全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Evaluation of Cytotoxic Effects of Different Concentrations of Porous Hollow Au Nanoparticles (PHAuNPs) on Cells

DOI: 10.1155/2014/631248

Full-Text   Cite this paper   Add to My Lib

Abstract:

Nanoparticles (NPs) have been introduced as a suitable alternative in many in vivo bioapplications. The risks of utilizing nanoparticles continue to be an ongoing research. Furthermore, the various chemicals used in their synthesis influence the cytotoxic effects of nanoparticles. We have investigated the cytotoxicity of Porous Hollow Au Nanoparticles (PHAuNPs) on cancer cell lines PC-3, PC-3ML, and MDA-MB-231 and the normal cell line PNT1A. Cell proliferation for the different cells in the presence of different concentrations of the PHAuNPs was assessed after 24 hours and 72 hours of incubation using MTT assay. The study also included the cytotoxic evaluation of pegylated PHAuNPs. Identical cell seeding densities, particle concentrations, and incubation times were employed for these two types of Au nanoparticles. Our results indicated that (1) impact on cell proliferation was concentration dependent and was different for the different cell types without cellular necrosis and (b) cellular proliferation might be impacted more based on the cell line. 1. Introduction Nanoparticles (NPs) exhibit unique properties compared to their constituent bulk materials. These include quantum confinement in semiconductor particles, surface plasmon resonance [1–8] in metal particles, and superparamagnetism in magnetic particles. Such unique properties allow the use of nanoparticles in a wide variety of applications. Nanoparticles such as fullerenes, liquid crystals, liposomes, Au nanoparticles, and quantum dots have been demonstrated in biomedical applications [5, 6], chemotherapy [7, 8], drug delivery [8–11], imaging [11] and cosmetics [12–14]. Gold nanoparticles (AuNPs) have been subjects of intensive research in the last decade. In vivo applications of AuNPs include photothermal ablation treatment and optical imaging. Imaging utilizes the surface plasmon resonance (SPR) effect of AuNPs, a strong enhancement of absorption, and scattering of light in resonant with the SPR frequency. AuNPs possess two additional important merits for in vivo applications: (1) they are generally considered as biocompatible; (2) they can be easily functionalized with well-established thiol-Au linkage. However, the SPR wavelength for solid AuNPs lies around 520 nm, which excludes their in vivo applications since this wavelength is strongly absorbed by tissues and blood. In the last decade, several engineered Au nanoparticles such as nanoshells and nanocages have been developed to tune SPR wavelength of these nanoparticles to near-infrared region that is commonly regarded as a “clear window”

References

[1]  Z. Fang, C. Lin, R. Ma, S. Huang, and X. Zhu, “Planar plasmonic focusing and optical transport using CdS nanoribbon,” ACS Nano, vol. 4, no. 1, pp. 75–82, 2010.
[2]  Z. Fang, J. Cai, Z. Yan, P. Nordlander, N. J. Halas, and X. Zhu, “Removing a wedge from a metallic nanodisk reveals a fano resonance,” Nano Letters, vol. 11, no. 10, pp. 4475–4479, 2011.
[3]  Z. Fang, Q. Peng, W. Song et al., “Plasmonic focusing in symmetry broken nanocorrals,” Nano Letters, vol. 11, no. 2, pp. 893–897, 2011.
[4]  Z. Fang, Y. -R. Zhen, L. Fan, X. Zhu, and P. Nordlander, “Tunable wide-angle plasmonic perfect absorber at visible frequencies,” Physical Review B, vol. 85, Article ID 245401, 2012.
[5]  G. Han, P. Ghosh, and V. M. Rotello, “Multifunctional gold nanoparticles for drug delivery,” in Advances in Experimental Medicine and Biology, vol. 620, pp. 48–56, 2007.
[6]  K. K. Jain, “Applications of nanobiotechnology in clinical diagnostic,” Clinical Chemistry, vol. 53, no. 11, pp. 2002–2009, 2007.
[7]  S. Gelperina, K. Kisich, M. D. Iseman, and L. Heifets, “The potential advantages of nanoparticle drug delivery systems in chemotherapy of tuberculosis,” American Journal of Respiratory and Critical Care Medicine, vol. 172, no. 12, pp. 1487–1490, 2005.
[8]  O. C. Farokhzad, J. Cheng, B. A. Teply et al., “Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 16, pp. 6315–6320, 2006.
[9]  S. Dhar, N. Kolishetti, S. J. Lippard, and O. C. Farokhzad, “Targeted delivery of a cisplatin prodrug for safer and more effective prostate cancer therapy in vivo,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 5, pp. 1850–1855, 2011.
[10]  G. F. Paciotti, L. Myer, D. Weinreich et al., “Colloidal gold: a novel nanoparticle vector for tumor directed drug delivery,” Drug Delivery, vol. 11, no. 3, pp. 169–183, 2004.
[11]  G. F. Paciotti, D. G. I. Kingston, and L. Tamarkin, “Colloidal gold nanoparticles: a novel nanoparticle platform for developing multifunctional tumor-targeted drug delivery vectors,” Drug Development Research, vol. 67, no. 1, pp. 47–54, 2006.
[12]  M. Modo, J. W. M. Bulte, M. Mathias, and J. J. Modo, Nanoparticles in Biomedical Imaging Emerging Technologies and Applications, Springer Science+Business Media, New York, NY, USA, 2008.
[13]  A. Dingler, R. P. Blum, H. Niehus, R. H. Müller, and S. Gohla, “Solid lipid nanoparticles (SLN(TM)/Lipopearls(TM))—a pharmaceutical and cosmetic carrier for the application of vitamin E in dermal products,” Journal of Microencapsulation, vol. 16, no. 6, pp. 751–767, 1999.
[14]  V. L. Colvin, “The potential environmental impact of engineered nanomaterials,” Nature Biotechnology, vol. 21, pp. 1166–1170, 2003.
[15]  A. Nel, T. Xia, L. M?dler, and N. Li, “Toxic potential of materials at the nanolevel,” Science, vol. 311, no. 5761, pp. 622–627, 2006.
[16]  R. D. Averitt, D. Sarkar, and N. J. Halas, “Plasmon resonance shifts of Au-coated Au2S nanoshells: insight into multicomponent nanoparticle growth,” Physical Review Letters, vol. 78, no. 22, pp. 4217–4220, 1997.
[17]  L. R. Hirsch, R. J. Stafford, J. A. Bankson et al., “Nanoshell-mediated near-infrared thermal therapy of tumors under magnetic resonance guidance,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 23, pp. 13549–13554, 2003.
[18]  A. M. Gobin, M. H. Lee, N. J. Halas, W. D. James, R. A. Drezek, and J. L. West, “Near-infrared resonant nanoshells for combined optical imaging and photothermal cancer therapy,” Nano Letters, vol. 7, no. 7, pp. 1929–1934, 2007.
[19]  K. Fu, J. Sun, L. R. Bickford et al., “Measurement of immunotargeted plasmonic nanoparticles' cellular binding: a key factor in optimizing diagnostic efficacy,” Nanotechnology, vol. 19, no. 4, Article ID 045103, 2008.
[20]  S. Lal, S. E. Clare, and N. J. Halas, “Nanoshell-enabled photothermal cancer therapy: impending clinical impact,” Accounts of Chemical Research, vol. 41, no. 12, pp. 1842–1851, 2008.
[21]  S. Lal, N. K. Grady, J. Kundu, C. S. Levin, J. B. Lassiter, and N. J. Halas, “Tailoring plasmonic substrates for surface enhanced spectroscopies,” Chemical Society Reviews, vol. 37, no. 5, pp. 898–911, 2008.
[22]  R. Bardhan, W. Chen, C. Perez-Torres et al., “Nanoshells with targeted simultaneous enhancement of magnetic and optical imaging and photothermal therapeutic response,” Advanced Functional Materials, vol. 19, no. 24, pp. 3901–3909, 2009.
[23]  J. R. Cole, N. A. Mirin, M. W. Knight, G. P. Goodrich, and N. J. Halas, “Photothermal efficiencies of nanoshells and nanorods for clinical therapeutic applications,” The Journal of Physical Chemistry C, vol. 113, no. 28, pp. 12090–12094, 2009.
[24]  Y. Wang, W. Qian, Y. Tan, and S. Ding, “A label-free biosensor based on gold nanoshell monolayers for monitoring biomolecular interactions in diluted whole blood,” Biosensors and Bioelectronics, vol. 23, no. 7, pp. 1166–1170, 2008.
[25]  A. F. Low, G. J. Tearney, B. E. Bouma, and I. K. Jang, “Technology Insight: optical coherence tomography-current status and future development,” Nature Clinical Practice Cardiovascular Medicine, vol. 3, pp. 154–162, 2006.
[26]  J. Chen, F. Saeki, B. J. Wiley et al., “Gold nanocages: bioconjugation and their potential use as optical imaging contrast agents,” Nano Letters, vol. 5, no. 3, pp. 473–477, 2005.
[27]  R. A. Jensen, J. Sherin, and S. R. Emory, “Single nanoparticle based optical pH probe,” Applied Spectroscopy, vol. 61, no. 8, pp. 832–838, 2007.
[28]  S. Keren, C. Zavaleta, Z. Cheng, A. De La Zerda, O. Gheysens, and S. S. Gambhir, “Noninvasive molecular imaging of small living subjects using Raman spectroscopy,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 15, pp. 5844–5849, 2008.
[29]  M. Rycenga, Z. Wang, E. Gordon et al., “Probing the photothermal effect of gold-based nanocages with surface-enhanced Raman scattering (SERS),” Angewandte Chemie, vol. 48, no. 52, pp. 9769–9927, 2009.
[30]  M. S. Yavuz, Y. Cheng, J. Chen et al., “Gold nanocages covered by smart polymers for controlled release with near-infrared light,” Nature Materials, vol. 8, no. 12, pp. 935–939, 2009.
[31]  L. Au, J. Chen, L. V. Wang, and Y. Xia, “Gold nanocages for cancer imaging and therapy,” Methods Molecular Biology, vol. 624, pp. 83–99, 2010.
[32]  V. P. Zharov, K. E. Mercer, E. N. Galitovskaya, and M. S. Smeltzer, “Photothermal nanotherapeutics and nanodiagnostics for selective killing of bacteria targeted with gold nanoparticles,” Biophysical Journal, vol. 90, no. 2, pp. 619–627, 2006.
[33]  C. Loo, A. Lowery, N. Halas, J. West, and R. Drezek, “Immunotargeted nanoshells for integrated cancer imaging and therapy,” Nano Letters, vol. 5, no. 4, pp. 709–711, 2005.
[34]  X. Huang, I. H. El-Sayed, W. Qian, and M. A. El-Sayed, “Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods,” Journal of the American Chemical Society, vol. 128, no. 6, pp. 2115–2120, 2006.
[35]  X. Huang, P. K. Jain, I. H. El-Sayed, and M. A. El-Sayed, “Determination of the minimum temperature required for selective photothermal destruction of cancer cells with the use of immunotargeted gold nanoparticles,” Photochemistry and Photobiology, vol. 82, no. 2, pp. 412–417, 2006.
[36]  X. Huang, W. Qian, I. H. El-Sayed, and M. A. El-Sayed, “The potential use of the enhanced nonlinear properties of gold nanospheres in photothermal cancer therapy,” Lasers in Surgery and Medicine, vol. 39, no. 9, pp. 747–753, 2007.
[37]  J. Chen, D. Wang, J. Xi et al., “Immuno gold nanocages with tailored optical properties for targeted photothermal destruction of cancer cells,” Nano Letters, vol. 7, no. 5, pp. 1318–1322, 2007.
[38]  L. Tong, Y. Zhao, T. B. Huff, M. N. Hansen, A. Wei, and J.-X. Cheng, “Gold nanorods mediate tumor cell death by compromising membrane integrity,” Advanced Materials, vol. 19, no. 20, pp. 3136–3141, 2007.
[39]  W. Cai, T. Gao, H. Hong J, and Sun, “Applications of gold nanoparticles in cancer nanotechnology,” Nanotechnology, Science and Applications, vol. 1, pp. 17–32, 2008.
[40]  B. Merchant, “Gold, the Noble metal and the paradoxes of its toxicology,” Biologicals, vol. 26, no. 1, pp. 49–59, 1998.
[41]  C. Huang, Y. Hao, J. Nyagilo, D. P. Dave, L. Xu, and X. Sun, “Porous Hollow gold nanoparticles for cancer SERS imaging,” Journal of Nano Research, vol. 10, pp. 137–148, 2010.
[42]  C. Huang, J. Jiangs, M. Lu, L. Sun, E. I. Meletis, and Y. Hao, “Capturing electrochemically evolved nanobubbles by electroless deposition. A facile route to the synthesis of hollow nanoparticles,” Nano Letters, vol. 9, no. 12, pp. 4297–4301, 2009.
[43]  Y. Sun and Y. Xia, “Shape-controlled synthesis of gold and silver nanoparticles,” Science, vol. 298, no. 5601, pp. 2176–2179, 2002.
[44]  N. R. Jana, L. Gearheart, and C. J. Murphy, “Wet chemical synthesis of high aspect ratio cylindrical gold nanorods,” The Journal of Physical Chemistry B, vol. 105, no. 19, pp. 4065–4067, 2001.
[45]  E. C. Cho, Q. Zhang, and Y. Xia, “The effect of sedimentation and diffusion on cellular uptake of gold nanoparticles,” Nature Nanotechnolgy, vol. 6, pp. 385–391, 2011.
[46]  B. C. Heng, X. Zhao, S. Xiong, K. W. Ng, F. Y.-C. Boey, and J. S.-C. Loo, “Cytotoxicity of zinc oxide (ZnO) nanoparticles is influenced by cell density and culture format,” Archives of Toxicology, vol. 85, no. 6, pp. 695–704, 2011.
[47]  C. Uboldi, D. Bonacchi, G. Lorenzi et al., “Gold nanoparticles induce cytotoxicity in the alveolar type-II cell lines A549 and NCIH441,” Particle and Fibre Toxicology, vol. 6, article 18, 2009.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413