Tetrandrine induces cell death in SAS human oral cancer cells through caspase activation-dependent apoptosis and LC3-I and LC3-II activation-dependent autophagy

  • Authors:
    • An-Cheng Huang
    • Jin-Cherng Lien
    • Meng-Wei Lin
    • Jai-Sing Yang
    • Ping-Ping Wu
    • Shu-Jen Chang
    • Tung-Yuan Lai
  • View Affiliations

  • Published online on: May 21, 2013     https://doi.org/10.3892/ijo.2013.1952
  • Pages: 485-494
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Numerous studies have demonstrated that autophagy is associated with cancer development. Thus, agents to induce autophagy could be employed in some cases for the treatment of cancer. Our results showed that tetrandrine significantly decreased the viability of SAS cells in a concentration- and time-dependent manner. Tetrandrine induced nuclear condensation, demonstrated by DAPI staining. The early events in apoptosis analysed by Annexin V/PI staining indicated that the percentage of cells staining positive for Annexin V was slightly increased in SAS cells with tetrandrine treatment but was much lower following bafilomycin A1 pre-treatment. Tetrandrine caused AVO and MDC induction in SAS cells in a concentration-dependent manner by fluorescence microscopy. Tetrandrine also caused LC-3 expression in SAS cells in a time-dependent manner. Our results show that tetrandrine treatment induced the levels of cleaved caspase-3 in a concentration- and time-dependent manner. Tetrandrine treatment induced the levels of LC-3 II, Atg-5, beclin-1, p-S6, p-ULK, p-mTOR, p-Akt (S473) and raptor. Tetrandrine decreased cell viability, but bafilomycin A1, 3-MA, chloroquine and NAC protected tetrandrine-treated SAS cells against decrease of cell viability. Atg-5, beclin-1 siRNA decreased tetrandrine-induced cleaved caspase-3 and cleaved PARP in SAS cells and protected tetrandrine-treated SAS cells against decrease in cell viability. Chloroquine, NAC and bafilomycin A1 also decreased tetrandrine-induced cleaved caspase-3 and cleaved PARP in SAS cells. Our results indicate the tetrandrine induces apoptosis and autophagy of SAS human cancer cells via caspase-dependent and LC3-I and LC3-II‑dependent pathways.

Introduction

Oral cancer is one of the common cancers and is the fourth leading cause of cancer death in Taiwan male population but the 16th in females (13). Based on the 2008 report from the Department of Health, R.O.C. (Taiwan) indicated that 7.9 individuals per 100,000 die annually from oral cancer (1). It has been demonstrated that about 95% of these oral cavity cancers are squamous cell carcinomas and human oral cancer is largely associated with the risk factors of chronic smoking or alcohol consumption (46). Furthermore, the susceptibility of an individual to oral cancer is associated with the individual genetic factors and carcinogen-exposure behavior (79). In Taiwan, it was suggested that smoking and betel nut chewing are the two most important risk factors (1,10). Based on the above report the government indicated that approximately 85% of the oral cancer patients are regular users of betel quid (areca nut). The current treatments for oral cancer are inadequate and a significant proportion of oral cancer patients may develop local invasion and metastases (11). Numerous studies have been performed to find novel agents (especial from natural plant) which can trigger programmed cell death (apoptosis) in tumor cells and hopefully to provide a new therapeutic approach for anticancer design (1214).

It is well documented that numerous natural compounds present anticancer effects and some of them play important roles in cancer treatment clinically. Tetrandrine (C38H42O8N2), a bisbenzylisoquinoline alkaloid, was isolated from the root of Stephania tetrandra S. Moore, has been used as an anti-inflammatory, antipyretic and analgesic herb in Chinese medicine (1518), furthermore, in myocyte and vascular smooth muscle cells, tetrandrine inhibits the ICa-L and reduces Ca2+ flows into the cytosol (19,20). Tetrandrine has been found to induce cytotoxic effect such as the inhibiting proliferation and inducing apoptosis of various cancer cells such as breast cancer, lung cancer, neuroblastoma, Burkitt’s lymphoma, hepatoma, glioma, leukemia and colon cancer (2125). Tetrandrine induced G0/G1 phase arrest in Neuro 2a mouse neuroblastoma cells (26) and human Hep G2 cells (27). Tetrandrine have been demonstrated to inhibit lipid peroxidation and platelet aggregation and reduces ischemia/reperfusion injury (2830) and tetrandrine suppressed T and B cells and inhibited the production of cytokines (29). It was reported that tetrandrine is cytotoxic to RT-2 glioma cells and it has antitumor effects on subcutaneous and intra-cerebral gliomas, and inhibits angiogenesis in subcutaneous gliomas (3134). Tetradine induced apoptosis by activating reactive oxygen species and repressing Akt activity in human hepatocellular carcinoma HCC cells (35,36). Recently, it was reported that tetrandrine induced apoptosis and triggers caspase cascade in human bladder cancer cells (37).

There is no previous report on studies investigating the effects of tetrandrine on the cytotoxic on human oral cancer cells. In the present study, we investigated the antitumor effect of tetrandrine against human oral squamous carcinoma cells and the mechanism by which the association occurs. Using human oral cancer SAS cells, we demonstrated that tetrandrine exhibits antitumor activity on the cells by inhibiting the activity of the caspase pathway and inducing autophagy and apoptosis. Our study suggests that tetrandrine may be a potential agent to combat oral cancer cells.

Materials and methods

Cell culture and chemicals

Tetrandrine, dimethyl sulfoxide (DMSO), propidium iodide (PI), potassium phosphates, ribonuclease-A, Triton X-100, Tris-HCl and trypan blue were obtained from Sigma Chemical Co. (St. Louis, MO, USA). Dulbecco’s Modified Eagle’s medium (DMEM), L-glutamine, fetal bovine serum (FBS), penicillin, streptomycin and trypsin-EDTA were obtained from Gibco-BRL/Invitrogen Corp (Grand Island, NY, USA). The SAS cell line (human oral squamous cell carcinoma) was obtained from Dr Pei-Jung Lu (Graduate Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan). Cells were cultured in DMEM containing 10% FBS, 2 mM L-glutamine, 100 U/ml penicillin and 100 μg/ml streptomycin in 75 cm2 tissue culture flasks at 37°C under a humidified 5% CO2 and 95% air atmosphere.

Assessment of cell morphology and viability

Tetrandrine was prepared and dissolved in DMSO. SAS cells (2.5×105) were plated in 24-well plates in DMEM and incubated at 37°C for 24 h. Cells were then treated with 0, 1, 5, 10, 15, 20, 25, 30, 35 and 40 μM tetrandrine for 24 and 48 h. DMSO was used as a vehicle control. For inhibitor treatment, cells were pre-treated with autophagic inhibitor (bafilomycin A1, 3-MA, chloroquine), caspase inhibitor (pan-caspase, caspase-3, caspase-8 and caspase-9 inhibitor) or NAC (Sigma Chemical Co) and then treated with 0, 5, 10, 15, 20, 25, 30, 35 and 40 μM tetrandrine for 24 and 48 h. At the end of the incubation period, cells were photographed with a phase-contrast microscope. They were then harvested, stained with PI (5 μg/ml) and analyzed by flow cytometry (Becton-Dickinson, San Jose, CA, USA) (3840).

DAPI staining

SAS cells were treated with or without 0, 1, 10, 20, 25 and 30 μM tetrandrine for 24 h. They were then isolated, stained with DAPI and photographed using a fluorescence microscope (3840).

Annexin V and PI staining assay

SAS cells were treated with or without 0 and 25 μM tetrandrine for 24 h then were re-suspended in Annexin V-FITC (Becton-Dickinson, San Jose, CA, USA) alone or in combination with 10 ml of PI (50 mg/ml) and were incubated at room temperature for 15 min. The staining was analysed by flow cytometry (3840).

Electron microscopy

SAS cells were treated with or without (0 and 25 μM) tetrandrine for 24 h then were fixed with a solution containing 2.5% glutaraldehyde and 2% paraformaldehyde (in 0.1 M cacodylate buffer, pH 7.3) for 1 h. After fixation, the samples were postfixed for 30 min in the same buffer containing 1% OsO4. Ultra-thin sections were observed under a transmission electron microscope (JEM-1200EX, JEOL Ltd., Tokyo, Japan) at 100 kV (41).

Acridine orange staining

SAS cells were treated with 25 μM tetrandrine for 0, 2, 4 and 6 h and then cells were harvested and stained with 1 mg/ml acridine orange for a period of 20 min and analysed by a fluorescence microscope (41).

Monodansyl cadaverine MDC staining

SAS cells were treated with 0, 10, 20 and 30 μM tetrandrine for 6 h and then cells were harvested and stained with 1 mg/ml MDC for a period of 20 min and analysed by a fluorescence microscope (42).

GFP-LC3 expression

SAS cells were transfected with pEGFP-LC3 for 16 h and then were treated with 0, 10, 20 and 30 μM tetrandrine for 6 h. The cells were harvested, fixed with 4% paraformaldehyde for 10 min at room temperature. Cells were stained with LC-3-FITC for a period of 20 min and analysed by a fluorescence microscope (41).

Western blot analysis

SAS cells were pre-treated with Atg-5 or beclin-1 siRNA bafilomycin A1 then treated with 0, 10, 15, 20, 25 and 30 μM tetrandrine and then harvested by scraping into RIPA buffer and sonicated for 15 min with 30 sec pulses. The protein concentration was measured using the BCA assay (Thermo Scientific) following the instructions of the manufacturer. Incubation with primary antibodies was done overnight at 4°C. Immunoreactive proteins were visualised with the ECL chemiluminescent detection system (Perkin Elmer Life Sciences, MA, USA) and BioMax LightFilm (Eastman Kodak, New Heaven, CT, USA) according to the manufacturer’s instructions (43).

Statistical analysis

Data of control and experimental groups were expressed as mean ± standard deviation for at least three separate experiments. Statistical analyses of the data were performed using Student’s t-test and one-way analysis of variance (ANOVA). Statistical significance was set at P<0.05 (43).

Results

Effects of tetrandrine on cell morphology and viability in SAS cells

Cells were morphologically altered by tetrandrine treatment as shown in Fig. 1A. We tested the cell viability of tetrandrine in SAS cell lines. SAS cells were treated with 0, 5, 10, 15, 20, 25, 30, 35 and 40 μM tetrandrine for 24 and 48 h. Tetrandrine significantly decreased the growth of cells in a concentration- and time-dependent manner (Fig. 1B).

Tetrandrine-induced apoptosis in SAS cells

Induction of apoptosis by tetrandrine in SAS cells was confirmed by DAPI staining, as seen in Fig. 2B, which showed that tetrandrine induced nuclei condensation. These effects were dose-dependent as noted in Fig. 2B. Higher concentrations of tetrandrine resulted in a greater number of apoptotic cells being stained. We analysed surface exposure of phosphatidylserine, an early event in apoptosis by Annexin V/PI staining. The percentage of cells staining positive for Annexin V was slightly increased in SAS cells types after 24 h of tetrandrine treatment but was much lower following bafilomycin A1 of pre-treatment.

Tetrandrine induced autophagy in SAS cells

Autophagic vacuoles containing cellular membranous structures were increased in SAS-treated with tetrandrine for 24 h compared with untreated cells, as determined by electron microscopy (Fig. 3A). We used Acridine orange staining to AVOs, including autophagic vacuoles and lysosomes. Cells with AVOs had enhanced red fluorescence that was detected by fluorescence microscopy (Fig. 3B). Tetrandrine caused MDC induction in SAS cells in a concentration-dependent manner (Fig. 3B). Tetrandrine also caused LC-3 expression in SAS cells in a time-dependent manner.

Effects of tetrandrine on levels of proteins associated with apoptosis and autophagy

Results are presented in Fig. 4A, tetrandrine treatment induced the levels of cleaved-caspase-3 in a concentration- and time-dependent manner. Tetrandrine treatment induced the levels of LC-3 II, Atg-5, beclin-1, p-S6, p-ULK, p-mTOR, p-Akt (S473), and raptor in a concentration- and time-dependent manner (Fig. 4B).

Autophagy inhibitors decreased tetrandrine-induced apoptosis in SAS cells

Results are presented in Fig. 5A, tetrandrine decreased the cell viability, but bafilomycin A1, 3-MA, chlorobafilomycin A1, 3-MA, chloro-A1, 3-MA, chloroquine and NAC protected tetrandrine-treated SAS cells against the decrease of cell viability. On the other hand, pan-caspase, caspase-3, caspase-8 and caspase-9 inhibitor also protected tetrandrine-treated SAS cells against the decrease of cell viability (Fig. 5B).

Atg-5, beclin-1 siRNA, bafilomycin A1 and NAC decrease tetrandrine-induced cleaved caspase-3 and cleaved PARP in SAS cells

Results are presented in Fig. 6A, tetrandrine treatment induced the levels of cleaved caspase-3 and cleaved PARP, however, Atg-5, beclin-1 siRNA decreased tetrandrine-induced cleaved caspase-3 and cleaved PARP in SAS cells and protected tetrandrine-treated SAS cells against decrease of cell viability. In Fig. 6B, chloroquine, NAC and bafilomycin A1 also decreased tetrandrine-induced cleaved caspase-3 and cleaved PARP in SAS cells. In Fig. 6C, bafilomycin A1 decreased tetrandrine-induced LC3 expression in SAS cells.

Discussion

Oral cancer is most frequent in men between 55 and 65 and in women between 50 and 75, and in men this disease is the fourth most common cause of cancer death (44,45). The treatment for oral cancer is still unsatisfactory. Thus, it is necessary to identify and develop new anticancer agents that will selectively target the tumor. It was reported that the induction of apoptosis through the caspase cascade triggered by tetrandrine has inhibitory effects to various tumor cells (20,21,27,37). Many studies have demonstrated that tetrandrine has pharmacologic potential in cancer therapy but to date the effects of tetrandrine on human oral cancer is little known and have not been identified and reported in oral cancer cells. In the present study, we demonstrated the anticancer effect of tetrandrine on oral SAS cancer and elucidate the underlying mechanisms of action. Tetrandrine treatment showed growth inhibitory effect and cell apoptosis and autophagy induction on oral cancer SAS cells, which was associated with caspase activation (Fig. 4) and autophagic vesicle formation (Fig. 3). To the best of our knowledge, this is the first report of the effects of tetrandrine on human oral cancer.

The current studies indicate that SAS cells are sensitive to the cytotoxic actions of the tetrandrine and the sensitivity was largely similar to that observed previously in the liver and bladder tumor cell lines (27,31,35,37). Based on the observation and results of Annexin V and electro-microscope examination indicated that tetrandrine induced cell death may be through the formation of autophagy and the induction of apoptosis in SAS cells (Fig. 2). Autophagy from several complementary assays provided evidence for the formation of autophagic vesicles in SAS cells and LC3-I and II expression which confirmed the occurrence of autophagic flux. This is in agreement with a report on human hepatocellular carcinoma (HCC) cells after exposure to tetrandrine inducing autophagy (17). Tetrandrine also promoted apoptosis in SAS cells, although at low levels, however, both may lead to cell death. In contrast, tetrandrine appears to preferentially promote autophagy in SAS cells.

Based on other reports autophagy is quite controversial, for example, substantial evidence shows autophagy may occur through cytoprotective or through cytotoxic functions in different cancer cells (46,47). Herein, our results confirm that autophagy mediates the cell-killing effects of tetrandrine on SAS cells. The main reason is that the observations of autophagy from tetrandrine treated SAS cells was blocked by using preatment of 3-MA, NAC, chloroquine or bafilomycin that demonstrated only a modest degree of protection from the tetrandrine (Fig. 5). It may be due to autophagy being blocked thus leading to the tetrandrine-treated cells to cause apoptosis. These reasons are also seen from other studies (48,49). These observations indicate that SAS cells exposed to tetrandrine may be lethal to the tumor cells and that the cells may die by an alternative pathway if the primary pathway is blocked. Thus, further investigation is needed by in vivo experiments to investigate whether or not tetrandrine can be used as a potential anti-oral cancer agent. Other reports have demonstrated that if an agent is capable of promoting autophagy it is likely to have clinical utility (5054).

We demonstrated that tetrandrine induced apoptosis in SAS cells based on the results from flow cytometric assay (Fig. 2) and this may involve the activations of caspase-8, -9 and -3 (Fig. 4). Therefore, the possibility of mitophagy exists, at the low dose of tetrandrine and then selective removal of damaged mitochondria. However, other studies demonstrated that tetrandrine induced the release of cytochrome c but did not cause the activation of the caspases or apoptosis in human hepatocellular carcinoma cells (27). It was reported that during mitophagy (55), cell did not show caspase activation in apoptotic cell death. Our results show that tetrandrine promoted the expressions of LC3-I and -II (Fig. 4) and it is well documented that LC3-I and -II are involved in the development of autophagy and both proteins play an important role during autophagy (5658). Furthermore, other proteins such as Atg-5, Atg-7 and beclin-1 are also involved in the development, especial Atg-5 is essential for autophagy (5962). In this study, we found that protein levels of Atg-5 and beclin-1 (Fig. 4) were increased after tetrandrine treatment in SAS cells. As shown in Fig. 6, tetrandrine-induced autophagy in SAS cells is dependent on the upregulation of Atg-5 and beclin-1. However, tetrandrine did not affect the levels of Atg-7 in SAS cells, but other reports show that tetrandrine promoted the expression of Atg-5 and Atg-7 in HCC cells (17). The detailed mechanism of Atg-5 and Atg-7 in tetrandrine treated SAS cells will also require further investigation as other factors may be involved. We also used siRNA to knock down Atg-5, beclin-1, and LC3-1 and -II. The results show affect on Atg-5 and beclin-1 but not on LC3-I and LC3-II in SAS cells. After the knockdown of Atg-5 and beclin-1 by siRNA the cell proliferation was decreased; however, no significant reduction was observed in LC3-1 and LC3-II siRNA treatment (Fig. 6).

Taken together, we found that tetrandrine may be a promising chemotherapeutic agent with a variety of anticancer effects such as induction of autophagy and apoptosis in human oral cancer SAS cells in vitro. Tetrandrine treatment induced cancer cells to undergo apoptosis and autophagy. The upregulation of Atg-5, and beclin-1 was essential to the induction of tetrandrine-induced autophagy in SAS cells (Fig. 4). These findings suggest that tetrandrine may be a potential clinical candidate for the treatment of SAS.

References

1. 

Tseng CH: Oral cancer in Taiwan: is diabetes a risk factor? Clin Oral Investig. Aug 16–2012.(Epub ahead of print).

2. 

Bau DT, Tseng HC, Wang CH, et al: Oral cancer and genetic polymorphism of DNA double strand break gene Ku70 in Taiwan. Oral Oncol. 44:1047–1051. 2008. View Article : Google Scholar : PubMed/NCBI

3. 

Chie WC, Li CY, Huang CS, Chang KJ, Yen ML and Lin RS: Oral contraceptives and breast cancer risk in Taiwan, a country of low incidence of breast cancer and low use of oral contraceptives. Int J Cancer. 77:219–223. 1998. View Article : Google Scholar : PubMed/NCBI

4. 

Zavras AI, Douglass CW, Joshipura K, et al: Smoking and alcohol in the etiology of oral cancer: gender-specific risk profiles in the south of Greece. Oral Oncol. 37:28–35. 2001. View Article : Google Scholar : PubMed/NCBI

5. 

Moreno-Lopez LA, Esparza-Gomez GC, Gonzalez-Navarro A, Cerero-Lapiedra R, Gonzalez-Hernandez MJ and Dominguez-Rojas V: Risk of oral cancer associated with tobacco smoking, alcohol consumption and oral hygiene: a case-control study in Madrid, Spain. Oral Oncol. 36:170–174. 2000. View Article : Google Scholar : PubMed/NCBI

6. 

Kerawala CJ: Oral cancer, smoking and alcohol: the patients’ perspective. Br J Oral Maxillofac Surg. 37:374–376. 1999.

7. 

Shukla D, Dinesh Kale A, Hallikerimath S, Vivekanandhan S and Venkatakanthaiah Y: Genetic polymorphism of drug metabolizing enzymes (GSTM1 and CYP1A1) as risk factors for oral premalignant lesions and oral cancer. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 156:253–259. 2012. View Article : Google Scholar : PubMed/NCBI

8. 

Plu-Bureau G, Bossard N and Thalabard JC: Oral contraception and genetic factors in breast cancer: characteristics and limits of case-only studies. Rev Epidemiol Sante Publique. 48:294–303. 2000.(In French).

9. 

Hung HC, Chuang J, Chien YC, et al: Genetic polymorphisms of CYP2E1, GSTM1, and GSTT1; environmental factors and risk of oral cancer. Cancer Epidemiol Biomarkers Prev. 6:901–905. 1997.PubMed/NCBI

10. 

Yang YY, Koh LW, Tsai JH, et al: Involvement of viral and chemical factors with oral cancer in Taiwan. Jpn J Clin Oncol. 34:176–183. 2004. View Article : Google Scholar : PubMed/NCBI

11. 

Jung J, Cho NH, Kim J, et al: Significant invasion depth of early oral tongue cancer originated from the lateral border to predict regional metastases and prognosis. Int J Oral Maxillofac Surg. 38:653–660. 2009. View Article : Google Scholar : PubMed/NCBI

12. 

Hour MJ, Lee KT, Wu YC, et al: A novel antitubulin agent, DPQZ, induces cell apoptosis in human oral cancer cells through Ras/Raf inhibition and MAP kinases activation. Arch Toxicol. Dec 5–2012.(Epub ahead of print).

13. 

Shin JA, Kim JS, Hong IS and Cho SD: Bak is a key molecule in apoptosis induced by methanol extracts of Codonopsis lanceolata and Tricholoma matsutake in HSC-2 human oral cancer cells. Oncol Lett. 4:1379–1383. 2012.PubMed/NCBI

14. 

Tsai SC, Lu CC, Lee CY, et al: AKT serine/threonine protein kinase modulates bufalin-triggered intrinsic pathway of apoptosis in CAL 27 human oral cancer cells. Int J Oncol. 41:1683–1692. 2012.PubMed/NCBI

15. 

Xu XH, Gan YC, Xu GB, et al: Tetrandrine citrate eliminates imatinib-resistant chronic myeloid leukemia cells in vitro and in vivo by inhibiting Bcr-Abl/β-catenin axis. J Zhejiang Univ Sci B. 13:867–874. 2012.PubMed/NCBI

16. 

Gao S, Cui YL, Yu CQ, Wang QS and Zhang Y: Tetrandrine exerts antidepressant-like effects in animal models: role of brain-derived neurotrophic factor. Behav Brain Res. 238:79–85. 2013. View Article : Google Scholar : PubMed/NCBI

17. 

Gong K, Chen C, Zhan Y, Chen Y, Huang Z and Li W: Autophagy-related gene 7 (ATG7) and reactive oxygen species/extracellular signal-regulated kinase regulate tetrandrine-induced autophagy in human hepatocellular carcinoma. J Biol Chem. 287:35576–35588. 2012. View Article : Google Scholar

18. 

Shan QQ, Gong YP, Guo Y, Lin J, Zhou RQ and Yang X: Anti-tumor effect of tanshinone II A, tetrandrine, honokiol, curcumin, oridonin and paeonol on leukemia cell lines. Sichuan Da Xue Xue Bao Yi Xue Ban. 43:362–366. 2012.(In Chinese).

19. 

Wang G, Lemos JR and Iadecola C: Herbal alkaloid tetrandrine: fron an ion channel blocker to inhibitor of tumor proliferation. Trends Pharmacol Sci. 25:120–123. 2004. View Article : Google Scholar : PubMed/NCBI

20. 

Rao MR: Effects of tetrandrine on cardiac and vascular remodeling. Acta Pharmacol Sin. 23:1075–1085. 2002.PubMed/NCBI

21. 

Li X, Lu X, Xu H, et al: Paclitaxel/tetrandrine coloaded nanoparticles effectively promote the apoptosis of gastric cancer cells based on ‘oxidation therapy’. Mol Pharm. 9:222–229. 2012.PubMed/NCBI

22. 

Wu JM, Chen Y, Chen JC, Lin TY and Tseng SH: Tetrandrine induces apoptosis and growth suppression of colon cancer cells in mice. Cancer Lett. 287:187–195. 2010. View Article : Google Scholar : PubMed/NCBI

23. 

Liu B, Wang T, Qian X, Liu G, Yu L and Ding Y: Anticancer effect of tetrandrine on primary cancer cells isolated from ascites and pleural fluids. Cancer Lett. 268:166–175. 2008. View Article : Google Scholar : PubMed/NCBI

24. 

Chen YJ: Potential role of tetrandrine in cancer therapy. Acta Pharmacol Sin. 23:1102–1106. 2002.PubMed/NCBI

25. 

Ye Z, Sun A, Li L, Cao X and Ye W: Reversal of adriamycin or vincristine resistance by tetrandrine in human cancer cells in vitro. Zhongguo Zhong Yao Za Zhi. 21:369–371. 3841996.(In Chinese).

26. 

Jin Q, Kang C, Soh Y, et al: Tetrandrine cytotoxicity and its dual effect on oxidative stress-induced apoptosis through modulating cellular redox states in Neuro 2a mouse neuroblastoma cells. Life Sci. 71:2053–2066. 2002. View Article : Google Scholar

27. 

Kuo PL and Lin CC: Tetrandrine-induced cell cycle arrest and apoptosis in Hep G2 cells. Life Sci. 73:243–252. 2003. View Article : Google Scholar : PubMed/NCBI

28. 

Cheng F, Li Y, Feng L and Li S: Effects of tetrandrine on ischemia/reperfusion injury in mouse liver. Transplant Proc. 40:2163–2166. 2008. View Article : Google Scholar : PubMed/NCBI

29. 

Zhang P, Tang Iu N and Zhao YC: The effects of Tetrandrine on the changes of IL-1beta, TNF-alpha and IL-8 after cerebral ischemia/reperfusion in rats. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 22:443–444. 4782006.(In Chinese).

30. 

Liu Z, Xu Z, Shen W, Li Y, Zhang J and Ye X: Effect of pharmacologic preconditioning with tetrandrine on subsequent ischemia/ reperfusion injury in rat liver. World J Surg. 28:620–624. 2004.PubMed/NCBI

31. 

Chen Y, Chen JC and Tseng SH: Tetrandrine suppresses tumor growth and angiogenesis of gliomas in rats. Int J Cancer. 124:2260–2269. 2009. View Article : Google Scholar : PubMed/NCBI

32. 

Qian XP, Liu BR, Hu J, et al: Inhibitory effect of tetrandrine on angiogenesis. Ai Zheng. 27:1050–1055. 2008.(In Chinese).

33. 

Kobayashi S, Kimura I, Fukuta M, et al: Inhibitory effects of tetrandrine and related synthetic compounds on angiogenesis in streptozotocin-diabetic rodents. Biol Pharm Bull. 22:360–365. 1999. View Article : Google Scholar : PubMed/NCBI

34. 

Kobayashi S, Inaba K, Kimura I and Kimura M: Inhibitory effects of tetrandrine on angiogenesis in adjuvant-induced chronic inflammation and tube formation of vascular endothelial cells. Biol Pharm Bull. 21:346–349. 1998. View Article : Google Scholar : PubMed/NCBI

35. 

Liu C, Gong K, Mao X and Li W: Tetrandrine induces apoptosis by activating reactive oxygen species and repressing Akt activity in human hepatocellular carcinoma. Int J Cancer. 129:1519–1531. 2011. View Article : Google Scholar : PubMed/NCBI

36. 

Chen XL, Ren KH, He HW and Shao RG: Involvement of PI3K/ AKT/GSK3beta pathway in tetrandrine-induced G1 arrest and apoptosis. Cancer Biol Ther. 7:1073–1078. 2008. View Article : Google Scholar : PubMed/NCBI

37. 

Li X, Su B, Liu R, Wu D and He D: Tetrandrine induces apoptosis and triggers caspase cascade in human bladder cancer cells. J Surg Res. 166:e45–e51. 2011. View Article : Google Scholar : PubMed/NCBI

38. 

Lu KW, Chen JC, Lai TY, et al: Gypenosides suppress growth of human oral cancer SAS cells in vitro and in a murine xenograft model: the role of apoptosis mediated by caspase-dependent and caspase-independent pathways. Integr Cancer Ther. 11:129–140. 2012. View Article : Google Scholar : PubMed/NCBI

39. 

Lu KW, Chen JC, Lai TY, et al: Gypenosides causes DNA damage and inhibits expression of DNA repair genes of human oral cancer SAS cells. In Vivo. 24:287–291. 2010.PubMed/NCBI

40. 

Lu KW, Chen JC, Lai TY, et al: Gypenosides inhibits migration and invasion of human oral cancer SAS cells through the inhibition of matrix metalloproteinase-2 -9 and urokinaseplasminogen by ERK1/2 and NF-kappa B signaling pathways. Hum Exp Toxicol. 30:406–415. 2011. View Article : Google Scholar : PubMed/NCBI

41. 

Xie CM, Chan WY, Yu S, Zhao J and Cheng CH: Bufalin induces autophagy-mediated cell death in human colon cancer cells through reactive oxygen species generation and JNK activation. Free Radic Biol Med. 51:1365–1375. 2011. View Article : Google Scholar : PubMed/NCBI

42. 

Vazquez CL and Colombo MI: Assays to assess autophagy induction and fusion of autophagic vacuoles with a degradative compartment, using monodansylcadaverine (MDC) and DQ-BSA. Methods Enzymol. 452:85–95. 2009. View Article : Google Scholar : PubMed/NCBI

43. 

Chueh FS, Chen YL, Hsu SC, et al: Triptolide induced DNA damage in A375.S2 human malignant melanoma cells is mediated via reduction of DNA repair genes. Oncol Rep. 29:613–618. 2013.PubMed/NCBI

44. 

Revonta M, Raitanen J, Sihvo S, et al: Health and life style among infertile men and women. Sex Reprod Healthc. 1:91–98. 2010. View Article : Google Scholar : PubMed/NCBI

45. 

Palefsky JM, Gillison ML and Strickler HD: Chapter 16: HPV vaccines in immunocompromised women and men. Vaccine. 24(Suppl 3): S3/140–146. 2006. View Article : Google Scholar : PubMed/NCBI

46. 

Gondi CS and Rao JS: Cathepsin B as a cancer target. Expert Opin Ther Targets. 17:281–291. 2013. View Article : Google Scholar : PubMed/NCBI

47. 

Tekpli X, Holme JA, Sergent O and Lagadic-Gossmann D: Role for membrane remodeling in cell death: Implication for health and disease. Toxicology. 304:141–157. 2013. View Article : Google Scholar : PubMed/NCBI

48. 

Zhang XY, Xi XY and Zhao ZD: Autophagy inhibitor 3-MA decreases the production and release of infectious enterovirus 71 particles. Zhonghua Shi Yan He Lin Chuang Bing Du Xue Za Zhi. 25:176–178. 2011.(In Chinese).

49. 

Liu D, Yang Y, Liu Q and Wang J: Inhibition of autophagy by 3-MA potentiates cisplatin-induced apoptosis in esophageal squamous cell carcinoma cells. Med Oncol. 28:105–111. 2011. View Article : Google Scholar : PubMed/NCBI

50. 

He Y, Zhao X, Gao J, et al: Quantum dots-based immuno-fluorescent imaging of stromal fibroblasts caveolin-1 and light chain 3b expression and identification of their clinical significance in human gastric cancer. Int J Mol Sci. 13:13764–13780. 2012. View Article : Google Scholar

51. 

Liu GH, Zhong Q, Ye YL, et al: Expression of beclin 1 in bladder cancer and its clinical significance. Int J Biol Markers. Nov 5–2012.(Epub ahead of print).

52. 

Levy JM and Thorburn A: Targeting autophagy during cancer therapy to improve clinical outcomes. Pharmacol Ther. 131:130–141. 2011. View Article : Google Scholar : PubMed/NCBI

53. 

Schrump DS: Cytotoxicity mediated by histone deacetylase inhibitors in cancer cells: mechanisms and potential clinical implications. Clin Cancer Res. 15:3947–3957. 2009. View Article : Google Scholar : PubMed/NCBI

54. 

Shen Y, Liang LZ, Hong MH, Xiong Y, Wei M and Zhu XF: Expression and clinical significance of microtubule-associated protein 1 light chain 3 (LC3) and Beclin1 in epithelial ovarian cancer. Ai Zheng. 27:595–599. 2008.(In Chinese).

55. 

Pavlides S, Vera I, Gandara R, et al: Warburg meets autophagy: cancer-associated fibroblasts accelerate tumor growth and metastasis via oxidative stress, mitophagy, and aerobic glycolysis. Antioxid Redox Signal. 16:1264–1284. 2012. View Article : Google Scholar

56. 

McLeland CB, Rodriguez J and Stern ST: Autophagy monitoring assay: qualitative analysis of MAP LC3-I to II conversion by immunoblot. Methods Mol Biol. 697:199–206. 2011. View Article : Google Scholar : PubMed/NCBI

57. 

Reggiori F, Monastyrska I, Verheije MH, et al: Coronaviruses Hijack the LC3-I-positive EDEMosomes, ER-derived vesicles exporting short-lived ERAD regulators, for replication. Cell Host Microbe. 7:500–508. 2010. View Article : Google Scholar : PubMed/NCBI

58. 

Gimenez-Xavier P, Francisco R, Platini F, Perez R and Ambrosio S: LC3-I conversion to LC3-II does not necessarily result in complete autophagy. Int J Mol Med. 22:781–785. 2008.PubMed/NCBI

59. 

Cao Y, Nair U, Yasumura-Yorimitsu K and Klionsky DJ: A multiple ATG gene knockout strain for yeast two-hybrid analysis. Autophagy. 5:699–705. 2009. View Article : Google Scholar : PubMed/NCBI

60. 

Noda NN, Ohsumi Y and Inagaki F: ATG systems from the protein structural point of view. Chem Rev. 109:1587–1598. 2009. View Article : Google Scholar : PubMed/NCBI

61. 

Stasyk OV, Nazarko TY and Sibirny AA: Methods of plate pexophagy monitoring and positive selection for ATG gene cloning in yeasts. Methods Enzymol. 451:229–239. 2008. View Article : Google Scholar : PubMed/NCBI

62. 

Chung T, Suttangkakul A and Vierstra RD: The ATG autophagic conjugation system in maize: ATG transcripts and abundance of the ATG8-lipid adduct are regulated by development and nutrient availability. Plant Physiol. 149:220–234. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August 2013
Volume 43 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang A, Lien J, Lin M, Yang J, Wu P, Chang S and Lai T: Tetrandrine induces cell death in SAS human oral cancer cells through caspase activation-dependent apoptosis and LC3-I and LC3-II activation-dependent autophagy. Int J Oncol 43: 485-494, 2013
APA
Huang, A., Lien, J., Lin, M., Yang, J., Wu, P., Chang, S., & Lai, T. (2013). Tetrandrine induces cell death in SAS human oral cancer cells through caspase activation-dependent apoptosis and LC3-I and LC3-II activation-dependent autophagy. International Journal of Oncology, 43, 485-494. https://doi.org/10.3892/ijo.2013.1952
MLA
Huang, A., Lien, J., Lin, M., Yang, J., Wu, P., Chang, S., Lai, T."Tetrandrine induces cell death in SAS human oral cancer cells through caspase activation-dependent apoptosis and LC3-I and LC3-II activation-dependent autophagy". International Journal of Oncology 43.2 (2013): 485-494.
Chicago
Huang, A., Lien, J., Lin, M., Yang, J., Wu, P., Chang, S., Lai, T."Tetrandrine induces cell death in SAS human oral cancer cells through caspase activation-dependent apoptosis and LC3-I and LC3-II activation-dependent autophagy". International Journal of Oncology 43, no. 2 (2013): 485-494. https://doi.org/10.3892/ijo.2013.1952