253
Views
15
CrossRef citations to date
0
Altmetric
Original Research

Zinc Oxide Nanoparticles Induce Mitochondrial Biogenesis Impairment and Cardiac Dysfunction in Human iPSC-Derived Cardiomyocytes

, , ORCID Icon, , , , , & show all
Pages 2669-2683 | Published online: 21 Apr 2020

References

  • Liu J, Feng X, Wei L, Chen L, Song B, Shao L. The toxicology of ion-shedding zinc oxide nanoparticles. Crit Rev Toxicol. 2016;46(4):348–384. doi:10.3109/10408444.2015.113786426963861
  • Jacobsen NR, Stoeger T, van den Brule S, et al. Acute and subacute pulmonary toxicity and mortality in mice after intratracheal instillation of ZnO nanoparticles in three laboratories. Food Chem Toxicol. 2015;85:84–95. doi:10.1016/j.fct.2015.08.00826260750
  • Danielsen PH, Cao Y, Roursgaard M, Moller P, Loft S. Endothelial cell activation, oxidative stress and inflammation induced by a panel of metal-based nanomaterials. Nanotoxicology. 2015;9(7):813–824. doi:10.3109/17435390.2014.98044925405261
  • Konduru NV, Murdaugh KM, Sotiriou GA, et al. Bioavailability, distribution and clearance of tracheally-instilled and gavaged uncoated or silica-coated zinc oxide nanoparticles. Part Fibre Toxicol. 2014;11:44. doi:10.1186/s12989-014-0044-625183210
  • Esmaeillou M, Moharamnejad M, Hsankhani R, Tehrani AA, Maadi H. Toxicity of ZnO nanoparticles in healthy adult mice. Environ Toxicol Pharmacol. 2013;35(1):67–71. doi:10.1016/j.etap.2012.11.00323262039
  • Li CH, Shen CC, Cheng YW, et al. Organ biodistribution, clearance, and genotoxicity of orally administered zinc oxide nanoparticles in mice. Nanotoxicology. 2012;6(7):746–756. doi:10.3109/17435390.2011.62071721950449
  • Xiaoli F, Junrong W, Xuan L, et al. Prenatal exposure to nanosized zinc oxide in rats: neurotoxicity and postnatal impaired learning and memory ability. Nanomedicine (Lond). 2017;12(7):777–795. doi:10.2217/nnm-2016-039728322126
  • Kielbik P, Kaszewski J, Rosowska J, et al. Biodegradation of the ZnO:Eu nanoparticles in the tissues of adult mouse after alimentary application. Nanomedicine. 2017;13(3):843–852. doi:10.1016/j.nano.2016.11.00227884640
  • Cao Y, Gong Y, Liao W, et al. A review of cardiovascular toxicity of TiO2, ZnO and Ag nanoparticles (NPs). Biometals. 2018;31(4):457–476. doi:10.1007/s10534-018-0113-729748744
  • Monse C, Hagemeyer O, Raulf M, et al. Concentration-dependent systemic response after inhalation of nano-sized zinc oxide particles in human volunteers. Part Fibre Toxicol. 2018;15(1):8. doi:10.1186/s12989-018-0246-429429408
  • Baky NA, Faddah LM, Al-Rasheed NM, Al-Rasheed NM, Fatani AJ. Induction of inflammation, DNA damage and apoptosis in rat heart after oral exposure to zinc oxide nanoparticles and the cardioprotective role of alpha-lipoic acid and vitamin E. Drug Res. 2013;63(5):228–236. doi:10.1055/s-0033-1334923
  • Chuang HC, Juan HT, Chang CN, et al. Cardiopulmonary toxicity of pulmonary exposure to occupationally relevant zinc oxide nanoparticles. Nanotoxicology. 2014;8(6):593–604. doi:10.3109/17435390.2013.80980923738974
  • Bengalli R, Gualtieri M, Capasso L, Urani C, Camatini M. Impact of zinc oxide nanoparticles on an in vitro model of the human air-blood barrier. Toxicol Lett. 2017;279:22–32. doi:10.1016/j.toxlet.2017.07.87728709982
  • Yan Z, Wang W, Wu Y, et al. Zinc oxide nanoparticle-induced atherosclerotic alterations in vitro and in vivo. Int J Nanomedicine. 2017;12:4433–4442. doi:10.2147/IJN.S13489728652743
  • Bessemer RA, Butler KM, Tunnah L, et al. Cardiorespiratory toxicity of environmentally relevant zinc oxide nanoparticles in the freshwater fish Catostomus commersonii. Nanotoxicology. 2015;9(7):861–870. doi:10.3109/17435390.2014.98273725427894
  • Chuang KJ, Lee KY, Pan CH, et al. Effects of zinc oxide nanoparticles on human coronary artery endothelial cells. Food Chem Toxicol. 2016;93:138–144. doi:10.1016/j.fct.2016.05.00827185063
  • Liang S, Sun K, Wang Y, et al. Role of Cyt-C/caspases-9,3, Bax/Bcl-2 and the FAS death receptor pathway in apoptosis induced by zinc oxide nanoparticles in human aortic endothelial cells and the protective effect by alpha-lipoic acid. Chem Biol Interact. 2016;258:40–51. doi:10.1016/j.cbi.2016.08.01327544635
  • Handral HK, Tong HJ, Islam I, Sriram G, Rosa V, Cao T. Pluripotent stem cells: an in vitro model for nanotoxicity assessments. J Appl Toxicol. 2016;36(10):1250–1258. doi:10.1002/jat.334727241574
  • Magdy T, Schuldt AJT, Wu JC, Bernstein D, Burridge PW. Human induced pluripotent stem cell (hiPSC)-derived cells to assess drug cardiotoxicity: opportunities and problems. Annu Rev Pharmacol Toxicol. 2018;58:83–103. doi:10.1146/annurev-pharmtox-010617-05311028992430
  • Karakikes I, Ameen M, Termglinchan V, Wu JC. Human induced pluripotent stem cell-derived cardiomyocytes: insights into molecular, cellular, and functional phenotypes. Circ Res. 2015;117(1):80–88. doi:10.1161/CIRCRESAHA.117.30536526089365
  • Yoshida Y, Yamanaka S. Induced pluripotent stem cells 10 years later: for cardiac applications. Circ Res. 2017;120(12):1958–1968. doi:10.1161/CIRCRESAHA.117.31108028596174
  • Cui N, Wu F, Lu WJ, et al. Doxorubicin-induced cardiotoxicity is maturation dependent due to the shift from topoisomerase IIalpha to IIbeta in human stem cell derived cardiomyocytes. J Cell Mol Med. 2019;23(7):4627–4639. doi:10.1111/jcmm.1434631106979
  • Yin J, Guo J, Zhang Q, et al. Doxorubicin-induced mitophagy and mitochondrial damage is associated with dysregulation of the PINK1/parkin pathway. Toxicol in Vitro. 2018;51:1–10. doi:10.1016/j.tiv.2018.05.00129729358
  • Bostan HB, Rezaee R, Valokala MG, et al. Cardiotoxicity of nano-particles. Life Sci. 2016;165:91–99. doi:10.1016/j.lfs.2016.09.01727686832
  • Guo C, Wang J, Jing L, et al. Mitochondrial dysfunction, perturbations of mitochondrial dynamics and biogenesis involved in endothelial injury induced by silica nanoparticles. Environ Pollut. 2018;236:926–936. doi:10.1016/j.envpol.2017.10.06029074197
  • Gustafsson CM, Falkenberg M, Larsson NG. Maintenance and expression of mammalian mitochondrial DNA. Annu Rev Biochem. 2016;85:133–160. doi:10.1146/annurev-biochem-060815-01440227023847
  • Wenz T. Regulation of mitochondrial biogenesis and PGC-1alpha under cellular stress. Mitochondrion. 2013;13(2):134–142. doi:10.1016/j.mito.2013.01.00623347985
  • Birket MJ, Casini S, Kosmidis G, et al. PGC-1alpha and reactive oxygen species regulate human embryonic stem cell-derived cardiomyocyte function. Stem Cell Rep. 2013;1(6):560–574. doi:10.1016/j.stemcr.2013.11.008
  • Sun J, Wang S, Zhao D, Hun FH, Weng L, Liu H. Cytotoxicity, permeability, and inflammation of metal oxide nanoparticles in human cardiac microvascular endothelial cells: cytotoxicity, permeability, and inflammation of metal oxide nanoparticles. Cell Biol Toxicol. 2011;27(5):333–342. doi:10.1007/s10565-011-9191-921681618
  • Wang Y, Wu Y, Quadri F, Prox JD, Guo L. Cytotoxicity of ZnO nanowire arrays on excitable cells. Nanomaterials (Basel). 2017;7(4):80. doi:10.3390/nano7040080
  • Blinova K, Dang Q, Millard D, et al. International multisite study of human-induced pluripotent stem cell-derived cardiomyocytes for drug proarrhythmic potential assessment. Cell Rep. 2018;24(13):3582–3592. doi:10.1016/j.celrep.2018.08.07930257217
  • Wang M, Wang J, Liu Y, et al. Subcellular targets of zinc oxide nanoparticles during the aging process: role of cross-talk between mitochondrial dysfunction and endoplasmic reticulum stress in the genotoxic response. Toxicol Sci. 2019;171(1):159–171. doi:10.1093/toxsci/kfz132
  • Wang MM, Wang YC, Wang XN, et al. Mutagenicity of ZnO nanoparticles in mammalian cells: role of physicochemical transformations under the aging process. Nanotoxicology. 2015;9(8):972–982. doi:10.3109/17435390.2014.99281625676621
  • Wang Y, Ding L, Yao C, et al. Toxic effects of metal oxide nanoparticles and their underlying mechanisms. Sci China Mater. 2017;60(2):93–108. doi:10.1007/s40843-016-5157-0
  • Pfanner N, Warscheid B, Wiedemann N. Mitochondrial proteins: from biogenesis to functional networks. Nat Rev Mol Cell Biol. 2019;20(5):267–284. doi:10.1038/s41580-018-0092-030626975
  • Park YH, Bae HC, Kim J, Jeong SH, Yang SI, Son SW. Zinc oxide nanoparticles induce HIF-1alpha protein stabilization through increased reactive oxygen species generation from electron transfer chain complex III of mitochondria. J Dermatol Sci. 2018;91(1):104–107. doi:10.1016/j.jdermsci.2018.03.01029622477
  • Babele PK, Thakre PK, Kumawat R, Tomar RS. Zinc oxide nanoparticles induce toxicity by affecting cell wall integrity pathway, mitochondrial function and lipid homeostasis in Saccharomyces cerevisiae. Chemosphere. 2018;213:65–75. doi:10.1016/j.chemosphere.2018.09.02830212720
  • Zhao X, Ren X, Zhu R, Luo Z, Ren B. Zinc oxide nanoparticles induce oxidative DNA damage and ROS-triggered mitochondria-mediated apoptosis in zebrafish embryos. Aquat Toxicol. 2016;180:56–70. doi:10.1016/j.aquatox.2016.09.01327658222
  • Pohjoismaki JL, Goffart S. The role of mitochondria in cardiac development and protection. Free Radic Biol Med. 2017;106:345–354. doi:10.1016/j.freeradbiomed.2017.02.03228216385
  • Guo J, Guo Q, Fang H, et al. Cardioprotection against doxorubicin by metallothionein Is associated with preservation of mitochondrial biogenesis involving PGC-1alpha pathway. Eur J Pharmacol. 2014;737:117–124. doi:10.1016/j.ejphar.2014.05.01724858368
  • Yuan H, Zhang Q, Guo J, et al. A PGC-1alpha-mediated transcriptional network maintains mitochondrial redox and bioenergetic homeostasis against doxorubicin-induced toxicity in human cardiomyocytes: implementation of TT21C. Toxicol Sci. 2016;150(2):400–417. doi:10.1093/toxsci/kfw00626781513
  • Kussauer S, David R, Lemcke H. hiPSCs derived cardiac cells for drug and toxicity screening and disease modeling: what micro- electrode-array analyses can tell us. Cells. 2019;8(11):1331. doi:10.3390/cells8111331
  • Millard D, Dang Q, Shi H, et al. Cross-site reliability of human induced pluripotent stem cell-derived cardiomyocyte based safety assays using microelectrode arrays: results from a blinded CiPA pilot study. Toxicol Sci. 2018;164(2):550–562. doi:10.1093/toxsci/kfy11029718449
  • Du J, Wang S, You H, Jiang R, Zhuang C, Zhang X. Developmental toxicity and DNA damage to zebrafish induced by perfluorooctane sulfonate in the presence of ZnO nanoparticles. Environ Toxicol. 2016;31(3):360–371. doi:10.1002/tox.2205025258305
  • Haberl N, Hirn S, Holzer M, Zuchtriegel G, Rehberg M, Krombach F. Effects of acute systemic administration of TiO2, ZnO, SiO2, and Ag nanoparticles on hemodynamics, hemostasis and leukocyte recruitment. Nanotoxicology. 2015;9(8):963–971. doi:10.3109/17435390.2014.99281525670207
  • Li S, Pan H, Tan C, et al. Mitochondrial dysfunctions contribute to hypertrophic cardiomyopathy in patient iPSC-derived cardiomyocytes with MT-RNR2 mutation. Stem Cell Rep. 2018;10(3):808–821. doi:10.1016/j.stemcr.2018.01.013