Skip to main content
Log in

Curcumin-encapsulated polymeric nanoparticles for metastatic osteosarcoma cells treatment

负载姜黄素的聚合物纳米粒子抑制转移性骨肉瘤细胞增值和迁移

  • Articles
  • Published:
Science China Materials Aims and scope Submit manuscript

Abstract

Osteosarcoma is a high-class malignant bone cancer with a less than 20% five-year survival rate due to its early metastasis potential. There is an urgent need to develop a versatile and innoxious drug to treat metastatic osteosarcoma. Curcumin (Cur) has shown its potential for the treatment of many cancers; however, the clinical implication of native curcumin is severely hindered by its intrinsic property. In this study, a mixed system of monomethoxy (polyethylene glycol)-poly(d, l-lactide-co-glycolide)/poly(ε-caprolactone) (mPEG-PLGA/PCL) was used to build a formulation of curcumin-encapsulated nanoparticles (Cur-NPs), which significantly improved the solubility, stability and cellular uptake of curcumin. Moreover, the Cur-NPs were superior to free curcumin in the matter of inhibition on the proliferation, migration and invasion of osteosarcoma 143B cells. It was found that both free curcumin and Cur-NPs could decrease the expressions of c-Myc and MMP7 in the level of mRNA and protein, which explained why free curcumin and Cur-NPs could inhibit the proliferation and invasion of metastatic osteosarcoma 143B cells. The Cur-NPs provided a promising strategy for metastatic osteosarcoma treatment.

摘要

骨肉瘤是一种高转移性的恶性肿瘤, 5年生存率不到20%.姜黄素(Cur)具有治疗癌症的潜在功效, 但其自身水溶性和稳定性差等性质限制了其临床使用. 本文用聚乙二醇-聚(D, L-丙交酷-乙交酷)/聚(ε-已内酷)负载姜黄素形成纳米粒子, 可以改善姜黄素的水溶性、 稳定性和细胞内吞. 与非负载的姜黄素相比, 姜黄素纳米粒子在抑制骨肉瘤细胞增殖和迁移方面显示出明显的优势. 研究结果表明, 姜黄素和其纳米粒子均可降低c-Myc和MMP7表达, 这可以很好地解释姜黄素纳米粒子抑制骨肉瘤细胞的机制.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Kansara M, Teng MW, Smyth MJ, et al. Translational biology of osteosarcoma. Nat Rev Cancer, 2014, 14: 722–735

    Article  Google Scholar 

  2. Grimer RJ. Surgical options for children with osteosarcoma. Lancet Oncology, 2005, 6: 85–92

    Article  Google Scholar 

  3. Luetke A, Meyers PA, Lewis I, et al. Osteosarcoma treatment–Where do we stand? A state of the art review. Cancer Treatment Rev, 2014, 40: 523–532

    Article  Google Scholar 

  4. Li Y, Rogoff HA, Keates S, et al. Suppression of cancer relapse and metastasis by inhibiting cancer stemness. Proc Natl Acad Sci USA, 2015, 112: 1839–1844

    Article  Google Scholar 

  5. Benjamin RS. Osteosarcoma: better treatment through better trial design. Lancet Oncology, 2015, 16: 12–13

    Article  Google Scholar 

  6. Kunnumakkara AB, Anand P, Aggarwal BB. Curcumin inhibits proliferation, invasion, angiogenesis and metastasis of different cancers through interaction with multiple cell signaling proteins. Cancer Lett, 2008, 269: 199–225

    Article  Google Scholar 

  7. Heger M, van Golen RF, Broekgaarden M, et al. The molecular basis for the pharmacokinetics and pharmacodynamics of curcumin and its metabolites in relation to cancer. Pharmacol Rev, 2014, 66: 222–307

    Article  Google Scholar 

  8. Cheng AL, Hsu HC, Lin JK, et al. Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions. Anticancer Res, 2001, 21: 2895–2900

    Google Scholar 

  9. Dhillon N, Aggarwal BB, Newman RA, et al. Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin Cancer Res, 2008, 14: 4491–4499

    Article  Google Scholar 

  10. Liu L, Sun L, Wu Q, et al. Curcumin loaded polymeric micelles inhibit breast tumor growth and spontaneous pulmonary metastasis. Int J Pharm, 2013, 443: 175–182

    Article  Google Scholar 

  11. Gao X, Zheng F, Guo G, et al. Improving the anti-colon cancer activity of curcumin with biodegradable nano-micelles. J Mater Chem B, 2013, 1: 5778

    Article  Google Scholar 

  12. Yallapu MM, Nagesh PKB, Jaggi M, et al. Therapeutic applications of curcumin nanoformulations. AAPS J, 2015, 17: 1341–1356

    Article  Google Scholar 

  13. Chang Z, Xing J, Yu X. Curcumin induces osteosarcoma MG63 cells apoptosis via ROS/Cyto-C/Caspase-3 pathway. Tumor Biol, 2014, 35: 753–758

    Article  Google Scholar 

  14. Chang R, Sun L, Webster TJ. Selective inhibition of MG-63 osteosarcoma cell proliferation induced by curcumin-loaded selfassembled arginine-rich-RGD nanospheres. Int J Nanomedicine, 2015, 1: 3351

    Google Scholar 

  15. Chen P, Wang H, Yang F, et al. Curcumin promotes osteosarcoma cell death by activating MiR-125a/ERRα signal pathway. J Cell Biochem, 2017, 118: 74–81

    Article  Google Scholar 

  16. Walters DK, Muff R, Langsam B, et al. Cytotoxic effects of curcumin on osteosarcoma cell lines. Invest New Drugs, 2008, 26: 289–297

    Article  Google Scholar 

  17. Jin S, Xu H, Shen J, et al. Apoptotic effects of curcumin on human osteosarcoma U2OS cells. Orthop Surg, 2009, 1: 144–152

    Article  Google Scholar 

  18. Fossey SL, Bear MD, Lin J, et al. The novel curcumin analog FLLL32 decreases STAT3 DNA binding activity and expression, and induces apoptosis in osteosarcoma cell lines. BMC Cancer, 2011, 11: 112

    Article  Google Scholar 

  19. Peng SF, Lee CY, Hour MJ, et al. Curcumin-loaded nanoparticles enhance apoptotic cell death of U2OS human osteosarcoma cells through the Akt-Bad signaling pathway. Int J Oncol, 2014, 44: 238–246

    Article  Google Scholar 

  20. Fatima MT, Chanchal A, Yavvari PS, et al. Cell permeating nanocomplexes of amphiphilic polyelectrolytes enhance solubility, stability, and anti-cancer efficacy of curcumin. Biomacromolecules, 2016, 17: 2375–2383

    Article  Google Scholar 

  21. Si M, Zhao J, Li X, et al. Reversion effects of curcumin on multidrug resistance of MNNG/HOS human osteosarcoma cells in vitro and in vivo through regulation of P-glycoprotein. Chin Med J, 2013, 126: 4116–4123

    Google Scholar 

  22. Dhule SS, Penfornis P, Frazier T, et al. Curcumin-loaded γ-cyclodextrin liposomal nanoparticles as delivery vehicles for osteosarcoma. Nanomed-Nanotech Biol Med, 2012, 8: 440–451

    Article  Google Scholar 

  23. Luu HH, Kang Q, Park JK, et al. An orthotopic model of human osteosarcoma growth and spontaneous pulmonary metastasis. Clin Exp Metastasis, 2005, 22: 319–329

    Article  Google Scholar 

  24. He N, Zhang Z. Baicalein suppresses the viability of MG-63 osteosarcoma cells through inhibiting c-MYC expression via Wnt signaling pathway. Mol Cell Biochem, 2015, 405: 187–196

    Article  Google Scholar 

  25. Lin CY, Lovén J, Rahl PB, et al. Transcriptional amplification in tumor cells with elevated c-Myc. Cell, 2012, 151: 56–67

    Article  Google Scholar 

  26. Dang CV. MYC on the path to cancer. Cell, 2012, 149: 22–35

    Article  Google Scholar 

  27. Wu CH, van Riggelen J, Yetil A, et al. Cellular senescence is an important mechanism of tumor regression upon c-Myc inactivation. Proc Natl Acad Sci USA, 2007, 104: 13028–13033

    Article  Google Scholar 

  28. Lin L, Zhang JH, Panicker LM, et al. The parafibromin tumor suppressor protein inhibits cell proliferation by repression of the cmyc proto-oncogene. Proc Natl Acad Sci USA, 2008, 105: 17420–17425

    Article  Google Scholar 

  29. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell, 2010, 141: 52–67

    Article  Google Scholar 

  30. Szarvas T, Becker M, vom Dorp F, et al. Matrix metalloproteinase-7 as a marker of metastasis and predictor of poor survival in bladder cancer. Cancer Sci, 2010, 101: 1300–1308

    Article  Google Scholar 

  31. Hu XT, Zhang FB, Fan YC, et al. Phospholipase C delta 1 is a novel 3p22.3 tumor suppressor involved in cytoskeleton organization, with its epigenetic silencing correlated with high-stage gastric cancer. Oncogene, 2009, 28: 2466–2475

    Article  Google Scholar 

  32. Sakamoto N, Naito Y, Oue N, et al. MicroRNA-148a is downregulated in gastric cancer, targets MMP7, and indicates tumor invasiveness and poor prognosis. Cancer Sci, 2014, 105: 236–243

    Article  Google Scholar 

  33. Song N, Liu H, Ma X, et al. Placental growth factor promotes metastases of ovarian cancer through MiR-543-regulated MMP7. Cell Physiol Biochem, 2015, 37: 1104–1112

    Article  Google Scholar 

  34. Han G, Wang Y, Bi W. c-Myc overexpression promotes osteosarcoma cell invasion via activation of MEK-ERK pathway. Oncol Res Feat Preclin Clin Cancer Therap, 2012, 20: 149–156

    Google Scholar 

  35. Song W, Tang Z, Li M, et al. Polypeptide-based combination of paclitaxel and cisplatin for enhanced chemotherapy efficacy and reduced side-effects. Acta Biomater, 2014, 10: 1392–1402

    Article  Google Scholar 

  36. Song W, Tang Z, Lei T, et al. Stable loading and delivery of disulfiram with mPEG-PLGA/PCL mixed nanoparticles for tumor therapy. Nanomed-Nanotech Biol Med, 2016, 12: 377–386

    Article  Google Scholar 

  37. Franken NAP, Rodermond HM, Stap J, et al. Clonogenic assay of cells in vitro. Nat Protoc, 2006, 1: 2315–2319

    Article  Google Scholar 

  38. Yallapu MM, Khan S, Maher DM, et al. Anti-cancer activity of curcumin loaded nanoparticles in prostate cancer. Biomaterials, 2014, 35: 8635–8648

    Article  Google Scholar 

  39. Tian J, Min Y, Rodgers Z, et al. Nanoparticle delivery of chemotherapy combination regimen improves the therapeutic efficacy in mouse models of lung cancer. Nanomed-Nanotech Biol Med, 2017, 13: 1301–1307

    Article  Google Scholar 

  40. Wang Z, Tan J, McConville C, et al. Poly lactic-co-glycolic acid controlled delivery of disulfiram to target liver cancer stem-like cells. Nanomed-Nanotech Biol Med, 2017, 13: 641–657

    Article  Google Scholar 

  41. Shen Y. Elastin-like polypeptide fusion for precision design of protein-polymer conjugates with improved pharmacology. Sci China Mater, 2015, 58: 767–768

    Article  Google Scholar 

  42. Zhang Y, Xiao CS, Li MQ, et al. Co-delivery of doxorubicin and paclitaxel with linear-dendritic block copolymer for enhanced anticancer efficacy. Sci China Chem, 2014, 57: 624–632

    Article  Google Scholar 

  43. Xu C, Tian H, Chen X. Recent progress in cationic polymeric gene carriers for cancer therapy. Sci China Chem, 2017, 60: 319–328

    Article  Google Scholar 

  44. Wang J, Liu Y, Ma Y, et al. NIR-activated supersensitive drug release using nanoparticles with a flow core. Adv Funct Mater, 2016, 26: 7516–7525

    Article  Google Scholar 

  45. Li D, Ma Y, Du J, et al. Tumor acidity/NIR controlled interaction of transformable nanoparticle with biological systems for cancer therapy. Nano Lett, 2017, 17: 2871–2878

    Article  Google Scholar 

  46. Leung MHM, Kee TW. Effective stabilization of curcumin by association to plasma proteins: human serum albumin and fibrinogen. Langmuir, 2009, 25: 5773–5777

    Article  Google Scholar 

  47. Cabral H, Matsumoto Y, Mizuno K, et al. Accumulation of sub-100 nm polymeric micelles in poorly permeable tumours depends on size. Nat Nanotech, 2011, 6: 815–823

    Article  Google Scholar 

  48. Li Y. Realize molecular surgical knife in tumor therapy by nanotechnology. Sci China Mater, 2015, 58: 851–851

    Article  Google Scholar 

  49. Zhen M, Shu C, Li J, et al. A highly efficient and tumor vasculartargeting therapeutic technique with size-expansible gadofullerene nanocrystals. Sci China Mater, 2015, 58: 799–810

    Article  Google Scholar 

  50. Yang X, Li Z, Wang N, et al. Curcumin-encapsulated polymeric micelles suppress the development of colon cancer in vitro and in vivo. Sci Rep, 2015, 5: 10322

    Article  Google Scholar 

  51. Wang YJ, Pan MH, Cheng AL, et al. Stability of curcumin in buffer solutions and characterization of its degradation products. J Pharm Biomed Anal, 1997, 15: 1867–1876

    Article  Google Scholar 

  52. Kaur K, Kumar R, Mehta SK. Nanoemulsion: a new medium to study the interactions and stability of curcumin with bovine serum albumin. J Mol Liquids, 2015, 209: 62–70

    Article  Google Scholar 

  53. Mohanty C, Acharya S, Mohanty AK, et al. Curcumin-encapsulated MePEG/PCL diblock copolymeric micelles: a novel controlled delivery vehicle for cancer therapy. Nanomedicine, 2010, 5: 433–449

    Article  Google Scholar 

  54. Li C, Luo T, Zheng Z, et al. Curcumin-functionalized silk materials for enhancing adipogenic differentiation of bone marrow-derived human mesenchymal stem cells. Acta Biomater, 2015, 11: 222–232

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of China (51520105004, 51673189, 51390484, 51403204, 51673185, 51473029 and 51503202), Science and Technology Service Network Initiative (KFJ-SW-STS-166), and the Chinese Academy of Sciences Youth Innovation Promotion Association.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Zhaohui Tang  (汤朝晖) or Xueqi Fu  (付学奇).

Additional information

Guanyi Wang was born in 1990, a PhD candidate in Jilin university. Currently she is a joint PhD student in Professor Zhaohui Tang and Xuesi Chen’s group. She is majoring in biochemistry and molecular biology. Her research interests mainly focus on anti-tumor therapeutics.

Zhaohui Tang was born in 1976. Currently he is a professor at the Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences. His research interests include fabrication of nano-medicine, anti-tumor therapeutics and industrial development of biodegradable medical polymer materials.

Xueqi Fu was born in 1960. Currently he is a professor at Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University. His research interests focus on the cell signal transduction and anti-tumor drug screening.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, G., Song, W., Shen, N. et al. Curcumin-encapsulated polymeric nanoparticles for metastatic osteosarcoma cells treatment. Sci. China Mater. 60, 995–1007 (2017). https://doi.org/10.1007/s40843-017-9107-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40843-017-9107-x

Keywords

Navigation