Skip to main content
Top
Published in: Tumor Biology 2/2014

01-02-2014 | Research Article

Artemisinin inhibits gastric cancer cell proliferation through upregulation of p53

Authors: Hong-Tao Zhang, Yun-Long Wang, Jie Zhang, Qin-Xian Zhang

Published in: Tumor Biology | Issue 2/2014

Login to get access

Abstract

Recent population studies suggest that the use of artemisinin is associated with reduced incidence and improved prognosis of certain cancers. In the current study, we assessed the effect of artemisinin on gastric cancer cells (AGS and MKN74 cells). We found that artemisinin inhibited growth and modulated expression of cell-cycle regulators in these cells. Treatment with artemisinin was also associated with induction of p27kip1 and p21kip1, two negative cell-cycle regulators. Furthermore, we revealed that artemisinin treatment led to an increased expression of p53. Taken together, these results provide evidence for a mechanism that may contribute to the antineoplastic effects of artemisinin suggested by recent population studies and justify further work to explore potential roles for it in gastric cancer prevention and treatment.
Appendix
Available only for authorised users
Literature
1.
go back to reference Rugge M, Fassan M, Graham DY. Clinical guidelines: secondary prevention of gastric cancer. Nature Reviews Gastroenterology & Hepatology. 2012;9(3):128–9.CrossRef Rugge M, Fassan M, Graham DY. Clinical guidelines: secondary prevention of gastric cancer. Nature Reviews Gastroenterology & Hepatology. 2012;9(3):128–9.CrossRef
2.
go back to reference Smyth EC, Cunningham D. Gastric cancer in 2012: defining treatment standards and novel insights into disease biology. Nature Reviews Clinical Oncology. 2013;10(2):73–4.PubMedCrossRef Smyth EC, Cunningham D. Gastric cancer in 2012: defining treatment standards and novel insights into disease biology. Nature Reviews Clinical Oncology. 2013;10(2):73–4.PubMedCrossRef
3.
go back to reference Ali R, Mirza Z, Ashraf GM, Kamal MA, Ansari SA, Damanhouri GA, et al. New anticancer agents: recent developments in tumor therapy. Anticancer Research. 2012;32(7):2999–3005.PubMed Ali R, Mirza Z, Ashraf GM, Kamal MA, Ansari SA, Damanhouri GA, et al. New anticancer agents: recent developments in tumor therapy. Anticancer Research. 2012;32(7):2999–3005.PubMed
4.
go back to reference Albini A, Tosetti F, Li VW, Noonan DM, Li WW. Cancer prevention by targeting angiogenesis. Nature Reviews Clinical Oncology. 2012;9(9):498–509.PubMedCrossRef Albini A, Tosetti F, Li VW, Noonan DM, Li WW. Cancer prevention by targeting angiogenesis. Nature Reviews Clinical Oncology. 2012;9(9):498–509.PubMedCrossRef
5.
go back to reference Javeshghani S, Zakikhani M, Austin S, et al. Carbon source and myc expression influence the antiproliferative actions of metformin. Cancer Research. 2012;72(23):6257–67.PubMedCrossRef Javeshghani S, Zakikhani M, Austin S, et al. Carbon source and myc expression influence the antiproliferative actions of metformin. Cancer Research. 2012;72(23):6257–67.PubMedCrossRef
6.
go back to reference Zhang X, Gu L, Li J, et al. Degradation of MDM2 by the interaction between berberine and DAXX leads to potent apoptosis in MDM2-overexpressing cancer cells. Cancer Research. 2010;70(23):9895–904.PubMedCentralPubMedCrossRef Zhang X, Gu L, Li J, et al. Degradation of MDM2 by the interaction between berberine and DAXX leads to potent apoptosis in MDM2-overexpressing cancer cells. Cancer Research. 2010;70(23):9895–904.PubMedCentralPubMedCrossRef
7.
go back to reference Kato K, Gong J, Iwama H, et al. The antidiabetic drug metformin inhibits gastric cancer cell proliferation in vitro and in vivo. Molecular Cancer Therapeutics. 2012;11(3):549–60.PubMedCrossRef Kato K, Gong J, Iwama H, et al. The antidiabetic drug metformin inhibits gastric cancer cell proliferation in vitro and in vivo. Molecular Cancer Therapeutics. 2012;11(3):549–60.PubMedCrossRef
8.
go back to reference Efferth T, Dunstan H, Sauerbrey A, Miyachi H, Chitambar CR. The anti-malarial artesunate is also active against cancer. International Journal of Oncology. 2001;18(4):767–7739.PubMed Efferth T, Dunstan H, Sauerbrey A, Miyachi H, Chitambar CR. The anti-malarial artesunate is also active against cancer. International Journal of Oncology. 2001;18(4):767–7739.PubMed
9.
go back to reference Utzinger J, Xiao SH, Tanner M, Keiser J. Artemisinins for schistosomiasis and beyond. Current Opinion in Investigational Drugs. 2007;8(2):105–16.PubMed Utzinger J, Xiao SH, Tanner M, Keiser J. Artemisinins for schistosomiasis and beyond. Current Opinion in Investigational Drugs. 2007;8(2):105–16.PubMed
10.
go back to reference Willoughby Sr JA, Sundar SN, Cheung M, et al. Artemisinin blocks prostate cancer growth and cell cycle progression by disrupting Sp1 interactions with the cyclin-dependent kinase-4 (CDK4) promoter and inhibiting CDK4 gene expression. Journal of Biological Chemistry. 2009;284(4):2203–13.PubMedCrossRef Willoughby Sr JA, Sundar SN, Cheung M, et al. Artemisinin blocks prostate cancer growth and cell cycle progression by disrupting Sp1 interactions with the cyclin-dependent kinase-4 (CDK4) promoter and inhibiting CDK4 gene expression. Journal of Biological Chemistry. 2009;284(4):2203–13.PubMedCrossRef
11.
go back to reference Kong R, Jia G, Cheng ZX, et al. Dihydroartemisinin enhances Apo2L/TRAIL-mediated apoptosis in pancreatic cancer cells via ROS-mediated up-regulation of death receptor 5. PLoS One. 2012;7(5):e37222.PubMedCentralPubMedCrossRef Kong R, Jia G, Cheng ZX, et al. Dihydroartemisinin enhances Apo2L/TRAIL-mediated apoptosis in pancreatic cancer cells via ROS-mediated up-regulation of death receptor 5. PLoS One. 2012;7(5):e37222.PubMedCentralPubMedCrossRef
12.
go back to reference Roy S, Gu M, Ramasamy K, et al. p21/Cip1 and p27/Kip1 Are essential molecular targets of inositol hexaphosphate for its antitumor efficacy against prostate cancer. Cancer Research. 2009;69(3):1166–73.PubMedCentralPubMedCrossRef Roy S, Gu M, Ramasamy K, et al. p21/Cip1 and p27/Kip1 Are essential molecular targets of inositol hexaphosphate for its antitumor efficacy against prostate cancer. Cancer Research. 2009;69(3):1166–73.PubMedCentralPubMedCrossRef
13.
go back to reference Akli S, Van Pelt CS, Bui T, et al. Overexpression of the low molecular weight cyclin E in transgenic mice induces metastatic mammary carcinomas through the disruption of the ARF-p53 pathway. Cancer Research. 2007;67(15):7212–22.PubMedCrossRef Akli S, Van Pelt CS, Bui T, et al. Overexpression of the low molecular weight cyclin E in transgenic mice induces metastatic mammary carcinomas through the disruption of the ARF-p53 pathway. Cancer Research. 2007;67(15):7212–22.PubMedCrossRef
14.
go back to reference Yamada Y, Yoshida T, Hayashi K, et al. p53 gene mutations in gastric cancer metastases and in gastric cancer cell lines derived from metastases. Cancer Research. 1991;51(21):5800–5.PubMed Yamada Y, Yoshida T, Hayashi K, et al. p53 gene mutations in gastric cancer metastases and in gastric cancer cell lines derived from metastases. Cancer Research. 1991;51(21):5800–5.PubMed
15.
go back to reference Tin AS, Sundar SN, Tran KQ, et al. Antiproliferative effects of artemisinin on human breast cancer cells requires the downregulated expression of the E2F1 transcription factor and loss of E2F1-target cell cycle genes. Anti-Cancer Drugs. 2012;23(4):370–9.PubMedCrossRef Tin AS, Sundar SN, Tran KQ, et al. Antiproliferative effects of artemisinin on human breast cancer cells requires the downregulated expression of the E2F1 transcription factor and loss of E2F1-target cell cycle genes. Anti-Cancer Drugs. 2012;23(4):370–9.PubMedCrossRef
16.
go back to reference Xiao F, Gao W, Wang X, Chen T. Amplification activation loop between caspase-8 and -9 dominates artemisinin-induced apoptosis of ASTC-a-1 cells. Apoptosis. 2012;17(6):600–11.PubMedCrossRef Xiao F, Gao W, Wang X, Chen T. Amplification activation loop between caspase-8 and -9 dominates artemisinin-induced apoptosis of ASTC-a-1 cells. Apoptosis. 2012;17(6):600–11.PubMedCrossRef
17.
go back to reference Cerezo M, Tichet M, Abbe P, et al. Metformin blocks melanoma invasion and metastasis development in AMPK/p53-dependent manner. Molecular Cancer Therapeutics. 2013;12(8):1605–15.PubMedCrossRef Cerezo M, Tichet M, Abbe P, et al. Metformin blocks melanoma invasion and metastasis development in AMPK/p53-dependent manner. Molecular Cancer Therapeutics. 2013;12(8):1605–15.PubMedCrossRef
18.
go back to reference Lee J, van Hummelen P, Go C, et al. High-throughput mutation profiling identifies frequent somatic mutations in advanced gastric adenocarcinoma. PLoS One. 2012;7(6):e38892.PubMedCentralPubMedCrossRef Lee J, van Hummelen P, Go C, et al. High-throughput mutation profiling identifies frequent somatic mutations in advanced gastric adenocarcinoma. PLoS One. 2012;7(6):e38892.PubMedCentralPubMedCrossRef
19.
go back to reference Cotter TG. Apoptosis and cancer: the genesis of a research field. Nature Reviews Cancer. 2009;9(7):501–7.PubMedCrossRef Cotter TG. Apoptosis and cancer: the genesis of a research field. Nature Reviews Cancer. 2009;9(7):501–7.PubMedCrossRef
20.
go back to reference Vakifahmetoglu-Norberg H, Kim M, Xia HG, et al. Chaperone-mediated autophagy degrades mutant p53. Genes and Development. 2013;27(15):1718–30.PubMedCrossRef Vakifahmetoglu-Norberg H, Kim M, Xia HG, et al. Chaperone-mediated autophagy degrades mutant p53. Genes and Development. 2013;27(15):1718–30.PubMedCrossRef
21.
go back to reference Wang Y, Huang ZQ, Wang CQ, et al. Artemisinin inhibits extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinase-9 expression via a protein kinase Cδ/p38/extracellular signal-regulated kinase pathway in phorbol myristate acetate-induced THP-1 macrophages. Clinical and Experimental Pharmacology and Physiology. 2011;38(1):11–8.PubMedCrossRef Wang Y, Huang ZQ, Wang CQ, et al. Artemisinin inhibits extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinase-9 expression via a protein kinase Cδ/p38/extracellular signal-regulated kinase pathway in phorbol myristate acetate-induced THP-1 macrophages. Clinical and Experimental Pharmacology and Physiology. 2011;38(1):11–8.PubMedCrossRef
22.
go back to reference Sarina, Yagi Y, Nakano O, et al. Induction of neurite outgrowth in PC12 cells by artemisinin through activation of ERK and p38 MAPK signaling pathways. Brain Research. 2013;1490:61–71.PubMedCrossRef Sarina, Yagi Y, Nakano O, et al. Induction of neurite outgrowth in PC12 cells by artemisinin through activation of ERK and p38 MAPK signaling pathways. Brain Research. 2013;1490:61–71.PubMedCrossRef
Metadata
Title
Artemisinin inhibits gastric cancer cell proliferation through upregulation of p53
Authors
Hong-Tao Zhang
Yun-Long Wang
Jie Zhang
Qin-Xian Zhang
Publication date
01-02-2014
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 2/2014
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-013-1193-1

Other articles of this Issue 2/2014

Tumor Biology 2/2014 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