AccScience Publishing / GTM / Volume 1 / Issue 1 / DOI: 10.36922/gtm.v1i1.34
ORIGINAL RESEARCH ARTICLE

Green synthesized zinc oxide nanoparticles induce apoptosis by suppressing PI3K/Akt/mTOR signaling pathway in osteosarcoma MG63 cells

Satheeshkuma Subramaniyan1 Yoganathan Kamaraj1 Veenayohini Kumaresan1 Muthulakshmi Kannaiyan2 Ernest David3 Babujanarthanam Ranganathan3 Vijayanand Selvaraj3 Agilan Balupillai3
Show Less
1 Department of Microbiology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, India
2 Department of Microbiology, Faculty of Science, Idhaya College for Women, Tiruvannamalai, Tamil Nadu, India
3 Department of Biotechnology, Thiruvalluvar University, Serkkadu, Vellore, Tamil Nadu, India
Global Translational Medicine 2022, 1(1), 34 https://doi.org/10.36922/gtm.v1i1.34
Submitted: 3 March 2022 | Accepted: 15 April 2022 | Published: 23 May 2022
© 2022 by the Authors. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

This study aimed to assess the apoptosis-inducing mechanism of zinc oxide nanoparticles (ZnO NPs) stabilized by Solanum xanthocarpum plant extract in human osteosarcoma MG63 cells. In the present study, we synthesized ZnO NPs from S. xanthocarpum extract and evaluated its anticancer mechanism on MG 63 cells. The synthesized ZnO NPs were characterized by ultraviolet spectroscopy, X-ray crystallography, transmission electron microscopy, energy dispersive X-ray, and Fourier-transform infrared spectroscopy analysis. The mean size of the synthesized ZnO NPs was 21.62 ± 7.45 nm and spherical in shape. The cytotoxicity of ZnO NPs on MG63 cells was determined by MTT assay. The Western blot analysis was carried out to examine the expression of apoptotic and autophagy-related proteins in MG63 cells. The findings of the study reveal that ZnO NPs treatment showed concentration-dependent cytotoxicity, increased lipid peroxidation, decreased antioxidant activity, increased reactive oxygen species generation, and increased DNA damage. In addition, ZnO NPs treatment increased the expression of apoptotic members such as p53, Bax, caspase-3, -8, and -9 while downregulating Bcl-2 expression in MG63 cells. Furthermore, ZnO NPs treatment suppressed the P13K/AKT/mTOR signaling pathway and increased the expression of LC3 and beclin-1 in MG63 cells. The present study demonstrated that ZnO NPs induced apoptosis and autophagy in MG63 cells through modifying apoptotic and autophagy-related proteins.

Keywords
Osteosarcoma
Solanum xanthocarpum
Zinc oxide nanoparticles
Apoptosis
Autophagy
References
[1]

Cai ST, Zhang D, Zhang G, et al., 2015, Volume-sensitive chloride channels are involved in cisplatin treatment of osteosarcoma. Mol Med Rep, 11: 2465–2470. https://doi.org/10.3892/mmr.2014.3068

[2]

Zhang YL, Zhang G, Zhang S, et al., 2014, Osteosarcoma metastasis: Prospective role of ezrin. Tumor Biol, 35: 5055–5059. https://doi.org/10.1007/s13277-014-1799-y

[3]

Hattinger CM, Pasello M, Ferrari S, et al., 2010, Emerging drugs for high-grade osteosarcoma. Expert Opin Emerg Drugs, 15: 615–634. https://doi.org/10.1517/14728214.2010.505603 

[4]

Brasseur K, Gévry N, Asselin E, 2017, Chemoresistance and targeted therapies in ovarian and endometrial cancers. Oncotarget, 8: 4008–4042. https://doi.org/10.18632/oncotarget.14021 

[5]

Jayarambabu N, Kumari BS, 2015, Beneficial role of zinc oxide nanoparticles on green crop production. Int J Multidiscip Adv Res Trends, 2: 273–282.

[6]

Senthilkumar SR, Sivakumar T, 2014, Green tea (Camellia sinensis) mediated synthesis of zinc oxide nanoparticles and studies on their antimicrobial activities. Int J Pharm Pharm Sci, 6: 461–465. 

[7]

Rasmussen JW, Martinez E, Louka P, et al., 2010, Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug delivery applications. Expert Opin Drug Deliv, 7: 1063–1077. https://doi.org/10.1517/17425247.2010.502560 

[8]

Aminuzzaman M, Ying LP, Goh WS, et al., 2018, Green synthesis of zinc oxide nanoparticles using aqueous extract of Garcinia mangostana fruit pericarp and their photocatalytic activity. Bull Mater Sci, 41: 50. https://doi.org/10.1007/s12034-018-1568-4

[9]

Elumalai K, Velmurugan S, Ravi S, et al., 2015, Facile, eco-friendly and template free phytosynthesis of cauliflower like ZnO nanoparticles using leaf extract of Tamarindus indica (L.) and its biological evolution of antibacterial and antifungal activities. Spectrochim Acta A Mol Biomol Spectrosc, 136: 1052–1057. https://doi.org/10.1016/j.saa.2018.09.018 

[10]

Jayaseelan C, Rahuman AA, Kirthi VA, et al., 2012, Novel microbial route to synthesise ZnO nanoparticles using Aeromonas hydrophila and their activity against pathogenic bacteria and fungi. Spectrochim Acta A Mol Biomol Spectrosc, 90: 78–84. https://doi.org/10.1016/j.saa.2012.01.006

[11]

Sangeetha G, Rajeshwari S, Venckatesh R, 2011, Green synthesis of zinc oxide nanoparticles by Aloe barbadensis miller leaf extract: structure and optical properties. Mater Res Bull, 46: 2560–2566. https://doi.org/10.1016/j.materresbull.2011.07.046 

[12]

Sreenivasan CV, Jovitta J, Suja S, 2012, Synthesis of ZnO nanoparticles from Alpinia purpurata and their antimicrobial properties. Res J Pharm Biol Chem Sci, 3: 1206–1213.

[13]

Okhawa H, Ohishi N, Yagi K, 1979, Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem, 95: 351–358. https://doi.org/10.1016/0003-2697(79)90738-3

[14]

Kakkar P, Das B, Viswanathan PN, 1984, A modified spectrophotometric assay of superoxide dismutase Indian. J Biochem Biophys, 21: 130–132. 

[15]

Sinha KA, 1972, Colorimetric assay of catalase. Anal Biochem, 47: 389–394. https://doi.org/10.1016/0003-2697(72)90132-7

[16]

Wang CP, Myung E, Lau BH, 1993, An automated micro-fluorometric assay for monitoring oxidative burst activity of phagocytes. J Immunol Methods, 159: 131–138. https://doi.org/10.1016/0022-1759(93)90150-6

[17]

Karthikeyan S, Kanimozhi G, Prasad NR, et al., 2011, Radiosensitizing effect of ferulic acid on human cervical carcinoma cells in vitro. Toxicol In Vitro, 25: 136675. https://doi.org/10.1016/j.tiv.2011.05.007 

[18]

Huang WW, Ko SW, Tsai HY, et al., 2011, Cantharidin induces G2/M phase arrest and apoptosis in human colorectal cancer colo 205 cells through inhibition of CDK1 activity and caspase-dependent signaling pathways. Int J Oncol, 38: 1067–1073. https://doi.org/10.3892/ijo.2011.922

[19]

Le Marchand L, 2002, Cancer preventive effects of flavonoids a review. Biomed Pharmacother, 56: 296301.

[20]

Guo D, Wu C, Jiang H, et al., 2008, Synergistic cytotoxic effect of different-sized ZnO nanoparticles and daunorubicin against leukemia cancer cells under UV irradiation. J Photochem Photobiol B, 93: 119–126. https://doi.org/10.1016/j.jphotobiol.2008.07.009

[21]

Ryter SW, Kim HP, Hoetzel A, et al., 2007, Mechanisms of cell death in oxidative stress. Antioxid Redox Signal, 9: 49–89.

[22]

Santhoshkumar J, Kumar SV, Rajeshkumar S, 2017, Synthesis of zinc oxide nanoparticles using plant leaf extract against urinary tract infection pathogen. Resour Efficient Technol, 3: 459–465. https://doi.org/10.1016/j.reffit.2017.05.001

[23]

Amudha P, Prabakaran R, Senthil Kumar S, et al., 2017, Phytochemical analysis of Albizia chinensis (Osbeck) Merr medicinal plant. J Pharm Biol Sci, 12: 89–92.

[24]

Fakhari S, Jamzad M, Fard HK, 2019, Green synthesis of zinc oxide nanoparticles: A comparison. Green Chem Let Rev, 12: 19–24. https://doi.org/10.1080/17518253.2018.1547925

[25]

Shilpa PN, Krishnan SS, Niranjali D, 2012, Induction of apoptosis by methanolic extract of Rubia cordifolia Linn in HEp-2cell line is mediated by reactive oxygen species. Asian Pac J Cancer Prev, 13: 27538. https://doi.org/10.7314/apjcp.2012.13.6.2753 

[26]

Yoganathan K, Sangeetha D, Uma C, et al., 2021, Triterpenoid compound betulin attenuates allergic airway inflammation by modulating antioxidants, inflammatory cytokines and tissue transglutaminase in ovalbumin-induced asthma mice model. J Pharm Pharmacol, 73: 968–978. https://doi.org/10.1093/jpp/rgab015

[27]

Kuo CH, Michael HH, 2010, Morphologically controlled the synthesis of Cu2O nanocrystals and their properties. Nano Today, 5: 10616. https://doi.org/10.1016/j.nantod.2010.02.001

[28]

Sharma V, Anderson D, Dhawan A, 2012, Zinc oxide nanoparticles induce oxidative DNA damage and ROS- triggered mitochondria mediated apoptosis in human liver cells (HepG2). Apoptosis, 17: 85270. https://doi.org/10.1007/s10495-012-0705-6

[29]

Carmody RJ, Cotter TG, 2001, Signalling apoptosis: A radical approach. Redox Rep, 6: 77-90. 

[30]

Collis SJ, DeWeese TJ, Jeggo PA, et al., 2005, The life and death of DNA-PK. Oncogene, 24: 94961. https://doi.org/10.1038/sj.onc.1208332

[31]

Youle RJ, Strasser A, 2008, The BCL-2 protein family: Opposing activities that mediate cell death. Nat Rev Mol Cell Biol, 9: 47–59. https://doi.org/10.1038/nrm2308

[32]

Burger H, Nooter K, Boersma AW, et al., 1998, Expression of p53, Bcl-2 and Bax in cisplatin-induced apoptosis in testicular germ cell tumour cell lines. Br J Cancer, 77: 1562–1567. https://doi.org/10.1038/bjc.1998.257

[33]

Priya K, Vijayakumar M, Janani B, 2020, Chitosan-mediated synthesis of biogenic silver nanoparticles (AgNPs), nanoparticle characterisation and in vitro assessment of anticancer activity in human hepatocellular carcinoma HepG2 cells. Int J Biol Macromol, 149: 844–852. https://doi.org/10.1016/j.ijbiomac.2020.02.007

[34]

Patnaik A, Appleman LJ, Mountz JM, et al., 2011, A first-in-human phase I study of intravenous PI3K inhibitor BAY 80-6946 in patients with advanced solid tumors: Results of dose-escalation phase. J Clin Oncol, 29: 3035. https://doi.org/10.1200/jco.2011.29.15_suppl.3035

[35]

Yip PY, 2015, Phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin (PI3K-Akt-mTOR) signaling pathway in non-small cell lung cancer. Lung Cancer Res, 4: 165. https://doi.org/10.1016/j.ctrv.2014.06.006

[36]

Liu J, Chen W, Zhang H, et al., 2017, miR-214 targets the PTEN-mediated PI3K/Akt signaling pathway and regulates cell proliferation and apoptosis in ovarian cancer. Oncol Lett, 14: 5711–5718. https://doi.org/10.3892/ol.2017.6953

[37]

Chen S, Rehman SK, Zhang W, et al., 2010, Autophagy is a therapeutic target in anticancer drug resistance. Biochim Biophys Acta; 1806: 220–9. https://doi.org/10.1016/j.bbcan.2010.07.003

[38]

Pattingre S, Espert L, Biard-Piechaczyk M, et al., 2008, Regulation of macro autophagy by mTOR and Beclin 1 complexes. Biochimie, 90: 313–323. https://doi.org/10.1016/j.biochi.2007.08.014

[39]

Kabeya Y, Mizushima N, Ueno T, et al., 2000, LC3, a mammalian homologue of yeast Apg8p, is localised in autophagosome membranes after processing. EMBO J, 19: 5720–5728. https://doi.org/10.1093/emboj/19.21.5720

[40]

Mathew R, Karp CM, Beaudoin B, et al., 2009, Autophagy suppresses tumorigenesis through elimination of p62. Cell, 137: 1062–1075. https://doi.org/10.1016/j.cell.2009.03.048

Conflict of interest
The authors declare no conflict of interest.
Share
Back to top
Global Translational Medicine, Electronic ISSN: 2811-0021 Published by AccScience Publishing