618
Views
58
CrossRef citations to date
0
Altmetric
Original Research

Selenium Nanoparticles Pre-Treatment Reverse Behavioral, Oxidative Damage, Neuronal Loss and Neurochemical Alterations in Pentylenetetrazole-Induced Epileptic Seizures in Mice

, , , , , , ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 6339-6353 | Published online: 24 Aug 2020

References

  • Engelborghs S, D’Hooge R, Paul D. Pathophysiology of epilepsy. Acta neurologicaBelgica. 2001;100:201–213.
  • Jennum P, Sabers A, Christensen J, Ibsen R, Kjellberg J. Socioeconomic outcome of epilepsy surgery: a controlled national study. Seizure. 2016;42:52–56. doi:10.1016/j.seizure.2016.09.01627770730
  • Liu S, Yu W, Lu Y. The causes of new-onset epilepsy and seizures in the elderly. Neuropsychiatr Dis Treat. 2016;12:1425–1434. doi:10.2147/NDT.S10790527382285
  • Mao XY, Zhou HH, Jin WL. Redox-related neuronal death and crosstalk as drug targets: focus on epilepsy. Front Neurosci. 2019;13:512. doi:10.3389/fnins.2019.0051231191222
  • da Fonseca DV, da Silva Maia Bezerra Filho C, Lima TC, de Almeida RN, de Sousa DP. Anticonvulsant essential oils and their relationship with oxidative stress in epilepsy. Biomolecules. 2019;9:12. doi:10.3390/biom9120835
  • Ryan K, Liang LP, Rivard C, Patel M. Temporal and spatial increase of reactive nitrogen species in the kainate model of temporal lobe epilepsy. Neurobiol Dis. 2014;64:8–15. doi:10.1016/j.nbd.2013.12.00624361554
  • Attia GM, Elmansy RA, Elsaed WM. Neuroprotective effect of nilotinib on pentylenetetrazol-induced epilepsy in adult rat hippocampus: involvement of oxidative stress, autophagy, inflammation, and apoptosis. Folia Neuropathologica. 2019;57(2):146–160. doi:10.5114/fn.2019.8442331556574
  • Hashemian M, Anissian D, Ghasemi-Kasman M, et al. Curcumin-loaded chitosan-alginate-STPP nanoparticles ameliorate memory deficits and reduce glial activation in pentylenetetrazol-induced kindling model of epilepsy. Prog Neuropsychopharmacol Biol Psychiatry. 2017;79(Pt B):462–471. doi:10.1016/j.pnpbp.2017.07.02528778407
  • Arafa NM, Abdel-Rahman M, El-khadragy MF, Kassab RB. Evaluation of the possible epileptogenic activity of ciprofloxacin: the role of Nigella sativa on amino acids neurotransmitters. Neurochem Res. 2013;38(1):174–185. doi:10.1007/s11064-012-0905-z23086472
  • Abdel-Rahman M, Arafa N, El-Khadragy M, Kassab R. The neuroprotective role of Nigella sativa extract on ciprofloxacin and pentylenetetrazole treated rats. Afr J Pharm Pharmacol. 2013;7:660–1670. doi:10.5897/AJPP12.897
  • de Kinderen RJ, Evers SM, Rinkens R, et al. Side-effects of antiepileptic drugs: the economic burden. Seizure. 2014;23(3):184–190. doi:10.1016/j.seizure.2013.11.00924315662
  • Gupta J, Fatima MT, Islam Z, Khan RH, Uversky VN, Salahuddin P. Nanoparticle formulations in the diagnosis and therapy of Alzheimer’s disease. Int J Biol Macromol. 2019;130:515–526. doi:10.1016/j.ijbiomac.2019.02.15630826404
  • Anissian D, Ghasemi-Kasman M, Khalili-Fomeshi M, et al. Piperine-loaded chitosan-STPP nanoparticles reduce neuronal loss and astrocytes activation in chemical kindling model of epilepsy. Int J Biol Macromol. 2018;107(Pt A):973–983. doi:10.1016/j.ijbiomac.2017.09.07328939512
  • Rawat M, Yadukrishnan P, Kumar N. Mechanisms of action of nanoparticles in living systems In: Microbial Biotechnology in Environmental Monitoring and Cleanup. IGI Global; 2018:220–236.
  • Loeschner K, Hadrup N, Hansen M, et al. Absorption, distribution, metabolism and excretion of selenium following oral administration of elemental selenium nanoparticles or selenite in rats. Metallomics. 2014;6(2):330–337. doi:10.1039/c3mt00309d24413471
  • Alajmi RA, Al-Megrin WA, Metwally D, et al. Anti-Toxoplasma activity of silver nanoparticles green synthesized with Phoenix dactylifera and Ziziphus spina-christi extracts which inhibits inflammation through liver regulation of cytokines in Balb/c mice. Biosci Rep. 2019;39(5):5. doi:10.1042/BSR20190379
  • Huo X, Zhang Y, Jin X, Li Y, Zhang L. A novel synthesis of selenium nanoparticles encapsulated PLGA nanospheres with curcumin molecules for the inhibition of amyloid beta aggregation in Alzheimer’s disease. J Photochem Photobiol B. 2019;190:98–102. doi:10.1016/j.jphotobiol.2018.11.00830504054
  • Al-Quraishy S, Dkhil MA, Abdel Moneim AE. Anti-hyperglycemic activity of selenium nanoparticles in streptozotocin-induced diabetic rats. Int J Nanomedicine. 2015;10:6741–6756. doi:10.2147/IJN.S9137726604749
  • Pillai R, Uyehara-Lock JH, Bellinger FP. Selenium and selenoprotein function in brain disorders. IUBMB Life. 2014;66(4):229–239. doi:10.1002/iub.126224668686
  • Amani H, Habibey R, Shokri F, et al. Selenium nanoparticles for targeted stroke therapy through modulation of inflammatory and metabolic signaling. Sci Rep. 2019;9(1):6044. doi:10.1038/s41598-019-42633-930988361
  • Wadhwani SA, Shedbalkar UU, Singh R, Chopade BA. Biogenic selenium nanoparticles: current status and future prospects. Appl Microbiol Biotechnol. 2016;100(6):2555–2566. doi:10.1007/s00253-016-7300-726801915
  • Zhang J, Zhou X, Yu Q, et al. Epigallocatechin-3-gallate (EGCG)-stabilized selenium nanoparticles coated with Tet-1 peptide to reduce amyloid-β aggregation and cytotoxicity. ACS Appl Mater Interfaces. 2014;6(11):8475–8487. doi:10.1021/am501341u24758520
  • Abdelfattah MS, Badr SEA, Lotfy SA, et al. Rutin and selenium co-administration reverse 3-nitropropionic acid-induced neurochemical and molecular impairments in a mouse model of Huntington’s disease. Neurotox Res. 2019.
  • El-Missiry MA, Othman AI, Amer MA, Sedki M, Ali SM, El-Sherbiny IM. Nanoformulated ellagic acid ameliorates pentylenetetrazol-induced experimental epileptic seizures by modulating oxidative stress, inflammatory cytokines and apoptosis in the brains of male mice. Metab Brain Dis. 2019.
  • Dkhil MA, Zrieq R, Al-Quraishy S, Abdel Moneim AE. Selenium nanoparticles attenuate oxidative stress and testicular damage in streptozotocin-induced diabetic rats. Molecules. 2016;21:11. doi:10.3390/molecules21111517
  • Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem. 1951;193(1):265–275.14907713
  • Kim YS, Kim SH, Shin J, et al. Luteolin suppresses cancer cell proliferation by targeting vaccinia-related kinase 1. PLoS One. 2014;9(10):e109655. doi:10.1371/journal.pone.010965525310002
  • Shi F, Zhao P, Li X, Pan H, Ma S, Ding L. Cytotoxicity of luteolin in primary rat hepatocytes: the role of CYP3A-mediated ortho-benzoquinone metabolite formation and glutathione depletion. J Appl Toxicol. 2015;35(11):1372–1380. doi:10.1002/jat.310625612170
  • Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem. 1979;95(2):351–358. doi:10.1016/0003-2697(79)90738-336810
  • Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR. Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal Biochem. 1982;126(1):131–138. doi:10.1016/0003-2697(82)90118-X7181105
  • Ellman GL. Tissue sulfhydryl groups. Arch BiochemBiophys. 1959;82(1):70–77. doi:10.1016/0003-9861(59)90090-6
  • Factor VM, Kiss A, Woitach JT, Wirth PJ, Thorgeirsson SS. Disruption of redox homeostasis in the transforming growth factor-alpha/c-myc transgenic mouse model of accelerated hepatocarcinogenesis. J Biol Chem. 1998;273(25):15846–15853. doi:10.1074/jbc.273.25.158469624185
  • Paglia DE, Valentine WN. Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase. J Lab Clin Med. 1967;70(1):158–169.6066618
  • Aebi H. Catalase in vitro. Methods Enzymol. 1984;105:121–126.6727660
  • Misra HP, Fridovich I. The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. J Biol Chem. 1972;247(10):3170–3175.4623845
  • Abdel Moneim AE. Indigofera oblongifolia prevents lead acetate-induced hepatotoxicity, oxidative stress, fibrosis and apoptosis in rats. PLoS One. 2016;11(7):e0158965. doi:10.1371/journal.pone.015896527391413
  • Ellman GL, Courtney KD, Andres V Jr., Feather-Stone RM. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol. 1961;7:88–95. doi:10.1016/0006-2952(61)90145-913726518
  • Pagel P, Blome J, Wolf HU. High-performance liquid chromatographic separation and measurement of various biogenic compounds possibly involved in the pathomechanism of Parkinson’s disease. J Chromatogr B Biomed Sci Appl. 2000;746(2):297–304. doi:10.1016/S0378-4347(00)00348-011076082
  • Heinrikson RL, Meredith SC. Amino acid analysis by reverse-phase high-performance liquid chromatography: precolumn derivatization with phenylisothiocyanate. Anal Biochem. 1984;136(1):65–74. doi:10.1016/0003-2697(84)90307-56711815
  • Almeer RS, AlBasher GI, Alarifi S, Alkahtani S, Ali D, Abdel Moneim AE. Royal jelly attenuates cadmium-induced nephrotoxicity in male mice. Sci Rep. 2019;9(1):5825. doi:10.1038/s41598-019-42368-730967588
  • Al-Quraishy S, Dkhil MA, Abdel-Gaber R, Zrieq R, Hafez TA, Mubaraki MA, Abdel Moneim AE. Myristica fragrans seed extract reverses scopolamine-induced cortical injury via stimulation of HO-1 expression in male rats. Environ Sci Pollut Res. 2020;27(11):12395–12404. doi:10.1007/s11356-020-07686-8
  • Hasan A, Morshed M, Memic A, Hassan S, Webster TJ, Marei HE. Nanoparticles in tissue engineering: applications, challenges and prospects. Int J Nanomedicine 2018;13:5637-5655. doi:10.2147/IJN.S153758
  • Samokhina E, Samokhin A. Neuropathological profile of the pentylenetetrazol (PTZ) kindling model. Int J Neurosci. 2018;128(11):1086–1096. doi:10.1080/00207454.2018.148106429792126
  • Al Omairi NE, Radwan OK, Alzahrani YA, Kassab RB. Neuroprotective efficiency of Mangiferaindica leaves extract on cadmium-induced cortical damage in rats. Metab Brain Dis. 2018. doi:10.1007/s11011-018-0222-6
  • Zhu X, Dong J, Han B, et al. Neuronal nitric oxide synthase contributes to PTZ kindling epilepsy-induced hippocampal endoplasmic reticulum stress and oxidative damage. Front Cell Neurosci. 2017;11:377. doi:10.3389/fncel.2017.0037729234274
  • Zsurka G, Kunz W. Mitochondrial dysfunction and seizures: the neuronal energy crisis. Lancet Neurol. 2015;14:956–966. doi:10.1016/S1474-4422(15)00148-926293567
  • Uchida K, Shiraishi M, Naito Y, Torii Y, Nakamura Y, Osawa T. Activation of stress signaling pathways by the end product of lipid peroxidation. 4-hydroxy-2-nonenal is a potential inducer of intracellular peroxide production. J Biol Chem. 1999;274(4):2234–2242. doi:10.1074/jbc.274.4.22349890986
  • Kassab RB, Lokman MS, Essawy EA. Neurochemical alterations following the exposure to di-n-butyl phthalate in rats. Metab Brain Dis. 2019;34(1):235–244. doi:10.1007/s11011-018-0341-030446882
  • Ikwegbue PC, Masamba P, Oyinloye BE, Kappo AP. Roles of heat shock proteins in apoptosis, oxidative stress, human inflammatory diseases, and cancer. Pharmaceuticals. 2018;11(1):2. doi:10.3390/ph11010002
  • Hussein AM, Adel M, El-Mesery M, Abbas KM, Ali AN, Abulseoud OA. L-carnitine modulates epileptic seizures in pentylenetetrazole-kindled rats via suppression of apoptosis and autophagy and upregulation of hsp70. Brain Sci. 2018;8(3):45. doi:10.3390/brainsci8030045
  • Dericioglu N, Soylemezoglu F, Gursoy-Ozdemir Y, Akalan N, Saygi S, Dalkara T. Cell death and survival mechanisms are concomitantly active in the hippocampus of patients with mesial temporal sclerosis. Neuroscience. 2013;237:56–65. doi:10.1016/j.neuroscience.2013.01.05023384610
  • Banach M, Piskorska B, Czuczwar SJ, Borowicz KK. Nitric oxide, epileptic seizures, and action of antiepileptic drugs. CNS Neurol Disord Drug Targets. 2011;10(7):808–819. doi:10.2174/18715271179807234721999730
  • Mueller SG, Trabesinger AH, Boesiger P, Wieser HG. Brain glutathione levels in patients with epilepsy measured by in vivo (1)H-MRS. Neurology. 2001;57(8):1422–1427. doi:10.1212/WNL.57.8.142211673583
  • Lee SH, Choi BY, Kho AR, et al. Protective effects of protocatechuic acid on seizure-induced neuronal death. Int J Mol Sci. 2018;19:1.
  • Ahmed HH, Abd El-Maksoud MD, Abdel Moneim AE, Aglan HA. Pre-clinical study for the antidiabetic potential of selenium nanoparticles. Biol Trace Elem Res. 2016.
  • Shalby AB, El-Maksoud MDA, Moneim AEA, Ahmed HH. Antifibrotic candidates of Selenium nanoparticles and selenium in the experimental model. J Appl Pharm Sci. 2017;7(09):191–198.
  • Naziroglu M, Muhamad S, Pecze L. Nanoparticles as potential clinical therapeutic agents in Alzheimer’s disease: focus on selenium nanoparticles. Expert Rev Clin Pharmacol. 2017;10(7):773–782. doi:10.1080/17512433.2017.132478128463572
  • AlBasher G, Alfarraj S, Alarifi S, et al. Nephroprotective role of selenium nanoparticles against glycerol-induced acute kidney injury in rats. Biol Trace Elem Res. 2019.
  • Cao C, Li X, Qin L, et al. High selenium yeast mitigates aluminum-induced cerebral inflammation by increasing oxidative stress and blocking NO production. Biometals. 2018;31(5):835–843. doi:10.1007/s10534-018-0128-030014352
  • Tawfik KM, Moustafa YM, El-Azab MF. Neuroprotective mechanisms of sildenafil and selenium in PTZ-kindling model: implications in epilepsy. Eur J Pharmacol. 2018;833:131–144. doi:10.1016/j.ejphar.2018.05.03529807031
  • Malyar RM, Li H, Liu D, et al. Selenium/Zinc-enriched probiotics improve serum enzyme activity, antioxidant ability, inflammatory factors and related gene expression of Wistar rats inflated under heat stress. Life Sci. 2020;248:117464. doi:10.1016/j.lfs.2020.11746432097667
  • Huang WY, Lin S, Chen HY, et al. NADPH oxidases as potential pharmacological targets against increased seizure susceptibility after systemic inflammation. J Neuroinflammation. 2018;15(1):140. doi:10.1186/s12974-018-1186-529753328
  • Demirci K, Naziroglu M, Ovey IS, Balaban H. Selenium attenuates apoptosis, inflammation and oxidative stress in the blood and brain of aged rats with scopolamine-induced dementia. Metab Brain Dis. 2017;32(2):321–329. doi:10.1007/s11011-016-9903-127631101
  • Singh N, Saha L, Kumari P, et al. Effect of dimethyl fumarate on neuroinflammation and apoptosis in pentylenetetrazol kindling model in rats. Brain Res Bull. 2019;144:233–245. doi:10.1016/j.brainresbull.2018.11.01330472152
  • Yousef S, Omar A, Fahad A, et al. The neuroprotective role of coenzyme Q10 against lead acetate-induced neurotoxicity is mediated by antioxidant, anti-inflammatory and anti-apoptotic activities. Int J Environ Res Public Health. 2019;16(16):2895. doi:10.3390/ijerph16162895
  • Long M, Yang S, Wang Y, et al. The protective effect of selenium on chronic zearalenone-induced reproductive system damage in male mice. Molecules. 2016;21:12. doi:10.3390/molecules21121687
  • Pauletti A, Terrone G, Shekh-Ahmad T, et al. Targeting oxidative stress improves disease outcomes in a rat model of acquired epilepsy. Brain. 2019;142(7):e39. doi:10.1093/brain/awz13031145451
  • de Sales Santos IM, Feitosa CM, de Freitas RM. Pilocarpine-induced seizures produce alterations on choline acetyltransferase and acetylcholinesterase activities and deficit memory in rats. Cell Mol Neurobiol. 2010;30(4):569–575. doi:10.1007/s10571-009-9481-419941057
  • Kurian MA, Gissen P, Smith M, Heales S Jr., Clayton PT. The monoamine neurotransmitter disorders: an expanding range of neurological syndromes. Lancet Neurol. 2011;10(8):721–733. doi:10.1016/S1474-4422(11)70141-721777827
  • Statnick MA, Dailey JW, Jobe PC, Browning RA. Abnormalities in brain serotonin concentration, high-affinity uptake, and tryptophan hydroxylase activity in severe-seizure genetically epilepsy-prone rats. Epilepsia. 1996;37(4):311–321. doi:10.1111/j.1528-1157.1996.tb00565.x8603634
  • Tchekalarova J, Pechlivanova D, Atanasova T, Markova P, Lozanov V, Stoynev A. Diurnal variations in depression-like behavior of Wistar and spontaneously hypertensive rats in the kainate model of temporal lobe epilepsy. Epilepsy Behav. 2011;20(2):277–285. doi:10.1016/j.yebeh.2010.12.02121277833
  • Rezaei M, Sadeghian A, Roohi N, Shojaei A, Mirnajafi-Zadeh J. Epilepsy and dopaminergic system. Physiol Pharmacol. 2017;21:1–14.
  • Briere R, Sherwin AL, Robitaille Y, Olivier A, Quesney LF, Reader TA. Alpha-1 adrenoceptors are decreased in human epileptic foci. Ann Neurol. 1986;19(1):26–30. doi:10.1002/ana.4101901063004318
  • Giorgi F, Pizzanelli C, Biagioni F, Murri L, Fornai F. The role of norepinephrine in epilepsy: from the bench to the bedside. Neurosci Biobehav Rev. 2004;28:507–524. doi:10.1016/j.neubiorev.2004.06.00815465138
  • Cavalcante TMB, De Melo JMAJ, Lopes LB, et al. Ivabradine possesses anticonvulsant and neuroprotective action in mice. Biomed Pharmacother/Biomedecine&pharmacotherapie. 2019;109:2499–2512. doi:10.1016/j.biopha.2018.11.09630551511
  • Barker-Haliski M, White HS. Glutamatergic mechanisms associated with seizures and epilepsy. Cold Spring Harb Perspect Med. 2015;5(8):a022863. doi:10.1101/cshperspect.a02286326101204
  • Al Kahtani MA. Effect of both selenium and biosynthesized nanoselenium particles on cadmium-induced neurotoxicity in albino rats. Hum Exp Toxicol. 2019;960327119880589.
  • Ansari MA, Ahmad AS, Ahmad M, et al. Selenium protects cerebral ischemia in rat brain mitochondria. Biol Trace Elem Res. 2004;101(1):73–86. doi:10.1385/BTER:101:1:7315516704
  • Abozaid O, El-sonbaty S, Ezz W. Effect of Acrylamide on neurotransmitters and acetyl-cholinesterase activity in the brain of rats: therapeutic effect of ferulic acid and selenium nanoparticles. 2017.
  • Tang Y-L, Wang S-W, Lin S-M. Both inorganic and organic selenium supplements can decrease brain monoamine oxidase B enzyme activity in adult rats. Br J Nutri. 2008;100(3):660–665. doi:10.1017/S0007114508911594
  • Zafar KS, Siddiqui A, Sayeed I, Ahmad M, Salim S, Islam F. Dose-dependent protective effect of selenium in rat model of Parkinson’s disease: neurobehavioral and neurochemical evidences. J Neurochem. 2001;57(8):438–446. doi:10.1046/j.1471-4159.2003.01531.x
  • Solovyev ND. Importance of selenium and selenoprotein for brain function: from antioxidant protection to neuronal signalling. J Inorg Biochem. 2015;153:1–12. doi:10.1016/j.jinorgbio.2015.09.00326398431
  • Savaskan NE, Brauer AU, Kuhbacher M, et al. Selenium deficiency increases susceptibility to glutamate-induced excitotoxicity. FASEB J. 2003;17(1):112–114. doi:10.1096/fj.02-0067fje12424220
  • Ma YM, Ibeanu G, Wang LY, et al. Selenium suppresses glutamate-induced cell death and prevents mitochondrial morphological dynamic alterations in hippocampal HT22 neuronal cells. BMC Neurosci. 2017;18(1):15. doi:10.1186/s12868-017-0337-428103798