VM, Sadiq AMM. Naringin modulates the impairment of memory, anxiety, locomotor, and emotionality behaviors in rats exposed to deltamethrin; a possible mechanism association with oxidative stress, acetylcholinesterase and ATPase. Biomedicine & Preventive Nutrition, (2014); 4(4): 527–533.

Mohammadi H, Ghassemi-Barghi N, Malakshah O, Ashari S. Pyrethroid exposure and neurotoxicity: a mechanistic approach. Archives of Industrial Hygiene and Toxicology, (2019); 70(2): 74–89.

Park JH, Jung JW, Ahn Y-J, Kwon HW, Neuroprotective properties of phytochemicals against paraquat-induced oxidative stress and neurotoxicity in Drosophila melanogaster. Pesticide Biochemistry and Physiology, (2012); 104(2): 118–125.

Farombi EO, Abolaji AO, Farombi TH, Oropo AS, Owoje OA, Awunah MT. Garcinia kola seed biflavonoid fraction (Kolaviron), increases longevity and attenuates rotenone-induced toxicity in Drosophila melanogaster. Pesticide Biochemistry and Physiology (2018); 145: 39–45.

Seidl SE, Potashkin JA. The promise of neuroprotective agents in Parkinson’s disease. Frontiers in neurology, (2011); 2: 68.

de Andrade Teles RB, Diniz TC, Costa Pinto TC, de Oliveira Júnior RG, Gama E Silva M, de Lavor ÉM, Fernandes AWC, de Oliveira AP, de Almeida Ribeiro FPR, da Silva AAM, et al. Flavonoids as Therapeutic Agents in Alzheimer’s and Parkinson’s Diseases: A Systematic Review of Preclinical Evidences. Oxidative medicine and cellular longevity, (2018); 2018: 7043213.

Pandey KB, Rizvi SI. Plant polyphenols as dietary antioxidants in human health and disease. Oxidative medicine and cellular longevity, (2009); 2(5): 270–278.

Li X, Zhang J, Rong H, Zhang X, Dong M. Ferulic Acid Ameliorates MPP+/MPTP-Induced Oxidative Stress via ERK1/2-Dependent Nrf2 Activation: Translational Implications for Parkinson Disease Treatment. Molecular Neurobiology, (2020); 57(7): 2981–2995.

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 design, development and therapy, (2015); 9: 5499–5510.

Thapliyal S, Singh T, Handu S, Bisht M, Kumari P, Arya P, Srivastava P, Gandham R. A Review on Potential Footprints of Ferulic Acid for Treatment of Neurological Disorders. Neurochemical Research, (2021); 46(5): 1043–1057.

Ramadan MF. Nutritional value, functional properties and nutraceutical applications of black cumin (Nigella sativa L.): an overview. International Journal of Food Science & Technology, (2007); 42(10): 1208–1218.

Akram Khan M, Afzal M. Chemical composition of Nigella sativa Linn: Part 2 Recent advances. Inflammopharmacology, (2016); 24(2–3): 67–79.

Isaev NK, Chetverikov NS, Stelmashook E V, Genrikhs EE, Khaspekov LG, Illarioshkin SN. Thymoquinone as a Potential Neuroprotector in Acute and Chronic Forms of Cerebral Pathology. Biochemistry (Moscow), (2020); 85(2): 167–176.

Sedaghat R, Roghani M, Khalili M. Neuroprotective effect of thymoquinone, the nigella sativa bioactive compound, in 6-hydroxydopamine-induced hemi-parkinsonian rat model. Iranian journal of pharmaceutical research: IJPR (2014); 13(1): 227.

Farkhondeh T, Samarghandian S, Shahri AMP, Samini F. The Neuroprotective Effects of Thymoquinone: A Review. Dose-response : a publication of International Hormesis Society, (2018); 16(2): 1559325818761455–1559325818761455.

Radad K, Moldzio R, Taha M, Rausch W-D. Thymoquinone protects dopaminergic neurons against MPP+and rotenone. Phytotherapy Research, (2009); 23(5): 696–700.

Ardah MT, Merghani MM, Haque ME. Thymoquinone prevents neurodegeneration against MPTP in vivo and modulates α-synuclein aggregation in vitro. Neurochemistry International, (2019); 128: 115–126.

Caesar LK, Cech NB. Synergy and antagonism in natural product extracts: when 1 + 1 does not equal 2. Natural product reports, (2019); 36(6): 869–888.

Jana SC, Bettencourt-Dias M, Durand B, Megraw TL. Drosophila melanogaster as a model for basal body research. Cilia, (2016); 5: 22.

Ugur B, Chen K, Bellen HJ. Drosophila tools and assays for the study of human diseases. Disease models & mechanisms, (2016); 9(3): 235–244.

Pandey UB, Nichols CD. Human disease models in Drosophila melanogaster and the role of the fly in therapeutic drug discovery. Pharmacological reviews, (2011); 63(2): 411–436.

Sudati JH, Vieira FA, Pavin SS, Dias GRM, Seeger RL, Golombieski R, Athayde ML, Soares FA, Rocha JBT, Barbosa NV. Valeriana officinalis attenuates the rotenone-induced toxicity in Drosophila melanogaster. NeuroToxicology, (2013); 37: 118–126.

Hossain MS, Sapon MA. Drosophila as a Model for Analyzing of Human Genetic and Pathogenic Related Diseases. International Journal of Innovation and Scientific Research, (2014); 12: 126-134.

Panchal K, Tiwari AK. Drosophila melanogaster “a potential model organism” for identification of pharmacological properties of plants/plant-derived components. Biomedicine & Pharmacotherapy, (2017); 89: 1331–1345.

Cheng L, Baonza A, Grifoni D. Drosophila Models of Human Disease. BioMed research international, (2018); 2018: 7214974.

Abdulbaki H, Al-Deeb M. Chlorpyrifos-induced dopaminergic damage in Drosophila melanogaster assessed by gene expression , AChE assay , and negative geotaxis. Genetics and Molecular Reaserch, (2022); 21(3).

Livak KJ, Schmittgen TD. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2−ΔΔCT Method. Methods, (2001); 25(4): 402–408.

Norry FM, Larsen PF, Liu Y, Loeschcke V. Combined expression patterns of QTL-linked candidate genes best predict thermotolerance in Drosophila melanogaster. Journal of Insect Physiology, (2009); 55(11): 1050–1057.

Figueira FH, de Quadros Oliveira N, de Aguiar LM, Escarrone AL, Primel EG, Barros DM, da Rosa CE. Exposure to atrazine alters behaviour and disrupts the dopaminergic

system in Drosophila melanogaster. Comparative Biochemistry and Physiology Part C: Toxicology & Pharmacology, (2017); 202: 94–102.

Inagaki HK, Ben-Tabou de-Leon S, Wong AM, Jagadish S, Ishimoto H, Barnea G, Kitamoto T, Axel R, Anderson DJ. Visualizing neuromodulation in vivo: TANGO-mapping of dopamine signaling reveals appetite control of sugar sensing. Cell, (2012); 148(3): 583–595.

Ellman GL, Courtney KD, Andres V, Feather-Stone RM. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochemical pharmacology, (1961); 7: 88–95.

Anjum SI, Yousf MJ, Ayaz S, Siddiqui BS. Toxicological evaluation of chlorpyrifos and Neem extract (Biosal B) against 3RD instars larvae of Drosophila melanogaster. Journal of Animal & Plant Sciences, (2010); 20(1): 9–12.

Pérez JJ, Williams MK, Weerasekera G, Smith K, Whyatt RM, Needham LL, Barr DB. Measurement of pyrethroid, organophosphorus, and carbamate insecticides in human plasma using isotope dilution gas chromatography-high resolution mass spectrometry. Journal of chromatography. B, Analytical technologies in the biomedical and life sciences, (2010); 878(27): 2554–2562.

Mani VM, Gokulakrishnan A, Sadiq AM. Molecular Mechanism of Neu-rodevelopmental Toxicity Risks of Occupational Exposure of Pyrethroid Pesticide with Reference to Deltamethrin-A Critical Review. BAOJ Pathology, (2017); 1(008).

Wolansky MJ, Harrill JA. Neurobehavioral toxicology of pyrethroid insecticides in adult animals: A critical review. Neurotoxicology and Teratology, (2008); 30(2): 55–78.

Lazarini CA, Florio JC, Lemonica IP, Bernardi MM. Effects of prenatal exposure to deltamethrin on forced swimming behavior, motor activity, and striatal dopamine levels in male and female rats. Neurotoxicology and Teratology, (2001); 23(6): 665–673.

Johri A, Yadav S, Singh RL, Dhawan A, Ali M, Parmar D. Long lasting effects of prenatal exposure to deltamethrin on cerebral and hepatic cytochrome P450s and behavioral activity in rat offspring. European Journal of Pharmacology, (2006); 544(1–3): 58–68.

Richardson JR, Taylor MM, Shalat SL, Guillot 3rd TS, Caudle WM, Hossain MM, Mathews TA, Jones SR, Cory-Slechta DA, Miller GW. Developmental pesticide exposure reproduces features of attention deficit hyperactivity disorder. FASEB journal : official publication of the Federation of American Societies for Experimental Biology, (2015); 29(5): 1960–1972.

Kung TS, Richardson JR, Cooper KR, White LA. Developmental Deltamethrin Exposure Causes Persistent Changes in Dopaminergic Gene Expression, Neurochemistry, and Locomotor Activity in Zebrafish. Toxicological sciences : an official journal of the Society of Toxicology, (2015); 146(2): 235–243.

Liu G, Ma Q, Shi N. Tyrosine hydroxylase as a target for deltamethrin in the nigrostriatal dopaminergic pathway. Biomedical and Environmental Sciences, (2006); 19(1): 27.

Tayebati SK, Di Tullio MA, Ricci A, Amenta F. Influence of dermal exposure to the pyrethroid insecticide deltamethrin on rat brain microanatomy and cholinergic/dopaminergic neurochemistry. Brain Research, (2009); 1301: 180–188.

Robertson HA. Synergistic Interactions of D1- and D2-Selective Dopamine Agonists in Animal Models for Parkinson’s Disease: Sites of Action and Implications for the Pathogenesis of Dyskinesias. Canadian Journal of Neurological Sciences / Journal Canadien des Sciences Neurologiques, (1992); 19(S1): 147–152.

Hasbi A, O’Dowd BF, George SR. Dopamine D1-D2 receptor heteromer signaling pathway in the brain: emerging physiological relevance. Molecular brain, (2011); 4: 26.

Khan AM, Raina R, Dubey N, Verma PK. Effect of deltamethrin and fluoride co-exposure on the brain antioxidant status and cholinesterase activity in Wistar rats. Drug and Chemical Toxicology, (2017); 41(2): 123–127.

Soderlund DM, Clark JM, Sheets LP, Mullin LS, Piccirillo VJ, Sargent D, Stevens JT, Weiner ML. Mechanisms of pyrethroid neurotoxicity: implications for cumulative risk assessment. Toxicology, (2002); 171(1): 3–59.

Adefegha SA, Omojokun OS, Oboh G, Fasakin O, Ogunsuyi O. Modulatory Effects of Ferulic Acid on Cadmium-Induced Brain Damage. Journal of Evidence-Based Complementary & Alternative Medicine, (2016); 21(4): NP56–NP61.

Abdel-Daim MM, Abushouk AI, Bungău SG, Bin-Jumah M, El-kott AF, Shati AA, Aleya L, Alkahtani S. Protective effects of thymoquinone and diallyl sulphide against malathion-induced toxicity in rats. Environmental Science and Pollution Research, (2020); 27(10): 10228–10235.




DOI: http://dx.doi.org/10.62940/als.v10i2.1799

Refbacks

  • There are currently no refbacks.