Drug Res (Stuttg) 2017; 67(04): 211-216
DOI: 10.1055/s-0042-122136
Original Article
© Georg Thieme Verlag KG Stuttgart · New York

The Effect of Chrysin Loaded PLGA-PEG on Metalloproteinase Gene Expression in Mouse 4T1 Tumor Model

Zoheyr Mohammadi
1   Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
,
Mohsen Sharif Zak
2   Departments of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
,
Khaled Seidi
1   Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
,
Meisam Barati
3   Nutrition Research Center, School of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
,
Abolfazl Akbarzadeh
4   Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
6   Universal Scientific Education and Research Network (USERN),Tabriz, Iran
,
Nosratollah Zarghami
5   Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
1   Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
2   Departments of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
› Author Affiliations
Further Information

Publication History

received 31 July 2016

accepted 08 November 2016

Publication Date:
06 February 2017 (online)

Abstract

Aim:

Despite the advances in medicine, cancer remains as one of the leading causes of the death worldwide. Through our previous study, the Chrysin loaded PGLA/PEG has been synthesized, and its physico-chemical properties were characterized. The aim of the present study was to evaluate the Chrysin loaded PGLA/PEG nanoparticle therapeutic effects on TIMP-1, TIMP-2, MMP-2, MMP-9 and PI3k expression in Mouse 4T1 breast tumor model.

Materials and methods:

30 mice were enrolled in the current study, and the mice were randomly divided into 3 groups: untreated (n=10), Chrysin treatment (n=10) and Chrysin-loaded PLGA/PEG-based treatment (n=10). 1064T1 mammary carcinoma cells subcutaneously inoculated in the flank on mice orthotopically. After the treatments, the primary tumors were isolated from the Mice under anesthesia. For RNA extraction, the isolated tissues were frozen in −70°C. RNA extraction was performed by using RNA extraction kit. The expression of TIMP-1, TIMP-2, MMP-2 and MMP-9 were measured by the real time PCR.

Results:

The study results showed the expression of TIMP-1 and TIMP-2 in Chrysin-loaded PLGA/PEG treatment groups was higher than Chrysin receiving one. Also, the results showed that the MMP-9 and MMP-2 expressions were reduced after Chrysin loaded PLGA/PEG treatment. The reduction of the mentioned genes was greater in Chrysin-loaded PLGA/PEG treatment group in comparison with Chrysin receiving group.

Conclusion:

According to our present study, expression of the mentioned genes after treatments, Chrysin; especially, Chrysin-loaded PLGA/PEG could be proposed as a new component in the cancer therapy for reducing the progression and metastasis.

 
  • References

  • 1 DeSantis CE, Bray F, Ferlay J. et al. International variation in female breast cancer incidence and mortality rates. Cancer Epidemiology Biomarkers & Prevention 2015; 10: 1495-1506
  • 2 Ferlay J, Soerjomataram I, Dikshit R. et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015; 5: E359-E386
  • 3 Zarzecki MS, Araujo SM, Bortolotto VC et al. Hypolipidemic action of chrysin on Triton WR-1339-induced hyperlipidemia in female C57BL/6 mice. Toxicology Reportshttp://dx.doi.org/10.1016/j.toxrep.2014.02.003
  • 4 Sun X, Huo X, Luo T. et al. The anticancer flavonoid chrysin induces the unfolded protein response in hepatoma cells. J Cell Mol Med 2011; 11: 2389-2398
  • 5 Munin A, Edwards-Lévy F. Encapsulation of natural polyphenolic compounds; a review. Pharmaceutics 2011; 4: 793-829
  • 6 Shi J, Votruba AR, Farokhzad OC. et al. Nanotechnology in drug delivery and tissue engineering: from discovery to applications. Nano Lett 2010; 9: 3223-3230
  • 7 Cammas S, Suzuki K, Sone C. et al. Thermo-responsive polymer nanoparticles with a core-shell micelle structure as site-specific drug carriers. J Controlled Release 1997; 2: 157-164
  • 8 Jeong B, Bae YH, Kim SW. Thermoreversible gelation of PEG-PLGA-PEG triblock copolymer aqueous solutions. Macromolecules 1999; 21: 7064-7069
  • 9 Chung JE, Yokoyama M, Okano T. Inner core segment design for drug delivery control of thermo-responsive polymeric micelles. J Controlled Release 2000; 1: 93-103
  • 10 Xia W, Chang J. Well-ordered mesoporous bioactive glasses (MBG): a promising bioactive drug delivery system. J Controlled Release 2006; 3: 522-530
  • 11 Xia W, Chang J, Lin J. et al. The pH-controlled dual-drug release from mesoporous bioactive glass/polypeptide graft copolymer nanomicelle composites. Eur J Pharm Biopharm 2008; 2: 546-552
  • 12 Cho K, Wang X, Nie S. et al. Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res 2008; 5: 1310-1316
  • 13 Zhang Z, Feng S-S. Nanoparticles of poly (lactide)/vitamin E TPGS copolymer for cancer chemotherapy: synthesis, formulation, characterization and in vitro drug release. Biomaterials 2006; 2: 262-270
  • 14 Makadia HK, Siegel SJ. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers 2011; 3: 1377-1397
  • 15 Anderson JM, Shive MS. Biodegradation and biocompatibility of PLA and PLGA microspheres. Adv Drug Del Rev 2012; 72-82
  • 16 Cheng J, Teply BA, Sherifi I. et al. Formulation of functionalized PLGA–PEG nanoparticles for in vivo targeted drug delivery. Biomaterials 2007; 5: 869-876
  • 17 Zhang X, Zeng X, Liang X. et al. The chemotherapeutic potential of PEG-b-PLGA copolymer micelles that combine chloroquine as autophagy inhibitor and docetaxel as an anti-cancer drug. Biomaterials 2014; 33: 9144-9154
  • 18 Tao W, Zeng X, Zhang J. et al. Synthesis of cholic acid-core poly (ε-caprolactone-ran-lactide)-b-poly (ethylene glycol) 1000 random copolymer as a chemotherapeutic nanocarrier for liver cancer treatment. Biomaterials Science 2014; 9: 1262-1274
  • 19 Danhier F, Ansorena E, Silva JM. et al. PLGA-based nanoparticles: an overview of biomedical applications. J Controlled Release 2012; 2: 505-522
  • 20 Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev 2006; 1: 9-34
  • 21 Mohammadzadeh G, Zarghami N. Associations between single-nucleotide polymorphisms of the adiponectin gene, serum adiponectin levels and increased risk of type 2 diabetes mellitus in Iranian obese individuals. Scandinavian Journal of Clinical and Laboratory Investigation 2009; 69: 764-771
  • 22 Talvensaari-Mattila A, Pääkkö P, Höyhtyä M. et al. Matrix metalloproteinase-2 immunoreactive protein. Cancer 1998; 6: 1153-1162
  • 23 Pellikainen JM, Ropponen KM, Kataja VV. et al. Expression of matrix metalloproteinase (MMP)-2 and MMP-9 in breast cancer with a special reference to activator protein-2, HER2, and prognosis. Clin Cancer Res 2004; 22: 7621-7628
  • 24 Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nature Reviews Cancer 2002; 3: 161-174
  • 25 Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 2001; 463
  • 26 Li W, Sun YN, Yan XT. et al. Flavonoids from Astragalus membranaceus and their inhibitory effects on LPS-stimulated pro-inflammatory cytokine production in bone marrow-derived dendritic cells. Arch Pharmacal Res 2014; 2: 186-192
  • 27 Zarghami N, Yu H. Quantification of creatine kinase BB isoenzyme in tumor cytosols and serum with an ultrasensitive time-resolved immunofluorometric technique. Clinical Biochemistry 1995; 28: 243-253
  • 28 Khokha R. Suppression of the tumorigenic and metastatic abilities of murine B16-F10 melanoma cells in vivo by the overexpression of the tissue inhibitor of the metalloproteinases-1. J Natl Cancer Inst 1994; 4: 299-304
  • 29 Mellatyar H, Akbarzadeh A, Rahmati M. et al. Comparison of inhibitory effect of 17-DMAG nanoparticles and free 17-DMAG in HSP90 gene expression in lung cancer. Asian Pacific Journal of Cancer Prevention 2014; 15: 8693-8698
  • 30 Anari E, Akbarzadeh A, Zarghami N. Chrysin-loaded PLGA-PEG nanoparticles designed for enhanced effect on the breast cancer cell line. Artificial cells, nanomedicine, and biotechnology http://dx.doi.org/10.3109/21691401.2015.1029633
  • 31 Zhou R, Xu L, Ye M. et al. Formononetin inhibits migration and invasion of MDA-MB-231 and 4T1 breast cancer cells by suppressing MMP-2 and MMP-9 through PI3K/AKT signaling pathways. Horm Metab Res 2014; 11: 753-760
  • 32 Sugahara KN, Teesalu T, Karmali PP. et al. Coadministration of a tumor-penetrating peptide enhances the efficacy of cancer drugs. Science 2010; 5981: 1031-1035
  • 33 Yang L, Wu X, Liu F. et al. Novel biodegradable polylactide/poly (ethylene glycol) micelles prepared by direct dissolution method for controlled delivery of anticancer drugs. Pharm Res 2009; 10: 2332-2342
  • 34 Zheng H, Li S, Pu Y. et al. Nanoparticles generated by PEG-Chrysin conjugates for efficient anticancer drug delivery. Eur J Pharm Biopharm 2014; 3: 454-460
  • 35 Khoo BY, Chua SL, Balaram P. Apoptotic effects of chrysin in human cancer cell lines. Int J Mol Sci 2010; 5: 2188-2199
  • 36 Swellam T, Miyanaga N, Onozawa M. et al. Antineoplastic activity of honey in an experimental bladder cancer implantation model: in vivo and in vitro studies. Int J Urol 2003; 4: 213-219
  • 37 Zhou L, Zhang P, Yang G. et al. Solubility of chrysin in ethanol and water mixtures. J Chem Eng Data 2014; 7: 2215-2220
  • 38 Scully OJ, Bay B-H, Yip G. et al. Breast cancer metastasis. Cancer Genomics-Proteomics 2012; 5: 311-320
  • 39 Alix-Panabieres C, Müller V, Pantel K. Current status in human breast cancer micrometastasis. Curr Opin Oncol 2007; 6: 558-563
  • 40 Demaria S, Kawashima N, Yang AM. et al. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res 2005; 2: 728-734
  • 41 Jezierska A, Motyl T. Matrix metalloproteinase-2 involvement in breast cancer progression: a mini-review. Med Sci Monit 2009; 2: RA32-RA40
  • 42 Vizoso F, Gonzalez L, Corte M. et al. Study of matrix metalloproteinases and their inhibitors in breast cancer. Br J Cancer 2007; 6: 903-911
  • 43 Khokha R, Zimmer MJ, Graham CH. et al. Suppression of Invasion by Inducible Expression of Tissue Inhibitor of Metalloproteinase-1 (TIMP-1) in B 16-F10 Melanoma Cells. J Natl Cancer Inst 1992; 13: 1017-1022