State of the Art on Toxicological Mechanisms of Metal and Metal Oxide Nanoparticles and Strategies to Reduce Toxicological Risks
Abstract
:1. Introduction
2. Preparation Methods
2.1. Laser Ablation
2.2. Spark Discharge
2.3. Evaporation/Condensation
2.4. Mechanical Milling
2.5. Electrochemical Synthesis
2.6. Chemical Reduction Method
2.7. Microwave-Assisted Synthesis
2.8. Green-Synthesis
| Synthesis Method | Advantages | Disadvantages |
|---|---|---|
| Laser ablation | Simple and effective Easy to modify nanoparticles properties by changing synthesis parameters | The laser path can be blocked by the portions of material released from the surface, causing reduction in ablation rate |
| Spark discharge | Cost-efficient Environmentally-friendly No impurities | Pure gas is required |
| Evaporation/condensation | Control of size No solvents used | High energy required |
| Mechanical milling | Work at low temperatures No solvent used | High energy required Time consuming method Contamination from milling media |
| Electrochemical synthesis | Simple, fast and inexpensive method Control of size and morphology of nanoparticles | Impurities from liquid media |
| Chemical reduction method | Simple and effective | Impurities from reaction Toxicity issues of reactive agents |
| Microwave-assisted synthesis | More efficient use of energy Higher production rates | Less homogeneity of nanoparticles size and morphology |
| Green synthesis | Eco-friendly Less toxicity Reduction of energy consumption | Use of natural sources Less effective than other methods |
3. Toxicity Mechanisms of Metal Nanoparticles
3.1. Silver Nanoparticles
3.2. Gold Nanoparticles
3.3. Copper/Copper Oxide Nanoparticles
3.4. Zinc/Zinc Oxide Nanoparticles
3.5. Iron Oxide Nanoparticles
4. Strategies to Reduce Metal Nanoparticles Toxicity
4.1. Surface Functionalization
4.2. Antibody Functionalization
4.3. Coating Modification
4.4. Morphology
4.4.1. Size
4.4.2. Shape
| Stragey Employed | Type of Metal NP | Functionalitzation Stragey | Physicochemical Characteritzation | In Vitro Studies | In Vivo Studies | References |
|---|---|---|---|---|---|---|
| Surface functionalization | Gold nanoparticles | PEG-SH and PG-NH2 groups | TEM/HRTEM, UV-Vis spectroscopy | Citotoxicity assay in SAOS-2 cell line cultivated in McCoy’s 5A medium with 15% heat-inactivated FBS, penicillin and streptomycin | ND | [134] |
| Surface functionalization | Zinc nanoparticles | PEG | FTIR | Citotoxicity assay in THP-1 immune cells | ND | [135] |
| Surface functionalization | Gold nanoparticles | Anionic ligands | ND | ND | ND | [137] |
| Antibody funtionalization | Gold coated magnetite | Antibody raanibizumab | SEM, DLS, XRD, TGA | Citotoxicity test by MTT assay | ND | [138] |
| Coating modification | Zinc nanoparticles | Silica coating | TEM, XPS, EDX, FTIR | Citotoxicity assessment in both colorectal epithelial cell lines (SW480 and DLD-1) | ND | [142] |
| Coating modification | Iron oxide nanoparticles | Silica coating | TEM, DLS and potential measurements | Citotoxicity assay in HeLa and A549 cells | ND | [143] |
| Coating modification | Silver nanoparticles | Silica coating | TEM and SEM images, optical absortion | Toxicity evaluation with E. coli bacteria | ND | [26] |
| Coating modification | Copper nanoparticles | Chitosan coating | XPS, XRD, TEM, DLS | Citotoxicity with human alveolar epithelial cell (A549) using standard MTS assay | In vivo study using mice by nasal administration to investigate inflammatory responses | [146] |
| Size modification | Silver Nanoparticles | ND | TEM, DLS, Z-potential | Citotoxicity study with murine peritoneal macrophage cell line (RAW 264.7) and L929 fibroblasts | ND | [149] |
| Size modification | Gold nanoparticles | ND | TEM, ICP-MS | In vitro study with HeLa cells by MTT assay | Mice intraperitoneal injection into BALB/C at a dose of 8 mg/kg/week | [151] |
| Shape modification | Gold nanoparticles | chitosan | HR Tem images | In vitro study into four cancer cell lines: AGS, HepG2, HT29, HeLa by MTT assay | ND | [156] |
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 2019, 12, 908–931. [Google Scholar] [CrossRef]
- Canaparo, R.; Foglietta, F.; Limongi, T.; Serpe, L. Biomedical applications of reactive oxygen species generation by metal nanoparticles. Materials 2021, 14, 53. [Google Scholar] [CrossRef] [PubMed]
- Sachin, K.; Karn, S.K. Microbial Fabricated Nanosystems: Applications in Drug Delivery and Targeting. Front. Chem. 2021, 9, 617353. [Google Scholar] [CrossRef]
- Gu, X.; Xu, Z.; Gu, L.; Xu, H.; Han, F.; Chen, B.; Pan, X. Preparation and antibacterial properties of gold nanoparticles: A review. Environ. Chem. Lett. 2021, 19, 167–187. [Google Scholar] [CrossRef]
- AlNadhari, S.; Al-Enazi, N.M.; Alshehrei, F.; Ameen, F. A review on biogenic synthesis of metal nanoparticles using marine algae and its applications. Environ. Res. 2021, 194, 110672. [Google Scholar] [CrossRef] [PubMed]
- Engin, A.B. Combined Toxicity of Metal Nanoparticles: Comparison of Individual and Mixture Particles Effect. In Protein Kinase-mediated Decisions Between Life and Death; Engin, A.B., Engin, A., Eds.; Springer International Publishing: Cham, Switzerland, 2021; pp. 165–193. ISBN 978-3-030-49844-3. [Google Scholar]
- Turan, N.B.; Erkan, H.S.; Engin, G.O.; Bilgili, M.S. Nanoparticles in the aquatic environment: Usage, properties, transformation and toxicity—A review. Process Saf. Environ. Prot. 2019, 130, 238–249. [Google Scholar] [CrossRef]
- Pinzaru, I.; Coricovac, D.; Dehelean, C.; Moacă, E.A.; Mioc, M.; Baderca, F.; Sizemore, I.; Brittle, S.; Marti, D.; Calina, C.D.; et al. Stable PEG-coated silver nanoparticles—A comprehensive toxicological profile. Food Chem. Toxicol. 2018, 111, 546–556. [Google Scholar] [CrossRef]
- Parsai, T.; Kumar, A. Weight-of-evidence process for assessing human health risk of mixture of metal oxide nanoparticles and corresponding ions in aquatic matrices. Chemosphere 2021, 263, 128289. [Google Scholar] [CrossRef] [PubMed]
- De Oliveira, P.F.M.; Torresi, R.M.; Emmerling, F.; Camargo, P.H.C. Challenges and opportunities in the bottom-up mechanochemical synthesis of noble metal nanoparticles. J. Mater. Chem. A 2020, 8, 16114–16141. [Google Scholar] [CrossRef]
- Isaacoff, B.P.; Brown, K.A. Progress in Top-Down Control of Bottom-Up Assembly. Nano Lett. 2017, 17, 6508–6510. [Google Scholar] [CrossRef]
- Kargozar, S.; Mozafari, M. Nanotechnology and Nanomedicine: Start small, think big. Mater. Today Proc. 2018, 5, 15492–15500. [Google Scholar] [CrossRef]
- Zhang, J.; Claverie, J.; Chaker, M.; Ma, D. Colloidal Metal Nanoparticles Prepared by Laser Ablation and their Applications. ChemPhysChem 2017, 18, 986–1006. [Google Scholar] [CrossRef] [PubMed]
- Alhamid, M.Z.; Hadi, B.S.; Khumaeni, A. Synthesis of silver nanoparticles using laser ablation method utilizing Nd:YAG laser. In AIP Conference Proceedings; AIP Publishing LLC: Melville, NY, USA, 2019; Volume 2202, p. 020013. [Google Scholar] [CrossRef]
- Vahabzadeh, E.; Torkamany, M.J. Iron Oxide Nanocrystals Synthesis by Laser Ablation in Water: Effect of Laser Wavelength. J. Clust. Sci. 2014, 25, 959–968. [Google Scholar] [CrossRef]
- Kim, M.; Osone, S.; Kim, T.; Higashi, H.; Seto, T. Synthesis of nanoparticles by laser ablation: A review. KONA Powder Part. J. 2017, 2017, 80–90. [Google Scholar] [CrossRef] [Green Version]
- Al-Nassar, S.I.; Hussein, F.I.; Ma, A.K. The effect of laser pulse energy on ZnO nanoparticles formation by liquid phase pulsed laser ablation. J. Mater. Res. Technol. 2019, 8, 4026–4031. [Google Scholar] [CrossRef]
- Amendola, V.; Polizzi, S.; Meneghetti, M. Laser ablation synthesis of gold nanoparticles in organic solvents. J. Phys. Chem. B 2006, 110, 7232–7237. [Google Scholar] [CrossRef]
- Boutinguiza, M.; Comesaña, R.; Lusquiños, F.; Riveiro, A.; Del Val, J.; Pou, J. Production of silver nanoparticles by laser ablation in open air. Appl. Surf. Sci. 2015, 336, 108–111. [Google Scholar] [CrossRef]
- Tabrizi, N.S.; Ullmann, M.; Vons, V.A.; Lafont, U.; Schmidt-Ott, A. Generation of nanoparticles by spark discharge. J. Nanoparticle Res. 2009, 11, 315–332. [Google Scholar] [CrossRef] [Green Version]
- Singh, A.; Ghosh, A. A thermo-electric model of material removal during electric discharge machining. Int. J. Mach. Tools Manuf. 1999, 39, 669–682. [Google Scholar] [CrossRef]
- Messing, M.E.; Dick, K.A.; Wallenberg, L.R.; Deppert, K. Generation of size-selected gold nanoparticles by spark discharge—For growth of epitaxial nanowires. Gold Bull. 2009, 42, 20–26. [Google Scholar] [CrossRef] [Green Version]
- Oh, H.-C.; Jung, J.-H.; Park, H.-H.; Ji, J.-H.; Kim, S.-S. Generation of Silver Nanoparticles by Spark Discharge Aerosol Generator Using Air as a Carrier Gas. Trans. Korean Soc. Mech. Eng. B 2006, 30, 170–176. [Google Scholar] [CrossRef]
- Tabrizi, N.S.; Xu, Q.; Van Der Pers, N.M.; Schmidt-Ott, A. Generation of mixed metallic nanoparticles from immiscible metals by spark discharge. J. Nanoparticle Res. 2010, 12, 247–259. [Google Scholar] [CrossRef]
- Harra, J.; Mäkitalo, J.; Siikanen, R.; Virkki, M.; Genty, G.; Kobayashi, T.; Kauranen, M.; Mäkelä, J.M. Size-controlled aerosol synthesis of silver nanoparticles for plasmonic materials. J. Nanoparticle Res. 2012, 14, 870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sotiriou, G.A.; Sannomiya, T.; Teleki, A.; Krumeich, F.; Vörös, J.; Pratsinis, S.E. Non-toxic dry-coated nanosilver for plasmonic biosensors. Adv. Funct. Mater. 2010, 20, 4250–4257. [Google Scholar] [CrossRef] [Green Version]
- Prasad Yadav, T.; Manohar Yadav, R.; Pratap Singh, D. Mechanical Milling: A Top Down Approach for the Synthesis of Nanomaterials and Nanocomposites. Nanosci. Nanotechnol. 2012, 2, 22–48. [Google Scholar] [CrossRef] [Green Version]
- Mancillas-Salas, S.; Hernández-Rodríguez, P.; Reynosa-Martínez, A.C.; López-Honorato, E. Production of aluminum nanoparticles by wet mechanical milling. MRS Adv. 2020, 5, 3133–3140. [Google Scholar] [CrossRef]
- Arbain, R.; Othman, M.; Palaniandy, S. Preparation of iron oxide nanoparticles by mechanical milling. Miner. Eng. 2011, 24, 1–9. [Google Scholar] [CrossRef]
- Saravanan, A.; Kumar, P.S.; Karishma, S.; Vo, D.V.N.; Jeevanantham, S.; Yaashikaa, P.R.; George, C.S. A review on biosynthesis of metal nanoparticles and its environmental applications. Chemosphere 2021, 264, 128580. [Google Scholar] [CrossRef]
- Jeun, Y.E.; Baek, B.; Lee, M.W.; Ahn, H.S. Surfactant-free electrochemical synthesis of metallic nanoparticles via stochastic collisions of aqueous nanodroplet reactors. Chem. Commun. 2018, 54, 10052–10055. [Google Scholar] [CrossRef] [PubMed]
- McDarby, S.P.; Wang, C.J.; King, M.E.; Personick, M.L. An Integrated Electrochemistry Approach to the Design and Synthesis of Polyhedral Noble Metal Nanoparticles. J. Am. Chem. Soc. 2020, 142, 21322–21335. [Google Scholar] [CrossRef]
- Therese, G.H.A.; Kamath, P.V. Electrochemical synthesis of metal oxides and hydroxides. Chem. Mater. 2000, 12, 1195–1204. [Google Scholar] [CrossRef]
- Yanilkin, V.V.; Nasretdinova, G.R.; Kokorekin, V.A. Mediated electrochemical synthesis of metal nanoparticles. Russ. Chem. Rev. 2018, 87, 1080–1110. [Google Scholar] [CrossRef]
- Khandel, P.; Yadaw, R.K.; Soni, D.K.; Kanwar, L.; Shahi, S.K. Biogenesis of Metal Nanoparticles and Their Pharmacological Applications: Present Status and Application Prospects. J. Nanostruct. Chem. 2018, 12, 217–254. [Google Scholar] [CrossRef] [Green Version]
- Khan, Z.; Al-Thabaiti, S.A.; Obaid, A.Y.; Al-Youbi, A.O. Preparation and characterization of silver nanoparticles by chemical reduction method. Colloids Surf. B Biointerfaces 2011, 82, 513–517. [Google Scholar] [CrossRef]
- Lidor-Shalev, O.; Zitoun, D. Reaction mechanism of “amine-borane route” towards Sn, Ni, Pd, Pt nanoparticles. RSC Adv. 2014, 4, 63603–63610. [Google Scholar] [CrossRef]
- Pelletier, F.; Ciuculescu, D.; Mattei, J.G.; Lecante, P.; Casanove, M.J.; Yaacoub, N.; Greneche, J.M.; Schmitz-Antoniak, C.; Amiens, C. On the use of amine-borane complexes to synthesize iron nanoparticles. Chem. Eur. J. 2013, 19, 6021–6026. [Google Scholar] [CrossRef] [PubMed]
- Sanyal, U.; Jagirdar, B.R. Metal and alloy nanoparticles by amine-borane reduction of metal salts by solid-phase synthesis: Atom economy and green process. Inorg. Chem. 2012, 51, 13023–13033. [Google Scholar] [CrossRef] [PubMed]
- Kalidindi, S.B.; Sanyal, U.; Jagirdar, B.R. Chemical synthesis of metal nanoparticles using amine-boranes. ChemSusChem 2011, 4, 317–324. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, T.D. From formation mechanisms to synthetic methods toward shape-controlled oxide nanoparticles. Nanoscale 2013, 5, 9455–9482. [Google Scholar] [CrossRef]
- Carroll, K.J.; Reveles, J.U.; Shultz, M.D.; Khanna, S.N.; Carpenter, E.E. Preparation of elemental Cu and Ni nanoparticles by the polyol method: An experimental and theoretical approach. J. Phys. Chem. C 2011, 115, 2656–2664. [Google Scholar] [CrossRef]
- Mahamuni, P.P.; Patil, P.M.; Dhanavade, M.J.; Badiger, M.V.; Shadija, P.G.; Lokhande, A.C.; Bohara, R.A. Synthesis and characterization of zinc oxide nanoparticles by using polyol chemistry for their antimicrobial and antibiofilm activity. Biochem. Biophys. Rep. 2019, 17, 71–80. [Google Scholar] [CrossRef] [PubMed]
- Ungelenk, J.; Speldrich, M.; Dronskowski, R.; Feldmann, C. Polyol-mediated low-temperature synthesis of crystalline tungstate nanoparticles MWO4 (M = Mn, Fe, Co, Ni, Cu, Zn). Solid State Sci. 2014, 31, 62–69. [Google Scholar] [CrossRef]
- Favier, I.; Pla, D.; Gómez, M. Palladium Nanoparticles in Polyols: Synthesis, Catalytic Couplings, and Hydrogenations. Chem. Rev. 2020, 120, 1146–1183. [Google Scholar] [CrossRef]
- Nüchter, M.; Ondruschka, B.; Bonrath, W.; Gum, A. Microwave assisted synthesis—A critical technology overview. Green Chem. 2004, 6, 128–141. [Google Scholar] [CrossRef]
- Blosi, M.; Albonetti, S.; Dondi, M.; Martelli, C.; Baldi, G. Microwave-assisted polyol synthesis of Cu nanoparticles. J. Nanoparticle Res. 2011, 13, 127–138. [Google Scholar] [CrossRef]
- Li, D.; Komarneni, S. Microwave-assisted polyol process for synthesis of Ni nanoparticles. J. Am. Ceram. Soc. 2006, 89, 1510–1517. [Google Scholar] [CrossRef]
- Jadoun, S.; Arif, R.; Jangid, N.K.; Meena, R.K. Green synthesis of nanoparticles using plant extracts: A review. Environ. Chem. Lett. 2021, 19, 355–374. [Google Scholar] [CrossRef]
- Salem, S.S.; Fouda, A. Green Synthesis of Metallic Nanoparticles and Their Prospective Biotechnological Applications: An Overview. Biol. Trace Elem. Res. 2021, 199, 344–370. [Google Scholar] [CrossRef] [PubMed]
- Singh, J.; Dutta, T.; Kim, K.H.; Rawat, M.; Samddar, P.; Kumar, P. “Green” synthesis of metals and their oxide nanoparticles: Applications for environmental remediation. J. Nanobiotechnol. 2018, 16, 84. [Google Scholar] [CrossRef]
- Nadaroglu, H.; Alayli, A.; Nadaroğlu, H.; Alayli Güngör, A.; İnce, S. Synthesis of Nanoparticles by Green Synthesis Method. Int. J. Innov. Res. Rev. 2017, 1, 6–9. [Google Scholar]
- Nasrollahzadeh, M.; Ghorbannezhad, F.; Issaabadi, Z.; Sajadi, S.M. Recent Developments in the Biosynthesis of Cu-Based Recyclable Nanocatalysts Using Plant Extracts and their Application in the Chemical Reactions. Chem. Rec. 2019, 19, 601–643. [Google Scholar] [CrossRef]
- Iravani, S. Green synthesis of metal nanoparticles using plants. Green Chem. 2011, 13, 2638–2650. [Google Scholar] [CrossRef]
- Kuppusamy, P.; Yusoff, M.M.; Maniam, G.P.; Govindan, N. Biosynthesis of metallic nanoparticles using plant derivatives and their new avenues in pharmacological applications—An updated report. Saudi Pharm. J. 2016, 24, 473–484. [Google Scholar] [CrossRef]
- Dauthal, P.; Mukhopadhyay, M. Noble Metal Nanoparticles: Plant-Mediated Synthesis, Mechanistic Aspects of Synthesis, and Applications. Ind. Eng. Chem. Res. 2016, 55, 9557–9577. [Google Scholar] [CrossRef]
- Sánchez-López, E.; Gomes, D.; Esteruelas, G.; Bonilla, L.; Lopez-Machado, A.L.; Galindo, R.; Cano, A.; Espina, M.; Ettcheto, M.; Camins, A.; et al. Metal-based nanoparticles as antimicrobial agents: An overview. Nanomaterials 2020, 10, 292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sengul, A.B.; Asmatulu, E. Toxicity of metal and metal oxide nanoparticles: A review. Environ. Chem. Lett. 2020, 18, 1659–1683. [Google Scholar] [CrossRef]
- Pujalté, I.; Passagne, I.; Daculsi, R.; De Portal, C.; Ohayon-Courtès, C.; L’Azou, B. Cytotoxic effects and cellular oxidative mechanisms of metallic nanoparticles on renal tubular cells: Impact of particle solubility. Toxicol. Res. 2015, 4, 409–422. [Google Scholar] [CrossRef]
- Lozano, T.; Rey, M.; Rojas, E.; Moya, S.; Fleddermann, J.; Estrela-Lopis, I.; Donath, E.; Wang, B.; Mao, Z.; Gao, C.; et al. Cytotoxicity effects of metal oxide nanoparticles in human tumor cell lines. J. Phys. Conf. Ser. 2011, 304, 012046. [Google Scholar] [CrossRef] [Green Version]
- Taniyama, Y.; Griendling, K.K. Reactive Oxygen Species in the Vasculature: Molecular and Cellular Mechanisms. Hypertension 2003, 42, 1075–1081. [Google Scholar] [CrossRef] [Green Version]
- Volpe, C.M.O.; Villar-Delfino, P.H.; Dos Anjos, P.M.F.; Nogueira-Machado, J.A. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death Dis. 2018, 9, 119. [Google Scholar] [CrossRef]
- Prasad, S.; Gupta, S.C.; Tyagi, A.K. Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett. 2017, 387, 95–105. [Google Scholar] [CrossRef] [PubMed]
- Rice-Evans, C.A. Formation of free radicals and mechanisms of action in normal biochemical processes and pathological states. New Compr. Biochem. 1994, 28, 131–153. [Google Scholar] [CrossRef]
- Greenberg, M.E.; Li, X.M.; Gugiu, B.G.; Gu, X.; Qin, J.; Salomon, R.G.; Hazen, S.L. The lipid whisker model of the structure of oxidized cell membranes. J. Biol. Chem. 2008, 283, 2385–2396. [Google Scholar] [CrossRef] [Green Version]
- Klaunig, J.E.; Kamendulis, L.M.; Hocevar, B.A. Oxidative stress and oxidative damage in carcinogenesis. Toxicol. Pathol. 2010, 38, 96–109. [Google Scholar] [CrossRef] [Green Version]
- Oberley, T.D. Commentary Oxidative Damage and Cancer. Am. J. Pathol. 2002, 160, 403–408. [Google Scholar] [CrossRef] [Green Version]
- Liu, G.; Zou, H.; Luo, T.; Long, M.; Bian, J.; Liu, X.; Gu, J.; Yuan, Y.; Song, R.; Wang, Y.; et al. Caspase-dependent and caspase-independent pathways are involved in cadmium-induced apoptosis in primary rat proximal tubular cell culture. PLoS ONE 2016, 11, e0166823. [Google Scholar] [CrossRef] [Green Version]
- Brentnall, M.; Rodriguez-Menocal, L.; De Guevara, R.L.; Cepero, E.; Boise, L.H. Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 2013, 14, 32. [Google Scholar] [CrossRef] [Green Version]
- Vazquez-Muñoz, R.; Borrego, B.; Juárez-Moreno, K.; García-García, M.; Mota Morales, J.D.; Bogdanchikova, N.; Huerta-Saquero, A. Toxicity of silver nanoparticles in biological systems: Does the complexity of biological systems matter? Toxicol. Lett. 2017, 276, 11–20. [Google Scholar] [CrossRef]
- Smith, J.N.; Thomas, D.G.; Jolley, H.; Kodali, V.K.; Littke, M.H.; Munusamy, P.; Baer, D.R.; Gaffrey, M.J.; Thrall, B.D.; Teeguarden, J.G. All that is silver is not toxic: Silver ion and particle kinetics reveals the role of silver ion aging and dosimetry on the toxicity of silver nanoparticles. Part. Fibre Toxicol. 2018, 15, 47. [Google Scholar] [CrossRef] [PubMed]
- Cho, Y.M.; Mizuta, Y.; Akagi, J.I.; Toyoda, T.; Sone, M.; Ogawa, K. Size-dependent acute toxicity of silver nanoparticles in mice. J. Toxicol. Pathol. 2018, 31, 73–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hadrup, N.; Sharma, A.K.; Loeschner, K. Toxicity of silver ions, metallic silver, and silver nanoparticle materials after in vivo dermal and mucosal surface exposure: A review. Regul. Toxicol. Pharmacol. 2018, 98, 257–267. [Google Scholar] [CrossRef] [Green Version]
- Greulich, C.; Braun, D.; Peetsch, A.; Diendorf, J.; Siebers, B.; Epple, M.; Köller, M. The toxic effect of silver ions and silver nanoparticles towards bacteria and human cells occurs in the same concentration range. RSC Adv. 2012, 2, 6981–6987. [Google Scholar] [CrossRef]
- Gaillet, S.; Rouanet, J.M. Silver nanoparticles: Their potential toxic effects after oral exposure and underlying mechanisms—A review. Food Chem. Toxicol. 2015, 77, 58–63. [Google Scholar] [CrossRef]
- Asharani, P.V.; Lian Wu, Y.; Gong, Z.; Valiyaveettil, S. Toxicity of silver nanoparticles in zebrafish models. Nanotechnology 2008, 19, 255102. [Google Scholar] [CrossRef]
- DiFonzo, N.; Bordia, P. Reproduced with permission of the copyright owner. Further reproduction prohibited without. J. Allergy Clin. Immunol. 1998, 130, 556. [Google Scholar]
- Carlson, C.; Hussein, S.M.; Schrand, A.M.; Braydich-Stolle, L.K.; Hess, K.L.; Jones, R.L.; Schlager, J.J. Unique cellular interaction of silver nanoparticles: Size-dependent generation of reactive oxygen species. J. Phys. Chem. B 2008, 112, 13608–13619. [Google Scholar] [CrossRef] [PubMed]
- Almofti, M.R.; Ichikawa, T.; Yamashita, K.; Terada, H.; Shinohara, Y. Silver ion induces a cyclosporine A-insensitive permeability transition in rat liver mitochondria and release of apoptogenic cytochrome c. J. Biochem. 2003, 134, 43–49. [Google Scholar] [CrossRef]
- Hwang, M.G.; Katayama, H.; Ohgaki, S. Inactivation of Legionella pneumophila and Pseudomonas aeruginosa: Evaluation of the bactericidal ability of silver cations. Water Res. 2007, 41, 4097–4104. [Google Scholar] [CrossRef]
- Uygur, B.; Craig, G.; Mason, M.D.; Ng, A.K. Cytotoxicity and genotoxicity of silver nanomaterials. NSTI Nanotechnol. 2009, 2, 383–386. [Google Scholar]
- Chi, Z.; Liu, R.; Zhao, L.; Qin, P.; Pan, X.; Sun, F.; Hao, X. A new strategy to probe the genotoxicity of silver nanoparticles combined with cetylpyridine bromide. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2009, 72, 577–581. [Google Scholar] [CrossRef]
- Kumari, M.; Mukherjee, A.; Chandrasekaran, N. Genotoxicity of silver nanoparticles in Allium cepa. Sci. Total Environ. 2009, 407, 5243–5246. [Google Scholar] [CrossRef]
- Wang, X.; Ji, Z.; Chang, C.H.; Zhang, H.; Wang, M.; Liao, Y.P.; Lin, S.; Meng, H.; Li, R.; Sun, B.; et al. Use of coated silver nanoparticles to understand the relationship of particle dissolution and bioavailability to cell and lung toxicological potential. Small 2014, 10, 385–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gliga, A.R.; Di Bucchianico, S.; Lindvall, J.; Fadeel, B.; Karlsson, H.L. RNA-sequencing reveals long-term effects of silver nanoparticles on human lung cells. Sci. Rep. 2018, 8, 6668. [Google Scholar] [CrossRef]
- Gliga, A.R.; Skoglund, S.; Wallinder, I.O.; Fadeel, B.; Karlsson, H.L. Size-dependent cytotoxicity of silver nanoparticles in human lung cells: The role of cellular uptake, agglomeration and Ag release. Part. Fibre Toxicol. 2014, 11, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samberg, M.E.; Oldenburg, S.J.; Monteiro-Riviere, N.A. Evaluation of silver nanoparticle toxicity in skin in vivo and keratinocytes in vitro. Environ. Health Perspect. 2010, 118, 407–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hackenberg, S.; Scherzed, A.; Kessler, M.; Hummel, S.; Technau, A.; Froelich, K.; Ginzkey, C.; Koehler, C.; Hagen, R.; Kleinsasser, N. Silver nanoparticles: Evaluation of DNA damage, toxicity and functional impairment in human mesenchymal stem cells. Toxicol. Lett. 2011, 201, 27–33. [Google Scholar] [CrossRef]
- Ghosh, P.; Han, G.; De, M.; Kim, C.K.; Rotello, V.M. Gold nanoparticles in delivery applications. Adv. Drug Deliv. Rev. 2008, 60, 1307–1315. [Google Scholar] [CrossRef]
- Paciotti, G.F.; Myer, L.; Weinreich, D.; Goia, D.; Pavel, N.; McLaughlin, R.E.; Tamarkin, L. Colloidal gold: A novel nanoparticle vector for tumor directed drug delivery. Drug Deliv. J. Deliv. Target. Ther. Agents 2004, 11, 169–183. [Google Scholar] [CrossRef]
- Durr, N.J.; Larson, T.; Smith, D.K.; Korgel, B.A.; Sokolov, K.; Ben-yakar, A. Two-Photon Luminescence Imaging of Cancer Cells Using Molecularly Targeted Gold Nanorods. Nano Lett. 2007, 7, 941–945. [Google Scholar] [CrossRef]
- Li, J.; Hu, J.; Wang, Z. Gold Nanoparticles With Special Shapes: Controlled Synthesis, Surface-enhanced Raman Scattering, and The Application in Biodetection. Sensors 2007, 7, 3299–3311. [Google Scholar]
- Goodman, C.M.; McCusker, C.D.; Yilmaz, T.; Rotello, V.M. Toxicity of gold nanoparticles functionalized with cationic and anionic side chains. Bioconjug. Chem. 2004, 15, 897–900. [Google Scholar] [CrossRef]
- Schaeublin, N.M.; Braydich-Stolle, L.K.; Schrand, A.M.; Miller, J.M.; Hutchison, J.; Schlager, J.J.; Hussain, S.M. Surface charge of gold nanoparticles mediates mechanism of toxicity. Nanoscale 2011, 3, 410–420. [Google Scholar] [CrossRef] [PubMed]
- Connor, E.E.; Mwamuka, J.; Gole, A.; Murphy, C.J.; Wyatt, M.D. Gold nanoparticles are taken up by human cells but do not cause acute cytotoxicity. Small 2005, 1, 325–327. [Google Scholar] [CrossRef]
- Jia, H.Y.; Liu, Y.; Zhang, X.J.; Han, L.; Du, L.B.; Tian, Q.; Xu, Y.C. Potential oxidative stress of gold nanoparticles by induced-NO releasing in serum. J. Am. Chem. Soc. 2009, 131, 40–41. [Google Scholar] [CrossRef] [PubMed]
- Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef] [Green Version]
- Li, J.J.; Zou, L.; Hartono, D.; Ong, C.N.; Bay, B.H.; Yung, L.Y.L. Gold nanoparticles induce oxidative damage in lung fibroblasts in vitro. Adv. Mater. 2008, 20, 138–142. [Google Scholar] [CrossRef]
- Chaicherd, S.; Killingsworth, M.C.; Pissuwan, D. Toxicity of gold nanoparticles in a commercial dietary supplement drink on connective tissue fibroblast cells. SN Appl. Sci. 2019, 1, 336. [Google Scholar] [CrossRef] [Green Version]
- Li, T.; Albee, B.; Alemayehu, M.; Diaz, R.; Ingham, L.; Kamal, S.; Rodriguez, M.; Whaley Bishnoi, S. Comparative toxicity study of Ag, Au, and Ag-Au bimetallic nanoparticles on Daphnia magna. Anal. Bioanal. Chem. 2010, 398, 689–700. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Wu, Y.; Jin, S.; Tian, Y.; Zhang, X.; Zhao, Y.; Yu, L.; Liang, X.J. Gold nanoparticles induce autophagosome accumulation through size-dependent nanoparticle uptake and lysosome impairment. ACS Nano 2011, 5, 8629–8639. [Google Scholar] [CrossRef]
- Li, S.; Zhang, C.; Cao, W.; Ma, B.; Ma, X.; Jin, S.; Zhang, J.; Wang, P.C.; Li, F.; Liang, X.J. Anchoring effects of surface chemistry on gold nanorods: Modulating autophagy. J. Mater. Chem. B 2015, 3, 3324–3330. [Google Scholar] [CrossRef] [PubMed]
- Lim, S.M.; Mohamad Hanif, E.A.; Chin, S.F. Is targeting autophagy mechanism in cancer a good approach? The possible double-edge sword effect. Cell Biosci. 2021, 11, 56. [Google Scholar] [CrossRef]
- Pérez-Hernández, M.; Arias, A.; Martínez-García, D.; Pérez-Tomás, R.; Quesada, R.; Soto-Cerrato, V. Targeting Autophagy for Cancer Treatment and Tumor Chemosensitization. Cancers 2019, 11, 1599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sahoo, S.K.; Panyam, J.; Prabha, S.; Labhasetwar, V. Residual polyvinyl alcohol associated with poly (D,L-lactide-co-glycolide) nanoparticles affects their physical properties and cellular uptake. J. Control. Release 2002, 82, 105–114. [Google Scholar] [CrossRef]
- Forouhar Vajargah, M.; Mohamadi Yalsuyi, A.; Hedayati, A.; Faggio, C. Histopathological lesions and toxicity in common carp (Cyprinus carpio L. 1758) induced by copper nanoparticles. Microsc. Res. Tech. 2018, 81, 724–729. [Google Scholar] [CrossRef] [PubMed]
- Ameh, T.; Sayes, C.M. The potential exposure and hazards of copper nanoparticles: A review. Environ. Toxicol. Pharmacol. 2019, 71, 103220. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, A.; Das, J.; Manna, P.; Sil, P.C. Nano-copper induces oxidative stress and apoptosis in kidney via both extrinsic and intrinsic pathways. Toxicology 2011, 290, 208–217. [Google Scholar] [CrossRef] [PubMed]
- Edlich, F. BCL-2 proteins and apoptosis: Recent insights and unknowns. Biochem. Biophys. Res. Commun. 2018, 500, 26–34. [Google Scholar] [CrossRef] [PubMed]
- Assadian, E.; Zarei, M.H.; Gilani, A.G.; Farshin, M.; Degampanah, H.; Pourahmad, J. Toxicity of Copper Oxide (CuO) Nanoparticles on Human Blood Lymphocytes. Biol. Trace Elem. Res. 2018, 184, 350–357. [Google Scholar] [CrossRef]
- Siddiqui, M.A.; Alhadlaq, H.A.; Ahmad, J.; Al-Khedhairy, A.A.; Musarrat, J.; Ahamed, M. Copper Oxide Nanoparticles Induced Mitochondria Mediated Apoptosis in Human Hepatocarcinoma Cells. PLoS ONE 2013, 8, e69534. [Google Scholar] [CrossRef] [Green Version]
- Doudi, M.; Setorki, M. Acute effect of nano-copper on liver tissue and function in rat Acute effect of nano-copper on liver tissue and function in rat ffect of nano-copper on function and tissue liver of rat E. Nanomed. J. 2015, 1, 331–338. [Google Scholar]
- Lei, R.; Wu, C.; Yang, B.; Ma, H.; Shi, C.; Wang, Q.; Wang, Q.; Yuan, Y.; Liao, M. Integrated metabolomic analysis of the nano-sized copper particle-induced hepatotoxicity and nephrotoxicity in rats: A rapid in vivo screening method for nanotoxicity. Toxicol. Appl. Pharmacol. 2008, 232, 292–301. [Google Scholar] [CrossRef] [PubMed]
- Singh, S. Zinc oxide nanoparticles impacts: Cytotoxicity, genotoxicity, developmental toxicity, and neurotoxicity. Toxicol. Mech. Methods 2019, 29, 300–311. [Google Scholar] [CrossRef] [PubMed]
- Pasquet, J.; Chevalier, Y.; Pelletier, J.; Couval, E.; Bouvier, D.; Bolzinger, M.A. The contribution of zinc ions to the antimicrobial activity of zinc oxide. Colloids Surf. A Physicochem. Eng. Asp. 2014, 457, 263–274. [Google Scholar] [CrossRef]
- Singh, N.; Manshian, B.; Jenkins, G.J.S.; Griffiths, S.M.; Williams, P.M.; Maffeis, T.G.G.; Wright, C.J.; Doak, S.H. NanoGenotoxicology: The DNA damaging potential of engineered nanomaterials. Biomaterials 2009, 30, 3891–3914. [Google Scholar] [CrossRef] [PubMed]
- Song, W.; Zhang, J.; Guo, J.; Zhang, J.; Ding, F.; Li, L.; Sun, Z. Role of the dissolved zinc ion and reactive oxygen species in cytotoxicity of ZnO nanoparticles. Toxicol. Lett. 2010, 199, 389–397. [Google Scholar] [CrossRef] [PubMed]
- Tang, Y.; Xin, H.; Yang, S.; Guo, M.; Malkoske, T.; Yin, D.; Xia, S. Environmental risks of ZnO nanoparticle exposure on Microcystis aeruginosa: Toxic effects and environmental feedback. Aquat. Toxicol. 2018, 204, 19–26. [Google Scholar] [CrossRef] [PubMed]
- Lai, X.; Wei, Y.; Zhao, H.; Chen, S.; Bu, X.; Lu, F.; Qu, D.; Yao, L.; Zheng, J.; Zhang, J. The effect of Fe2O3 and ZnO nanoparticles on cytotoxicity and glucose metabolism in lung epithelial cells. J. Appl. Toxicol. 2015, 35, 651–664. [Google Scholar] [CrossRef]
- Pasupuleti, S.; Alapati, S.; Ganapathy, S.; Anumolu, G.; Pully, N.R.; Prakhya, B.M. Toxicity of zinc oxide nanoparticles through oral route. Toxicol. Ind. Health 2012, 28, 675–686. [Google Scholar] [CrossRef]
- Mohammed, L.; Gomaa, H.G.; Ragab, D.; Zhu, J. Magnetic nanoparticles for environmental and biomedical applications: A review. Particuology 2017, 30, 1–14. [Google Scholar] [CrossRef]
- Ameta, R.; Chohadia, A.K.; Jain, A.; Punjabi, P.B. Chapter 3—Fenton and Photo-Fenton Processes. In Advanced Oxidation Processes for Waste Water Treatment; Ameta, S.C., Ameta, R., Eds.; Academic Press: Cambridge, MA, USA; Elsevier: Amsterdam, The Netherlands, 2018; pp. 49–87. [Google Scholar] [CrossRef]
- Wang, S.; Luo, J.; Zhang, Z.; Dong, D.; Shen, Y.; Fang, Y.; Hu, L.; Liu, M.; Dai, C.; Peng, S.; et al. Iron and magnetic: New research direction of the ferroptosis-based cancer therapy. Am. J. Cancer Res. 2018, 8, 1933–1946. [Google Scholar]
- Naqvi, S.; Samim, M.; Abdin, M.Z.; Ahmed, F.J.; Maitra, A.N.; Prashant, C.K.; Dinda, A.K. Concentration-dependent toxicity of iron oxide nanoparticles mediated by increased oxidative stress. Int. J. Nanomed. 2010, 5, 983–989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ran, Q.; Xiang, Y.; Liu, Y.; Xiang, L.; Li, F.; Deng, X.; Xiao, Y.; Chen, L.; Chen, L.; Li, Z. Eryptosis Indices as a Novel Predictive Parameter for Biocompatibility of Fe3O4 Magnetic Nanoparticles on Erythrocytes. Sci. Rep. 2015, 5, 16209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soenen, S.J.H.; Nuytten, N.; De Meyer, S.F.; De Smedt, S.C.; De Cuyper, M. High intracellular iron oxide nanoparticle concentrations affect cellular cytoskeleton and focal adhesion kinase-mediated signaling. Small 2010, 6, 832–842. [Google Scholar] [CrossRef] [PubMed]
- Morris, J.B.; Olzinski, A.R.; Bernard, R.E.; Aravindhan, K.; Mirabile, R.C.; Boyce, R.; Willette, R.N.; Jucker, B.M. p38 MAPK inhibition reduces aortic ultrasmall superparamagnetic iron oxide uptake in a mouse model of atherosclerosis: MRI assessment. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 265–271. [Google Scholar] [CrossRef] [Green Version]
- Shubayev, V.I.; Pisanic, T.R.; Jin, S. Magnetic nanoparticles for theragnostics. Adv. Drug Deliv. Rev. 2009, 61, 467–477. [Google Scholar] [CrossRef] [Green Version]
- Buyukhatipoglu, K.; Clyne, A.M. Superparamagnetic iron oxide nanoparticles change endothelial cell morphology and mechanics via reactive oxygen species formation. J. Biomed. Mater. Res. Part A 2011, 96, 186–195. [Google Scholar] [CrossRef]
- Sánchez-López, E.; Esteruelas, G.; Ortiz, A.; Espina, M.; Prat, J.; Muñoz, M.; Cano, A.; Calpena, A.C.; Ettcheto, M.; Camins, A.; et al. Dexibuprofen biodegradable nanoparticles: One step closer towards a better ocular interaction study. Nanomaterials 2020, 10, 720. [Google Scholar] [CrossRef] [Green Version]
- Jose, A.; Sunaja Devi, K.R.; Pinheiro, D.; Lakshmi Narayana, S. Electrochemical synthesis, photodegradation and antibacterial properties of PEG capped zinc oxide nanoparticles. J. Photochem. Photobiol. B Biol. 2018, 187, 25–34. [Google Scholar] [CrossRef] [PubMed]
- Wuelfing, W.P.; Gross, S.M.; Miles, D.T.; Murray, R.W. Nanometer gold clusters protected by surface-bound monolayers of thiolated poly(ethylene glycol) polymer electrolyte. J. Am. Chem. Soc. 1998, 120, 12696–12697. [Google Scholar] [CrossRef]
- Karakoti, A.S.; Das, S.; Thevuthasan, S.; Seal, S. PEGylated inorganic nanoparticles. Angew. Chem. Int. Ed. 2011, 50, 1980–1994. [Google Scholar] [CrossRef]
- Reznickova, A.; Slavikova, N.; Kolska, Z.; Kolarova, K.; Belinova, T.; Hubalek Kalbacova, M.; Cieslar, M.; Svorcik, V. PEGylated gold nanoparticles: Stability, cytotoxicity and antibacterial activity. Colloids Surf. A Physicochem. Eng. Asp. 2019, 560, 26–34. [Google Scholar] [CrossRef]
- Luo, M.; Shen, C.; Feltis, B.N.; Martin, L.L.; Hughes, A.E.; Wright, P.F.A.; Turney, T.W. Reducing ZnO nanoparticle cytotoxicity by surface modification. Nanoscale 2014, 6, 5791–5798. [Google Scholar] [CrossRef] [PubMed]
- Otsuka, H.; Nagasaki, Y.; Kataoka, K. PEGylated nanoparticles for biological and pharmaceutical applications. Adv. Drug Deliv. Rev. 2003, 55, 403–419. [Google Scholar] [CrossRef]
- Salassi, S.; Canepa, E.; Ferrando, R.; Rossi, G. Anionic nanoparticle-lipid membrane interactions: The protonation of anionic ligands at the membrane surface reduces membrane disruption. RSC Adv. 2019, 9, 13992–13997. [Google Scholar] [CrossRef] [Green Version]
- Ayata, N.; Sezer, A.D.; Bucak, S.; Turanlı, E.T. Preparation and in vitro characterization of monoclonal antibody ranibizumab conjugated magnetic nanoparticles for ocular drug delivery. Braz. J. Pharm. Sci. 2020, 56, 1–15. [Google Scholar] [CrossRef]
- Sharma, A.; Goyal, A.K.; Rath, G. Recent advances in metal nanoparticles in cancer therapy. J. Drug Target. 2018, 26, 617–632. [Google Scholar] [CrossRef] [PubMed]
- Cecchi, F.; Rabe, D.C.; Bottaro, D.P. Targeting the HGF/Met signaling pathway in cancer therapy. Expert Opin. Ther. Targets 2012, 16, 553–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, S.; Bouchy, S.; Penninckx, S.; Marega, R.; Fichera, O.; Gallez, B.; Feron, O.; Martinive, P.; Heuskin, A.C.; Michiels, C.; et al. Antibody-functionalized gold nanoparticles as tumor-Targeting radiosensitizers for proton therapy. Nanomedicine 2019, 14, 317–333. [Google Scholar] [CrossRef] [PubMed]
- Chia, S.L.; Leong, D.T. Reducing ZnO nanoparticles toxicity through silica coating. Heliyon 2016, 2, e00177. [Google Scholar] [CrossRef] [Green Version]
- Malvindi, M.A.; De Matteis, V.; Galeone, A.; Brunetti, V.; Anyfantis, G.C.; Athanassiou, A.; Cingolani, R.; Pompa, P.P. Toxicity assessment of silica coated iron oxide nanoparticles and biocompatibility improvement by surface engineering. PLoS ONE 2014, 9, e85835. [Google Scholar] [CrossRef] [Green Version]
- Soumbo, M.; Scarangella, A.; Villeneuve-Faure, C.; Bonafos, C.; Roques, C.; Makasheva, K. Combined effect of proteins and AgNPs on the adhesion of yeast Candida albicans on solid silica surfaces. In Proceedings of the 2020 IEEE 20th International Conference on Nanotechnology (IEEE-NANO), Montreal, QC, Canada, 29–31 July 2020; pp. 242–245. [Google Scholar] [CrossRef]
- Santo-Orihuela, P.L.; Foglia, M.L.; Targovnik, A.M.; Miranda, V.M.; Desimone, M.F. Nanotoxicological Effects of SiO2 Nanoparticles on Spodoptera frugiperda Sf9 Cells. Curr. Pharm. Biotechnol. 2016, 17, 465–470. [Google Scholar] [CrossRef] [PubMed]
- Worthington, K.L.S.; Adamcakova-Dodd, A.; Wongrakpanich, A.; Mudunkotuwa, I.A.; Mapuskar, K.A.; Joshi, V.B.; Allan Guymon, C.; Spitz, D.R.; Grassian, V.H.; Thorne, P.S.; et al. Chitosan coating of copper nanoparticles reducesin vitrotoxicity and increases inflammation in the lung. Nanotechnology 2013, 24, 395101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boyles, M.S.P.; Kristl, T.; Andosch, A.; Zimmermann, M.; Tran, N.; Casals, E.; Himly, M.; Puntes, V.; Huber, C.G.; Lütz-Meindl, U.; et al. Chitosan functionalisation of gold nanoparticles encourages particle uptake and induces cytotoxicity and pro-inflammatory conditions in phagocytic cells, as well as enhancing particle interactions with serum components. J. Nanobiotechnol. 2015, 13, 84. [Google Scholar] [CrossRef] [Green Version]
- Wu, M.; Guo, H.; Liu, L.; Liu, Y.; Xie, L. Size-dependent cellular uptake and localization profiles of silver nanoparticles. Int. J. Nanomed. 2019, 14, 4247–4259. [Google Scholar] [CrossRef] [Green Version]
- Park, M.V.D.Z.; Neigh, A.M.; Vermeulen, J.P.; de la Fonteyne, L.J.J.; Verharen, H.W.; Briedé, J.J.; van Loveren, H.; de Jong, W.H. The effect of particle size on the cytotoxicity, inflammation, developmental toxicity and genotoxicity of silver nanoparticles. Biomaterials 2011, 32, 9810–9817. [Google Scholar] [CrossRef]
- Chithrani, B.D.; Ghazani, A.A.; Chan, W.C.W. Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. Nano Lett. 2006, 6, 662–668. [Google Scholar] [CrossRef]
- Chen, Y.S.; Hung, Y.C.; Liau, I.; Huang, G.S. Assessment of the in vivo toxicity of gold nanoparticles. Nanoscale Res. Lett. 2009, 4, 858–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khodashenas, B.; Ghorbani, H.R. Synthesis of silver nanoparticles with different shapes. Arab. J. Chem. 2019, 12, 1823–1838. [Google Scholar] [CrossRef] [Green Version]
- Lee, Y.J.; Ahn, E.Y.; Park, Y. Shape-dependent cytotoxicity and cellular uptake of gold nanoparticles synthesized using green tea extract. Nanoscale Res. Lett. 2019, 14, 129. [Google Scholar] [CrossRef] [PubMed]
- Chang, Y.; Li, K.; Feng, Y.; Cheng, Y.; Zhang, M.; Wang, Z.; Wu, Z.; Zhang, H. Achievement of safer palladium nanocrystals by enlargement of {100} crystallographic facets. Nanotoxicology 2017, 11, 907–922. [Google Scholar] [CrossRef]
- Yaqoob, S.B.; Adnan, R.; Rameez Khan, R.M.; Rashid, M. Gold, Silver, and Palladium Nanoparticles: A Chemical Tool for Biomedical Applications. Front. Chem. 2020, 8, 376. [Google Scholar] [CrossRef] [PubMed]
- Xie, X.; Liao, J.; Shao, X.; Li, Q.; Lin, Y. The Effect of shape on Cellular Uptake of Gold Nanoparticles in the forms of Stars, Rods, and Triangles. Sci. Rep. 2017, 7, 3827. [Google Scholar] [CrossRef] [PubMed]

Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
García-Torra, V.; Cano, A.; Espina, M.; Ettcheto, M.; Camins, A.; Barroso, E.; Vazquez-Carrera, M.; García, M.L.; Sánchez-López, E.; Souto, E.B. State of the Art on Toxicological Mechanisms of Metal and Metal Oxide Nanoparticles and Strategies to Reduce Toxicological Risks. Toxics 2021, 9, 195. https://doi.org/10.3390/toxics9080195
García-Torra V, Cano A, Espina M, Ettcheto M, Camins A, Barroso E, Vazquez-Carrera M, García ML, Sánchez-López E, Souto EB. State of the Art on Toxicological Mechanisms of Metal and Metal Oxide Nanoparticles and Strategies to Reduce Toxicological Risks. Toxics. 2021; 9(8):195. https://doi.org/10.3390/toxics9080195
Chicago/Turabian StyleGarcía-Torra, Victor, Amanda Cano, Marta Espina, Miren Ettcheto, Antoni Camins, Emma Barroso, Manel Vazquez-Carrera, Maria Luisa García, Elena Sánchez-López, and Eliana B. Souto. 2021. "State of the Art on Toxicological Mechanisms of Metal and Metal Oxide Nanoparticles and Strategies to Reduce Toxicological Risks" Toxics 9, no. 8: 195. https://doi.org/10.3390/toxics9080195
APA StyleGarcía-Torra, V., Cano, A., Espina, M., Ettcheto, M., Camins, A., Barroso, E., Vazquez-Carrera, M., García, M. L., Sánchez-López, E., & Souto, E. B. (2021). State of the Art on Toxicological Mechanisms of Metal and Metal Oxide Nanoparticles and Strategies to Reduce Toxicological Risks. Toxics, 9(8), 195. https://doi.org/10.3390/toxics9080195

