Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2017

Open Access 01-12-2017 | Research

Sophoridine induces apoptosis and S phase arrest via ROS-dependent JNK and ERK activation in human pancreatic cancer cells

Authors: Zihang Xu, Fei Zhang, Chao Bai, Chao Yao, Hairong Zhong, Chunpu Zou, Xiao Chen

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2017

Login to get access

Abstract

Background

Pancreatic cancer is generally acknowledged as the most common primary malignant tumor, and it is known to be resistant to conventional chemotherapy. Novel, selective antitumor agents are pressingly needed.

Methods

CCK-8 and colony formation assay were used to investigate the cell growth. Flow cytometry analysis was used to evaluate the cell cycle and cell apoptosis. The peroxide-sensitive fluorescent probe DCFH-DA was used to measure the intracellular ROS levels. Western blot assay was used to detect the levels of cell cycle and apoptosis related proteins. Xenografts in nude mice were used to evaluate the effect of Sophoridine on pancreatic cancer cell in vivo.

Results

Sophoridine killed cancer cells but had low cytotoxicity to normal cells. Pancreatic cancer cells were particularly sensitive. Sophoridine inhibited the proliferation of pancreatic cancer cells and induced cell cycle arrest at S phase and mitochondrial-related apoptosis. Moreover, Sophoridine induced a sustained activation of the phosphorylation of ERK and JNK. In addition, Sophoridine provoked the generation of reactive oxygen species (ROS) in pancreatic cancer cells. Finally, in vivo, Sophoridine suppressed tumor growth in mouse xenograft models.

Conclusion

These findings suggest Sophoridine is promising to be a novel, potent and selective antitumor drug candidate for pancreatic cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Garrido-Laguna I, Hidalgo M. Pancreatic cancer: from state-of-the-art treatments to promising novel therapies. Nat Rev Clin Oncol. 2015;12:319–34.CrossRef Garrido-Laguna I, Hidalgo M. Pancreatic cancer: from state-of-the-art treatments to promising novel therapies. Nat Rev Clin Oncol. 2015;12:319–34.CrossRef
2.
go back to reference Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5–29.CrossRef Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5–29.CrossRef
3.
go back to reference Zhang F, Liu B, Wang Z, Yu XJ, Ni QX, Yang WT, Mukaida N, Li YY. A novel regulatory mechanism of Pim-3 kinase stability and its involvement in pancreatic cancer progression. Mol Cancer Res. 2013;11:1508–20.CrossRef Zhang F, Liu B, Wang Z, Yu XJ, Ni QX, Yang WT, Mukaida N, Li YY. A novel regulatory mechanism of Pim-3 kinase stability and its involvement in pancreatic cancer progression. Mol Cancer Res. 2013;11:1508–20.CrossRef
4.
go back to reference Clardy J, Walsh C. Lessons from natural molecules. Nature. 2004;432:829–37.CrossRef Clardy J, Walsh C. Lessons from natural molecules. Nature. 2004;432:829–37.CrossRef
5.
go back to reference Koehn FE, Carter GT. The evolving role of natural products in drug discovery. Nat Rev Drug Discov. 2005;4:206–20.CrossRef Koehn FE, Carter GT. The evolving role of natural products in drug discovery. Nat Rev Drug Discov. 2005;4:206–20.CrossRef
6.
go back to reference Ji HF, Li XJ, Zhang HY. Natural products and drug discovery. Can thousands of years of ancient medical knowledge lead us to new and powerful drug combinations in the fight against cancer and dementia? EMBO Rep. 2009;10:194–200.CrossRef Ji HF, Li XJ, Zhang HY. Natural products and drug discovery. Can thousands of years of ancient medical knowledge lead us to new and powerful drug combinations in the fight against cancer and dementia? EMBO Rep. 2009;10:194–200.CrossRef
7.
go back to reference Huang X, Li B, Shen L. Studies on the anti-inflammatory effect and its mechanisms of sophoridine. J Anal Methods Chem. 2014;2014:502626.CrossRef Huang X, Li B, Shen L. Studies on the anti-inflammatory effect and its mechanisms of sophoridine. J Anal Methods Chem. 2014;2014:502626.CrossRef
8.
go back to reference Zhang X, Jin L, Cui Z, Zhang C, Wu X, Park H, Quan H, Jin C. Antiparasitic effects of oxymatrine and matrine against Toxoplasma gondii in vitro and in vivo. Exp Parasitol. 2016;165:95–102.CrossRef Zhang X, Jin L, Cui Z, Zhang C, Wu X, Park H, Quan H, Jin C. Antiparasitic effects of oxymatrine and matrine against Toxoplasma gondii in vitro and in vivo. Exp Parasitol. 2016;165:95–102.CrossRef
9.
go back to reference Li CQ, Zhu YT, Zhang FX, Fu LC, Li XH, Cheng Y, Li XY. Anti-HBV effect of liposome-encapsulated matrine in vitro and in vivo. World J Gastroenterol. 2005;11:426–8.CrossRef Li CQ, Zhu YT, Zhang FX, Fu LC, Li XH, Cheng Y, Li XY. Anti-HBV effect of liposome-encapsulated matrine in vitro and in vivo. World J Gastroenterol. 2005;11:426–8.CrossRef
10.
go back to reference Liang L, Wang XY, Zhang XH, Ji B, Yan HC, Deng HZ, Wu XR. Sophoridine exerts an anti-colorectal carcinoma effect through apoptosis induction in vitro and in vivo. Life Sci. 2012;91:1295–303.CrossRef Liang L, Wang XY, Zhang XH, Ji B, Yan HC, Deng HZ, Wu XR. Sophoridine exerts an anti-colorectal carcinoma effect through apoptosis induction in vitro and in vivo. Life Sci. 2012;91:1295–303.CrossRef
11.
go back to reference Bi C, Ye C, Li Y, Zhao W, Shao R, Song D. Synthesis and biological evaluation of 12-N-p-chlorobenzyl sophoridinol derivatives as a novel family of anticancer agents. Acta Pharm Sin B. 2016;6:222–8.CrossRef Bi C, Ye C, Li Y, Zhao W, Shao R, Song D. Synthesis and biological evaluation of 12-N-p-chlorobenzyl sophoridinol derivatives as a novel family of anticancer agents. Acta Pharm Sin B. 2016;6:222–8.CrossRef
12.
go back to reference Tan CJ, Zhao Y, Goto M, Hsieh KY, Yang XM, Morris-Natschke SL, Liu LN, Zhao BY, Lee KH. Alkaloids from Oxytropis Ochrocephala and antiproliferative activity of sophoridine derivatives against cancer cell lines. Bioorg Med Chem Lett. 2016;26:1495–7.CrossRef Tan CJ, Zhao Y, Goto M, Hsieh KY, Yang XM, Morris-Natschke SL, Liu LN, Zhao BY, Lee KH. Alkaloids from Oxytropis Ochrocephala and antiproliferative activity of sophoridine derivatives against cancer cell lines. Bioorg Med Chem Lett. 2016;26:1495–7.CrossRef
13.
go back to reference Zhou H, Xu M, Gao Y, Deng Z, Cao H, Zhang W, Wang Q, Zhang B, Song G, Zhan Y, Hu T. Matrine induces caspase-independent program cell death in hepatocellular carcinoma through bid-mediated nuclear translocation of apoptosis inducing factor. Mol Cancer. 2014;13:59.CrossRef Zhou H, Xu M, Gao Y, Deng Z, Cao H, Zhang W, Wang Q, Zhang B, Song G, Zhan Y, Hu T. Matrine induces caspase-independent program cell death in hepatocellular carcinoma through bid-mediated nuclear translocation of apoptosis inducing factor. Mol Cancer. 2014;13:59.CrossRef
14.
go back to reference Tan C, Qian X, Jia R, Wu M, Liang Z. Matrine induction of reactive oxygen species activates p38 leading to caspase-dependent cell apoptosis in non-small cell lung cancer cells. Oncol Rep. 2013;30:2529–35.CrossRef Tan C, Qian X, Jia R, Wu M, Liang Z. Matrine induction of reactive oxygen species activates p38 leading to caspase-dependent cell apoptosis in non-small cell lung cancer cells. Oncol Rep. 2013;30:2529–35.CrossRef
15.
go back to reference Zou C, Mallampalli RK. Regulation of histone modifying enzymes by the ubiquitin-proteasome system. Biochim Biophys Acta. 1843;2014:694–702. Zou C, Mallampalli RK. Regulation of histone modifying enzymes by the ubiquitin-proteasome system. Biochim Biophys Acta. 1843;2014:694–702.
16.
go back to reference Zhang F, Li M, Wu X, Hu Y, Cao Y, Wang X, Xiang S, Li H, Jiang L, Tan Z, et al. 20(S)-ginsenoside Rg3 promotes senescence and apoptosis in gallbladder cancer cells via the p53 pathway. Drug Des Devel Ther. 2015;9:3969–87.PubMedPubMedCentral Zhang F, Li M, Wu X, Hu Y, Cao Y, Wang X, Xiang S, Li H, Jiang L, Tan Z, et al. 20(S)-ginsenoside Rg3 promotes senescence and apoptosis in gallbladder cancer cells via the p53 pathway. Drug Des Devel Ther. 2015;9:3969–87.PubMedPubMedCentral
17.
go back to reference Zhang F, Ma Q, Xu Z, Liang H, Li H, Ye Y, Xiang S, Zhang Y, Jiang L, Hu Y, et al. Dihydroartemisinin inhibits TCTP-dependent metastasis in gallbladder cancer. J Exp Clin Cancer Res. 2017;36:68.CrossRef Zhang F, Ma Q, Xu Z, Liang H, Li H, Ye Y, Xiang S, Zhang Y, Jiang L, Hu Y, et al. Dihydroartemisinin inhibits TCTP-dependent metastasis in gallbladder cancer. J Exp Clin Cancer Res. 2017;36:68.CrossRef
18.
go back to reference Zhang W, Liu HT. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 2002;12:9–18.CrossRef Zhang W, Liu HT. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 2002;12:9–18.CrossRef
19.
go back to reference Chauhan D, Li G, Sattler M, Podar K, Mitsiades C, Mitsiades N, Munshi N, Hideshima T, Anderson KC. Superoxide-dependent and -independent mitochondrial signaling during apoptosis in multiple myeloma cells. Oncogene. 2003;22:6296–300.CrossRef Chauhan D, Li G, Sattler M, Podar K, Mitsiades C, Mitsiades N, Munshi N, Hideshima T, Anderson KC. Superoxide-dependent and -independent mitochondrial signaling during apoptosis in multiple myeloma cells. Oncogene. 2003;22:6296–300.CrossRef
20.
go back to reference Ballif BA, Blenis J. Molecular mechanisms mediating mammalian mitogen-activated protein kinase (MAPK) kinase (MEK)-MAPK cell survival signals. Cell Growth Differ. 2001;12:397–408.PubMed Ballif BA, Blenis J. Molecular mechanisms mediating mammalian mitogen-activated protein kinase (MAPK) kinase (MEK)-MAPK cell survival signals. Cell Growth Differ. 2001;12:397–408.PubMed
21.
go back to reference Kondo N, Nakamura H, Masutani H, Yodoi J. Redox regulation of human thioredoxin network. Antioxid Redox Signal. 2006;8:1881–90.CrossRef Kondo N, Nakamura H, Masutani H, Yodoi J. Redox regulation of human thioredoxin network. Antioxid Redox Signal. 2006;8:1881–90.CrossRef
22.
go back to reference Lee WY, Liu KW, Yeung JH. Reactive oxygen species-mediated kinase activation by dihydrotanshinone in tanshinones-induced apoptosis in HepG2 cells. Cancer Lett. 2009;285:46–57.CrossRef Lee WY, Liu KW, Yeung JH. Reactive oxygen species-mediated kinase activation by dihydrotanshinone in tanshinones-induced apoptosis in HepG2 cells. Cancer Lett. 2009;285:46–57.CrossRef
23.
go back to reference Moon DO, Kim MO, Kang SH, Choi YH, Kim GY. Sulforaphane suppresses TNF-alpha-mediated activation of NF-kappaB and induces apoptosis through activation of reactive oxygen species-dependent caspase-3. Cancer Lett. 2009;274:132–42.CrossRef Moon DO, Kim MO, Kang SH, Choi YH, Kim GY. Sulforaphane suppresses TNF-alpha-mediated activation of NF-kappaB and induces apoptosis through activation of reactive oxygen species-dependent caspase-3. Cancer Lett. 2009;274:132–42.CrossRef
24.
go back to reference Kim BC, Kim HG, Lee SA, Lim S, Park EH, Kim SJ, Lim CJ. Genipin-induced apoptosis in hepatoma cells is mediated by reactive oxygen species/c-Jun NH2-terminal kinase-dependent activation of mitochondrial pathway. Biochem Pharmacol. 2005;70:1398–407.CrossRef Kim BC, Kim HG, Lee SA, Lim S, Park EH, Kim SJ, Lim CJ. Genipin-induced apoptosis in hepatoma cells is mediated by reactive oxygen species/c-Jun NH2-terminal kinase-dependent activation of mitochondrial pathway. Biochem Pharmacol. 2005;70:1398–407.CrossRef
25.
go back to reference Xiao D, Herman-Antosiewicz A, Antosiewicz J, Xiao H, Brisson M, Lazo JS, Singh SV. Diallyl trisulfide-induced G(2)-M phase cell cycle arrest in human prostate cancer cells is caused by reactive oxygen species-dependent destruction and hyperphosphorylation of Cdc 25 C. Oncogene. 2005;24:6256–68.CrossRef Xiao D, Herman-Antosiewicz A, Antosiewicz J, Xiao H, Brisson M, Lazo JS, Singh SV. Diallyl trisulfide-induced G(2)-M phase cell cycle arrest in human prostate cancer cells is caused by reactive oxygen species-dependent destruction and hyperphosphorylation of Cdc 25 C. Oncogene. 2005;24:6256–68.CrossRef
Metadata
Title
Sophoridine induces apoptosis and S phase arrest via ROS-dependent JNK and ERK activation in human pancreatic cancer cells
Authors
Zihang Xu
Fei Zhang
Chao Bai
Chao Yao
Hairong Zhong
Chunpu Zou
Xiao Chen
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2017
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-017-0590-5

Other articles of this Issue 1/2017

Journal of Experimental & Clinical Cancer Research 1/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine