DOI QR코드

DOI QR Code

PLGA-Loaded Gold-Nanoparticles Precipitated with Quercetin Downregulate HDAC-Akt Activities Controlling Proliferation and Activate p53-ROS Crosstalk to Induce Apoptosis in Hepatocarcinoma Cells

  • Bishayee, Kausik (Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University) ;
  • Khuda-Bukhsh, Anisur Rahman (Cytogenetics and Molecular Biology Laboratory, Department of Zoology, University of Kalyani) ;
  • Huh, Sung-Oh (Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University)
  • Received : 2014.12.11
  • Accepted : 2015.03.04
  • Published : 2015.06.30

Abstract

Controlled release of medications remains the most convenient way to deliver drugs. In this study, we precipitated gold nanoparticles with quercetin. We loaded gold-quercetin into poly(DL-lactide-co-glycolide) nanoparticles (NQ) and tested the biological activity of NQ on HepG2 hepatocarcinoma cells to acquire the sustained release property. We determined by circular dichroism spectroscopy that NQ effectively caused conformational changes in DNA and modulated different proteins related to epigenetic modifications and c ell cycle control. The mitochondrial membrane potential (MMP), reactive oxygen species (ROS), cell cycle, apoptosis, DNA damage, and caspase 3 activity were analyzed by flow cytometry, and the expression profiles of different anti- and pro-apoptotic as well as epigenetic signals were studied by immunoblotting. A cytotoxicity assay indicated that NQ preferentially killed cancer cells, compared to normal cells. NQ interacted with HepG2 cell DNA and reduced histone deacetylases to control cell proliferation and arrest the cell cycle at the sub-G stage. Activities of cell cycle-related proteins, such as $p21^{WAF}$, cdk1, and pAkt, were modulated. NQ induced apoptosis in HepG2 cells by activating p53-ROS crosstalk and induces epigenetic modifications leading to inhibited proliferation and cell cycle arrest.

Keywords

References

  1. Ajazuddin, and Saraf, S. (2010). Applications of novel drug delivery system for herbal formulations. Fitoterapia 81, 680-689. https://doi.org/10.1016/j.fitote.2010.05.001
  2. Arvizo, R., Bhattacharya, R., and Mukherjee, P. (2010). Gold nanoparticles: opportunities and challenges in nanomedicine. Expert Opin. Drug Deliv. 7, 753-763. https://doi.org/10.1517/17425241003777010
  3. Bhadoriya, S.S., Mangal, A., Madoriya, N., and Dixit, P. (2011). Bioavailability and bioactivity enhancement of herbal drugs by "Nanotechnology": a review. J. Curr. Pharmaceutical. Res. 8, 1-7.
  4. Bhattacharyya, S.S., Paul, S., De, A., Das, D., Samadder, A., Boujedaini, N., and Khuda-Bukhsh, A.R. (2011). Poly (lactideco- glycolide) acid nanoencapsulation of a synthetic coumarin: Cytotoxicity and bio-distribution in mice, in cancer cell line and interaction with calf thymus DNA as target. Toxicol. Appl. Pharmacol. 253, 270-282. https://doi.org/10.1016/j.taap.2011.04.010
  5. Bishayee, K., Ghosh, S., Mukherjee, A., Sadhukhan, R., Mondal, J., and Khuda-Bukhsh, A.R. (2013a). Quercetin induces cytochrome c release and ROS accumulation to promote apoptosis and arrest the cell cycle in G2/M, in cervical carcinoma: signal cascade and drug-DNA interaction. Cell Prolif. 46, 153-163. https://doi.org/10.1111/cpr.12017
  6. Bishayee, K., Paul, A., Ghosh, S., Sikdar, S., Mukherjee, A., Biswas, R., Boujedaini, N., and Khuda-Bukhsh, A.R. (2013b). Condurango-glycoside-A fraction of Gonolobus condurango induces DNA damage associated senescence and apoptosis via ROS-dependent p53 signalling pathway in HeLa cells. Mol. Cell. Biochem. 382, 173-183. https://doi.org/10.1007/s11010-013-1732-5
  7. Brannon-Peppas, L., and Blanchette, O.J. (2004). Nanoparticle and targeted systems for cancer therapy. Adv. Drug Deliv. Rev. 56, 1649-1659. https://doi.org/10.1016/j.addr.2004.02.014
  8. Daugaard, M., Nitsch, R., Razaghi, B., McDonald, L., Jarrar, A., Torrino, S., Castillo-Lluva, S., Rotblat, B., Li, L., Malliri, A., et al. (2013). Hace1 controls ROS generation of vertebrate Rac1- dependent NADPH oxidase complexes. Nat. Commun. 4, 2180
  9. Duan, H., Heckman, C.A., and Boxer, L.M. (2005). Histone deacetylase inhibitors down-regulate bcl-2 expression and induce apoptosis in t(14;18) lymphomas. Mol. Cell. Biol. 25, 1608-1619. https://doi.org/10.1128/MCB.25.5.1608-1619.2005
  10. Falkenberg, K.J., and Johnstone, R.W. (2014). Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat. Rev. Drug Discov. 13, 673-691. https://doi.org/10.1038/nrd4360
  11. Fulda, S., and Debatin, K.M. (2006). Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 25, 4798-4811. https://doi.org/10.1038/sj.onc.1209608
  12. Ghosh, S., Bishayee, K., and Khuda-Bukhsh, A.R. (2014). graveoline isolated from ethanolic extract of ruta graveolens triggers apoptosis and autophagy in skin melanoma cells: a novel apoptosis-independent autophagic signaling pathway. Phytother. Res. 28, 1153-1162 https://doi.org/10.1002/ptr.5107
  13. Hagelkruys, A., Sawicka, A., Rennmayr, M., and Seiser, C. (2011). The biology of HDAC in cancer: the nuclear and epigenetic components. Handb. Exp. Pharmacol. 206, 13-37. https://doi.org/10.1007/978-3-642-21631-2_2
  14. Hanahan, D., and Weinberg, R.A. (2011). Hallmarks of cancer: the next generation. Cell 144, 646-674. https://doi.org/10.1016/j.cell.2011.02.013
  15. Harper, J.W., Adami, G.R., Wei, N., Keyomarsi, K., and Elledge, S.J. (1993). The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell. 75, 805-816. https://doi.org/10.1016/0092-8674(93)90499-G
  16. Havsteen, B. (2002). The biochemistry and medical significance of the flavonoids. Pharmacol. Ther. 6, 67-202. https://doi.org/10.1006/mthe.2002.0633
  17. Huang, B.H., Laban, M., Leung, C.H., Lee, L., Lee, C.K., and Salto- Tellez, M. (2005). Inhibition of histone deacetylase 2 increases apoptosis and p21Cip1/WAF1 expression, independent of histone deacetylase 1. Cell Death Differ. 12, 395-404. https://doi.org/10.1038/sj.cdd.4401567
  18. Hurley, L.H. (2002). DNA and its associated processes as targets for cancer therapy. Nat. Rev. Cancer 2, 188-200. https://doi.org/10.1038/nrc749
  19. Jain, S., Hirst, D.G., and O'Sullivan, J.M. (2012). Gold nanoparticles as novel agents for cancer therapy. Br. J. Radiol. 85, 101-113. https://doi.org/10.1259/bjr/59448833
  20. Karbowski, M., and Youle, R.J. (2003). Dynamics of mitochondrial morphology in healthy cells and during apoptosis. Cell Death Differ. 10, 870-880. https://doi.org/10.1038/sj.cdd.4401260
  21. Kim, M.J., Sun, Y., Yang, H., Kim, N.H., Jeon, S.H., and Huh, SO. (2012). Involvement of the cAMP response element binding protein, CREB, and cyclin D1 in LPA-induced proliferation of P19 embryonic carcinoma cells. Mol. Cells 34, 323-328. https://doi.org/10.1007/s10059-012-0163-6
  22. Kumari, A., Yadav, S.K., and Yadav, S.C. (2010). Biodegradable polymeric nanoparticles based drug delivery systems. Colloid. Surfaces. B. 75, 1-18. https://doi.org/10.1016/j.colsurfb.2009.09.001
  23. Lee, H., and Odom, T.W. (2015). Controlling ligand density on nanoparticles as a means to enhance biological activity. Nanomedicine 10, 177-180. https://doi.org/10.2217/nnm.14.204
  24. Lemarie, A., and Grimm, S. (2011). Mitochondrial respiratory chain complexes: apoptosis sensors mutated in cancer. Oncogene 300, 3985-4003.
  25. Liu, S. (2012). Epigenetics advancing personalied nanomedicine in cancer therapy. Adv. Drug Deliv. Rev. 64, 1532-1543. https://doi.org/10.1016/j.addr.2012.08.004
  26. Loo, D.T. (2002). TUNEL Assay: An Overview of Techniques. In in situ detection of DNA damage: methods and protocol, Didenko, V.V. eds. (Spinger), pp. 313-323.
  27. Maitland, M.L., and Schilsky, R.L. (2011). Clinical trials in the era of personalized oncology. CA Cancer J. Clin. 61, 365-381. https://doi.org/10.3322/caac.20135
  28. Marks, P.A., Richon, V.M., and Rifkind, R.A. (2000). Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J. Natl. Cancer Inst. 92, 1210-1216. https://doi.org/10.1093/jnci/92.15.1210
  29. Mikhailov, V., Mikhailova, M., Pulkrabek, D.J., Dong, Z., Venkatachalam, M.A., and Saikumar, P. (2001). Bcl-2 prevents Bax oligomerization in the mitochondrial outer membrane. J. Biol. Chem. 276, 18361-18374. https://doi.org/10.1074/jbc.M100655200
  30. Mosmann, T. (1983). Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Methods 65, 55-63. https://doi.org/10.1016/0022-1759(83)90303-4
  31. Noh, J.H., Jung, K.H., Kim, J.K., Eun, J.W., Bae, H.J., and Xie, H.J. (2011). Aberrant regulation of HDAC2 mediates proliferation of hepatocellular carcinoma cells by deregulating expression of G1/S cell cycle proteins. PLoS One 6, e28103. https://doi.org/10.1371/journal.pone.0028103
  32. Puhl, A.C., Fagundes, M., dos Santos, K.C., Polikarpov, I., das Gracas, M.F., da Silva, F., Fernandes, J.B., Vieira, P.C., and Forim, M.R. (2011). Preparation and characterization of polymeric nanoparticles loaded with the flavonoid luteolin, by using factorial design. Int. J. Drug Deliv. 3, 683-698.
  33. Shahabuddin, M.S., Gopal, M., and Raghavan, S.C. (2009). Intercalating cytotoxic, anti-tumour activity of 8-chloro and 4- morp96holinopyrimido [4',5':4.5]thieno(2,3-b)quinolone. J. Photochem. Photobiol. B. 94, 13-19. https://doi.org/10.1016/j.jphotobiol.2008.09.001
  34. Shahbazian, D., Sznol, J., and Kluger, H.M. (2012). Vertical pathway targeting in cancer therapy. Adv. Pharmacol. 65, 1-26. https://doi.org/10.1016/B978-0-12-397927-8.00001-4
  35. Shobha Devi, C., Anil Kumar, D., Singh, S.S., Gabra, N., Deepika, N., Kumar, Y.P., and Satyanarayana, S. (2013). Synthesis, interaction with DNA, cytotoxicity, cell cycle arrest and apoptotic inducing properties of ruthenium(II) molecular "light switch" complexes. Eur. J. Med. Chem. 64, 410-421. https://doi.org/10.1016/j.ejmech.2013.04.006
  36. Talluri, S., and Dick, F.A. (2012). Regulation of transcription and chromatin structure by pRB. Cell Cycle 11, 3189-3198. https://doi.org/10.4161/cc.21263
  37. Vijayababu, M.R., Kanagaraj, P., Arunkumar, A., Ilangovan, R., Dharmarajan, A., and Arunakaran, J. (2006). Quercetin induces p53-independent apoptosis in human prostate cancer cells by modulating Bcl-2-related proteins: a possible mediation by IGFBP-3. Oncol. Res. 16, 67-74. https://doi.org/10.3727/000000006783981224
  38. Yu, T., Robotham, J.L., and Yoon, Y. (2006). Increased production of reactive oxygen species in hyperglycemic conditions requires dynamic change of mitochondrial morphology. Proc. Natl. Acad. Sci. USA 103, 2653-2658. https://doi.org/10.1073/pnas.0511154103

Cited by

  1. CS-PEG decorated PLGA nano-prototype for delivery of bioactive compounds: A novel approach for induction of apoptosis in HepG2 cell line vol.62, pp.2, 2017, https://doi.org/10.1016/j.advms.2017.01.003
  2. Reactive Oxygen Species and Targeted Therapy for Pancreatic Cancer vol.2016, 2016, https://doi.org/10.1155/2016/1616781
  3. Oxidative Stress: A New Target for Pancreatic Cancer Prognosis and Treatment vol.6, pp.3, 2017, https://doi.org/10.3390/jcm6030029
  4. The inhibition of histone deacetylase 8 suppresses proliferation and inhibits apoptosis in gastric adenocarcinoma vol.47, pp.5, 2015, https://doi.org/10.3892/ijo.2015.3182
  5. Application of Bioactive Quercetin in Oncotherapy: From Nutrition to Nanomedicine vol.21, pp.1, 2016, https://doi.org/10.3390/molecules21010108
  6. Impact of Phytochemicals and Dietary Patterns on Epigenome and Cancer vol.69, pp.2, 2017, https://doi.org/10.1080/01635581.2017.1263746
  7. Hyaluronan suppresses lidocaine-induced apoptosis of human chondrocytes in vitro by inhibiting the p53-dependent mitochondrial apoptotic pathway vol.37, pp.5, 2016, https://doi.org/10.1038/aps.2015.151
  8. Sulforaphane potentiates growth-inhibiting and apoptosis-promoting activities of cisplatin following oxidative stress and mitochondrial dysfunction in malignant mesothelioma cells vol.12, pp.3, 2016, https://doi.org/10.1007/s13273-016-0034-x
  9. Effects of quercetin on pharmacokinetics of cefprozil in Chinese-Han male volunteers vol.46, pp.10, 2016, https://doi.org/10.3109/00498254.2015.1132792
  10. The effect of quercetin nanoparticle on cervical cancer progression by inducing apoptosis, autophagy and anti-proliferation via JAK2 suppression vol.82, 2016, https://doi.org/10.1016/j.biopha.2016.05.029
  11. Quercetin as an Emerging Anti-Melanoma Agent: A Four-Focus Area Therapeutic Development Strategy vol.3, 2016, https://doi.org/10.3389/fnut.2016.00048
  12. Cytotoxicity, Oxidative Stress, Cell Cycle Arrest, and Mitochondrial Apoptosis after Combined Treatment of Hepatocarcinoma Cells with Maleic Anhydride Derivatives and Quercetin vol.2017, pp.1942-0994, 2017, https://doi.org/10.1155/2017/2734976
  13. Food Bioactive HDAC Inhibitors in the Epigenetic Regulation of Heart Failure vol.10, pp.8, 2018, https://doi.org/10.3390/nu10081120
  14. Anticancer potential of quercetin: A comprehensive review pp.0951418X, 2018, https://doi.org/10.1002/ptr.6155
  15. Regulation of the Intracellular ROS Level Is Critical for the Antiproliferative Effect of Quercetin in the Hepatocellular Carcinoma Cell Line HepG2 pp.1532-7914, 2019, https://doi.org/10.1080/01635581.2018.1559929
  16. Quercetin induces cell apoptosis of myeloma and displays a synergistic effect with dexamethasone in vitro and in vivo xenograft models vol.7, pp.29, 2015, https://doi.org/10.18632/oncotarget.9993
  17. Gold-quercetin nanoparticles prevent metabolic endotoxemia-induced kidney injury by regulating TLR4/NF-κB signaling and Nrf2 pathway in high fat diet fed mice vol.12, pp.None, 2015, https://doi.org/10.2147/ijn.s116010
  18. Quercetin Loaded Nanoparticles in Targeting Cancer: Recent Development vol.19, pp.13, 2019, https://doi.org/10.2174/1871520619666190705150214
  19. Dietary Compounds as Epigenetic Modulating Agents in Cancer vol.10, pp.None, 2019, https://doi.org/10.3389/fgene.2019.00079
  20. Characterization and DNA methylation modulatory activity of gold nanoparticles synthesized by Pseudoalteromonas strain vol.44, pp.1, 2015, https://doi.org/10.1007/s12038-018-9842-6
  21. Quercetin shows anti‐tumor effect in hepatocellular carcinoma LM3 cells by abrogating JAK2/STAT3 signaling pathway vol.8, pp.10, 2019, https://doi.org/10.1002/cam4.2388
  22. Pharmaceutical Topical Delivery of Poorly Soluble Polyphenols: Potential Role in Prevention and Treatment of Melanoma vol.20, pp.6, 2019, https://doi.org/10.1208/s12249-019-1457-1
  23. Antitumor Effects of Quercetin in Hepatocarcinoma In Vitro and In Vivo Models: A Systematic Review vol.11, pp.12, 2015, https://doi.org/10.3390/nu11122875
  24. A New Histone Deacetylase Inhibitor, MHY4381, Induces Apoptosis via Generation of Reactive Oxygen Species in Human Prostate Cancer Cells vol.28, pp.2, 2020, https://doi.org/10.4062/biomolther.2019.074
  25. Local anesthetics counteract cell proliferation and migration of human triple‐negative breast cancer and melanoma cells vol.235, pp.4, 2020, https://doi.org/10.1002/jcp.29236
  26. Nanoencapsulated dietary polyphenols for cancer prevention and treatment: successes and challenges vol.15, pp.11, 2015, https://doi.org/10.2217/nnm-2019-0398
  27. Genome-Protecting Compounds as Potential Geroprotectors vol.21, pp.12, 2015, https://doi.org/10.3390/ijms21124484
  28. Quercetin-loaded nanoparticles enhance cytotoxicity and antioxidant activity on C6 glioma cells vol.25, pp.6, 2015, https://doi.org/10.1080/10837450.2020.1740933
  29. Cholesterol partially rescues the inhibition effect of pravastatin on keratinocytes proliferation by regulating cell cycle relative proteins through AKT and ERK pathway vol.33, pp.6, 2015, https://doi.org/10.1111/dth.14305
  30. Near IR responsive targeted integrated lipid polymer nanoconstruct for enhanced magnolol cytotoxicity in breast cancer vol.10, pp.None, 2020, https://doi.org/10.1038/s41598-020-65521-z
  31. New Insights Toward Nanostructured Drug Delivery of Plant-Derived Polyphenol Compounds: Cancer Treatment and Gene Expression Profiles vol.21, pp.None, 2021, https://doi.org/10.2174/1568009621666210525152802
  32. Decoding the Mechanism of Shen Qi Sha Bai Decoction in Treating Acute Myeloid Leukemia Based on Network Pharmacology and Molecular Docking vol.9, pp.None, 2015, https://doi.org/10.3389/fcell.2021.796757
  33. Targeting epigenetics in cancer: therapeutic potential of flavonoids vol.61, pp.10, 2015, https://doi.org/10.1080/10408398.2020.1763910
  34. Quercetin nanoformulations: a promising strategy for tumor therapy vol.12, pp.15, 2015, https://doi.org/10.1039/d1fo00851j
  35. Quercetin as a protective agent for liver diseases: A comprehensive descriptive review of the molecular mechanism vol.35, pp.9, 2015, https://doi.org/10.1002/ptr.7104
  36. Quercetin against MCF7 and CAL51 breast cancer cell lines: apoptosis, gene expression and cytotoxicity of nano-quercetin vol.16, pp.22, 2015, https://doi.org/10.2217/nnm-2021-0070
  37. Functional Depletion of HSP72 by siRNA and Quercetin Enhances Vorinostat-Induced Apoptosis in an HSP72-Overexpressing Cutaneous T-Cell Lymphoma Cell Line, Hut78 vol.22, pp.20, 2015, https://doi.org/10.3390/ijms222011258
  38. Nanotechnology Innovations to Enhance the Therapeutic Efficacy of Quercetin vol.11, pp.10, 2015, https://doi.org/10.3390/nano11102658
  39. Herbal and Natural Dietary Products: Upcoming Therapeutic Approach for Prevention and Treatment of Hepatocellular Carcinoma vol.73, pp.11, 2015, https://doi.org/10.1080/01635581.2020.1834591