Skip to main content
Top
Published in: Investigational New Drugs 6/2011

01-12-2011 | PRECLINICAL STUDIES

The anti-cancer activity of dihydroartemisinin is associated with induction of iron-dependent endoplasmic reticulum stress in colorectal carcinoma HCT116 cells

Authors: Jin-Jian Lu, Si-Meng Chen, Xiao-Wei Zhang, Jian Ding, Ling-Hua Meng

Published in: Investigational New Drugs | Issue 6/2011

Login to get access

Summary

Dihydroartemisinin (DHA), the main active metabolite of artemisinin derivatives, is among the artemisinin derivatives possessing potent anti-malarial and anti-cancer activities. In the present study, we found that DHA displayed significant anti-proliferative activity in human colorectal carcinoma HCT116 cells, which may be attributed to its induction of G1 phase arrest and apoptosis. To further elucidate the mechanism of action of DHA, a proteomic study employed two-dimensional gel electrophoresis (2-DE) and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) was performed. Glucose-regulated protein 78 (GRP78), which is related with endoplasmic reticulum stress (ER stress), was identified to be significantly up-regulated after DHA treatment. Further study demonstrated that DHA enhanced expression of GRP78 as well as growth arrest and DNA-damage-inducible gene 153 (GADD153, another ER stress-associated molecule) at both mRNA and protein levels. DHA treatment also led to accumulation of GADD153 in cell nucleus. Moreover, pretreatment of HCT116 cells with the iron chelator deferoxamine mesylate salt (DFO) abrogated induction of GRP78 and GADD153 upon DHA treatment, indicating iron is required for DHA-induced ER stress. This result is consistent with the fact that the anti-proliferative activity of DHA is also mediated by iron. We thus suggest the unbalance of redox may result in DHA-induced ER stress, which may contribute, at least in part, to its anti-cancer activity.
Literature
2.
go back to reference Lu JJ, Meng LH, Cai YJ et al (2008) Dihydroartemisinin induces apoptosis in HL-60 leukemia cells dependent of iron and p38 mitogen-activated protein kinase activation but independent of reactive oxygen species. Cancer Biol Ther 7:1017–1023PubMedCrossRef Lu JJ, Meng LH, Cai YJ et al (2008) Dihydroartemisinin induces apoptosis in HL-60 leukemia cells dependent of iron and p38 mitogen-activated protein kinase activation but independent of reactive oxygen species. Cancer Biol Ther 7:1017–1023PubMedCrossRef
3.
go back to reference Efferth T, Sauerbrey A, Olbrich A, Gebhart E, Rauch P, Weber HO et al (2003) Molecular modes of action of artesunate in tumor cell lines. Mol Pharmacol 64:382–394PubMedCrossRef Efferth T, Sauerbrey A, Olbrich A, Gebhart E, Rauch P, Weber HO et al (2003) Molecular modes of action of artesunate in tumor cell lines. Mol Pharmacol 64:382–394PubMedCrossRef
4.
go back to reference Efferth T, Olbrich A, Bauer R (2002) mRNA expression profiles for the response of human tumor cell lines to the antimalarial drugs artesunate, arteether, and artemether. Biochem Pharmacol 64:617–623PubMedCrossRef Efferth T, Olbrich A, Bauer R (2002) mRNA expression profiles for the response of human tumor cell lines to the antimalarial drugs artesunate, arteether, and artemether. Biochem Pharmacol 64:617–623PubMedCrossRef
5.
go back to reference Li LN, Zhang HD, Yuan SJ et al (2007) Artesunate attenuates the growth of human colorectal carcinoma and inhibits hyperactive Wnt/beta-catenin pathway. Int J Cancer 121:1360–1365PubMedCrossRef Li LN, Zhang HD, Yuan SJ et al (2007) Artesunate attenuates the growth of human colorectal carcinoma and inhibits hyperactive Wnt/beta-catenin pathway. Int J Cancer 121:1360–1365PubMedCrossRef
6.
go back to reference Jiao Y, Ge CM, Meng QH et al (2007) Dihydroartemisinin is an inhibitor of ovarian cancer cell growth. Acta Pharmacol Sin 28:1045–1056PubMedCrossRef Jiao Y, Ge CM, Meng QH et al (2007) Dihydroartemisinin is an inhibitor of ovarian cancer cell growth. Acta Pharmacol Sin 28:1045–1056PubMedCrossRef
7.
go back to reference Chen T, Li M, Zhang R et al (2009) Dihydroartemisinin induces apoptosis and sensitizes human ovarian cancer cells to carboplatin therapy. J Cell Mol Med 13:1358–1370PubMedCrossRef Chen T, Li M, Zhang R et al (2009) Dihydroartemisinin induces apoptosis and sensitizes human ovarian cancer cells to carboplatin therapy. J Cell Mol Med 13:1358–1370PubMedCrossRef
8.
go back to reference Lu YY, Chen TS, Qu JL et al (2009) Dihydroartemisinin (DHA) induces caspase-3-dependent apoptosis in human lung adenocarcinoma ASTC-a-1 cells. J Biomed Sci 16:16PubMedCrossRef Lu YY, Chen TS, Qu JL et al (2009) Dihydroartemisinin (DHA) induces caspase-3-dependent apoptosis in human lung adenocarcinoma ASTC-a-1 cells. J Biomed Sci 16:16PubMedCrossRef
9.
go back to reference Lu JJ, Meng LH, Shankavaram UT et al (2010) Dihydroartemisinin accelerates c-MYC oncoprotein degradation and induces apoptosis in c-MYC-overexpressing tumor cells. Biochem Pharmacol 80:22–30PubMedCrossRef Lu JJ, Meng LH, Shankavaram UT et al (2010) Dihydroartemisinin accelerates c-MYC oncoprotein degradation and induces apoptosis in c-MYC-overexpressing tumor cells. Biochem Pharmacol 80:22–30PubMedCrossRef
10.
go back to reference Chen HH, Zhou HJ, Wang WQ et al (2004) Antimalarial dihydroartemisinin also inhibits angiogenesis. Cancer Chemother Pharmacol 53:423–432PubMedCrossRef Chen HH, Zhou HJ, Wang WQ et al (2004) Antimalarial dihydroartemisinin also inhibits angiogenesis. Cancer Chemother Pharmacol 53:423–432PubMedCrossRef
11.
go back to reference Dell’Eva R, Pfeffer U, Vene R et al (2004) Inhibition of angiogenesis in vivo and growth of Kaposi’s sarcoma xenograft tumors by the anti-malarial artesunate. Biochem Pharmacol 68:2359–2366PubMedCrossRef Dell’Eva R, Pfeffer U, Vene R et al (2004) Inhibition of angiogenesis in vivo and growth of Kaposi’s sarcoma xenograft tumors by the anti-malarial artesunate. Biochem Pharmacol 68:2359–2366PubMedCrossRef
12.
go back to reference Mercer AE, Maggs JL, Sun XM et al (2007) Evidence for the involvement of carbon-centered radicals in the induction of apoptotic cell death by artemisinin compounds. J Biol Chem 282:9372–9382PubMedCrossRef Mercer AE, Maggs JL, Sun XM et al (2007) Evidence for the involvement of carbon-centered radicals in the induction of apoptotic cell death by artemisinin compounds. J Biol Chem 282:9372–9382PubMedCrossRef
13.
go back to reference Efferth T, Benakis A, Romero MR et al (2004) Enhancement of cytotoxicity of artemisinins toward cancer cells by ferrous iron. Free Radic Biol Med 37:998–1009PubMedCrossRef Efferth T, Benakis A, Romero MR et al (2004) Enhancement of cytotoxicity of artemisinins toward cancer cells by ferrous iron. Free Radic Biol Med 37:998–1009PubMedCrossRef
14.
go back to reference Efferth T (2006) Molecular pharmacology and pharmacogenomics of artemisinin and its derivatives in cancer cells. Curr Drug Targets 7:407–421PubMedCrossRef Efferth T (2006) Molecular pharmacology and pharmacogenomics of artemisinin and its derivatives in cancer cells. Curr Drug Targets 7:407–421PubMedCrossRef
15.
go back to reference Singh NP, Lai H (2001) Selective toxicity of dihydroartemisinin and holotransferrin toward human breast cancer cells. Life Sci 70:49–56PubMedCrossRef Singh NP, Lai H (2001) Selective toxicity of dihydroartemisinin and holotransferrin toward human breast cancer cells. Life Sci 70:49–56PubMedCrossRef
16.
go back to reference Efferth T, Oesch F (2004) Oxidative stress response of tumor cells: microarray-based comparison between artemisinins and anthracyclines. Biochem Pharmacol 68:3–10PubMedCrossRef Efferth T, Oesch F (2004) Oxidative stress response of tumor cells: microarray-based comparison between artemisinins and anthracyclines. Biochem Pharmacol 68:3–10PubMedCrossRef
17.
go back to reference Tao Z, Zhou Y, Lu J et al (2007) Caspase-8 preferentially senses the apoptosis-inducing action of NG-18, a Gambogic acid derivative, in human leukemia HL-60 cells. Cancer Biol Ther 6:691–696PubMedCrossRef Tao Z, Zhou Y, Lu J et al (2007) Caspase-8 preferentially senses the apoptosis-inducing action of NG-18, a Gambogic acid derivative, in human leukemia HL-60 cells. Cancer Biol Ther 6:691–696PubMedCrossRef
18.
go back to reference Yue QX, Cao ZW, Guan SH et al (2008) Proteomics characterization of the cytotoxicity mechanism of ganoderic acid D and computer-automated estimation of the possible drug target network. Mol Cell Proteomics 7:949–961PubMedCrossRef Yue QX, Cao ZW, Guan SH et al (2008) Proteomics characterization of the cytotoxicity mechanism of ganoderic acid D and computer-automated estimation of the possible drug target network. Mol Cell Proteomics 7:949–961PubMedCrossRef
19.
go back to reference Jiang XS, Tang LY, Cao XJ et al (2005) Two-dimensional gel electrophoresis maps of the proteome and phosphoproteome of primitively cultured rat mesangial cells. Electrophoresis 26:4540–4562PubMedCrossRef Jiang XS, Tang LY, Cao XJ et al (2005) Two-dimensional gel electrophoresis maps of the proteome and phosphoproteome of primitively cultured rat mesangial cells. Electrophoresis 26:4540–4562PubMedCrossRef
20.
go back to reference Lee AS (1992) Mammalian stress response: induction of the glucose-regulated protein family. Curr Opin Cell Biol 4:267–273PubMedCrossRef Lee AS (1992) Mammalian stress response: induction of the glucose-regulated protein family. Curr Opin Cell Biol 4:267–273PubMedCrossRef
21.
go back to reference Wang XZ, Lawson B, Brewer JW et al (1996) Signals from the stressed endoplasmic reticulum induce C/EBP-homologous protein (CHOP/GADD153). Mol Cell Biol 16:4273–4280PubMed Wang XZ, Lawson B, Brewer JW et al (1996) Signals from the stressed endoplasmic reticulum induce C/EBP-homologous protein (CHOP/GADD153). Mol Cell Biol 16:4273–4280PubMed
22.
go back to reference Chen H, Sun B, Pan S et al (2009) Dihydroartemisinin inhibits growth of pancreatic cancer cells in vitro and in vivo. Anticancer Drugs 20:131–140PubMedCrossRef Chen H, Sun B, Pan S et al (2009) Dihydroartemisinin inhibits growth of pancreatic cancer cells in vitro and in vivo. Anticancer Drugs 20:131–140PubMedCrossRef
23.
go back to reference Milli A, Cecconi D, Campostrini N et al (2008) A proteomic approach for evaluating the cell response to a novel histone deacetylase inhibitor in colon cancer cells. Biochim Biophys Acta 1784:1702–1710PubMed Milli A, Cecconi D, Campostrini N et al (2008) A proteomic approach for evaluating the cell response to a novel histone deacetylase inhibitor in colon cancer cells. Biochim Biophys Acta 1784:1702–1710PubMed
24.
go back to reference Disbrow GL, Baege AC, Kierpiec KA et al (2005) Dihydroartemisinin is cytotoxic to papillomavirus-expressing epithelial cells in vitro and in vivo. Cancer Res 65:10854–10861PubMedCrossRef Disbrow GL, Baege AC, Kierpiec KA et al (2005) Dihydroartemisinin is cytotoxic to papillomavirus-expressing epithelial cells in vitro and in vivo. Cancer Res 65:10854–10861PubMedCrossRef
25.
go back to reference Mu D, Zhang W, Chu D et al (2008) The role of calcium, P38 MAPK in dihydroartemisinin-induced apoptosis of lung cancer PC-14 cells. Cancer Chemother Pharmacol 61:639–645PubMedCrossRef Mu D, Zhang W, Chu D et al (2008) The role of calcium, P38 MAPK in dihydroartemisinin-induced apoptosis of lung cancer PC-14 cells. Cancer Chemother Pharmacol 61:639–645PubMedCrossRef
26.
go back to reference Kim I, Xu W, Reed JC (2008) Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities. Nat Rev Drug Discov 7:1013–1030PubMedCrossRef Kim I, Xu W, Reed JC (2008) Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities. Nat Rev Drug Discov 7:1013–1030PubMedCrossRef
27.
go back to reference Stockwin LH, Han B, Yu SX et al (2009) Artemisinin dimer anticancer activity correlates with heme-catalyzed reactive oxygen species generation and endoplasmic reticulum stress induction. Int J Cancer 125:1266–1275PubMedCrossRef Stockwin LH, Han B, Yu SX et al (2009) Artemisinin dimer anticancer activity correlates with heme-catalyzed reactive oxygen species generation and endoplasmic reticulum stress induction. Int J Cancer 125:1266–1275PubMedCrossRef
28.
go back to reference Brostrom CO, Brostrom MA (1998) Regulation of translational initiation during cellular responses to stress. Prog Nucleic Acid Res Mol Biol 58:79–125PubMedCrossRef Brostrom CO, Brostrom MA (1998) Regulation of translational initiation during cellular responses to stress. Prog Nucleic Acid Res Mol Biol 58:79–125PubMedCrossRef
29.
go back to reference Carlberg M, Larsson O (1993) Role of N-linked glycosylation in cell-cycle progression and initiation of DNA synthesis in tumor-transformed human fibroblasts. Anticancer Res 13:167–171PubMed Carlberg M, Larsson O (1993) Role of N-linked glycosylation in cell-cycle progression and initiation of DNA synthesis in tumor-transformed human fibroblasts. Anticancer Res 13:167–171PubMed
30.
go back to reference Brewer JW, Diehl JA (2000) PERK mediates cell-cycle exit during the mammalian unfolded protein response. Proc Natl Acad Sci USA 97:12625–12630PubMedCrossRef Brewer JW, Diehl JA (2000) PERK mediates cell-cycle exit during the mammalian unfolded protein response. Proc Natl Acad Sci USA 97:12625–12630PubMedCrossRef
31.
go back to reference McCullough KD, Martindale JL, Klotz LO et al (2001) Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol 21:1249–1259PubMedCrossRef McCullough KD, Martindale JL, Klotz LO et al (2001) Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol 21:1249–1259PubMedCrossRef
32.
go back to reference Hou J, Wang D, Zhang R et al (2008) Experimental therapy of hepatoma with artemisinin and its derivatives: in vitro and in vivo activity, chemosensitization, and mechanisms of action. Clin Cancer Res 14:5519–5530PubMedCrossRef Hou J, Wang D, Zhang R et al (2008) Experimental therapy of hepatoma with artemisinin and its derivatives: in vitro and in vivo activity, chemosensitization, and mechanisms of action. Clin Cancer Res 14:5519–5530PubMedCrossRef
33.
go back to reference Zhou HJ, Wang Z, Li A (2008) Dihydroartemisinin induces apoptosis in human leukemia cells HL60 via downregulation of transferrin receptor expression. Anticancer Drugs 19:247–255PubMedCrossRef Zhou HJ, Wang Z, Li A (2008) Dihydroartemisinin induces apoptosis in human leukemia cells HL60 via downregulation of transferrin receptor expression. Anticancer Drugs 19:247–255PubMedCrossRef
34.
go back to reference Wu Y, Fabritius M, Ip C (2009) Chemotherapeutic sensitization by endoplasmic reticulum stress: increasing the efficacy of taxane against prostate cancer. Cancer Biol Ther 8:146–152PubMedCrossRef Wu Y, Fabritius M, Ip C (2009) Chemotherapeutic sensitization by endoplasmic reticulum stress: increasing the efficacy of taxane against prostate cancer. Cancer Biol Ther 8:146–152PubMedCrossRef
Metadata
Title
The anti-cancer activity of dihydroartemisinin is associated with induction of iron-dependent endoplasmic reticulum stress in colorectal carcinoma HCT116 cells
Authors
Jin-Jian Lu
Si-Meng Chen
Xiao-Wei Zhang
Jian Ding
Ling-Hua Meng
Publication date
01-12-2011
Publisher
Springer US
Published in
Investigational New Drugs / Issue 6/2011
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-010-9481-8

Other articles of this Issue 6/2011

Investigational New Drugs 6/2011 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