We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Preliminary Communication

Nanoparticle-induced photocatalytic head and neck squamous cell carcinoma cell death is associated with autophagy

    Stephan Hackenberg

    Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital of Wuerzburg, Germany

    ,
    Agmal Scherzed

    Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital of Wuerzburg, Germany

    ,
    Antje Gohla

    Rudolf Virchow Center for Experimental Biomedicine, University of Wuerzburg, Germany

    Institute for Pharmacology & Toxicology, University of Wuerzburg, Germany

    ,
    Antje Technau

    Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital of Wuerzburg, Germany

    ,
    Katrin Froelich

    Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital of Wuerzburg, Germany

    ,
    Christian Ginzkey

    Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital of Wuerzburg, Germany

    ,
    Christian Koehler

    Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital of Wuerzburg, Germany

    ,
    Marc Burghartz

    Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital of Wuerzburg, Germany

    ,
    Rudolf Hagen

    Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital of Wuerzburg, Germany

    &
    Norbert Kleinsasser

    * Author for correspondence

    Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital of Wuerzburg, Germany.

    Published Online:https://doi.org/10.2217/nnm.13.41

    Aim: To characterize molecular mechanisms underlying photocatalytic cell death of head and neck squamous cell carcinoma (HNSCC) by zinc oxide nanoparticles (ZnO-NPs). Materials & methods: Human HNSCC-derived FaDu cells were incubated with ZnO-NPs followed by UVA-1 irradiation. Cytotoxicity was assessed by MTT assay and annexin-V propidium iodide test. Autophagy was detected by autophagosome accumulation, conversion of light chain 3 I to II, and lysosomal activity. The generation of reactive oxygen species was measured using the 2´,7´-dichlorofluorescein-diacetate test. Results: Apoptosis-independent cytotoxic effects were induced by 0.2- and 2-µg/ml ZnO-NPs and UVA-1. FaDu cells promoted autophagosome formation. Significantly elevated light chain 3 II and reactive oxygen species were seen after the combined application of both ZnO-NPs and UVA-1 as photocatalytic treatment. Autophagy probably mediates cell survival under UVA-1 or ZnO-NP exposure alone but induces self-digestive cell death after combined treatment. Conclusion: The effect of autophagy on HNSCC viability after nanoparticle-induced photocatalytic treatment seems to depend on the impact of the physicochemical trigger.

    Original submitted 14 June 2012; Revised submitted 19 November 2012; Published online 3 June 2013

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J. Clin.62,10–29 (2012).
    • Davis ME, Chen ZG, Shin DM. Nanoparticle therapeutics: an emerging treatment modality for cancer. Nat. Rev. Drug Discov.7,771–782 (2008).
    • Schroeder A, Heller DA, Winslow MM et al. Treating metastatic cancer with nanotechnology. Nat. Rev. Cancer12,39–50 (2011).▪ Summary of nanotherapeutic approaches in experimental oncology.
    • Yeber MC, Rodríguez J, Freer J, Duran N, Mansilla HD. Photocatalytic degradation of cellulose bleaching effluent by supported TiO2 and ZnO. Chemosphere41,1193–1197 (2000).
    • Theogaraj E, Riley S, Hughes L, Maier M, Kirkland D. An investigation of the photo-clastogenic potential of ultrafine titanium dioxide particles. Mutat. Res.634,205–219 (2007).
    • Hackenberg S, Scherzed A, Kessler M et al. 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 (2010).
    • Zhang AP, Sun YP. Photocatalytic killing effect of TiO2 nanoparticles on Ls-174-t human colon carcinoma cells. World J. Gastroenterol.10,3191–3193 (2004).
    • Seo JW, Chung H, Kim MY, Lee J, Choi IH, Cheon J. Development of water-soluble single-crystalline TiO2 nanoparticles for photocatalytic cancer-cell treatment. Small3,850–853 (2007).
    • Kelekar A. Autophagy. Ann. NY Acad. Sci.1066,259–271 (2005).
    • 10  Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science290,1717–1721 (2000).▪▪ Overview of autophagy pathways.
    • 11  Meijer AJ, Codogno P. Regulation and role of autophagy in mammalian cells. Int. J. Biochem. Cell Biol.36,2445–2462 (2004).
    • 12  Reggiori F, Klionsky DJ. Autophagosomes: biogenesis from scratch? Curr. Opin. Cell Biol.17,415–422 (2005).
    • 13  Paglin S, Hollister T, Delohery T et al. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res.61,439–444 (2001).▪ Summary of the role of autophagy in experimental oncology.
    • 14  Lambert LA, Qiao N, Hunt KK et al. Autophagy: a novel mechanism of synergistic cytotoxicity between doxorubicin and roscovitine in a sarcoma model. Cancer Res.68,7966–7974 (2008).
    • 15  Lin CI, Whang EE, Abramson MA et al. Autophagy: a new target for advanced papillary thyroid cancer therapy. Surgery146,1208–1214 (2009).
    • 16  Hussain SM, Hess KL, Gearhart JM, Geiss KT, Schlager JJ. In vitro toxicity of nanoparticles in BRL 3A rat liver cells. Toxicol. In Vitro19,975–983 (2005).▪▪ Important overview of the mechanisms of nanoparticle-induced cytotoxicity in mammalian cells.
    • 17  Rangan SR. A new human cell line (FaDu) from a hypopharyngeal carcinoma. Cancer29,117–121 (1972).
    • 18  Bihari P, Vippola M, Schultes S et al. Optimized dispersion of nanoparticles for biological in vitro and in vivo studies. Part. Fibre Toxicol.5,14 (2008).
    • 19  Seglen PO, Goron PB. 3-methyladenine: specific inhibitor of autophagic/lysosomal protein degradation in isolated rat hepatocytes. Proc. Natl Acad. Sci. USA79,1889–1892 (1982).
    • 20  Eskelinen EL. Fine structure of the autophagosome. Methods Mol. Biol.445,11–28 (2008).
    • 21  Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Methods65,55–63 (1983).
    • 22  Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the folin phenol reagent. J. Biol. Chem.193,265–275 (1951).
    • 23  Kyhse-Andersen J. Electroblotting of multiple gels: a simple apparatus without tank for rapid transfer of proteins from polyacrylamide to nitrocellulose. J. Biochem. Biophys. Methods10,203–210 (1984).
    • 24  Rodriguez-Enriquez S, Kim L, Currin RT, Lemasters JJ. Tracker dyes to probe mitochondrial autophagy (mitophagy) in rat hepatocytes. Autophagy2,29–46 (2006).
    • 25  Halamoda Kenzaoui B, Chapuis Bernasconi C, Guney-Ayra S, Juillerat-Jeanneret L. Induction of oxidative stress, lysosome activation and autophagy by nanoparticles in human brain-derived endothelial cells. Biochem. J.44,813–821 (2012).
    • 26  Ma X, Wu Y, Jin S et al. Gold nanoparticles induce autophagosome accumulation through size-dependent nanoparticle uptake and lysosome impairment. ACS Nano22,8629–8639 (2011).
    • 27  Johnson-Lyles DN, Peifley K, Lockett S et al. Fullerenol cytotoxicity in kidney cells is associated with cytoskeleton disruption, autophagic vacuole accumulation, and mitochondrial dysfunction. Toxicol. Appl. Pharmacol.248,249–258 (2010).
    • 28  Wu YN, Yang LX, Shi XY et al. The selective growth inhibition of oral cancer by iron core-gold shell nanoparticles through mitochondria-mediated autophagy. Biomaterials32,4565–4573 (2011).
    • 29  Li JJ, Hartono D, Ong CN, Bay BH, Yung LYL. Autophagy and oxidative stress associated with gold nanoparticles. Biomaterials31,5996–6003 (2010).
    • 30  Mizushima N, Yoshimori T, Levine B. Methods in mammalian autophagy research. Cell140,313–326 (2010).
    • 31  McLeland CB, Rodriguez J, Stern ST. Autophagy monitoring assay: qualitative analysis of MAP LC 3 I to LC 3 II conversion by immunoblot. Methods Mol. Biol.697,199–206 (2011).
    • 32  Lamore SD, Wondrak GT. Autophagic-lysosomal dysregulation downstream of cathepsin B inactivation in human skin fibroblasts exposed to UVA. Photochem. Photobiol. Sci.11,163–172 (2012).
    • 33  Neun BW, Stern ST. Monitoring lysosomal activity in nanoparticles-treated cells. Methods Mol. Biol.697,207–212 (2011).
    • 34  Stern ST, Zolnik BS, McLeland CB, Clogston J, Zheng J, McNeil SE. Induction of autophagy in porcine kidney cells by quantum dots: a common cellular response to nanomaterials? Toxicol. Sci.106,140–152 (2008).
    • 35  Lee CM, Huang ST, Huang SH et al. C60 fullerene-pentoxifylline dyad nanoparticles enhance autophagy to avoid cytotoxic effects caused by the β-amyloid peptide. Nanomedicine7,107–114 (2011).
    • 36  Chen Y, McMillan-Ward E, Kong J, Israels SJ, Gibson SB. Oxidative stress induces autophagic cell death independent of apoptosis in transformed and cancer cells. Cell Death Differ.15,171–182 (2008).
    • 37  Guo D, Wu C, Jiang H, Li Q, Wang X, Chen B. Synergistic cytotoxic effect of different sized ZnO nanoparticles and daunorubicin against leukemia cancer cells under UV irradiation. J. Photochem. Photobiol. B.93,119–126 (2008).
    • 38  Wang H, Wingett D, Engelhard MH et al. Fluorescent dye encapsulated ZnO particles with cell-specific toxicity for potential use in biomedical applications. J. Mater. Sci. Mater. Med.20,11–22 (2009).
    • 39  Hanley C, Layne J, Punnoose A et al. Preferential killing of cancer cells and activated human T cells using ZnO nanoparticles. Nanotechnology19,295103 (2008).▪ Describes the specific cytotoxic properties of zinc oxide nanoparticles in cancer cells.
    • 40  Akhtar MJ, Ahamed M, Kumar S, Khan MAM, Ahmad J, Alrokyan SA. Zinc oxide nanoparticles selectively induce apoptosis in human cancer cells through reactive oxygen species. Int. J. Nanomedicine7,845–857 (2012).
    • 41  Karataş OF, Sezgin E, Aydin O, Culha M. Interaction of gold nanoparticles with mitochondria. Colloids Surf. B Biointerfaces71,315–318 (2009).
    • 42  Khan MI, Mohammad A, Patil G, Naqvi SA, Chauhan LK, Ahmad I. Induction of ROS, mitochondrial damage and autophagy in lung epithelial cancer cells by iron oxide nanoparticles. Biomaterials33,1477–1488 (2012).▪ Summary of the role of reactive oxygen species and autophagy in cancer-specific cytotoxicity of nanomaterials.
    • 43  Sun L, Li Y, Liu X et al. Cytotoxicity and mitochondrial damage caused by silica nanoparticles. Toxicol. In Vitro25,1619–1629 (2011).
    • 44  Griguer CE, Oliva CR, Gillespie GY. Glucose metabolism heterogeneity in human and mouse malignant glioma cell lines. J. Neurooncol.74,123–133 (2005).