155
Views
10
CrossRef citations to date
0
Altmetric
Original Research

Copper Nanoparticles Induce Oxidative Stress via the Heme Oxygenase 1 Signaling Pathway in vitro Studies

ORCID Icon, , , , , , , & show all
Pages 1565-1573 | Published online: 26 Feb 2021

References

  • Nel A, Xia T, Madler L, Li N. Toxic potential of materials at the nanolevel. Science. 2006;311(5761):622–627. doi:10.1126/science.1114397
  • Dong C, Feng W, Xu W, et al. The coppery age: copper (Cu)-involved nanotheranostics. Adv Sci (Weinh). 2020;7(21):2001549. doi:10.1002/advs.202001549
  • Hassan S, Hassan FU, Rehman MS. Nano-particles of trace minerals in poultry nutrition: potential applications and future prospects. Biol Trace Elem Res. 2020;195(2):591–612. doi:10.1007/s12011-019-01862-9
  • Deka P, Borah BJ, Saikia H, Bharali P. Cu-based nanoparticles as emerging environmental catalysts. Chem Rec. 2019;19(2–3):462–473. doi:10.1002/tcr.201800055
  • Ameh T, Sayes CM. The potential exposure and hazards of copper nanoparticles: a review. Environ Toxicol Pharmacol. 2019;71:103220. doi:10.1016/j.etap.2019.103220
  • Anjum NA, Adam V, Kizek R, et al. Nanoscale copper in the soil-plant system - toxicity and underlying potential mechanisms. Environ Res. 2015;138:306–325. doi:10.1016/j.envres.2015.02.019
  • Chen Z, Meng H, Xing G, et al. Acute toxicological effects of copper nanoparticles in vivo. Toxicol Lett. 2006;163(2):109–120. doi:10.1016/j.toxlet.2005.10.003
  • Tang H, Xu M, Zhou X, et al. Acute toxicity and biodistribution of different sized copper nano-particles in rats after oral administration. Mater Sci Eng C Mater Biol Appl. 2018;93:649–663. doi:10.1016/j.msec.2018.08.032
  • Lee IC, Ko JW, Park SH, et al. Comparative toxicity and biodistribution of copper nanoparticles and cupric ions in rats. Int J Nanomedicine. 2016;11:2883–2900. doi:10.2147/IJN.S106346
  • Bai R, Zhang L, Liu Y, et al. The dose-dependent toxicological effects and potential perturbation on the neurotransmitter secretion in brain following intranasal instillation of copper nanoparticles. Nanotoxicology. 2012;6(5):562–575. doi:10.3109/17435390.2011.590906
  • Yang J, Hu S, Rao M, et al. Copper nanoparticle-induced ovarian injury, follicular atresia, apoptosis, and gene expression alterations in female rats. Int J Nanomedicine. 2017;12:5959–5971. doi:10.2147/IJN.S139215
  • Sirotkin AV, Radosová M, Tarko A, Martín-García I, Alonso F. Effect of morphology and support of copper nanoparticles on basic ovarian granulosa cell functions. Nanotoxicology. 2020;14(5):683–695. doi:10.1080/17435390.2020.1736680
  • Roychoudhury S, Nath S, Massanyi P, Stawarz R, Kacaniova M, Kolesarova A. Copper-induced changes in reproductive functions: in vivo and in vitro effects. Physiol Res. 2016;65(1):11–22. doi:10.33549/physiolres.933063
  • Midander K, Cronholm P, Karlsson HL, et al. Surface characteristics, copper release, and toxicity of nano- and micrometer-sized copper and copper(II) oxide particles: a cross-disciplinary study. Small. 2009;5(3):389–399. doi:10.1002/smll.200801220
  • Meng H, Chen Z, Xing G, et al. Ultrahigh reactivity provokes nanotoxicity: explanation of oral toxicity of nano-copper particles. Toxicol Lett. 2007;175(1–3):102–110. doi:10.1016/j.toxlet.2007.09.015
  • Strauch BM, Niemand RK, Winkelbeiner NL, Hartwig A. Comparison between micro- and nanosized copper oxide and water soluble copper chloride: interrelationship between intracellular copper concentrations, oxidative stress and DNA damage response in human lung cells. Part Fibre Toxicol. 2017;14(1):28. doi:10.1186/s12989-017-0209-1
  • Xu P, Xu J, Liu S, Yang Z. Nano copper induced apoptosis in podocytes via increasing oxidative stress. J Hazard Mater. 2012;241–242:279–286. doi:10.1016/j.jhazmat.2012.09.041
  • Denluck L, Wu F, Crandon LE, Harper BJ, Harper SL. Reactive oxygen species generation is likely a driver of copper based nanomaterial toxicity. Environ Sci Nano. 2018;5(6):1473–1481. doi:10.1039/C8EN00055G
  • Manna P, Ghosh M, Ghosh J, Das J, Sil PC. Contribution of nano-copper particles to in vivo liver dysfunction and cellular damage: role of IκBα/NF-κB, MAPKs and mitochondrial signal. Nanotoxicology. 2012;6(1):1–21. doi:10.3109/17435390.2011.552124
  • Sarkar A, Das J, Manna P, Sil PC. Nano-copper induces oxidative stress and apoptosis in kidney via both extrinsic and intrinsic pathways. Toxicology. 2011;290(2–3):208–217. doi:10.1016/j.tox.2011.09.086
  • Zhou X, Zhao L, Luo J, et al. The toxic effects and mechanisms of nano-cu on the spleen of rats. Int J Mol Sci. 2019;20(6):1469. doi:10.3390/ijms20061469
  • Da Clarke HJ. Regulation of germ cell development by intercellular signaling in the mammalian ovarian follicle. Wiley Interdiscip Rev Dev Biol. 2018;7(1):1. doi:10.1002/wdev.294
  • Lushchak VI. Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact. 2014;224:164–175. doi:10.1016/j.cbi.2014.10.016
  • Newsholme P, Cruzat VF, Keane KN, Carlessi R, de Bittencourt PI Jr. Molecular mechanisms of ROS production and oxidative stress in diabetes. Biochem J. 2016;473(24):4527–4550.
  • Nitti M, Piras S, Marinari UM, Moretta L, Pronzato MA, Furfaro AL. HO-1 induction in cancer progression: a matter of cell adaptation. Antioxidants (Basel). 2017;6(2):29. doi:10.3390/antiox6020029
  • Wang Y, Yang C, Elsheikh NAH, et al. HO-1 reduces heat stress-induced apoptosis in bovine granulosa cells by suppressing oxidative stress. Aging (Albany NY). 2019;11(15):5535–5547. doi:10.18632/aging.102136
  • Song C, Heping H, Shen Y, et al. AMPK/p38/Nrf2 activation as a protective feedback to restrain oxidative stress and inflammation in microglia stimulated with sodium fluoride. Chemosphere. 2020;244:125495. doi:10.1016/j.chemosphere.2019.125495
  • Campbell JH, Heikkila JJ. Effect of hemin, baicalein and heme oxygenase-1 (HO-1) enzyme activity inhibitors on Cd-induced accumulation of HO-1, HSPs and aggresome-like structures in Xenopus kidney epithelial cells. Comp Biochem Physiol C Toxicol Pharmacol. 2018;210:1–17. doi:10.1016/j.cbpc.2018.04.003
  • Zhang CH, Wang Y, Sun QQ, et al. Copper nanoparticles show obvious in vitro and in vivo reproductive toxicity via ERK mediated signaling pathway in female mice. Int J Biol Sci. 2018;14(13):1834–1844. doi:10.7150/ijbs.27640
  • Dhupal M, Oh JM, Tripathy DR, Kim SK, Koh SB, Park KS. Immunotoxicity of titanium dioxide nanoparticles via simultaneous induction of apoptosis and multiple toll-like receptors signaling through ROS-dependent SAPK/JNK and p38 MAPK activation. Int J Nanomedicine. 2018;13:6735–6750. doi:10.2147/IJN.S176087
  • Moniczewski A, Gawlik M, Smaga I, et al. Oxidative stress as an etiological factor and a potential treatment target of psychiatric disorders. Part 1. Chemical aspects and biological sources of oxidative stress in the brain. Pharmacol Rep. 2015;67(3):560–568. doi:10.1016/j.pharep.2014.12.014
  • Cuadrado A, Nebreda AR. Mechanisms and functions of p38 MAPK signalling. Biochem J. 2010;429(3):403–417. doi:10.1042/BJ20100323
  • Ono K, Han J. The p38 signal transduction pathway: activation and function. Cell Signal. 2000;12(1):1–13. doi:10.1016/S0898-6568(99)00071-6
  • Ahmad MI, Zou X, Ijaz MU, et al. Processed meat protein promoted inflammation and hepatic lipogenesis by upregulating Nrf2/Keap1 signaling pathway in Glrx-deficient mice. J Agric Food Chem. 2019;67(32):8794–8809. doi:10.1021/acs.jafc.9b03136
  • Aueviriyavit S, Phummiratch D, Maniratanachote R. Mechanistic study on the biological effects of silver and gold nanoparticles in Caco-2 cells–induction of the Nrf2/HO-1 pathway by high concentrations of silver nanoparticles. Toxicol Lett. 2014;224(1):73–83. doi:10.1016/j.toxlet.2013.09.020
  • Liu W, Hu T, Zhou L, et al. Nrf2 protects against oxidative stress induced by SiO2 nanoparticles. Nanomedicine (Lond). 2017;12(19):2303–2318. doi:10.2217/nnm-2017-0046
  • Wang J, Li N, Zheng L, et al. P38-Nrf-2 signaling pathway of oxidative stress in mice caused by nanoparticulate TiO2. Biol Trace Elem Res. 2011;140(2):186–197. doi:10.1007/s12011-010-8687-0
  • Ko WC, Shieh JM, Wu WB. P38 MAPK and Nrf2 activation mediated naked gold nanoparticle induced heme oxygenase-1 expression in rat aortic vascular smooth muscle cells. Arch Med Res. 2020;51(5):388–396. doi:10.1016/j.arcmed.2020.04.015
  • Loboda A, Damulewicz M, Pyza E, Jozkowicz A, Dulak J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: an evolutionarily conserved mechanism. Cell Mol Life Sci. 2016;73(17):3221–3247.
  • Chen S, Wang X, Nisar MF, Lin M, Zhong JL. Heme oxygenases: cellular multifunctional and protective molecules against UV-induced oxidative stress. Oxid Med Cell Longev. 2019;2019:5416728. doi:10.1155/2019/5416728
  • Liu X, Lin X, Zhang S, et al. Lycopene ameliorates oxidative stress in the aging chicken ovary via activation of Nrf2/HO-1 pathway. Aging (Albany NY). 2018;10(8):2016–2036. doi:10.18632/aging.101526