Demethoxycurcumin induces the apoptosis of human lung cancer NCI-H460 cells through the mitochondrial-dependent pathway

  • Authors:
    • Yang-Ching Ko
    • Jin-Cherng Lien
    • Hsin-Chung Liu
    • Shu-Chun Hsu
    • Bin-Chuan Ji
    • Mei-Due Yang
    • Wu-Huei Hsu
    • Jing-Gung Chung
  • View Affiliations

  • Published online on: March 18, 2015     https://doi.org/10.3892/or.2015.3865
  • Pages: 2429-2437
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung cancer is the most common cause of cancer-related mortality in the US as well as other regions of the world. Curcumin, demethoxycurcumin (DMC) and bisdemethoxycurcumin (BDMC) are the major components of Curcuma longa L. It has been reported that curcumin inhibits the growth of various types of cancer cells in vitro and in vivo. However, the mechanisms involved in the inhibition of cell growth and induced apoptosis by DMC in human lung cancer cells remain unclear. In the present study, we investigated the effect of DMC on cell death via the induction of apoptosis in NCI-H460 human lung cancer cells. Flow cytometric assay was used to examine the total percentage of viable cells, the population of cells in the sub-G1 phase of the cell cycle, the level of reactive oxygen species (ROS), Ca2+ production, mitochondrial membrane potential (ΔΨm) and caspase activity. Western blotting was used to examine the changes in the expression of cell cycle- and apoptosis‑associated proteins. Confocal microscopy was used to examine the translocation of apoptosis-associated proteins. The results indicated that DMC significantly induced cell morphological changes and decreased the percentage of viable NCI-H460 cells and DMC induced apoptosis based on the cell distribution in the sub-G1 phase. Moreover, DMC promoted ROS and Ca2+ production and decreased the level of ΔΨm and promoted the activities of caspase-3, -8 and -9. The Western blotting results showed that DMC promoted the expression of AIF, Endo G and PARP. The levels of Fas ligand (Fas L) and Fas were also upregulated. Furthermore, DMC promoted expression of ER stress-associated proteins such as GRP78, GADD153, IRE1β, ATF-6α, ATF-6β and caspase-4. Based on the findings, we suggest that DMC may be used as a novel anticancer agent for the treatment of lung cancer in the future.

Introduction

Non-small cell lung carcinoma is one of the leading causes of cancer-related mortality worldwide (1). Standard platinum-based chemotherapies provide marginal improvement in survival at the expense of substantial toxicity (2). Even with the addition of targeted therapy, the median survival of metastatic non-small cell lung cancer patients is approximately one year (3). Due to the unsatisfactory results of standard chemotherapy, the identification of new drugs is crucial.

In recent years, the public has become increasingly aware of alternative medical therapies through all forms of media in Western countries, and the use of complementary and alternative medicine has also increased, particularly among oncology patients. Curcumin, isolated from turmeric (Curcuma longa L.), contains curcumin as a major component but also contains demethoxycurcumin (DMC) and bisdemethoxycurcumin (BDMC), respectively (4). Extensive research during the last half century has revealed several important functions of curcumin such as antioxidant, anti-inflammatory and anticancer properties (57). Recently, more and more studies have demonstrated the stronger activity of DMC when compared with curcumin in many aspects (810). Numerous studies have shown that DMC induces cytotoxic effects in many cancer cell lines such as colon (11) and renal cell cancer (12), glioma (9) and leukemic cell lines (13). However, no study exists which shows the effects of DMC on human lung cancer cells, and the role of DMC in inducing cell cycle arrest and apoptosis has never been investigated in detail.

The NCI-H460 cell line is derived from human large cell lung cancer, which is one of the major types of non-small cell lung carcinoma. In the present study, we investigated the cytotoxic effects of DMC on human lung cancer NCI-H460 cells and we found that DMC induced cell death through the induction of apoptosis in vitro.

Materials and methods

Chemicals and reagents

DMC, dimethyl sulfoxide (DMSO), propidium iodide (PI) and Trypsin-EDTA were purchased from Sigma Chemical Co. (St. Louis, MO, USA). Culture medium RPMI-1640, fetal bovine serum (FBS), L-glutamine and penicillin-streptomycin were purchased from Gibco/Invitrogen Life Technologies (Carlsbad, CA, USA). Primary antibodies (anti-AIF, -Endo G, -GRP78, -GADD153, -IRE1β, -ATF-6α, -ATF-6β, and -caspase-4) were obtained from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA).

Cell culture

The human lung cancer cell line NCI-H460 was purchased from the Food Industry Research and Development Institute (Hsinchu, Taiwan). The cells were cultured in RPMI-1640 medium supplemented with 10% FBS, 100 U/ml penicillin, 100 μg/ml streptomycin and 2 mM glutamine and were incubated in a 5% CO2 humidified incubator at 37°C in a 75-cm2 tissue culture flask.

Assessment of cell morphological changes and viability

NCI-H460 cells were plated onto 12-well plates at a density of 2×105 cells/well, and DMC was added at final concentrations of 0, 15, 20, 25, 30 or 35 μM. In control wells, only DMSO (solvent) was added. The cells were exposed for 24 and 48 h. A phase-contrast microscope was used to observe morphological changes in the examined cells at the end of the time period. Cell viability was estimated through flow cytometric methods as described elsewhere (14,15).

Cell cycle distribution and sub-G1 assays

Approximately 2×105 cells/well of NCI-H460 cells in 12-well plates were incubated with 0, 15, 20, 25, 30, 35 and 40 μM of DMC for 48 h. The cells were harvested by centrifugation, washed with PBS and fixed in 70% ethanol at −20°C overnight. The cells were then resuspended in PBS containing 40 μg/ml of PI, 0.1 mg/ml RNase and 0.1% Triton X-100 in a dark room for 30 min and were subsequently analyzed by a flow cytometer (FACSCalibur; Becton-Dickinson, San Jose, CA, USA) (16,17). The cell cycle distribution and sub-G1 groups (apoptosis) were calculated and analyzed by CellQuest (Becton-Dickinson) and ModFit LT software (Verity Software House Inc., Topsham, ME, USA).

Reactive oxygen species (ROS), intracellular Ca2+ and mitochondrial membrane potential (ΔΨm) assays

NCI-H460 cells (2×105 cells/well) were treated with 35 μM of DMC for different time intervals. At the end of the incubation, cells from each treatment and time-points were collected, washed, counted and then were resuspended in 500 μl of DCFH-DA (10 μM) for 30 min for ROS (H2O2) measurement, resuspended in 500 μl of Fluo-3/AM (2.5 μg/ml) for 30 min for intracellular Ca2+ concentration measurement and resuspended in 500 μl of DiOC6 (4 μmol/l) for 30 min to determine the levels of ΔΨm. All samples were then individually analyzed by flow cytometry as described previously (16).

Caspase-3, -8 and -9 activity assay

NCI-H460 cells were plated onto 12-well plates at a density of 2×105 cells/well and incubated with or without 35 μM DMC. The cells were then incubated for 0, 6, 24 and 48 h and harvested, washed and resuspended in 25 μl of 10 μM substrate solution (PhiPhiLux and CaspaLux kit; OncoImmunin, Inc., Gaithersburg, MD, USA) before being incubated at 37°C for 60 min. The cells were washed again in PBS and were analyzed by flow cytometry as described previously (16,18,19).

Effects of DMC on expression of apoptosis-associated proteins as determined by western blot analysis

Cells (2×106/dish) were treated with 35 μM DMC and incubated for 0, 6, 24 and 48 h. The abundance of selective proteins associated with apoptosis was determined by western blotting. Briefly, at the end of the incubation, the cells were harvested and lysed as described previously (18,19). The levels of apoptosis-associated proteins were determined in the cell lysates using primary antibodies (those associated with the cell cycle such as anti-p21, p27, CDC25c, CDK2, cyclin A and cyclin E; those associated with apoptosis such as anti-AIF, Endo G, PARP, Fas-L and Fas; those associated with ER stress such as ATF6α, ATF-6β, IRE1β, GRP78, GADD153, caspase-4 and -12). For equal protein loading, each membrane was stripped and reprobed with the anti-β-actin antibody (18,19).

Confocal laser scanning microscopy

NCI-H460 cells (3×105 cells/well) were placed on 6-well chamber slides and incubated with or without 35 μM DMC for 48 h. The cells were then fixed, washed and permeabilized as described previously (16,17). Then anti-AIF, Endo G, ATF6β, IRE1α and p-PERK (all in green fluorescence) were individually used for staining each sample overnight, followed by washing and then staining with the secondary antibody (FITC-conjugated goat anti-mouse IgG). The cells were then stained using PI (red fluorescence) for nuclear examination under a Leica TCS SP2 confocal spectral microscope as described previously (16,17).

Statistical analysis

All data are expressed as the mean ± SD of 3 experiments. Statistical analysis was performed using the Student’s t-test, with a value of P<0.05 considered to indicate a statistically significant difference between the DMC-treated and untreated (control) group.

Results

DMC induces cell morphological changes and decreases the cell viability of NCI-H460 cells

The NCI-H460 cells were treated with various concentrations of DMC for 24 and 48 h and then were photographed to examine morphological changes. The percentage of total viable cells was then determined. DMC significantly induced cell morphological changes in a concentration-dependent manner and these changes were based on an increase in cell death and debris (Fig. 1A and B). The flow cytometric assay indicated that DMC decreased the percentage of viable cells in a concentration-dependent manner (Fig. 1C).

Cell cycle arrest and apoptosis of the NCI-H460 cells after treatment with DMC

The cell cycle distribution of the NCI-H460 cells after treatment with 0, 15, 20, 25, 30, 35 and 40 μM of DMC for 48 h is shown in Fig. 2A. Cell cycle arrest appeared to occur at the S stage after exposure to DMC. The sub-G1 peak, indicating the proportion of apoptosis, increased in a dose-dependent manner when the concentration of DMC was increased (Fig. 2B).

Effects of DMC on ROS production, ΔΨm and intracellular Ca2+ levels in the NCI-H460 cells

Following DMC treatment for different time intervals, ROS (Fig. 3A) and intracellular Ca2+ (Fig. 3C) were significantly increased as compared with the control group. In addition, there was a significant loss of ΔΨm after treatment with 35 μM of DMC (Fig. 3B).

DMC promotes the activity of caspase-3, -8 and -9 in the NCI-H460 cells

After treatment with DMC for different durations, the NCI-H460 cells exhibited increased caspase-3 activity. The caspase-3 activity reached its maximum when the duration of treatment was 48 h. The caspase-8 and -9 activities in the NCI-H460 cells were also increased following treatment of DMC at 35 μM (Fig. 4). These effects were time-dependent with the exception of the 24-h incubation, which showed a reduction in activity compared with the 6-h treatment.

DMC affects cell cycle arrest and levels of apoptosis-associated proteins and protein translocation in the NCI-H460 cells

In order to investigate whether DMC induces apoptosis in NCI-H460 cells via changes in the cell cycle and levels of apoptosis-associated proteins, the NCI-H460 cells were treated with 35 μM of DMC for 0, 6, 24 and 48 h and then levels of total proteins from the samples were quantitated and the levels of apoptosis-associated proteins were measured by western blotting. DMC significantly promoted the expression of p21 and p27 but reduced the expression of CDC25A, cyclin E, cyclin A and CDK2 (Fig. 5A). AIF, Endo G, PARP, Fas ligand (Fas L) and Fas were also upregulated (Fig. 5B). Furthermore, DMC promoted expression of ER stress-associated proteins GRP78, GADD153, IRE1β, ATF6α, ATF6β, caspase-12 and -4 (Fig. 5C). Moreover, DMC promoted the expression of calpain 1 (Fig. 5C), which is associated with apoptotic pathways. The results revealed that DMC induced apoptosis in the NCI-H460 cells through caspase-, ER stress- and mitochondrial-dependent pathways. The results from the confocal laser microscopy also revealed that DMC promoted the release of AIF (Fig. 6A), Endo G (Fig. 6B), ATF6β (Fig. 6C), IRE1α (Fig. 6D) and p-PERK (Fig. 6E) from the mitochondria to the cytosol and nuclei, respectively.

Discussion

Much evidence has shown that stimulating or inducing tumor cell apoptosis is one possibility for tumor treatment in patients with cancer. Although a few studies have shown that DMC induces cell death and apoptosis in human cancer cells as described previously, there is no study to show that DMC affects human lung cancer cells. The results of the present study revealed that DMC induced cell morphological changes (Fig. 1A and B) and decreased the percentage of viable cells (Fig. 1C) via the induction of the sub-G1 phase (apoptosis) and cell cycle arrest (Fig. 2A). DMC-induced apoptosis in the NCI-H460 cells was dose-dependent (Fig. 2B).

Dysregulation of the cell cycle is associated with tumorigenesis (18). We found that NCI-H460 cells were arrested at the S phase after treatment with DMC (Fig. 2A). Downregulation of CDK2, CDC25A, cyclin A and cyclin E as shown by western blotting (Fig. 5A), may be involved in the mechanism of this arrest. Based on the findings, DMC may exert its anticancer effects on NCI-H460 cells through both cell cycle arrest and apoptotic induction.

To further examine the molecular mechanism of DMC in NCI-H460 cells, we used flow cytometry and found that DMC significantly decreased the levels of ΔΨm (Fig. 3B) following a 24-h treatment and increased ROS and Ca2+ levels (Fig. 3A and C) at all treatment time periods. DMC also promoted the activities of caspase-3, -9 and -8 (Fig. 4).

Numerous studies have demonstrated that apoptotic cell death can occur through the extrinsic or the intrinsic apoptotic pathway (2022). The agents (Fas L) connected with Fas (CD95) then trigger the extrinsic pathway followed by the activation of caspase-8 and then activation of effector caspase-3 to cause cell apoptosis (23). Thus, we hypothesized that the extrinsic apoptotic pathway in NCI-H460 cells was activated following exposure to DMC. Thus, we used western blotting to examine the expression of FAS/CD95 in the NCI-H460 cells. The results revealed that DMC increased the expression of FAS/CD95 and Fas L (Fig. 5B) accompanied by increased caspase-8 activity (Fig. 4B).

It is currently known that cancer cell survival or death following exposure to anticancer drugs is associated with mitochondrial function. Thus, anticancer drugs may induce cancer cell apoptosis through mitochondrial-dependent and -independent pathways. Anticancer drugs may induce mitochondrial dysfunction in cells via dissipation of ΔΨm leading to liberation of numerous cell death proteins from the mitochondria (24). Therefore, it was reported that the intrinsic pathway depends on the dysfunction of the mitochondria resulting from an increase in the ratio of Bak:Bcl-2 which is caused by anticancer drugs thus leading to AIF and Endo G release from the mitochondria before inducing apoptosis (2527) which is termed caspase-independent pathways or alternatively causing cytochrome c release, activation of caspase-9 and -3 resulting in apoptosis termed the caspase-dependent pathway (28). Our results showed that DMC induced mitochondrial dysfunction (decreased levels of ΔΨm) (Fig. 3B) and increased caspase-8, -9 and -3 activity (Fig. 4) and increased the protein levels of AIF and Endo G (Fig. 5B) in the NCI-H460 cells. We also used confocal laser microscopy to confirm that DMC increased the expression of AIF and Endo G (Fig. 6A and B). Based on the findings, we suggest that DMC induced apoptosis in NCI-H460 cells through the mitochondrial-dependent and -independent pathways.

DMC induced ROS production in the NCI-H460 cells (Fig. 3A) and ROS have been shown to be involved in cell growth and apoptosis. An appropriate level of intracellular ROS promotes cellular proliferation (29), while excessive production of ROS may cause oxidative stress leading to cell apoptosis (29,30).

It has been reported that ROS may induce ER stress which leads to the release of stored Ca2+ from the ER leading to mitochondrial Ca2+ loading from ER stores (31). Both ROS and ER Ca2+ are required to initiate mitochondrial dysfunction (31). Our results revealed that DMC increased the ROS and Ca2+ production (Fig. 3A and C). Furthermore, it was reported that mitochondria take up Ca2+ and initiate apoptosis through opening of their permeability transition pores (32). This then results in the release of cytochrome c from the mitochondrial membrane which then activates caspase-9 and triggers the effector caspase-3 for causing apoptosis (33). It was reported that markers of ER stress such as transcriptionally induced GRP78 and GADD153 are produced (31). We found that DMC increased the protein levels of GRP78, GADD153, IRE1β, ATF6α, ATF6β, caspase-12 and -4 and the expression of calpain 1 (Fig. 5C), that are associated with apoptosis pathways.

In conclusion, DMC induced cell death (cytotoxic effect) in human lung cancer NCI-H460 cells, mediated through the induction of phase S cell cycle arrest by inhibition of the check-point proteins CDC25A, cyclin A, cyclin E and CDK2 and induced cell apoptosis associated with caspase-or mitochondrial-dependent pathways. Furthermore, DMC induced cell apoptosis also through cross-talk between the extrinsic and the intrinsic pathway as summarized in Fig. 7. DMC appears to have multiple molecular targets, and its application in the treatment of lung cancer patients warrants further investigation.

Acknowledgments

This study was supported in part by a research grant from China Medical University (no. CMU102-ASIA-20). Experiments and data analysis were performed in part through the use of the Medical Research Core Facilities Center, Office of Research and Development at China Medical University, Taichung, Taiwan, R.O.C.

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Non-small Cell Lung Cancer Collaborative Group: Chemotherapy in non-small cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials. BMJ. 311:899–909. 1995. View Article : Google Scholar

3 

Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, Lilenbaum R and Johnson DH: Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 355:2542–2550. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Jayaprakasha GK, Jagan Mohan Rao L and Sakariah KK: Improved HPLC method for the determination of curcumin, demethoxycurcumin, and bisdemethoxycurcumin. J Agric Food Chem. 50:3668–3672. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Balasubramanyam M, Koteswari AA, Kumar RS, Monickaraj SF, Maheswari JU and Mohan V: Curcumin-induced inhibition of cellular reactive oxygen species generation: novel therapeutic implications. J Biosci. 28:715–721. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Chan WH, Wu HJ and Hsuuw YD: Curcumin inhibits ROS formation and apoptosis in methylglyoxal-treated human hepatoma G2 cells. Ann NY Acad Sci. 1042:372–378. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Priyadarsini KI, Maity DK, Naik GH, Kumar MS, Unnikrishnan MK, Satav JG and Mohan H: Role of phenolic O-H and methylene hydrogen on the free radical reactions and antioxidant activity of curcumin. Free Radic Biol Med. 35:475–484. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Ahmed T and Gilani AH: Inhibitory effect of curcuminoids on acetylcholinesterase activity and attenuation of scopolamine-induced amnesia may explain medicinal use of turmeric in Alzheimer’s disease. Pharmacol Biochem Behav. 91:554–559. 2009. View Article : Google Scholar

9 

Luthra PM, Kumar R and Prakash A: Demethoxycurcumin induces Bcl-2 mediated G2/M arrest and apoptosis in human glioma U87 cells. Biochem Biophys Res Commun. 384:420–425. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Yodkeeree S, Chaiwangyen W, Garbisa S and Limtrakul P: Curcumin, demethoxycurcumin and bisdemethoxycurcumin differentially inhibit cancer cell invasion through the down-regulation of MMPs and uPA. J Nutr Biochem. 20:87–95. 2009. View Article : Google Scholar

11 

Tamvakopoulos C, Dimas K, Sofianos ZD, Hatziantoniou S, Han Z, Liu ZL, Wyche JH and Pantazis P: Metabolism and anticancer activity of the curcumin analogue, dimethoxycurcumin. Clin Cancer Res. 13:1269–1277. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Lee JW, Hong HM, Kwon DD, Pae HO and Jeong HJ: Dimethoxycurcumin, a structural analogue of curcumin, induces apoptosis in human renal carcinoma caki cells through the production of reactive oxygen species, the release of cytochrome c, and the activation of caspase-3. Korean J Urol. 51:870–878. 2010. View Article : Google Scholar

13 

Anuchapreeda S, Tima S, Duangrat C and Limtrakul P: Effect of pure curcumin, demethoxycurcumin, and bisdemethoxycurcumin on WT1 gene expression in leukemic cell lines. Cancer Chemother Pharmacol. 62:585–594. 2008. View Article : Google Scholar

14 

Ji BC, Hsu WH, Yang JS, et al: Gallic acid induces apoptosis via caspase-3 and mitochondrion-dependent pathways in vitro and suppresses lung xenograft tumor growth in vivo. J Agric Food Chem. 57:7596–7604. 2009. View Article : Google Scholar

15 

Lin SY, Lai WW, Ho CC, et al: Emodin induces apoptosis of human tongue squamous cancer SCC-4 cells through reactive oxygen species and mitochondria-dependent pathways. Anticancer Res. 29:327–335. 2009.PubMed/NCBI

16 

Gorczyca W, Melamed MR and Darzynkiewicz Z: Laser scanning cytometer (LSC) analysis of fraction of labelled mitoses (FLM). Cell Prolif. 29:539–547. 1996. View Article : Google Scholar : PubMed/NCBI

17 

Hsia TC, Yang JS, Chen GW, et al: The roles of endoplasmic reticulum stress and Ca2+ on rhein-induced apoptosis in A-549 human lung cancer cells. Anticancer Res. 29:309–318. 2009.PubMed/NCBI

18 

Diehl JA: Cycling to cancer with cyclin D1. Cancer Biol Ther. 1:226–231. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Liu DD, Ye YL, Zhang J, Xu JN, Qian XD and Zhang Q: Distinct pro-apoptotic properties of Zhejiang saffron against human lung cancer via a caspase-8-9-3 cascade. Asian Pac J Cancer Prev. 15:6075–6080. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Kumar S: Caspase function in programmed cell death. Cell Death Differ. 14:32–43. 2007. View Article : Google Scholar

21 

Wilson MR: Apoptotic signal transduction: Emerging pathways. Biochem Cell Biol. 76:573–582. 1998. View Article : Google Scholar

22 

Xu G and Shi Y: Apoptosis signaling pathways and lymphocyte homeostasis. Cell Res. 17:759–771. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Kim HJ, Yang KM, Park YS, Choi YJ, Yun JH, Son CH, Suh HS, Jeong MH and Jo WS: The novel resveratrol analogue HS-1793 induces apoptosis via the mitochondrial pathway in murine breast cancer cells. Int J Oncol. 41:1628–1634. 2012.PubMed/NCBI

24 

Tait SW and Green DR: Mitochondria and cell death: Outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 11:621–632. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Baek SH, Bae ON, Kim EK and Yu SW: Induction of mitochondrial dysfunction by poly(ADP-ribose) polymer: implication for neuronal cell death. Mol Cells. 36:258–266. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Liou GY and Storz P: Reactive oxygen species in cancer. Free Radic Res. 44:479–496. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Liu KC, Huang YT, Wu PP, Ji BC, Yang JS, Yang JL, Chiu TH, Chueh FS and Chung JG: The roles of AIF and Endo G in the apoptotic effects of benzyl isothiocyanate on DU 145 human prostate cancer cells via the mitochondrial signaling pathway. Int J Oncol. 38:787–796. 2011.PubMed/NCBI

28 

Yoo JO, Lim YC, Kim YM and Ha KS: Transglutaminase 2 promotes both caspase-dependent and caspase-independent apoptotic cell death via the calpain/Bax protein signaling pathway. J Biol Chem. 287:14377–14388. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Circu ML and Aw TY: Reactive oxygen species, cellular redox systems, and apoptosis. Free Radic Biol Med. 48:749–762. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Hajnóczky G, Csordás G, Das S, Garcia-Perez C, Saotome M, Sinha Roy S and Yi M: Mitochondrial calcium signalling and cell death: approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis. Cell Calcium. 40:553–560. 2006. View Article : Google Scholar

31 

Jacobson J and Duchen MR: Mitochondrial oxidative stress and cell death in astrocytes - requirement for stored Ca2+ and sustained opening of the permeability transition pore. J Cell Sci. 115:1175–1188. 2002.PubMed/NCBI

32 

Garrido C, Galluzzi L, Brunet M, Puig PE, Didelot C and Kroemer G: Mechanisms of cytochrome c release from mitochondria. Cell Death Differ. 13:1423–1433. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Rao RV, Ellerby HM and Bredesen DE: Coupling endoplasmic reticulum stress to the cell death program. Cell Death Differ. 11:372–380. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 33 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ko Y, Lien J, Liu H, Hsu S, Ji B, Yang M, Hsu W and Chung J: Demethoxycurcumin induces the apoptosis of human lung cancer NCI-H460 cells through the mitochondrial-dependent pathway. Oncol Rep 33: 2429-2437, 2015
APA
Ko, Y., Lien, J., Liu, H., Hsu, S., Ji, B., Yang, M. ... Chung, J. (2015). Demethoxycurcumin induces the apoptosis of human lung cancer NCI-H460 cells through the mitochondrial-dependent pathway. Oncology Reports, 33, 2429-2437. https://doi.org/10.3892/or.2015.3865
MLA
Ko, Y., Lien, J., Liu, H., Hsu, S., Ji, B., Yang, M., Hsu, W., Chung, J."Demethoxycurcumin induces the apoptosis of human lung cancer NCI-H460 cells through the mitochondrial-dependent pathway". Oncology Reports 33.5 (2015): 2429-2437.
Chicago
Ko, Y., Lien, J., Liu, H., Hsu, S., Ji, B., Yang, M., Hsu, W., Chung, J."Demethoxycurcumin induces the apoptosis of human lung cancer NCI-H460 cells through the mitochondrial-dependent pathway". Oncology Reports 33, no. 5 (2015): 2429-2437. https://doi.org/10.3892/or.2015.3865