簡易檢索 / 詳目顯示

研究生: 潘娟利
Rosita Pranata
論文名稱: 以替代毒理方法探討食品奈米物質之基因毒性機轉
Application of alternative methods to assess the genotoxic mechanisms of nanoparticles in food products
指導教授: 陳容甄
Chen, Rong-Jane
學位類別: 碩士
Master
系所名稱: 醫學院 - 食品安全衛生暨風險管理研究所
Department of Food Safety / Hygiene and Risk Management
論文出版年: 2022
畢業學年度: 110
語文別: 英文
論文頁數: 73
外文關鍵詞: titanium dioxide nanoparticles, zinc oxide nanoparticles, metal oxide nanoparticles, genotoxicity, autophagy, mitochondrial damage, food safety
相關次數: 點閱:53下載:3
分享至:
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報

隨著科技發展的進步,奈米物質獨特物化特性使其常被應用於各大工業,且具有極大的的發展空間。在食品工業中,奈米物質常作為一食品改良劑,使食品的品質進一步提升,例如:奈米氧化鋅(ZnO NPs)和奈米二氧化鈦(TiO2 NPs)分別被作為抗菌劑和增白劑來使用。但隨著奈米物質應用範圍擴大,使得人體暴露之途徑增加,故其獨特之物化特性對於人體之健康危害也逐漸被關注,但卻仍缺少相對應的風險評估方式。隨著大眾對動物福祉之意識提升,符合3R(替代、減少、優化)原則意念之體外試驗和電腦模擬方法,對於奈米物質毒性之危害風險評估更具前瞻性。因此,本研究目的為調查奈米顆粒之物化特性是如何造成基因毒性,並同時探討其毒性誘導機制。在細胞實驗中,使用具有不同化學組成及表面改質之羧酸奈米二氧化鈦(COOH-coated TiO2 NPs)、羧酸奈米氧化鋅(COOH-coated ZnO NPs)和胺基奈米氧化鋅(NH2-coated ZnO NPs)對HepaRG cells進行暴露。結果顯示,在HepaRG細胞中所有的奈米物質皆會在以不造成細胞毒殺效應之濃度範圍暴露下誘導基因毒性反應並使細胞存活率下降,同時也會使粒線體活性氧物質生成上升、膜電位下降及DNA損傷,促使細胞自噬和粒線體自噬作用。在三種奈米物質中,毒性強度由高至低依序為羧酸奈米氧化鋅、胺基奈米氧化鋅及羧酸奈米二氧化鈦。利用替代動物實驗方法探討奈米物質之潛在毒性風險是為現代研究極具前景之趨勢,本研究也應用替代動物毒性測試方法進行探討,並發現結果與先前動物模式之研究結果相符合,表明未來可依此作為符合3R原則之奈米物質毒性測試方法發展之基礎,同時,因奈米物質誘導之毒性機制仍未知,企盼未來能進行更深入的研究。

With the recent advances in technology, nanoparticles with their unique properties hold great promise in their current and future applications. In the food industry, the application of nanoparticles improves the quality of food products, e.g. ZnO NPs as anti-microbial additives and TiO2 NPs that serves as a whitening agent. Accordingly, different physicochemical properties of nanoparticles will lead to different characteristics. The increasing worldwide applications of nanoparticles in various fields have inevitably increased the risks of exposure to humans through various routes. Unfortunately, the rapid development is not accompanied by the respective risk assessment. Under the 3R principle (Replacement, Reduction, and Refinement), in vitro and in silico approaches are the alternative testing methods that are more suited to investigate nanoparticle-induced toxicity. Therefore, the purpose of this study is to investigate how the physicochemical properties of nanoparticles influence the resulting genotoxicity and the underlying mechanism. In this study, HepaRG cells were subjected to nanoparticles with different chemical composition and surface modification, which is COOH-coated TiO2 NPs, COOH-coated ZnO NPs, and NH2-coated ZnO NPs. All nanoparticles induced genotoxicity at a range of concentrations that is not cytotoxic in the cells, which were shown by the reduction in cell viability, increase in mitochondrial ROS production, MMP decrease, activation of DNA damage signaling pathway, and the induction of autophagy and mitophagy. Out of the three nanoparticles, COOH-coated ZnO NPs were found to be the most toxic, followed by NH2-coated ZnO NPs, and COOH-coated TiO2 NPs were the least toxic. The alternative toxicity testing methods for the analysis of nanoparticle-induced toxicity show a promising future. No experimental animals were used in this study, however, the results were consistent with the previous studies. Testing methods used in this study contribute to the validation of the alternative testing methods for nanoparticle toxicity studies. Further investigation of the nanoparticle-induced toxicity mechanism should be performed, as it remains to be elucidated.

摘要 I Abstract II Acknowledgments III Table of Contents V List of Tables VII List of Figures VIII List of Abbreviations IX Chapter 1 Introduction 1 Chapter 2 Literature Review 2 2.1 Nanoparticles 2 2.1.1 Nanoparticles in the food industry and personal care products 2 2.2 Nanoparticle-induced toxicity 3 2.2.1 Titanium dioxide nanoparticles (TiO2 NPs)-induced toxicity 5 2.2.2 Zinc oxide nanoparticles (ZnO NPs)-induced toxicity 6 2.3 Genotoxicity 12 2.4 Nanoparticle-induced autophagy 14 2.4.1 Crosstalk between DNA damage response (DDR) and autophagy pathway 15 2.4.2 Nanoparticle-induced mitophagy 16 2.4.3 Crosstalk between mitochondrial and autophagic pathways 17 2.5 Adverse Outcome Pathway (AOP) 18 2.6 Alternative methods in toxicity testing 18 Chapter 3 Study Design 20 3.1 Research objectives 20 3.2 Experimental design 20 Chapter 4 Materials and Methods 21 4.1 Cell line 21 4.2 Physicochemical characterization of the nanoparticles used in this study 21 4.3 Methods 21 4.3.1 Cell culture 21 4.3.2 Cell freezing 22 4.3.3 Cell thawing 22 4.3.4 Cell viability assay 22 4.3.5 Measurement of mitochondrial reactive oxygen species (ROS) production 23 4.3.6 Mitochondrial membrane potential (MMP) analysis 24 4.3.7 Immunofluorescence (IF) staining 24 4.3.8 Acidic vesicular organelles detection 25 4.3.9 Mitophagy detection 26 4.3.10 Western blotting 27 4.3.11 ToxPi analysis 30 4.4 Statistical analysis 30 Chapter 5 Results 31 5.1 Nanoparticles characterization 31 5.2 Exposure to nanoparticles decreased HepaRG cell viability 31 5.3 Exposure to NPs increased mitochondrial ROS production in HepaRG cells 32 5.4 Exposure to NPs induced changes in the mitochondrial membrane potential (MMP) in HepaRG cells 33 5.5 Exposure to nanoparticles induced DNA damage in HepaRG cells 35 5.6 Exposure to nanoparticles induced autophagy in HepaRG cells 37 5.7 Exposure to nanoparticles induced mitophagy in HepaRG cells 37 5.8 Toxicity comparison and ranking by ToxPi 39 Chapter 6 Discussion 40 Chapter 7 Conclusions and Suggestions 46 References 47 Tables 56 Figures 57

1. The European Commission, Commission Recommendation of 18 October 2011 on the definition of nanomaterial. 2011, Official Journal of the European Union: Brussels. p. 275/38-40.
2. Oberdörster, G., E. Oberdörster, and J. Oberdörster, Nanotoxicology: an emerging discipline evolving from studies of ultrafine particles. Environ Health Perspect, 2005. 113(7): p. 823-39.
3. Haque, E. and A.C. Ward, Zebrafish as a Model to Evaluate Nanoparticle Toxicity. Nanomaterials, 2018. 8(7): p. 561.
4. Chen, R.-J., et al., The Current Understanding of Autophagy in Nanomaterial Toxicity and Its Implementation in Safety Assessment-Related Alternative Testing Strategies. International Journal of Molecular Sciences, 2020. 21(7): p. 2387.
5. Di Bucchianico, S., et al., Calcium-dependent cyto- and genotoxicity of nickel metal and nickel oxide nanoparticles in human lung cells. Particle and Fibre Toxicology, 2018. 15(1): p. 32.
6. Djurišić, A.B., et al., Toxicity of Metal Oxide Nanoparticles: Mechanisms, Characterization, and Avoiding Experimental Artefacts. Small, 2015. 11(1): p. 26-44.
7. Warheit, D., Hazard and risk assessment strategies for nanoparticle exposures: how far have we come in the past 10 years? [version 1; peer review: 2 approved]. F1000Research, 2018. 7(376).
8. Cui, L., et al., Predictive Metabolomic Signatures for Safety Assessment of Metal Oxide Nanoparticles. ACS Nano, 2019. 13(11): p. 13065-13082.
9. Magdolenova, Z., et al., Safety Assessment of Nanoparticles Cytotoxicity and Genotoxicity of Metal Nanoparticles In Vitro. Journal of biomedical nanotechnology, 2011. 7: p. 20-1.
10. McClements, D.J. and H. Xiao, Is nano safe in foods? Establishing the factors impacting the gastrointestinal fate and toxicity of organic and inorganic food-grade nanoparticles. npj Science of Food, 2017. 1(1): p. 6.
11. Osman, I.F., et al., Genotoxicity and cytotoxicity of zinc oxide and titanium dioxide in HEp-2 cells. Nanomedicine, 2010. 5(8): p. 1193-1203.
12. Petrarca, C., et al., Engineered metal based nanoparticles and innate immunity. Clinical and molecular allergy : CMA, 2015. 13(1): p. 13-13.
13. Magdolenova, Z., et al., Mechanisms of genotoxicity. A review of in vitro and in vivo studies with engineered nanoparticles. Nanotoxicology, 2014. 8(3): p. 233-278.
14. Wang, R., et al., Potential adverse effects of nanoparticles on the reproductive system. International journal of nanomedicine, 2018. 13: p. 8487-8506.
15. Avery, V.M., et al., 3.07 - The Identification of Bioactive Natural Products by High Throughput Screening (HTS), in Comprehensive Natural Products II, H.-W. Liu and L. Mander, Editors. 2010, Elsevier: Oxford. p. 177-203.
16. Attene-Ramos, M.S., C.P. Austin, and M. Xia, High Throughput Screening, in Encyclopedia of Toxicology (Third Edition), P. Wexler, Editor. 2014, Academic Press: Oxford. p. 916-917.
17. He, C., et al., High-content analysis for mitophagy response to nanoparticles: A potential sensitive biomarker for nanosafety assessment. Nanomedicine: Nanotechnology, Biology and Medicine, 2019. 15(1): p. 59-69.
18. Deitersen, J., et al., High-throughput screening for natural compound-based autophagy modulators reveals novel chemotherapeutic mode of action for arzanol. Cell Death & Disease, 2021. 12: p. 560.
19. Kohl, Y., et al., Genotoxicity of Nanomaterials: Advanced In Vitro Models and High Throughput Methods for Human Hazard Assessment—A Review. Nanomaterials, 2020. 10(10): p. 1911.
20. Geppert, M., et al., Interactions of TiO2 Nanoparticles with Ingredients from Modern Lifestyle Products and Their Effects on Human Skin Cells. Chemical Research in Toxicology, 2020. 33(5): p. 1215-1225.
21. Horie, M., et al., Does photocatalytic activity of TiO2 nanoparticles correspond to photo-cytotoxicity? Cellular uptake of TiO2 nanoparticles is important in their photo-cytotoxicity. Toxicology Mechanisms and Methods, 2016. 26(4): p. 284-294.
22. Zhang, L.W. and N.A. Monteiro-Riviere, Toxicity assessment of six titanium dioxide nanoparticles in human epidermal keratinocytes. Cutaneous and Ocular Toxicology, 2019. 38(1): p. 66-80.
23. IARC, Carbon Black, Titanium Dioxide, and Talc, in IARC Monographs. 2010, WHO IARC: Lyon, France.
24. Shi, Y., et al., Titanium dioxide nanoparticles cause apoptosis in BEAS-2B cells through the caspase 8/t-Bid-independent mitochondrial pathway. Toxicology letters, 2010. 196(1): p. 21-27.
25. Guichard, Y., et al., Cytotoxicity and Genotoxicity of Nanosized and Microsized Titanium Dioxide and Iron Oxide Particles in Syrian Hamster Embryo Cells. The Annals of Occupational Hygiene, 2012. 56(5): p. 631-644.
26. Skocaj, M., et al., Titanium dioxide in our everyday life; is it safe? Radiol Oncol, 2011. 45(4): p. 227-47.
27. Shabbir, S., et al., Toxicological Consequences of Titanium Dioxide Nanoparticles (TiO(2)NPs) and Their Jeopardy to Human Population. Bionanoscience, 2021. 11(2): p. 621-632.
28. Hou, J., et al., Toxicity and mechanisms of action of titanium dioxide nanoparticles in living organisms. J Environ Sci (China), 2019. 75: p. 40-53.
29. Luo, Z., et al., Rethinking Nano-TiO2 Safety: Overview of Toxic Effects in Humans and Aquatic Animals. Small, 2020. 16(36): p. 2002019.
30. Chen, Z.-Y., et al., The Effect of the Chorion on Size-Dependent Acute Toxicity and Underlying Mechanisms of Amine-Modified Silver Nanoparticles in Zebrafish Embryos. International Journal of Molecular Sciences, 2020. 21(8).
31. Dunford, R., et al., Chemical oxidation and DNA damage catalysed by inorganic sunscreen ingredients. FEBS letters, 1997. 418(1-2): p. 87-90.
32. Mohammed, Y.H., et al., Support for the Safe Use of Zinc Oxide Nanoparticle Sunscreens: Lack of Skin Penetration or Cellular Toxicity after Repeated Application in Volunteers. Journal of Investigative Dermatology, 2019. 139(2): p. 308-315.
33. Ginzburg, A.L., et al., Zinc oxide-induced changes to sunscreen ingredient efficacy and toxicity under UV irradiation. Photochemical & Photobiological Sciences, 2021. 20(10): p. 1273-1285.
34. Sharma, V., et al., Induction of oxidative stress, DNA damage and apoptosis in mouse liver after sub-acute oral exposure to zinc oxide nanoparticles. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 2012. 745(1): p. 84-91.
35. Hou, J., et al., Toxic effects of different types of zinc oxide nanoparticles on algae, plants, invertebrates, vertebrates and microorganisms. Chemosphere, 2018. 193: p. 852-860.
36. Liou, S.-H., et al., Global DNA methylation and oxidative stress biomarkers in workers exposed to metal oxide nanoparticles. Journal of Hazardous Materials, 2017. 331: p. 329-335.
37. Coll, C., et al., Probabilistic environmental risk assessment of five nanomaterials (nano-TiO2, nano-Ag, nano-ZnO, CNT, and fullerenes). Nanotoxicology, 2016. 10(4): p. 436-444.
38. Shukla, R.K., et al., Genotoxic Potential of Nanoparticles: Structural and Functional Modifications in DNA. Front Genet, 2021. 12: p. 728250.
39. Singh, S., Zinc oxide nanoparticles impacts: cytotoxicity, genotoxicity, developmental toxicity, and neurotoxicity. Toxicology Mechanisms and Methods, 2019. 29(4): p. 300-311.
40. Almansour, M.I., et al., Zinc oxide nanoparticles hepatotoxicity: Histological and histochemical study. Environmental Toxicology and Pharmacology, 2017. 51: p. 124-130.
41. Kuang, H., et al., Size dependent effect of ZnO nanoparticles on endoplasmic reticulum stress signaling pathway in murine liver. Journal of Hazardous Materials, 2016. 317: p. 119-126.
42. Jia, X., et al., The potential liver, brain, and embryo toxicity of titanium dioxide nanoparticles on mice. Nanoscale research letters, 2017. 12(1): p. 1-14.
43. Patel, S., P. Patel, and S.R. Bakshi, Titanium dioxide nanoparticles: an in vitro study of DNA binding, chromosome aberration assay, and comet assay. Cytotechnology, 2017. 69(2): p. 245-263.
44. Chen, Z., et al., Hepatotoxicity and the role of the gut-liver axis in rats after oral administration of titanium dioxide nanoparticles. Particle and Fibre Toxicology, 2019. 16(1): p. 48.
45. Zhao, Y., et al., Foodborne TiO(2) Nanoparticles Induced More Severe Hepatotoxicity in Fructose-Induced Metabolic Syndrome Mice via Exacerbating Oxidative Stress-Mediated Intestinal Barrier Damage. Foods, 2021. 10(5).
46. Kongseng, S., et al., Cytotoxic and inflammatory responses of TiO2 nanoparticles on human peripheral blood mononuclear cells. Journal of Applied Toxicology, 2016. 36(10): p. 1364-1373.
47. Petković, J., et al., DNA damage and alterations in expression of DNA damage responsive genes induced by TiO2 nanoparticles in human hepatoma HepG2 cells. Nanotoxicology, 2011. 5(3): p. 341-353.
48. Jalili, P., et al., Chronic effects of two rutile TiO2 nanomaterials in human intestinal and hepatic cell lines. Particle and Fibre Toxicology, 2022. 19(1): p. 37.
49. He, Z., et al., Size-controlled lipid nanoparticle production using turbulent mixing to enhance oral DNA delivery. Acta Biomater, 2018. 81: p. 195-207.
50. Bai, W., et al., Toxicity of zinc oxide nanoparticles to zebrafish embryo: a physicochemical study of toxicity mechanism. Journal of Nanoparticle Research, 2010. 12(5): p. 1645-1654.
51. Punnoose, A., et al., Cytotoxicity of ZnO Nanoparticles Can Be Tailored by Modifying Their Surface Structure: A Green Chemistry Approach for Safer Nanomaterials. ACS Sustain Chem Eng, 2014. 2(7): p. 1666-1673.
52. Wang, J., et al., Zinc oxide nanoparticles induce toxicity in CAL 27 oral cancer cell lines by activating PINK1/Parkin-mediated mitophagy. Int J Nanomedicine, 2018. 13: p. 3441-3450.
53. Liu, Z., et al., Zinc oxide nanoparticles effectively regulate autophagic cell death by activating autophagosome formation and interfering with their maturation. Particle and Fibre Toxicology, 2020. 17(1): p. 46.
54. Kruszewski, M., et al., Chapter Five - Toxicity of Silver Nanomaterials in Higher Eukaryotes, in Advances in Molecular Toxicology, J.C. Fishbein, Editor. 2011, Elsevier. p. 179-218.
55. Zhao, H., et al., Mechanistic Understanding of the Engineered Nanomaterial-Induced Toxicity on Kidney. Journal of Nanomaterials, 2019. 2019: p. 2954853.
56. Liu, J., et al., The toxicology of ion-shedding zinc oxide nanoparticles. Crit Rev Toxicol, 2016. 46(4): p. 348-84.
57. Cohen, P.E., Meiosis Overview, in Encyclopedia of Reproduction (Second Edition), M.K. Skinner, Editor. 2018, Academic Press: Oxford. p. 5-12.
58. Eliopoulos, A.G., S. Havaki, and V.G. Gorgoulis, DNA Damage Response and Autophagy: A Meaningful Partnership. Frontiers in Genetics, 2016. 7.
59. OECD, Test No. 473: In vitro Mammalian Chromosome Aberration Test. 2016.
60. OECD, Test No. 487: In Vitro Mammalian Cell Micronucleus Test. 2016.
61. OECD, Test No. 489: In Vivo Mammalian Alkaline Comet Assay. 2016.
62. Edens, B.M., N. Miller, and Y.-C. Ma, Impaired Autophagy and Defective Mitochondrial Function: Converging Paths on the Road to Motor Neuron Degeneration. Frontiers in Cellular Neuroscience, 2016. 10.
63. Feng, X., et al., Nanomaterial-mediated autophagy: coexisting hazard and health benefits in biomedicine. Particle and Fibre Toxicology, 2020. 17(1): p. 53.
64. González-Rodríguez, Á., et al., Impaired autophagic flux is associated with increased endoplasmic reticulum stress during the development of NAFLD. Cell Death & Disease, 2014. 5(4): p. e1179-e1179.
65. Chakravorty, A., C.T. Jetto, and R. Manjithaya, Dysfunctional Mitochondria and Mitophagy as Drivers of Alzheimer’s Disease Pathogenesis. Frontiers in Aging Neuroscience, 2019. 11.
66. Oliveira, J.M.A., Mitochondrial Membrane Potential and Dynamics, in Mitochondrial Dysfunction in Neurodegenerative Disorders, A.K. Reeve, et al., Editors. 2012, Springer London: London. p. 127-139.
67. Zhang, Y., et al., Titanium dioxide nanoparticles induce proteostasis disruption and autophagy in human trophoblast cells. Chem Biol Interact, 2018. 296: p. 124-133.
68. Twig, G. and O.S. Shirihai, The interplay between mitochondrial dynamics and mitophagy. Antioxid Redox Signal, 2011. 14(10): p. 1939-51.
69. Chistiakov, D.A., et al., Mitochondrial Aging and Age-Related Dysfunction of Mitochondria. BioMed Research International, 2014. 2014: p. 238463.
70. Luo, C., et al., Mitochondrial accumulation under oxidative stress is due to defects in autophagy. Journal of Cellular Biochemistry, 2013. 114(1): p. 212-219.
71. Li, L., Y. Chen, and S.B. Gibson, Starvation-induced autophagy is regulated by mitochondrial reactive oxygen species leading to AMPK activation. Cell Signal, 2013. 25(1): p. 50-65.
72. Murugadoss, S., et al., A strategy towards the generation of testable adverse outcome pathways for nanomaterials. Altex, 2021. 38(4): p. 580-594.
73. Huang, H.J., et al., Current Strategies in Assessment of Nanotoxicity: Alternatives to In Vivo Animal Testing. Int J Mol Sci, 2021. 22(8).
74. Russell, W.M.S. and R.L. Burch, The principles of humane experimental technique. 1959: Methuen.
75. van Meerloo, J., G.J. Kaspers, and J. Cloos, Cell sensitivity assays: the MTT assay. Methods Mol Biol, 2011. 731: p. 237-45.
76. Reif, D., et al., Endocrine Profiling and Prioritization of Environmental Chemicals Using ToxCast Data. Environmental health perspectives, 2010. 118: p. 1714-20.
77. Zorova, L.D., et al., Mitochondrial membrane potential. Anal Biochem, 2018. 552: p. 50-59.
78. Ordureau, A., et al., Defining roles of PARKIN and ubiquitin phosphorylation by PINK1 in mitochondrial quality control using a ubiquitin replacement strategy. Proceedings of the National Academy of Sciences, 2015. 112(21): p. 6637-6642.
79. Muller, B., et al., Application of extracellular flux analysis for determining mitochondrial function in mammalian oocytes and early embryos. Scientific Reports, 2019. 9(1): p. 16778.
80. Chen, Y.-Y., et al., Skin damage induced by zinc oxide nanoparticles combined with UVB is mediated by activating cell pyroptosis via the NLRP3 inflammasome–autophagy–exosomal pathway. Particle and Fibre Toxicology, 2022. 19(1): p. 2.
81. Mizushima, N. and B. Levine, Autophagy in mammalian development and differentiation. Nat Cell Biol, 2010. 12(9): p. 823-30.
82. Tang, C., et al., Mitochondrial quality control in kidney injury and repair. Nature Reviews Nephrology, 2021. 17(5): p. 299-318.
83. Chan, N., et al., Broad activation of the ubiquitin–proteasome system by Parkin is critical for mitophagy. Human molecular genetics, 2011. 20: p. 1726-37.
84. Gatoo, M.A., et al., Physicochemical Properties of Nanomaterials: Implication in Associated Toxic Manifestations. BioMed Research International, 2014. 2014: p. 498420.
85. Marvel, S.W., et al., ToxPi Graphical User Interface 2.0: Dynamic exploration, visualization, and sharing of integrated data models. BMC Bioinformatics, 2018. 19(1): p. 80.
86. Shi, J., et al., Advances in genotoxicity of titanium dioxide nanoparticles in vivo and in vitro. NanoImpact, 2022. 25: p. 100377.
87. Yao, Y., et al., The Toxicity Of Metallic Nanoparticles On Liver: The Subcellular Damages, Mechanisms, And Outcomes. International Journal of Nanomedicine, 2019. Volume 14: p. 8787-8804.
88. Bouwmeester, H., et al., Characterization of translocation of silver nanoparticles and effects on whole-genome gene expression using an in vitro intestinal epithelium coculture model. ACS Nano, 2011. 5(5): p. 4091-103.
89. Kamiloglu, S., et al., Guidelines for cell viability assays. Food Frontiers, 2020. 1(3): p. 332-349.
90. Pujalte, I., et al., Cytotoxicity and oxidative stress induced by different metallic nanoparticles on human kidney cells. Particle and fibre toxicology, 2011. 8: p. 10.
91. Liou, G.Y. and P. Storz, Detecting reactive oxygen species by immunohistochemistry. Methods Mol Biol, 2015. 1292: p. 97-104.
92. Krylatov, A.V., et al., Reactive Oxygen Species as Intracellular Signaling Molecules in the Cardiovascular System. Curr Cardiol Rev, 2018. 14(4): p. 290-300.
93. An, P., et al., Hepatocyte mitochondria-derived danger signals directly activate hepatic stellate cells and drive progression of liver fibrosis. Nature Communications, 2020. 11(1): p. 2362.
94. Miranda, R.R., et al., Toxicological interactions of silver nanoparticles and non-essential metals in human hepatocarcinoma cell line. Toxicology in Vitro, 2017. 40: p. 134-143.
95. Zhang, L., et al., Redox signaling: Potential arbitrator of autophagy and apoptosis in therapeutic response. Free Radical Biology and Medicine, 2015. 89: p. 452-465.
96. Rodney, G.G., R. Pal, and R. Abo-Zahrah, Redox regulation of autophagy in skeletal muscle. Free Radic Biol Med, 2016. 98: p. 103-112.
97. Zhao, H., et al., Titanium Dioxide Nanoparticles Induce Mitochondrial Dynamic Imbalance and Damage in HT22 Cells. Journal of Nanomaterials, 2019. 2019: p. 4607531.
98. Wang, J., et al., ZnO nanoparticle-induced oxidative stress triggers apoptosis by activating JNK signaling pathway in cultured primary astrocytes. Nanoscale Research Letters, 2014. 9(1): p. 117.
99. Cho, H.M. and W. Sun, Molecular cross talk among the components of the regulatory machinery of mitochondrial structure and quality control. Experimental & Molecular Medicine, 2020. 52(5): p. 730-737.
100. Natarajan, V., et al., Titanium Dioxide Nanoparticles Trigger Loss of Function and Perturbation of Mitochondrial Dynamics in Primary Hepatocytes. PLOS ONE, 2015. 10(8): p. e0134541.
101. Guo, C., et al., Mitochondrial dysfunction, perturbations of mitochondrial dynamics and biogenesis involved in endothelial injury induced by silica nanoparticles. Environmental Pollution, 2018. 236: p. 926-936.
102. Giedt, R.J., et al., Mitochondrial Dynamics and Motility Inside Living Vascular Endothelial Cells: Role of Bioenergetics. Annals of Biomedical Engineering, 2012. 40(9): p. 1903-1916.
103. Yang, H., et al., Comparative study of cytotoxicity, oxidative stress and genotoxicity induced by four typical nanomaterials: the role of particle size, shape and composition. J Appl Toxicol, 2009. 29(1): p. 69-78.
104. Bhattacharya, D., et al., Differential toxicity of rod and spherical zinc oxide nanoparticles on human peripheral blood mononuclear cells. J Biomed Nanotechnol, 2014. 10(4): p. 707-16.
105. Fang, E.F., et al., Nuclear DNA damage signalling to mitochondria in ageing. Nature Reviews Molecular Cell Biology, 2016. 17(5): p. 308-321.
106. Stern, S.T., P.P. Adiseshaiah, and R.M. Crist, Autophagy and lysosomal dysfunction as emerging mechanisms of nanomaterial toxicity. Particle and Fibre Toxicology, 2012. 9(1): p. 20.
107. Popp, L., et al., Autophagic response to cellular exposure to titanium dioxide nanoparticles. Acta Biomater, 2018. 79: p. 354-363.
108. Chen, Y. and W. Dorn Gerald, PINK1-Phosphorylated Mitofusin 2 Is a Parkin Receptor for Culling Damaged Mitochondria. Science, 2013. 340(6131): p. 471-475.
109. Zhang, Y., et al., Mitochondria-targeted nanoparticles in treatment of neurodegenerative diseases. Exploration, 2021. 1(3): p. 20210115.

下載圖示 校內:2025-09-05公開
校外:2025-09-05公開
QR CODE