1,139
Views
67
CrossRef citations to date
0
Altmetric
Review

Nanoparticles as potential clinical therapeutic agents in Alzheimer’s disease: focus on selenium nanoparticles

, &
Pages 773-782 | Received 05 Oct 2016, Accepted 26 Apr 2017, Published online: 16 May 2017

References

  • Song B, Zhang Y, Liu J, et al. Is neurotoxicity of metallic nanoparticles the cascades of oxidative stress? Nanoscale Res Lett. 2016;11:291.
  • Parveen A, Sh R, Sushma, et al. Intranasal exposure to silica nanoparticles induce alterations in pro-inflammatory environment of rat brain, involvement of oxidative stress. Toxicol Ind Health. 2017;33(2):119–132.
  • Zhang R, Niu YJ, Li YW, et al. Acute toxicity study of the interaction between titanium dioxide nanoparticles and lead acetate in mice. Environ Toxicol Pharmacol. 2010;30:52–60.
  • Shinohara N, Oshima Y, Kobayashi T, et al. Pulmonary clearance kinetics and extrapulmonary translocation of seven titanium dioxide nano- and submicron materials following intratracheal administration in rats. Nanotoxicology. 2015;9(8):1050–1058.
  • Hritcu L, Stefan M, Ursu L, et al. Exposure to silver nanoparticles induces oxidative stress and memory deficits in laboratory rats. Cen Euro J Biol. 2011;6(4):497–509.
  • Feng XL, Chen AJ, Zhang YL, et al. Central nervous system toxicity of metallic nanoparticles. Int J Nanomed. 2015;10:4321–4340.
  • Wang JY, Rahman MF, Duhart HM, et al. Expression changes of dopaminergic system-related genes in PC12 cells induced by manganese, silver, or copper nanoparticles. Neurotoxicology. 2009;30(6):926–933.
  • Rahman MF, Wang J, Patterson TA, et al. Expression of genes related to oxidative stress in the mouse brain after exposure to silver-25 nanoparticles. Toxicol Lett. 2009;187:15–21.
  • Jeon YM, Park SK, Lee MY. Toxicoproteomic identification of TiO2 nanoparticle-induced protein expression changes in mouse brain. Anim Cells Syst. 2011;15(2):107–114.
  • Vedagiri A, Thangarajan S. Mitigating effect of chrysin loaded solid lipid nanoparticles against amyloid β25-35 induced oxidative stress in rat hippocampal region, an efficient formulation approach for Alzheimer’s disease. Neuropeptides. 2016;58:111–125.
  • Halliwell B. Oxidative stress and neurodegeneration, where are we now? J Neurochem. 2006;97(6):1634–1658.
  • Nazıroğlu M. Role of selenium on calcium signaling and oxidative stress-induced molecular pathways in epilepsy. Neurochem Res. 2009;34:2181–2191.
  • Nogueira CW, Rocha JB. Toxicology and pharmacology of selenium, emphasis on synthetic organoselenium compounds. Arch Toxicol. 2011;85(11):1313–1359.
  • Weekley CM, Harris HH. Which form is that? The importance of selenium speciation and metabolism in the prevention and treatment of disease. Chem Soc Rev. 2013;42:8870–8894.
  • Nazıroğlu M. TRPM2 cation channels, oxidative stress and neurological diseases, where are we now? Neurochem Res. 2011;36:355–366.
  • Nazıroğlu M, Demirdaş A. Psychiatric disorders and TRP channels, focus on psychotropic drugs. Curr Neuropharmacol. 2015;13(2):248–257.
  • Ceballos-Picot I, Merad-Boudia M, Nicole A, et al. Peripheral antioxidant enzyme activities and selenium in elderly subjects and in dementia of Alzheimer’s type – place of the extracellular glutathione peroxidase. Free Radic Biol Med. 1996;20(4):579–587.
  • Vural H, Demirin H, Kara Y, et al. Alterations of plasma magnesium, copper, zinc, iron and selenium concentrations and some related erythrocyte antioxidant enzyme activities in patients with Alzheimer’s disease. J Trace Elem Med Biol. 2010;24(3):169–173.
  • Rita Cardoso B, Silva Bandeira V, Jacob-Filho W, et al. Selenium status in elderly: relation to cognitive decline. J Trace Elem Med Biol. 2014;28(4):422–426.
  • Koç ER, Ilhan A, Aytürk Z, et al. A comparison of hair and serum trace elements in patients with Alzheimer disease and healthy participants. Turk J Med Sci. 2015;45:1034–1039.
  • Kosik KS. Alzheimer’s disease, a cell biological perspective. Science. 1992;256:780–783.
  • Balaban H, Nazıroğlu M, Demirci K, et al. The protective role of selenium on scopolamine-induced memory impairment, oxidative stress, and apoptosis in aged rats, the involvement of TRPM2 and TRPV1 channels. Mol Neurobiol. 2017;54:2852–2868.
  • Schweizer U, Bräuer AU, Köhrle J, et al. Selenium and brain function, a poorly recognized liaison. Brain Res Brain Res Rev. 2004;45:164–178.
  • Zhang J, Gao X, Zhang L, et al. Biological effects of a nano red elemental selenium. BioFactors. 2001;15:27–38.
  • Fernandes AP, Gandin V. Selenium compounds as therapeutic agents in cancer. Biochim Biophys Acta. 2015;1850(8):1642–1660.
  • Busquets MA, Sabaté R, Estelrich J. Potential applications of magnetic particles to detect and treat Alzheimer’s disease. Nanoscale Res Lett. 2014;9(1):538.
  • Gupta S, Babu P, Surolia A. Biphenyl ethers conjugated CdSe/ZnS core/shell quantum dots and interpretation of the mechanism of amyloid fibril disruption. Biomaterials. 2010;31(26):6809–6822.
  • 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. Appl Mater Interfaces. 2014;6:8475–8487.
  • Yin T, Yang L, Liu Y, et al. Sialic acid (SA)-modified selenium nanoparticles coated with a high blood-brain barrier permeability peptide-B6 peptide for potential use in Alzheimer’s disease. Acta Biomater. 2015;25:172–183.
  • Wang Z, Wang Y, Li W, et al. Design, synthesis, and evaluation of multitarget-directed selenium-containing clioquinol derivatives for the treatment of Alzheimer’s disease. ACS Chem Neurosci. 2014;5:952–962.
  • Kahya MC, Nazıroğlu M, Çiğ B. Selenium reduces mobile phone (900 MHz)-induced oxidative stress, mitochondrial function, and apoptosis in breast cancer cells. Biol Trace Elem Res. 2014;160:285–293.
  • Heusinkveld HJ, Wahle T, Campbell A, et al. Neurodegenerative and neurological disorders by small inhaled particles. Neurotoxicology. 2016;56:94–106.
  • Nazıroğlu M. New molecular mechanisms on the activation of TRPM2 channels by oxidative stress and ADP-ribose. Neurochem Res. 2007;32:1990–2001.
  • Bush AI, Pettingell WH, Multhaup G, et al. Rapid induction of Alzheimer A beta amyloid formation by zinc. Science. 1994;265:1464–1467.
  • Yamin G, Ono K, Inayathullah M, et al. Amyloid-protein assembly as a therapeutic target of Alzheimer’s disease. Curr Pharm Des. 2008;14:3231–3246.
  • Ajith TA, Padmajanair G. Mitochondrial pharmaceutics, a new therapeutic strategy to ameliorate oxidative stress in Alzheimer’s disease. Curr Aging Sci. 2015;8:235–240.
  • Tran MH, Yamada K, Olariu A, et al. Amyloid beta peptide induces nitric oxide production in rat hippocampus, association with cholinergic dysfunction and amelioration by inducible nitric oxide synthase inhibitors. Faseb J. 2001;15:1407–1409.
  • Choi J, Malakowsky CA, Talent JM, et al. Anti-apoptotic proteins are oxidized by Abeta25-35 in Alzheimer’s fibroblasts. Biochim Biophys Acta. 2003;1637:135–141.
  • Zhao QF, Yu JT, Tan L. S-nitrosylation in Alzheimer’s disease. Mol Neurobiol. 2015;51:268–280.
  • Durazo SA, Kompella UB. Functionalized nanosystems for targeted mitochondrial delivery. Mitochondrion. 2012;12:190–201.
  • Bonda DJ, Wang X, Perry G, et al. Oxidative stress in Alzheimer disease, a possibility for prevention. Neuropharmacology. 2010;59:290–294.
  • Jazvinšćak JM, Hof PR, Šimić G. Ceramides in Alzheimer’s disease, key mediators of neuronal apoptosis induced by oxidative stress and Aβ accumulation. Oxid Med Cell Longev. 2015;2015:346783.
  • Hayes JD, Flanagan JU, Jowsey IR. Glutathione transferases. Annu Rev Pharmacol Toxicol. 2005;45:51–88.
  • Loef M, Schrauzer GN, Selenium WH. Alzheimer’s disease, a systematic review. J Alzheimers Dis. 2011;26:81–104.
  • González-Domínguez R, García-Barrera T, Gómez-Ariza JL. Homeostasis of metals in the progression of Alzheimer’s disease. Biometals. 2014;27:539–549.
  • Ishrat T, Parveen K, Khan MM, et al. Selenium prevents cognitive decline and oxidative damage in rat model of streptozotocin-induced experimental dementia of Alzheimer’s type. Brain Res. 2009;1281:117–127.
  • Krishnan S, Rani P. Evaluation of selenium, redox status and their association with plasma amyloid/tau in Alzheimer’s disease. Biol Trace Elem Res. 2014;158:158–165.
  • Kumar VS, Gopalakrishnan A, Nazıroğlu M, et al. Calcium ion – the key player in cerebral ischemia. Curr Med Chem. 2014;21:2065–2075.
  • Yürüker V, Nazıroğlu M, Şenol N. Reduction in traumatic brain injury-induced oxidative stress, apoptosis, and calcium entry in rat hippocampus by melatonin, possible involvement of TRPM2 channels. Metab Brain Dis. 2015;30:223–231.
  • Ashraf GM, Tabrez S, Jabir NR, et al. An overview on global trends in nanotechnological approaches for Alzheimer therapy. Curr Drug Metab. 2015;16(8):719–727.
  • Soursou G, Alexiou A, Ashraf GM, et al. Applications of nanotechnology in diagnostics and therapeutics of Alzheimer’s and Parkinson’s disease. Curr Drug Metab. 2015;16(8):705–712.
  • Kamal MA. Global trends in nanotechnological approaches for various health issues – volume II. Curr Drug Metab. 2015;16(8):598–601.
  • Ali A, Sheikh IA, Mirza Z, et al. Application of proteomic tools in modern nanotechnological approaches towards effective management of neurodegenerative disorders. Curr Drug Metab. 2015;16(5):376–388.
  • Ahmad MZ, Ahmad J, Amin S, et al. Role of nanomedicines in delivery of anti-acetylcholinesterase compounds to the brain in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2014;13(8):1315–1324.
  • Alam Q, ZubairAlam M, Karim S, et al. A nanotechnological approach to the management of Alzheimer disease and type 2 diabetes. CNS Neurol Disord Drug Targets. 2014;13(3):478–486.
  • Bao P, Chen Z, Tai RZ, et al. Selenite-induced toxicity in cancer cells is mediated by metabolic generation of endogenous selenium nanoparticles. J Proteome Res. 2015;14:1127–1136.
  • Carrasco C, Rodríguez AB, Pariente JA. Melatonin as a stabilizer of mitochondrial function, role in diseases and aging. Turk J Biol. 2015;39:822–831.
  • Rayman MP. The importance of selenium to human health. Lancet. 2000;356:233–241.
  • Suzuki KT, Kurasaki K, Suzuki N. Selenocysteine beta-lyase and methylselenol demethylase in the metabolism of Se-methylated selenocompounds into selenide. Biochim Biophys Acta. 2007;1770:1053–1061.
  • Weekley CM, Aitken JB, Vogt S, et al. Metabolism of selenite in human lung cancer cells, X-ray absorption and fluorescence studies. J Am Chem Soc. 2011;133:18272–18279.
  • Verma A, Stellacci F. Effect of surface properties on nanoparticle-cell interactions. Small. 2010;6:12–21.
  • Huang B, Zhang J, Hou J, et al. Free radical scavenging efficiency of nano-Se in vitro. Free Radic Biol Med. 2003;35:805–813.
  • Zhang J, Wang H, Bao Y, et al. Nano red elemental selenium has no size effect in the induction of seleno-enzymes in both cultured cells and mice. Life Sci. 2004;75:237–244.
  • Qi L, Xu Z, Chen M. In vitro and in vivo suppression of hepatocellular carcinoma growth by chitosan nanoparticles. Eur J Cancer. 2007;43:184–193.
  • Peng D, Zhang J, Liu Q, et al. Size effect of elemental selenium nanoparticles (nano-Se) at supranutritional levels on selenium accumulation and glutathione S-transferase activity. Inorg Biochem. 2007;101:1457–1463.
  • Amin KA, Hashem KS, Alshehri FS, et al. Antioxidant and hepatoprotective efficiency of selenium nanoparticles against acetaminophen-induced hepatic damage. Biol Trace Elem Res. 2017;175:136–145.
  • Ze Y, Hu R, Wang X, et al. Neurotoxicity and gene expressed profile in brain-injured mice caused by exposure to titanium dioxide nanoparticles. J Biomed Mater Res Part A. 2014;102(2):470–478.
  • Choi J, Zheng QD, Katz HE, et al. Silica-based nanoparticle uptake and cellular response by primary microglia. Environ Health Perspect. 2010;118:589–595.
  • Li CH, Shen CC, Cheng YW, et al. Organ biodistribution, clearance, and genotoxicity of orally administered zinc oxide nanoparticles in mice. Nanotoxicology. 2012;6:746–756.
  • Tian L, Lin BC, Wu L, et al. Neurotoxicity induced by zinc oxide nanoparticles, age-related differences and interaction. Sci Rep. 2015;5:12.
  • Dziendzikowska K, Gromadzka-Ostrowska J, Lankoff A, et al. Time-dependent biodistribution and excretion of silver nanoparticles in male Wistar rats. J Appl Toxicol. 2012;32:920–928.
  • Yang Y, Mao Z, Huang W, et al. Redox enzyme-mimicking activities of CeO(2) nanostructures: intrinsic influence of exposed facets. Sci Rep. 2016;6:35344.
  • Dowding JM, Song W, Bossy K, et al. Cerium oxide nanoparticles protect against Aβ-induced mitochondrial fragmentation and neuronal cell death. Cell Death Differ. 2014;21(10):1622–1632.
  • Guan Y, Gao N, Ren J, et al. Rationally designed CeNP@MnMoS4 core-shell nanoparticles for modulating multiple facets of Alzheimer’s disease. Chem Eur J. 2016;22:14523–14526.
  • Guan Y, Li M, Dong K, et al. Ceria/POMs hybrid nanoparticles as a mimicking metallopeptidase for treatment of neurotoxicity of amyloid-β peptide. Biomaterials. 2016;98:92–102.
  • Du X, Wang C, Liu Q. Potential roles of selenium and selenoproteins in the prevention of Alzheimer’s disease. Curr Top Med Chem. 2016;16:835–848.
  • Huang CL, Hsiao IL, Lin HC, et al. Silver nanoparticles’ effect on gene expression of inflammatory and neurodegenerative responses in mouse brain neural cells. Environ Res. 2015;136:253–263.
  • Tang M, Wang M, Xing T, et al. Mechanisms of unmodified CdSe quantum dot-induced elevation of cytoplasmic calcium levels in primary cultures of rat hippocampal neurons. Biomaterials. 2008;29(33):4383–4391.
  • Pecze L, Blum W, Schwaller B. Routes of Ca2+ shuttling during Ca2+ oscillations: focus on the role of mitochondrial Ca2+ handling and cytosolic Ca2+ buffers. J Biol Chem. 2015;290(47):28214–28230.
  • Pecze L, Blum W, Henzi T, et al. Endogenous TRPV1 stimulation leads to the activation of the inositol phospholipid pathway necessary for sustained Ca(2+) oscillations. Biochim Biophys Acta. 2016;1863(12):2905–2915.
  • Pecze L, Winter Z, Jósvay K, et al. Divalent heavy metal cations block the TRPV1 Ca(2+) channel. Biol Trace Elem Res. 2013;151(3):451–461.
  • Pecze L, Jósvay K, Blum W, et al. Activation of endogenous TRPV1 fails to induce overstimulation-based cytotoxicity in breast and prostate cancer cells but not in pain-sensing neurons. Biochim Biophys Acta. 2016;1863(8):2054–2064.
  • Kahya MC, Nazıroğlu M, Övey İS. Modulation of diabetes-induced oxidative stress, apoptosis, and Ca2+ entry through TRPM2 and TRPV1 channels in dorsal root ganglion and hippocampus of diabetic rats by melatonin and selenium. Mol Neurobiol. 2017;54:2345–2360.
  • Nel A, Xia T, Mädler L, et al. Toxic potential of materials at the nano level. Science. 2006;311(5761):622–627.
  • Juillerat-Jeanneret L. The targeted delivery of cancer drugs across the blood-brain barrier, chemical modifications of drugs or drug-nanoparticles? Drug Discov Today. 2008;13:1099–1106.
  • Halamoda Kenzaoui B, Chapuis Bernasconi C, Guney-Ayra S, et al. Induction of oxidative stress, lysosome activation and autophagy by nanoparticles in human brain-derived endothelial cells. Biochem J. 2012;441:813–821.
  • Tang J, Xiong L, Zhou G, et al. Silver nanoparticles crossing through and distribution in the blood-brain barrier in vitro. J Nanosci Nanotechnol. 2010;10(10):6313–6317.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.