Browse > Article
http://dx.doi.org/10.5483/BMBRep.2019.52.8.058

Dihydroartemisinin inhibits HepG2.2.15 proliferation by inducing cellular senescence and autophagy  

Zou, Jiang (Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College)
Ma, Qiang (Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College)
Sun, Ru (Department of Laboratory Medicine, North Sichuan Medical College)
Cai, Jiajing (Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College)
Liao, Hebin (Translational Medicine Research Center, North Sichuan Medical College)
Xu, Lei (Translational Medicine Research Center, North Sichuan Medical College)
Xia, Jingruo (Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College)
Huang, Guangcheng (Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College)
Yao, Lihua (Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College)
Cai, Yan (Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College)
Zhong, Xiaowu (Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College)
Guo, Xiaolan (Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College)
Publication Information
BMB Reports / v.52, no.8, 2019 , pp. 520-525 More about this Journal
Abstract
Dihydroartemisinin (DHA) has been reported to possess anti-cancer activity against many cancers. However, the pharmacologic effect of DHA on HBV-positive hepatocellular carcinoma (HCC) remains unknown. Thus, the objective of the present study was to determine whether DHA could inhibit the proliferation of HepG2.2.15 cells and uncover the underlying mechanisms involved in the effect of DHA on HepG2.2.15 cells. We found that DHA effectively inhibited HepG2.2.15 HCC cell proliferation both in vivo and in vitro. DHA also reduced the migration and tumorigenicity capacity of HepG2.2.15 cells. Regarding the underlying mechanisms, results showed that DHA induced cellular senescence by up-regulating expression levels of proteins such as p-ATM, p-ATR, ${\gamma}-H_2AX$, P53, and P21 involved in DNA damage response. DHA also induced autophagy (green LC3 puncta gathered together and LC3II/LC3I ratio increased through AKT-mTOR pathway suppression). Results also revealed that DHA-induced autophagy was not linked to senescence or cell death. TPP1 (telomere shelterin) overexpression could not rescue DHA-induced anticancer activity (cell proliferation). Moreover, DHA down-regulated TPP1 expression. Gene knockdown of TPP1 caused similar phenotypes and mechanisms as DHA induced phenotypes and mechanisms in HepG2.2.15 cells. These results demonstrate that DHA might inhibit HepG2.2.15 cells proliferation through inducing cellular senescence and autophagy.
Keywords
Autophagy; Cellular senescence; Dihydroartemisinin; DNA damage response; HCC;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A (2018) Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68, 394-424   DOI
2 Mokdad AA, Lopez AD, Shahraz S et al (2014) Liver cirrhosis mortality in 187 countries between 1980 and 2010: a systematic analysis. BMC Med 12, 145   DOI
3 Eastman RT and Fidock DA (2009) Artemisinin-based combination therapies: a vital tool in efforts to eliminate malaria. Nat Rev Microbiol 7, 864-874   DOI
4 Tong Y, Liu Y, Zheng H et al (2016) Artemisinin and its derivatives can significantly inhibit lung tumorigenesis and tumor metastasis through Wnt/beta-catenin signaling. Oncotarget 7, 31413-31428   DOI
5 Ontikatze T, Rudner J, Handrick R, Belka C and Jendrossek V (2014) Dihydroartemisinin is a hypoxia-active anti-cancer drug in colorectal carcinoma cells. Front Oncol 4, 116   DOI
6 Thongchot S, Vidoni C, Ferraresi A et al (2018) Dihydroartemisinin induces apoptosis and autophagy-dependent cell death in cholangiocarcinoma through a DAPK1-BECLIN1 pathway. Mol Carcinog 57, 1735-1750   DOI
7 Li Y, Wang Y, Kong R et al (2016) Dihydroartemisinin suppresses pancreatic cancer cells via a microRNA-mRNA regulatory network. Oncotarget 7, 62460-62473   DOI
8 Liao K, Li J and Wang Z (2014) Dihydroartemisinin inhibits cell proliferation via AKT/GSK3beta/cyclinD1 pathway and induces apoptosis in A549 lung cancer cells. Int J Clin Exp Pathol 7, 8684-8691
9 Zhang S, Shi L, Ma H et al (2017) Dihydroartemisinin induces apoptosis in human gastric cancer cell line BGC-823 through activation of JNK1/2 and p38 MAPK signaling pathways. J Recept Signal Transduct Res 37, 174-180   DOI
10 Shi X, Wang L, Li X et al (2017) Dihydroartemisinin induces autophagy-dependent death in human tongue squamous cell carcinoma cells through DNA double-strand breakmediated oxidative stress. Oncotarget 8, 45981-45993   DOI
11 Amaravadi R, Kimmelman AC and White E (2016) Recent insights into the function of autophagy in cancer. Genes Dev 30, 1913-1930   DOI
12 Acosta JC and Gil J (2012) Senescence: a new weapon for cancer therapy. Trends Cell Biol 22, 211-219   DOI
13 Qin G, Zhao C, Zhang L et al (2015) Dihydroartemisinin induces apoptosis preferentially via a Bim-mediated intrinsic pathway in hepatocarcinoma cells. Apoptosis 20, 1072-1086   DOI
14 Chang BD, Xuan Y, Broude EV et al (1999) Role of p53 and p21waf1/cip1 in senescence-like terminal proliferation arrest induced in human tumor cells by chemotherapeutic drugs. Oncogene 18, 4808-4818   DOI
15 Zhang Z, Yao Z, Zhao S et al (2017) Interaction between autophagy and senescence is required for dihydroartemisinin to alleviate liver fibrosis. Cell Death Dis 8, e2886   DOI
16 Wang SY, Yu QJ, Zhang RD and Liu B (2011) Core signaling pathways of survival/death in autophagy-related cancer networks. Int J Biochem Cell Biol 43, 1263-1266   DOI
17 Gao X, Luo Z, Xiang T, Wang K, Li J and Wang P (2011) Dihydroartemisinin induces endoplasmic reticulum stressmediated apoptosis in HepG2 human hepatoma cells. Tumori 97, 771-780   DOI
18 Im E, Yeo C, Lee HJ and Lee EO (2018) Dihydroartemisinin induced caspase-dependent apoptosis through inhibiting the specificity protein 1 pathway in hepatocellular carcinoma SK-Hep-1 cells. Life Sci 192, 286-292   DOI
19 Zhang CZ, Zhang H, Yun J, Chen GG and Lai PB (2012) Dihydroartemisinin exhibits antitumor activity toward hepatocellular carcinoma in vitro and in vivo. Biochem Pharmacol 83, 1278-1289   DOI
20 Sun P, Yoshizuka N, New L et al (2007) PRAK is essential for ras-induced senescence and tumor suppression. Cell 128, 295-308   DOI
21 Guo X, Deng Y, Lin Y et al (2007) Dysfunctional telomeres activate an ATM-ATR-dependent DNA damage response to suppress tumorigenesis. EMBO J 26, 4709-4719   DOI
22 Itahana K, Campisi J and Dimri GP (2004) Mechanisms of cellular senescence in human and mouse cells. Biogerontology 5, 1-10   DOI
23 Kocak H, Ballew BJ, Bisht K et al (2014) Hoyeraal-Hreidarsson syndrome caused by a germline mutation in the TEL patch of the telomere protein TPP1. Genes Dev 28, 2090-2102   DOI
24 Liu X, Wu J, Fan M et al (2018) Novel dihydroartemisinin derivative DHA-37 induces autophagic cell death through upregulation of HMGB1 in A549 cells. Cell Death Dis 9, 1048   DOI
25 Qu C, Ma J, Liu X et al (2017) Dihydroartemisinin exerts anti-tumor activity by inducing mitochondrion and endoplasmic reticulum apoptosis and autophagic cell death in human glioblastoma cells. Mol Carcinog 11, 310
26 Chen C, Gu P and Wu J (2017) Structural insights into POT1-TPP1 interaction and POT1 C-terminal mutations in human cancer. Nat Commun 8, 14929   DOI