Silver nanoparticles: cytotoxic, apoptotic, and necrotic effects on MCF-7 cells

The present study was conducted to examine cytotoxic, apoptotic, and necrotic effects of silver nanoparticles (AgNPs) on MCF-7 cells. Colloidal AgNPs were fabricated in an alkaline pH environment via reduction of silver nitrate with hydroxylamine hydrochloride. The size of AgNPs was measured by atomic force microscopy (AFM), transmission electron microscopy (TEM), and dynamic light scattering. Zeta potential of AgNPs was determined by laser Doppler microelectrophoresis. After exposing MCF-7 cells to AgNPs for 24 h, cytotoxicity was measured by WST-1 assay. Apoptosis and necrosis in MCF-7 cells were detected by Annexin-V-FLUOS immunostaining and double staining of Hoechst dye with propidium iodide. In AFM and TEM analyses, the sizes of AgNPs varied from 16 nm to 20 nm. AgNPs were 80 nm in hydrodynamic diameter with a zeta potential of –38.2 mV. The WST-1 assay resulted in an IC50 value of 40 µg/mL. AgNPs caused apoptotic and necrotic effects in a dose-dependent manner. The apoptotic effect of AgNPs was marked up to a concentration of 80 µg/mL AgNPs. At higher concentrations, the apoptotic effect decreased while the necrotic effect became prominent. The results indicate that AgNPs with a zeta potential of –38.2 mV and hydrodynamic diameter of 80 nm can be used in vitro at concentrations of up to 40 µg/mL.

Silver nanoparticles: cytotoxic, apoptotic, and necrotic effects on MCF-7 cells

The present study was conducted to examine cytotoxic, apoptotic, and necrotic effects of silver nanoparticles (AgNPs) on MCF-7 cells. Colloidal AgNPs were fabricated in an alkaline pH environment via reduction of silver nitrate with hydroxylamine hydrochloride. The size of AgNPs was measured by atomic force microscopy (AFM), transmission electron microscopy (TEM), and dynamic light scattering. Zeta potential of AgNPs was determined by laser Doppler microelectrophoresis. After exposing MCF-7 cells to AgNPs for 24 h, cytotoxicity was measured by WST-1 assay. Apoptosis and necrosis in MCF-7 cells were detected by Annexin-V-FLUOS immunostaining and double staining of Hoechst dye with propidium iodide. In AFM and TEM analyses, the sizes of AgNPs varied from 16 nm to 20 nm. AgNPs were 80 nm in hydrodynamic diameter with a zeta potential of 38.2 mV. The WST-1 assay resulted in an IC50 value of 40 µg/mL. AgNPs caused apoptotic and necrotic effects in a dose-dependent manner. The apoptotic effect of AgNPs was marked up to a concentration of 80 µg/mL AgNPs. At higher concentrations, the apoptotic effect decreased while the necrotic effect became prominent. The results indicate that AgNPs with a zeta potential of 38.2 mV and hydrodynamic diameter of 80 nm can be used in vitro at concentrations of up to 40 µg/mL.

___

  • Ahmed M, Karns M, Goodson M, Rowe J, Hussain S, Schlager J, Hong Y (2008). DNA damage response to different surface chemistry of silver nanoparticles in mammalian cells. Toxicol Appl Pharmacol 233: 404–410.
  • Asharani PV, Hande MP, Valiyaveettil S (2009). Anti-proliferative activity of silver nanoparticles. BMC Cell Biol 10: 65–79.
  • Asharani, PV, Low Kah Mun G, Hande MP, Valiyaveettil S (2009). Cytotoxicity and genotoxicity of silver nanoparticles in human cells. ACS Nano 3: 279–290.
  • Boonstra J, Post JA (2004). Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells. Gene 337: 1–13.
  • Braydich-Stollen L, Hussain S, Schrand A, Schlager J, Hofmann M (2005). In vitro cytotoxicity of nanoparticles in mammalian germline stem cells. Toxicol Sci 88: 412–419.
  • Chaloupka K, Malam Y, Seifalian AM (2010). Nanosilver as a new generation of nanoproduct in biomedical applications. Trends Biotechnol 28: 580–588.
  • Chen X, Schluesener HJ (2008). Nanosilver: a nanoproduct in medical application. Toxicol Lett 176: 1–12.
  • Cho K, Park J, Osaka T, Park S (2005). The study of antimicrobial activity and preservative effects of nanosilver ingredients. Electrochim Acta 51: 956–960.
  • Foldbjerg R, Olesen P, Hougaard M, Dang DA, Hoffmann HJ, Autrup, H (2009). PVP-coated silver nanoparticles and silver ions induce reactive oxygen species, apoptosis, and necrosis in THP-1 monocytes. Toxicol Lett 190: 156–162.
  • Gopinath P, Gogoi SK, Chattopadhyay A, Ghosh SS (2008). Implications of silver nanoparticle induced cell apoptosis for in vitro gene therapy. Nanotechnology 19: 075104.
  • Hackenberg S. Scherzed A, Kessler M, Froelich K, Ginzkey C, Koehler C, Burghartz M, Hagen R, Kleinsasser N (2010). Zinc oxide nanoparticles induce photocatalytic cell death in human head and neck squamous cell carcinoma cell lines in vitro. Int J Oncol 37: 1583–1590.
  • Hackenberg S, Scherzed A, Kessler M, Hummel S, Technau A, Froelich K, Ginzkey C, Koehler C, Hagen R, Kleinsasser N (2011). Silver nanoparticles: evaluation of DNA damage, toxicity, and functional impairment in human mesenchymal stem cells. Toxicol Lett 201: 27–33.
  • Hess R, Jones L, Schlager JJ (2008). Unique cellular interaction of silver nanoparticles: size-dependent generation of reactive oxygen species. J Phys Chem B 112: 13608–13619.
  • Hsin Y, Chen C, Huang S, Shih T, Lai P, Chueh P (2008). The apoptotic effect of nanosilver is mediated by ROS- and JNK-dependent mechanism involving the mitochondrial pathway in NIH3T3 cells. Toxicol Lett 179: 130–139.
  • Hussain SM, Hess KL, Gearhart JM, Geiss KT, Schlager JJ (2005). In vitro toxicity of nanoparticles in BRL 3A rat liver cells. Toxicol In Vitro 1: 975–983.
  • Johnston HJ, Hutchison G, Christensen FM, Peters S, Hankin S, Stone V (2010). A review of the in vivo and in vitro toxicity of silver and gold particulates: particle attributes and biological mechanisms responsible for the observed toxicity. Crit Rev Toxicol 40: 328–346.
  • Kamphaus GD, Colorado PC, Panka DJ, Hopfer H, Ramchandran R, Torre A, Maeshima Y, Mier JW, Sukhatme VP, Kalluri R (2000). Canstatin, a novel matrix-derived inhibitor of angiogenesis and tumor growth. J Biol Chem 275: 1209–1215.
  • Kim J, Kuk E, Yuk N, Kim JH, Park SJ (2006). Mode of antibacterial action of silver nanoparticles. J Proteome Res 5: 916–924.
  • Kim YS, Song MY, Park JD, Song KS, Ryu HR, Chung YH, Chang HK, Lee JH, Oh KH, Kelman BJ et al. (2010). Subchronic oral toxicity of silver nanoparticles. Part Fibre Toxicol 7: 20–31.
  • Kreuter J, Gelperina S (2008). Use of nanoparticles for cerebral cancer. Tumori 94: 271–277.
  • Leopold N, Lendl B (2003). A new method for fast preparation of highly surface-enhanced Raman scattering (SERS) active silver colloids at room temperature by reduction of silver nitrate with hydroxylamine hydrochloride. J Phys Chem B 107: 5723–5727.
  • Li GY, Osborne NN (2008). Oxidative-induced apoptosis to an immortalized ganglion cell line is caspase independent but involves the activation of poly(ADP-ribose)polymerase and apoptosis-inducing factor. Brain Res 188: 35–43.
  • Liu W, Wu Y, Wang C, Li HC, Wang T, Liao CY, Cui L, Zhou QF, Yan B, Jiang GB (2010). Impact of silver nanoparticles on human cells: effect of particle size. Nanotoxicology 4: 319–330.
  • Liu X, Lee PY, Ho CM, Lui VC, Chen Y, Che CM, Tam PK, Wong KK (2010). Silver nanoparticles mediate differential responses in keratinocytes and fibroblasts during skin wound healing. Chem Med Chem 1: 468–475.
  • Mahmood M, Casciano DA, Mocan T, Iancu C, Xu Y, Mocan L, Iancu DT, Dervishi E, Li Z, Abdalmuhsen M et al. (2010). Cytotoxicity and biological effects of functional nanomaterials delivered to various cell lines. J Appl Toxicol 30: 74–83.
  • Oberdörster G, Oberdörster E, Oberdörster J (2005). Nanotoxicology: an emerging discipline evolving from studies of ultrafine particles. Environ Health Perspect 113: 823–839.
  • Oberley T, Sioutas C, Yeh JI, Wiesner MR, Nel AE (2006). Comparison of the abilities of ambient and manufactured nanoparticles to induce cellular toxicity according to an oxidative stress paradigm. Nano Lett 6: 1794–1807.
  • Pan Y, Neuss S, Leifert A, Fischer M, Wen F (2007). Size-dependent cytotoxicity of gold nanoparticles. Small 3: 1941–49.
  • Park S, Lee YK, Jung M, Kim KH, Chung N (2007). Cellular toxicity of various inhalable metal nanoparticles on human alveolar epithelial cells. Inhal Toxicol 19: 59–65.
  • Singh N, Manshian B, Jenkins GJ, Griffiths SM, Williams PM, Maffeis TG, Wright CJ, Doak SH (2009). NanoGenotoxicology: the DNA damaging potential of engineered nanomaterials. Biomaterials 30: 3891–3914.
  • Singh RP, Ramarao P (2012). Cellular uptake, intracellular trafficking, and cytotoxicity of silver nanoparticles. Toxicol Lett 213: 249– 2
  • Sohaebuddin SK, Thevenot PT, Baker D, Eaton JW, Tang L (2010). Nanomaterial cytotoxicity is composition, size, and cell type dependent. Part Fibre Toxicol 7: 22–44.
  • Sonoda E, Sasaki MS, Buerstedde JM, Bezzubova O, Shinohara A, Ogawa H, Takata M, Yamaguchi-Iwai Y, Takeda S (1998). Rad51-deficient vertebrate cells accumulate chromosomal breaks prior to cell death. EMBO J 15: 598–608.
  • Su J, Zhang J, Liu L, Huang Y, Mason RP (2008). Exploring feasibility of multicolored CdTe quantum dots for in vitro and in vivo fluorescent imaging. J Nanosci Nanotechnol 8: 1174–1177.
  • Sur I, Altunbek M, Kahraman M, Culha M (2012). The influence of the surface chemistry of silver nanoparticles on cell death. Nanotechnology 23: 375102.
  • Sur I, Cam D, Kahraman M, Baysal A, Culha M (2010). Interaction of multi-functional silver nanoparticles with living cells. Nanotechnology 21: 175104.
  • Tan WB, Jiang S, Zhang Y (2007). Quantum-dot based nanoparticles for targeted silencing of HER2/neu gene via RNA interference. Biomaterials 28: 1565–1571.
  • Tian, J, Wong KK, Ho CM, Lok CN, Yu WY, Che CM, Chiu JF, Tam PK (2007). Topical delivery of silver nanoparticles promotes wound healing. Chem Med Chem 2: 129–136.
  • Tice RR, Agurell E, Anderson D, Burlinson B, Hartmann A, Kobayashi H, Miyamae Y, Rojas E, Ryu JC, Saski YF (2000). Single cell gel/comet assay: guidelines for in vitro and in vivo genetic toxicology testing. Environ Mol Mutagen 35: 206–221.
  • Türk M, Rzaev ZMO, Kurucu G (2010). Bioengineering functional copolymers. XII. Interaction of boron-containing and PEO branched derivatives of poly(MA-alt-MVE) with HeLa cells. Health 2: 51–61.
  • Turrens J (2003). Mitochondrial formation of reactive oxygen species. J Physiol 552: 335–344. van Engeland M, Nieland LJ, Ramaekers FC, Schutte B, Reutelingsperger CP (1998). Annexin V-affinity assay: a review on an apoptosis detection system based on phosphatidylserine exposure. Cytometry 31: 1–9.
  • Wijnhoven SWP, Peijnenburg WJGM, Herberts CA, Hagens WI, Oomen AG, Heugens EHW, Roszek B, Bisschops J, Gosens I, Van De Meent D et al. (2009). Nano-silver – a review of available data and knowledge gaps in human and environmental risk assessment. Nanotoxicology 3: 109–138.
  • Yoon K, Hoon B, Park JH, Hwang J (2007). Susceptibility constants of Escherichia coli and Bacillus subtilis to silver and copper nanoparticles. Sci Total Environ 373: 572–575.
  • Zanette C, Pelin M, Crosera M, Adami G, Bovenzi M, Larese FF, Florio C (2011). Silver nanoparticles exert a long-lasting antiproliferative effect on human keratinocyte HaCaT cell line. Toxicol In Vitro 5: 1053–1060.
Turkish Journal of Biology-Cover
  • ISSN: 1300-0152
  • Yayın Aralığı: Yılda 6 Sayı
  • Yayıncı: TÜBİTAK
Sayıdaki Diğer Makaleler

Global assessment of network inference algorithms based on available literature of gene/protein interactions

Gökmen ALTAY, Nejla ALTAY, David NEAL

Morphological and molecular identification of pennate diatoms isolated from Urla, İzmir, coast of the Aegean Sea

Kutsal KESİCİ, İnci TÜNEY, Doğuş ZEREN, Mustafa GÜDEN, Atakan SUKATAR

Effect of asiatic and ursolic acids on growth and virulence factors of uropathogenic Escherichia coli strains

Dorota WOJNICZ, Dorota TICHACZEK-GOSKA, Marta KICIA

Lignification response for rolled leaves of Ctenanthe setosa under long-term drought stress

Rabiye TERZİ, Neslihan SARUHAN GÜLER, Nihal KUTLU ÇALIŞKAN, Asım KADIOĞLU

Differential expression analysis of defense-related genes responsive to Tilletia indica infection in wheat

Aradhika TRIPATHI, Rashmi AGGARWAL, Anita YADAV

Probing the architecture of testis and morphology of male germinal cells in the mud crab with the atomic force microscopy

Mahalingam ANBARASU, Ramangalingam KIRUBAGARAN, Gopal DHARANI, Thanumalaya SUBRAMONIAM

Transcriptional profiling of transferrin gene from Egyptian cotton leaf worm, Spodoptera littoralis

Nurper GÜZ, Aslı DAĞERİ, Tuğba ERDOĞAN, Mouzhgan MOUSAVI, Şerife BAYRAM, Mehmet Oktay GÜRKAN

Antimicrobial susceptibility patterns of environmental and hospital isolations of enterococci in Aydın

Erman ORYAŞIN, Hacı Halil BIYIK, Gamze BAŞBÜLBÜL, Bülent BOZDOĞAN

Activity of Brassica napus and Perilla frutescens microsomal w-3 desaturases expressed in yeast (Saccharomyces cerevisiae)

Mohammed ABDEL-REHEEM, David HILDEBRAND

Hepatopancreas mitochondria of Mytilus galloprovincialis: effect of zinc ions on mitochondrial bioenergetics and metabolism

Antonino DE LISI, Ermelinda PRATO, Francesca BIANDOLINO, Giulio SARLI, Donatella NEGRO, Gianluigi LA PIANA, Domenico MARZULLI