DOI QR코드

DOI QR Code

α-Mangostin and Apigenin Induced Cell Cycle Arrest and Programmed Cell Death in SKOV-3 Ovarian Cancer Cells

  • Ittiudomrak, Teeranai (Program in Biotechnology, Faculty of Science, Chulalongkorn University) ;
  • Puthong, Songchan (Institute of Biotechnology and Genetic Engineering, Chulalongkorn University) ;
  • Roytrakul, Sittiruk (National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency) ;
  • Chanchao, Chanpen (Department of Biology, Faculty of Science, Chulalongkorn University)
  • Received : 2018.07.16
  • Accepted : 2018.10.04
  • Published : 2019.04.15

Abstract

Ovarian cancer is the fifth main cause of pre-senescent death in women. Although chemotherapy is generally an efficient treatment, its side effects and the occurrence of chemotherapeutic resistance have prompted the need for alternative treatments. In this study, ${\alpha}$-mangostin and apigenin were evaluated as possible anticancer alternatives to the chemotherapeutic drug doxorubicin, used herein as a positive control. The ovarian adenocarcinoma cell line SKOV-3 (ATCC No. HTB77) was used as model ovarian cancer cells, whereas the skin fibroblast line CCD-986Sk (ATCC No. CRL-1947) and lung fibroblast line WI-38 (ATCC No. CCL-75) were used as model untransformed cells. Apigenin and doxorubicin inhibited the growth of SKOV-3 cells in a dose- and time-dependent manner. After 72 hr exposure, doxorubicin was mostly toxic to SKOV-3 cells, whereas apigenin was toxic to SKOV-3 cells but not CCD-986Sk and WI-38 cells. ${\alpha}$-Mangostin was more toxic to SKOV-3 cells than to CCD-986Sk cells. A lower cell density, cell shrinkage, and more unattached (floating round) cells were observed in all treated SKOV-3 cells, but the greatest effects were observed with ${\alpha}$-mangostin. With regard to programmed cell death, apigenin caused early apoptosis within 24 hr, whereas ${\alpha}$-mangostin and doxorubicin caused late apoptosis and necrosis after 72 hr of exposure. Caspase-3 activity was significantly increased in ${\alpha}$-mangostin-treated SKOV-3 cells after 12 hr of exposure, whereas only caspase-9 activity was significantly increased in apigenin-treated SKOV-3 cells at 24 hr. Both ${\alpha}$-mangostin and apigenin arrested the cell cycle at the $G_2/M$ phase, but after 24 and 48 hr, respectively. Significant upregulation of BCL2 (apoptosis-associated gene) and COX2 (inflammation-associated gene) transcripts was observed in apigenin- and ${\alpha}$-mangostin-treated SKOV-3 cells, respectively. ${\alpha}$-Mangostin and apigenin are therefore alternative options for SKOV-3 cell inhibition, with apigenin causing rapid early apoptosis related to the intrinsic apoptotic pathway, and ${\alpha}$-mangostin likely being involved with inflammation.

Keywords

References

  1. Reid, B.M., Permuth, J.B. and Sellers, T.A. (2017) Epidemiology of ovarian cancer: a review. Cancer Biol. Med., 14, 9-32. https://doi.org/10.20892/j.issn.2095-3941.2016.0084
  2. Li, J.X., Bi, Y.P., Wang, J., Yang, X., Tian, Y.F. and Sun, Z.F. (2018) JTC-801 inhibits the proliferation and metastasis of ovarian cancer cell SKOV3 through inhibition of the PI3K-AKT signaling pathway. Pharmazie, 73, 283-287.
  3. Zhang, H., Jiao, Y., Shi, C., Song, X., Chang, Y., Ren, Y. and Shi, X. (2018) Berbamine suppresses cell proliferation and promotes apoptosis in ovarian cancer partially via the inhibition of Wnt/${\beta}$-catenin signaling. Acta Biochim. Biophys. Sin. (Shanghai), 50, 532-539. https://doi.org/10.1093/abbs/gmy036
  4. Liotta, L.A., Steeg, P.S. and Stetler-Stevenson, W.G. (1991) Cancer metastasis and angiogenesis: animbalance of positive and negative regulation. Cell, 64, 327-336. https://doi.org/10.1016/0092-8674(91)90642-C
  5. Luvero, D., Plotti, F., Lopez, S., Scaletta, G., Capriglione, S., Montera, R., Antonelli, G., Ciuffreda, S., Carassiti, R., Oliveti, A. and Angioli, R. (2017) Antiangiogenics and immunotherapies in cervical cancer: an update and future's view. Med. Oncol., 34, 115. https://doi.org/10.1007/s12032-017-0972-8
  6. Ayla, S., Bilir, A., Erturkoglu, S., Tanriverdi, G., Soner, B.C., Sofuoglu, K., Ghisolfi, L. and Oktem, G. (2018) Effects of different drug treatments on the proliferation of human ovarian carcinoma cell line MDAH-774. Turk. J. Med. Sci., 48, 441-448.
  7. Rahman, N., Jeon, M., Song, H.Y. and Kim, Y.S. (2016) Cryptotanshinone, a compound of Salvia miltiorrhiza inhibits pre-adipocytes differentiation by regulation of adipogenesis-related genes expression via STAT3 signaling. Phytomedicine, 23, 58-67. https://doi.org/10.1016/j.phymed.2015.12.004
  8. Zhang, Y., Chen, S., Wei, C., Rankin, G.O., Rojanasakul, Y., Ren, N., Ye, X. and Chen, Y.C. (2018) Dietary Compound Proanthocyanidins from Chinese bayberry (Myrica rubra Sieb. et Zucc.) leaves inhibit angiogenesis and regulate cell cycle of cisplatin-resistant ovarian cancer cells via targeting Akt pathway. J. Funct. Foods, 40, 573-581. https://doi.org/10.1016/j.jff.2017.11.045
  9. Zhang, Q., Dong, J., Cui, J., Huang, G., Meng, Q. and Li, S. (2018) Cytotoxicity of synthesized 1,4-naphthoquinone oxime derivatives on selected human cancer cell lines. Chem. Pharm. Bull., 66, 612-619. https://doi.org/10.1248/cpb.c18-00013
  10. Puglisi, M.P., Bradaric, M.J., Pontikis, J., Cabai, J., Weyna, T., Tednes, P., Schretzman, R., Rickert, K., Cao, Z. and Andrei, D. (2018) Novel primary amine diazeniumdiolates-Chemical and biological characterization. Drug Dev. Res., 79, 136-143. https://doi.org/10.1002/ddr.21428
  11. Tsai, S.Y., Chung, P.C., Owaga, E.E., Tsai, I.J., Wang, P.Y., Tsai, J.I., Yeh, T.S. and Hsieh, R.H. (2016) Alpha-mangostin from mangosteen (Garcinia mangostana Linn.) pericarp extract reduces high fat-diet induced hepatic steatosis in rats by regulating mitochondria function and apoptosis. Nutr. Metab. (Lond.), 13, 88. https://doi.org/10.1186/s12986-016-0148-0
  12. Nugitrangson, P., Puthong, S., Iempridee, T., Pimtong, W., Pornpakakul, S. and Chanchao, C. (2016) In vitro and in vivo characterization of the anticancer activity of Thai stingless bee (Tetragonula laeviceps) cerumen. Exp. Biol. Med., 241, 166-176. https://doi.org/10.1177/1535370215600102
  13. Sandor, Z., Mottaghipisheh, J., Veres, K., Hohmann, J., Bencsik, T., Horvath, A., Kelemen, D., Papp, R., Bartho, L. and Csupor, D. (2018) Evidence supports tradition: the in vitro effects of Roman chamomile on smooth muscles. Front. Pharmacol., 9, 323. https://doi.org/10.3389/fphar.2018.00323
  14. Chantarudee, A., Phuwapraisirisan, P., Kimura, K., Okuyama, M., Mori, H., Kimura, A. and Chanchao, C. (2012) Chemical constituents and free radical scavenging activity of corn pollen collected from Apis mellifera hives compared to floral corn pollen at Nan, Thailand. BMC Complement. Altern. Med., 12, 45.
  15. Sivaranjani, M., Srinivasan, R., Aravindraja, C., KaruthaPandian, S. and Veera Ravi, A. (2018) Inhibitory effect of ${\alpha}$-mangostin on Acinetobacter baumannii biofilms-an in vitro study. Biofouling, 34, 579-593. https://doi.org/10.1080/08927014.2018.1473387
  16. Chen, G., Li, Y., Wang, W. and Deng, L. (2018) Bioactivity and pharmacological properties of ${\alpha}$-mangostin from the mangosteen fruit: a review. Expert Opin. Ther. Pat., 28, 415-427. https://doi.org/10.1080/13543776.2018.1455829
  17. Kumar, K.S., Sabu, V., Sindhu, G., Rauf, A.A. and Helen, A. (2018) Isolation, identification and characterization of apigenin from Justicia gendarussa and its anti-inflammatory activity. Int. Immunopharmacol., 59, 157-167. https://doi.org/10.1016/j.intimp.2018.04.004
  18. Abu Bakar, F.I., Abu Bakar, M.F., Rahmat, A., Abdullah, N., Sabran, S.F. and Endrini, S. (2018) Anti-gout potential of Malaysian medicinal plants. Front. Pharmacol., 9, 261. https://doi.org/10.3389/fphar.2018.00261
  19. El Habbash, A.I., Mohd Hashim, N., Ibrahim, M.Y., Yahayu, M., Omer, F.A.E., Abd Rahman, M., Nordin, N. and Lian, G.E.C. (2017) In vitro assessment of anti-proliferative effect induced by ${\alpha}$-mangostin from Cratoxylum arborescens on HeLa cells. PeerJ, 5, e3460. https://doi.org/10.7717/peerj.3460
  20. Suh, Y.A., Jo, S.Y., Lee, H.Y. and Lee, C. (2015) Inhibition of IL-6/STAT3 axis and targeting Axl and Tyro3 receptor tyrosine kinases by apigenin circumvent taxol resistance in ovarian cancer cells. Int. J. Oncol., 46, 1405-1411. https://doi.org/10.3892/ijo.2014.2808
  21. Tang, A.Q., Cao, X.C., Tian, L., He, L. and Liu, F.(2015) Apigenin inhibits the self-renewal capacity of human ovarian cancer SKOV3-derived sphere-forming cells. Mol. Med. Rep., 11, 2221-2226. https://doi.org/10.3892/mmr.2014.2974
  22. Niu, W., Chen, F., Wang, J., Qian, J. and Yan, S. (2017) Antitumor effect of sikokianin C, a selective cystathionine ${\beta}$-synthase inhibitor, against human colon cancer in vitro and in vivo. Medchemcomm, 9, 113-120. https://doi.org/10.1039/C7MD00484B
  23. Fang, A., Zhang, Q., Fan, H., Zhou, Y., Yao, Y., Zhang, Y. and Huang, X. (2017) Discovery of human lactate dehydrogenase A (LDHA) inhibitors as anticancer agents to inhibit the proliferation of MG-63 osteosarcoma cells. MedChem-Comm, 8, 1720-1726. https://doi.org/10.1039/C7MD00222J
  24. Xu, X., Rawling, T., Roseblade, A., Bishop, R. and Ung, A.T. (2017) Antiproliferative activities of alkaloid-like compounds. MedChemComm, 8, 2105-2114. https://doi.org/10.1039/C7MD00435D
  25. Koopman, G., Reutelingsperger, C.P., Kuijten, G.A., Keehnen, R.M., Pals, S.T. and van Oers, M.H. (1994) Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis. Blood, 84, 1415-1420. https://doi.org/10.1182/blood.V84.5.1415.bloodjournal8451415
  26. Schutte, B., Nuydens, R., Geerts, H. and Ramaekers, F. (1998) Annexin V binding assay as a tool to measure apoptosis in differentiated neuronal cells. J. Neurosci. Methods, 86, 63-69. https://doi.org/10.1016/S0165-0270(98)00147-2
  27. Easton, D.F., Bishop, D.T., Ford, D. and Crockford, G.P. (1993) Genetic linkage analysis in familial breast and ovarian cancer: results from 214 families. The Breast Cancer Linkage Consortium. Am. J. Hum. Genet., 52, 678-701.
  28. Yin, L., Li, J., Wei, Y., Ma, D., Sun, Y. and Sun, Y. (2018) Primary ovarian small cell carcinoma of pulmonary type with coexisting endometrial carcinoma in a breast cancer patient receiving tamoxifen: A case report and literature review. Medicine (Baltimore), 97, e10900. https://doi.org/10.1097/MD.0000000000010900
  29. Zhang, L., Shin, V.Y., Chai, X., Zhang, A., Chan, T.L., Ma, E.S., Rebbeck, T.R., Chen, J. and Kwong, A. (2018) Breast and ovarian cancer penetrance of BRCA1/2 mutations among Hong Kong women. Oncotarget, 9, 25025-25033. https://doi.org/10.18632/oncotarget.24382
  30. Reuter, S., Gupta, S.C., Chaturvedi, M.M. and Aggarwal, B.B. (2010) Oxidative stress, inflammation, and cancer: how are they linked? Free Radic. Biol. Med., 49, 1603-1616. https://doi.org/10.1016/j.freeradbiomed.2010.09.006
  31. Weinberg, F. and Chandel, N.S. (2009) Reactive oxygen species-dependent signaling regulates cancer. Cell. Mol. Life Sci., 66, 3663-3673. https://doi.org/10.1007/s00018-009-0099-y
  32. Chan, D.W., Liu, V.W., Tsao, G.S., Yao, K.M., Furukawa, T., Chan, K.K. and Ngan, H.Y. (2008) Loss of MKP3 mediated by oxidative stress enhances tumorigenicity and chemoresistance of ovarian cancer cells. Carcinogenesis, 29, 1742-1750. https://doi.org/10.1093/carcin/bgn167
  33. Kang, K.A. and Hyun, J.W. (2017) Oxidative stress, Nrf2 and epigenetic modification contribute to anticancer drug resistance. Toxicol. Res., 33, 1-5. https://doi.org/10.5487/TR.2017.33.1.001
  34. Yurdacan, B., Egeli, U., Eskiler, G.G., Eryilmaz, I.E., Cecener, G. and Tunca, B. (2018) The role of usnic acid-induced apoptosis and autophagy in hepatocellular carcinoma. Hum. Exp. Toxicol., doi:10.1177/0960327118792052 [Epub ahead of print].
  35. Boehning, A.L., Essien, S.A., Underwood, E.L., Dash, P.K. and Boehning, D. (2018) Cell type-dependent effects of ellagic acid on cellular metabolism. Biomed. Pharmacother., 106, 411-418. https://doi.org/10.1016/j.biopha.2018.06.142
  36. Hu,Y., Li, X., An, Y., Duan, J. and Yang, X.D. (2018) Selection of a novel CD19 aptamer for targeted delivery of doxorubicin to lymphoma cells. Oncotarget, 9, 26605-26615. https://doi.org/10.18632/oncotarget.24902
  37. Hupe, M.C., Hoda, M.R., Zengerling, F., Perner, S., Merseburger, A.S. and Cronauer, M.V. (2018) The BET-inhibitor PFI-1 diminishes AR/AR-V7 signaling in prostate cancer cells. World J. Urol., doi:10.1007/s00345-018-2382-8 [Epub ahead of print].
  38. Hanan, N.A., Chiu, H.I., Ramachandran, M.R., Tung, W.H., MohamadZain, N.N., Yahaya, N. and Lim, V. (2018) Cytotoxicity of plant-mediated synthesis of metallic nanoparticles:a systematic review. Int. J. Mol. Sci., 19, E1725. https://doi.org/10.3390/ijms19061725
  39. Vallee, A. and Lecarpentier, Y. (2018) Crosstalk between peroxisome proliferator-activated receptor gamma and the canonical wnt/${\beta}$-catenin pathway in chronic inflammation and oxidative stress during carcinogenesis. Front. Immunol., 9, 745. https://doi.org/10.3389/fimmu.2018.00745
  40. Otake, Y., Soundararajan, S., Sengupta, T.K., Kio, E.A., Smith, J.C., Pineda-Roman, M., Stuart, R.K., Spicer, E.K. and Fernandes, D.J. (2007) Overexpression of nucleolin in chronic lymphocytic leukemia cells induces stabilization of bcl2 mRNA. Blood, 109, 3069-3075. https://doi.org/10.1182/blood-2006-08-043257
  41. Shibata, M.A., Hamaoka, H., Morimoto, J., Kanayama, T., Maemura, K., Ito, Y., Iinuma, M. and Kondo, Y. (2018) Synthetic ${\alpha}$-mangostin dilaurate strongly suppresses wide-spectrum organ metastasis in a mouse model of mammary cancer. Cancer Sci., 109, 1660-1671. https://doi.org/10.1111/cas.13590
  42. Xia, Y. and Sun, J. (2018) Synergistic inhibition of cell proliferation by combined targeting with kinase inhibitors and dietary xanthone is a promising strategy for melanoma treatment. Clin. Exp. Dermatol., 43, 149-157. https://doi.org/10.1111/ced.13283
  43. Nurul, S.A.S., Hazilawati, H., Mohd, R.S., Mohd, F.H.R., Noordin, M.M. and Norhaizan, M.E. (2018) Subacute oral toxicity assessment of ethanol extract of Mariposa christiavespertilionis leaves in male Sprague Dawley rats. Toxicol. Res., 34, 85-95. https://doi.org/10.5487/TR.2018.34.2.085
  44. Lee, Y. (2017) Cancer chemopreventive potential of procyanidin. Toxicol. Res., 33, 273-282. https://doi.org/10.5487/TR.2017.33.4.273