Skip to main content
Top
Published in: Chinese Medicine 1/2019

Open Access 01-12-2019 | Ovarian Cancer | Research

Combination of shikonin with paclitaxel overcomes multidrug resistance in human ovarian carcinoma cells in a P-gp-independent manner through enhanced ROS generation

Authors: Zhu Wang, Jianhua Yin, Mingxing Li, Jing Shen, Zhangang Xiao, Yueshui Zhao, Chengliang Huang, Hanyu Zhang, Zhuo Zhang, Chi Hin Cho, Xu Wu

Published in: Chinese Medicine | Issue 1/2019

Login to get access

Abstract

Background

Shikonin (SKN), a naphthoquinone compound, is isolated from Chinese herbal medicine Lithospermum root and has been studied as an anticancer drug candidate in human tumor models. This study is designed to investigate whether SKN can sensitize the therapeutic effect of paclitaxel (PTX) in drug-resistant human ovarian carcinoma cells.

Methods

Human ovarian carcinoma A2780 cell along with the paired PTX-resistant A2780/PTX cells were used. The effects of SKN, PTX or their combination on cell viability were conducted using Sulforhodamine B assay. P-glycoprotein (P-gp) expression was analyzed by flow cytometry after staining with P-gp-FITC anti-body. P-gp activity was determined by a fluorometric MDR assay kit or a rhodamine 123-based efflux assay, respectively. Apoptosis was evaluated by flow cytometry after Annexin V-FITC/PI co-staining. The effect of SKN, PTX or their combination on reactive oxygen species (ROS) generation and expression of pyruvate kinase M2 (PKM2) were investigated using flow cytometry or western blotting, respectively. PKM2 activity was detected by a Pyruvate Kinase Assay Kit.

Results

SKN/PTX co-treatment led to synergistically enhanced cytotoxicity and apoptosis in PTX-resistant ovarian cancer cells, indicating the circumvention of multidrug resistance (MDR) of PTX by SKN. Further study indicated that the MDR reversal effect of SKN was independent of inhibiting activity of the efflux transporter P-gp. Notably, SKN/PTX significantly increased the generation of intracellular ROS in A2780/PTX cells, and scavenging intracellular ROS blocked the sensitizing effects of SKN in PTX-induced cytotoxicity and apoptosis in A2780/PTX cells, but not in A2780 cells. Furthermore, SKN/PTX-induced downregulation of PKM2 (a key enzyme in glycolysis) and the suppression of its activity were inhibited by a ROS scavenger N-acetyl cysteine (NAC), suggesting that the synergy of the SKN/PTX combination may be not rely on PKM2 suppression.

Conclusions

These results reveal a P-gp-independent mechanism through ROS generation for the SKN/PTX combination to overcome MDR in ovarian cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Bray F, Ferlay J, Soerjomataram I, Siegel R, Torre L, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRef Bray F, Ferlay J, Soerjomataram I, Siegel R, Torre L, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.CrossRef
2.
3.
go back to reference Ffrench B, Gasch C, O’Leary JJ, Gallagher MF. Developing ovarian cancer stem cell models: laying the pipeline from discovery to clinical intervention. Mol Cancer. 2014;13:262.CrossRef Ffrench B, Gasch C, O’Leary JJ, Gallagher MF. Developing ovarian cancer stem cell models: laying the pipeline from discovery to clinical intervention. Mol Cancer. 2014;13:262.CrossRef
4.
go back to reference Krishna R, Mayer LD. Multidrug resistance (MDR) in cancer. Mechanisms, reversal using modulators of MDR and the role of MDR modulators in influencing the pharmacokinetics of anticancer drugs. Eur J Pharm Sci. 2000;11:265–83.CrossRef Krishna R, Mayer LD. Multidrug resistance (MDR) in cancer. Mechanisms, reversal using modulators of MDR and the role of MDR modulators in influencing the pharmacokinetics of anticancer drugs. Eur J Pharm Sci. 2000;11:265–83.CrossRef
5.
go back to reference Lage H. An overview of cancer multidrug resistance: a still unsolved problem. Cell Mol Life Sci. 2008;65:3145.CrossRef Lage H. An overview of cancer multidrug resistance: a still unsolved problem. Cell Mol Life Sci. 2008;65:3145.CrossRef
6.
go back to reference An X, Sarmiento C, Tan T, Zhu H. Regulation of multidrug resistance by microRNAs in anti-cancer therapy. Acta Pharm Sin B. 2017;7:38–51.CrossRef An X, Sarmiento C, Tan T, Zhu H. Regulation of multidrug resistance by microRNAs in anti-cancer therapy. Acta Pharm Sin B. 2017;7:38–51.CrossRef
7.
go back to reference Pommier Y, Sordet O, Antony S, Hayward R, Kohn K. Apoptosis defects and chemotherapy resistance: molecular interaction maps and networks. Oncogene. 2004;23:2934–49.CrossRef Pommier Y, Sordet O, Antony S, Hayward R, Kohn K. Apoptosis defects and chemotherapy resistance: molecular interaction maps and networks. Oncogene. 2004;23:2934–49.CrossRef
8.
go back to reference Giménez-Bonafé P, Tortosa A, Pérez-Tomás R. Overcoming drug resistance by enhancing apoptosis of tumor cells. Curr Cancer Drug Targets. 2009;9:320–40.CrossRef Giménez-Bonafé P, Tortosa A, Pérez-Tomás R. Overcoming drug resistance by enhancing apoptosis of tumor cells. Curr Cancer Drug Targets. 2009;9:320–40.CrossRef
9.
go back to reference Domenichini A, Adamska A, Falasca M. ABC transporters as cancer drivers: potential functions in cancer development. Biochim Biophys Acta Gen Subj. 2018;1863:52–60.CrossRef Domenichini A, Adamska A, Falasca M. ABC transporters as cancer drivers: potential functions in cancer development. Biochim Biophys Acta Gen Subj. 2018;1863:52–60.CrossRef
10.
go back to reference Paškevičiūtė M, Petrikaitė V. Overcoming transporter-mediated multidrug resistance in cancer: failures and achievements of the last decades. Drug Deliv Transl Res. 2018;9:379–93.CrossRef Paškevičiūtė M, Petrikaitė V. Overcoming transporter-mediated multidrug resistance in cancer: failures and achievements of the last decades. Drug Deliv Transl Res. 2018;9:379–93.CrossRef
11.
go back to reference To K, Wu X, Yin C, Chai S, Yao S, Kadioglu O, Efferth T, Ye Y, Lin G. Reversal of multidrug resistance by Marsdenia tenacissima and its main active ingredients polyoxypregnanes. J Ethnopharmacol. 2017;203:110–9.CrossRef To K, Wu X, Yin C, Chai S, Yao S, Kadioglu O, Efferth T, Ye Y, Lin G. Reversal of multidrug resistance by Marsdenia tenacissima and its main active ingredients polyoxypregnanes. J Ethnopharmacol. 2017;203:110–9.CrossRef
12.
go back to reference Wu X, Ma J, Ye Y, Lin G. Transporter modulation by Chinese herbal medicines and its mediated pharmacokinetic herb-drug interactions. J Chromatogr B Analyt Technol Biomed Life Sci. 2016;1026:236–53.CrossRef Wu X, Ma J, Ye Y, Lin G. Transporter modulation by Chinese herbal medicines and its mediated pharmacokinetic herb-drug interactions. J Chromatogr B Analyt Technol Biomed Life Sci. 2016;1026:236–53.CrossRef
13.
go back to reference Hersey P, Xu DZ. Overcoming resistance of cancer cells to apoptosis. J Cell Physiol. 2010;196:9–18.CrossRef Hersey P, Xu DZ. Overcoming resistance of cancer cells to apoptosis. J Cell Physiol. 2010;196:9–18.CrossRef
14.
go back to reference Dinic J, Podolskirenic A, Stankovic T, Bankovic J, Pesic M. New Approaches with natural product drugs for overcoming multidrug resistance in cancer. Curr Pharm Design. 2015;21:5589–604.CrossRef Dinic J, Podolskirenic A, Stankovic T, Bankovic J, Pesic M. New Approaches with natural product drugs for overcoming multidrug resistance in cancer. Curr Pharm Design. 2015;21:5589–604.CrossRef
15.
go back to reference Han W, Xie J, Li L, Liu Z, Hu X. Necrostatin-1 reverts shikonin-induced necroptosis to apoptosis. Apoptosis. 2009;14:674.CrossRef Han W, Xie J, Li L, Liu Z, Hu X. Necrostatin-1 reverts shikonin-induced necroptosis to apoptosis. Apoptosis. 2009;14:674.CrossRef
16.
go back to reference Pc H, Huang YT, Tsai ML, Wang YJ, Lin JK, Pan MH. Induction of apoptosis by shikonin through coordinative modulation of the Bcl-2 family, p27, and p53, release of cytochrome c, and sequential activation of caspases in human colorectal carcinoma cells. J Agr Food Chem. 2004;52:6330–7.CrossRef Pc H, Huang YT, Tsai ML, Wang YJ, Lin JK, Pan MH. Induction of apoptosis by shikonin through coordinative modulation of the Bcl-2 family, p27, and p53, release of cytochrome c, and sequential activation of caspases in human colorectal carcinoma cells. J Agr Food Chem. 2004;52:6330–7.CrossRef
17.
go back to reference Guo XP, Zhang XY, Zhang SD. Clinical trial on the effects of shikonin mixture on later stage lung cancer. Chin J Modern Dev Tradit Med. 1991;11:598. Guo XP, Zhang XY, Zhang SD. Clinical trial on the effects of shikonin mixture on later stage lung cancer. Chin J Modern Dev Tradit Med. 1991;11:598.
18.
go back to reference Ko H, Kim SJ, Shim SH, Chang H, Ha CH. Shikonin induces apoptotic cell death via regulation of p53 and Nrf2 in AGS human stomach carcinoma cells. Biomol Ther. 2016;24:501–9.CrossRef Ko H, Kim SJ, Shim SH, Chang H, Ha CH. Shikonin induces apoptotic cell death via regulation of p53 and Nrf2 in AGS human stomach carcinoma cells. Biomol Ther. 2016;24:501–9.CrossRef
19.
go back to reference Singh F, Gao D, Lebwohl MG, Wei H. Shikonin modulates cell proliferation by inhibiting epidermal growth factor receptor signaling in human epidermoid carcinoma cells. Cancer Lett. 2003;200:115–21.CrossRef Singh F, Gao D, Lebwohl MG, Wei H. Shikonin modulates cell proliferation by inhibiting epidermal growth factor receptor signaling in human epidermoid carcinoma cells. Cancer Lett. 2003;200:115–21.CrossRef
20.
go back to reference Yang H, Zhou P, Huang H, Chen D, Ma N, Cui CQ, Shen S, Dong W, Zhang X, Lian W. Shikonin exerts antitumor activity via proteasome inhibition and cell death induction in vitro and in vivo. Int J Cancer. 2010;124:2450–9.CrossRef Yang H, Zhou P, Huang H, Chen D, Ma N, Cui CQ, Shen S, Dong W, Zhang X, Lian W. Shikonin exerts antitumor activity via proteasome inhibition and cell death induction in vitro and in vivo. Int J Cancer. 2010;124:2450–9.CrossRef
21.
go back to reference Duan D, Zhang B, Yao J, Liu Y, Fang J. Shikonin targets cytosolic thioredoxin reductase to induce ROS-mediated apoptosis in human promyelocytic leukemia HL-60 cells. Free Radical Biol Med. 2014;70:182–93.CrossRef Duan D, Zhang B, Yao J, Liu Y, Fang J. Shikonin targets cytosolic thioredoxin reductase to induce ROS-mediated apoptosis in human promyelocytic leukemia HL-60 cells. Free Radical Biol Med. 2014;70:182–93.CrossRef
22.
go back to reference Lu B, Wang Z, Ding Y, Wang X, Lu S, Wang C, He C, Piao M, Chi G, Luo Y. RIP1 and RIP3 contribute to shikonin-induced glycolysis suppression in glioma cells via increase of intracellular hydrogen peroxide. Cancer Lett. 2018;425:31–42.CrossRef Lu B, Wang Z, Ding Y, Wang X, Lu S, Wang C, He C, Piao M, Chi G, Luo Y. RIP1 and RIP3 contribute to shikonin-induced glycolysis suppression in glioma cells via increase of intracellular hydrogen peroxide. Cancer Lett. 2018;425:31–42.CrossRef
23.
go back to reference Lee MJ, Kao SH, Hunag JE, Sheu GT, Yeh CW, Hseu YC, Wang CJ, Hsu LS. Shikonin time-dependently induced necrosis or apoptosis in gastric cancer cells via generation of reactive oxygen species. Chem-Biol Interact. 2014;211:44–53.CrossRef Lee MJ, Kao SH, Hunag JE, Sheu GT, Yeh CW, Hseu YC, Wang CJ, Hsu LS. Shikonin time-dependently induced necrosis or apoptosis in gastric cancer cells via generation of reactive oxygen species. Chem-Biol Interact. 2014;211:44–53.CrossRef
24.
go back to reference Zhang CH, Wang J, Zhang LX, Lu YH, Ji TH, Xu L, Ling LJ. Shikonin reduces tamoxifen resistance through long non-coding RNA uc.57. Oncotarget. 2017;8:88658–69.PubMedCentral Zhang CH, Wang J, Zhang LX, Lu YH, Ji TH, Xu L, Ling LJ. Shikonin reduces tamoxifen resistance through long non-coding RNA uc.57. Oncotarget. 2017;8:88658–69.PubMedCentral
25.
go back to reference Wang SP, Wang L, Chen MW, Wang YT. Gambogic acid sensitizes resistant breast cancer cells to doxorubicin through inhibiting P-glycoprotein and suppressing survivin expression. Chem Biol Interact. 2015;235:76–84.CrossRef Wang SP, Wang L, Chen MW, Wang YT. Gambogic acid sensitizes resistant breast cancer cells to doxorubicin through inhibiting P-glycoprotein and suppressing survivin expression. Chem Biol Interact. 2015;235:76–84.CrossRef
26.
go back to reference Wang X, Cheng K, Han Y, Zhang G, Dong J, Cui Y, Yang Z. Effects of psoralen as an anti-tumor agent in human breast cancer MCF-7/ADR Cells. Biol Pharm Bull. 2016;39:815–22.CrossRef Wang X, Cheng K, Han Y, Zhang G, Dong J, Cui Y, Yang Z. Effects of psoralen as an anti-tumor agent in human breast cancer MCF-7/ADR Cells. Biol Pharm Bull. 2016;39:815–22.CrossRef
27.
go back to reference Chou TC, Talaly P. A simple generalized equation for the analysis of multiple inhibitions of Michaelis-Menten kinetic systems. J Biol Chem. 1977;252:6438–42. Chou TC, Talaly P. A simple generalized equation for the analysis of multiple inhibitions of Michaelis-Menten kinetic systems. J Biol Chem. 1977;252:6438–42.
28.
go back to reference Katsaros D, Arts HJG, Massobrio M, Zee AJHVD. Expression of drug resistance-associated markers P-GP, MRP1, MRP2 and LRP in ovarian cancer: clinical implications. Int J Gynecol Obstetrics. 2000;70:D73.CrossRef Katsaros D, Arts HJG, Massobrio M, Zee AJHVD. Expression of drug resistance-associated markers P-GP, MRP1, MRP2 and LRP in ovarian cancer: clinical implications. Int J Gynecol Obstetrics. 2000;70:D73.CrossRef
29.
go back to reference Garces AHI, Dias MSF, Paulino E, Ferreira CGM, de Melo AC. Treatment of ovarian cancer beyond chemotherapy: are we hitting the target? Cancer Chemoth Pharm. 2015;75:221–34.CrossRef Garces AHI, Dias MSF, Paulino E, Ferreira CGM, de Melo AC. Treatment of ovarian cancer beyond chemotherapy: are we hitting the target? Cancer Chemoth Pharm. 2015;75:221–34.CrossRef
30.
go back to reference Carpi S, Fogli S, Giannetti A, Adinolfi B, Tombelli S, Da Pozzo E, Vanni A, Martinotti E, Martini C, Breschi MC, Pellegrino M, Nieri P, Baldini F. Theranostic properties of a survivin-directed molecular beacon in human melanoma cells. PloS ONE. 2014;9:e114588.CrossRef Carpi S, Fogli S, Giannetti A, Adinolfi B, Tombelli S, Da Pozzo E, Vanni A, Martinotti E, Martini C, Breschi MC, Pellegrino M, Nieri P, Baldini F. Theranostic properties of a survivin-directed molecular beacon in human melanoma cells. PloS ONE. 2014;9:e114588.CrossRef
31.
go back to reference Lv X, Wang B, Xu X, Pan L, Wang B, Dong X, Zheng C, Du Q. Curcumin re-sensitizes multidrug resistant (MDR) breast cancer to cisplatin through inducing autophagy by decreasing CCAT1 expression. Rsc Adv. 2017;7:33572–9.CrossRef Lv X, Wang B, Xu X, Pan L, Wang B, Dong X, Zheng C, Du Q. Curcumin re-sensitizes multidrug resistant (MDR) breast cancer to cisplatin through inducing autophagy by decreasing CCAT1 expression. Rsc Adv. 2017;7:33572–9.CrossRef
32.
go back to reference Liu L, Ju Y, Wang J, Zhou R. Epigallocatechin-3-gallate promotes apoptosis and reversal of multidrug resistance in esophageal cancer cells. Pathol Res Pract. 2017;213:1242.CrossRef Liu L, Ju Y, Wang J, Zhou R. Epigallocatechin-3-gallate promotes apoptosis and reversal of multidrug resistance in esophageal cancer cells. Pathol Res Pract. 2017;213:1242.CrossRef
33.
go back to reference Raj L, Ide T, Gurkar AU, Foley M, Schenone M, Li X, Tolliday NJ, Golub TR, Carr SA, Shamji AF. Selective killing of cancer cells with a small molecule targeting stress response to ROS. Nature. 2011;475:231.CrossRef Raj L, Ide T, Gurkar AU, Foley M, Schenone M, Li X, Tolliday NJ, Golub TR, Carr SA, Shamji AF. Selective killing of cancer cells with a small molecule targeting stress response to ROS. Nature. 2011;475:231.CrossRef
34.
go back to reference He G, He G, Zhou R, Pi Z, Zhu T, Jiang L, Xie Y. Enhancement of cisplatin-induced colon cancer cells apoptosis by shikonin, a natural inducer of ROS in vitro and in vivo. Biochem Biophy Res Commun. 2016;469:1075–82.CrossRef He G, He G, Zhou R, Pi Z, Zhu T, Jiang L, Xie Y. Enhancement of cisplatin-induced colon cancer cells apoptosis by shikonin, a natural inducer of ROS in vitro and in vivo. Biochem Biophy Res Commun. 2016;469:1075–82.CrossRef
35.
go back to reference Chen J, Xie J, Jiang Z, Wang B, Wang Y, Hu X. Shikonin and its analogs inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2. Oncogene. 2011;30:4297.CrossRef Chen J, Xie J, Jiang Z, Wang B, Wang Y, Hu X. Shikonin and its analogs inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2. Oncogene. 2011;30:4297.CrossRef
36.
go back to reference Vander Heiden MG, Cantley LC, Thompson CB. Understanding the warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029.CrossRef Vander Heiden MG, Cantley LC, Thompson CB. Understanding the warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029.CrossRef
37.
go back to reference Anastasiou D, Yu Y, Israelsen WJ, Jiang JK, Boxer MB, Hong BS, Tempel W, Dimov S, Shen M, Jha A. Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis. Nat Chem Biol. 2012;8:839–47.CrossRef Anastasiou D, Yu Y, Israelsen WJ, Jiang JK, Boxer MB, Hong BS, Tempel W, Dimov S, Shen M, Jha A. Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis. Nat Chem Biol. 2012;8:839–47.CrossRef
38.
go back to reference Lim JY, Sun OY, Seol SY, Hong SW, Kim JW, Choi SH, Cho JY. Overexpression of the M2 isoform of pyruvate kinase is an adverse prognostic factor for signet ring cell gastric cancer. World J Gastroenterol. 2012;18:4037–43.CrossRef Lim JY, Sun OY, Seol SY, Hong SW, Kim JW, Choi SH, Cho JY. Overexpression of the M2 isoform of pyruvate kinase is an adverse prognostic factor for signet ring cell gastric cancer. World J Gastroenterol. 2012;18:4037–43.CrossRef
Metadata
Title
Combination of shikonin with paclitaxel overcomes multidrug resistance in human ovarian carcinoma cells in a P-gp-independent manner through enhanced ROS generation
Authors
Zhu Wang
Jianhua Yin
Mingxing Li
Jing Shen
Zhangang Xiao
Yueshui Zhao
Chengliang Huang
Hanyu Zhang
Zhuo Zhang
Chi Hin Cho
Xu Wu
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Chinese Medicine / Issue 1/2019
Electronic ISSN: 1749-8546
DOI
https://doi.org/10.1186/s13020-019-0231-3

Other articles of this Issue 1/2019

Chinese Medicine 1/2019 Go to the issue