Sedanolide induces autophagy through the PI3K, p53 and NF-κB signaling pathways in human liver cancer cells

  • Authors:
    • Shu-Ling Hsieh
    • Chi-Tsai Chen
    • Jyh-Jye Wang
    • Yu-Hao Kuo
    • Chien-Chun Li
    • Lan-Chi Hsieh
    • Chih-Chung Wu
  • View Affiliations

  • Published online on: October 15, 2015     https://doi.org/10.3892/ijo.2015.3206
  • Pages: 2240-2246
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sedanolide (SN), a phthalide-like compound from celery seed oil, possesses antioxidant effects. However, the effect of SN on cell death in human liver cancer cells has yet to be determined. In this study, cell viability determination, monodansylcadaverine (MDC) fluorescent staining and immunoblot analysis were performed to determine autophagy induction and autophagy-induced protein expression changes via molecular examination after human liver cancer (J5) cells were treated with SN. Our studies demonstrate that SN suppressed J5 cell viability by inducing autophagy. Phosphoinositide 3-kinase (PI3K)-I, mammalian target of rapamycin (mTOR) and Akt protein levels decreased, whereas PI3K-III, LC3-II and Beclin-1 protein levels increased following SN treatment in J5 cells. In addition, SN treatment upregulated nuclear p53 and damage-regulated autophagy modulator (DRAM) and downregulated cytosolic p53 and Tp53-induced glycolysis and apoptosis regulator (TIGAR) expression in J5 cells. Furthermore, the cytosolic phosphorylation of inhibitor of kappa B (IκB) and nuclear p65 and the DNA-binding activity of NF-κB increased after SN treatment. These results suggest that SN induces J5 cell autophagy by regulating PI3K, p53 and NF-κB autophagy-associated signaling pathways in J5 cells.

Introduction

Human hepatocellular carcinoma (HCC) is one of the most common malignant tumors and a significant cause of mortality in several regions of Africa and Asia (1,2). Triggering cancer cell death to reduce cancer cell number and inhibiting cancer cell proliferation by phytochemicals or chemotherapeutic agents represent some of the most effective current anticancer strategies (3). Cell death can occur through one of three pathways: necrosis, apoptosis or autophagy (4).

Autophagy is induced by various physiological conditions, such as mitochondrial damage, protein aggregation, pathogen infection and nutrient starvation (5,6). Autophagy is a multi-step cellular pathophysiological program, including initiation (pre-autophagosome formation), autophagosome formation, maturation and degradation (7). In these steps, phosphoinositide 3-kinases (PI3Ks) play key regulatory roles in many cellular processes, including cell survival, proliferation and differentiation (810). Both class I PI3K (PI3K-I)/Akt/ mammalian target of rapamycin (mTOR) and Beclin-1/class III PI3K (PI3K-III)/LC3-II signaling pathways are involved in pre-autophagosome formation (1114). Cytosolic and nuclear p53, damage-regulated autophagy modulator (DRAM) and the p53-induced glycolysis and apoptosis regulator (TIGAR) are involved in autophagosome formation (15). In addition, p53 initiates a cascade of starvation signals and triggers a starvation-like response by inhibiting mTOR (16). Furthermore, p53 activates both the DRAM and PI3K-III pathways upon autophagosome formation (15). Phytochemicals or chemical agents that regulate p53- and PI3K-mediated autophagy induction could serve as a potential starting point for novel methods of cancer prevention and treatment.

The transcription factor nuclear factor-kappa B (NF-κB) plays an important role in autophagy-induced signaling pathways (17,18). Several genes involved in autophagy, such as the genes encoding Beclin-1, LC3-II and DRAM, are regulated by NF-κB (15,19,20). Regulation of NF-κB activation affects autophagy-relevant gene expression, leading to the induction of autophagy.

Sedanolide (SN; 3-butyl-3a,4,5,6-tetrahydro-1(3H)-isobenzofuranone), is a phthalide-like compound from celery (Apium graveolens L.) seed oil (21,22). Celery seed contains 2% volatile oil. The typical aroma of celery seed essential oils arises due to SN, which is present at very low levels (1–3%) in the essential oil (23). Previous studies have demonstrated that SN protects against hydrogen peroxide- and tert-butyl hydroperoxide-induced toxicity in HepG2 and CaCo-2 cells (22). SN exhibits prostaglandin H endoperoxide synthase-I (COX-I) and prostaglandin H endoperoxide synthase-II (COX-II) inhibitory activities and topoisomerase-I and -II enzyme inhibitory activities (24). In benzo[α]pyrene-(BP) induced forestomach cancer in mice, SN reduced tumor incidence by 57% and tumor multiplicity by 83% by increasing glutathione S-transferase (GST) activity of mice (21). However, the role that SN plays in cancer prevention remains unclear, and research regarding its molecular mechanism through a role in cell death induction is limited. If SN is cytotoxic or induces cell death, its chemopreventative properties would be more favorable.

The aim of this study was to determine whether SN exhibits cytotoxic effects and whether these effects involve autophagy induction in human liver cancer cells. To investigate the effects of SN on cell viability and autophagy, an MTT assay and monodansylcadaverine (MDC) staining of J5 cells were performed. To understand the mechanism by which SN regulates pre-autophagosome and autophagosome formation in autophagy induction, Beclin-1, PI3K-III, LC3-II, PI3K-I, Akt and mTOR expression were investigated using an immunoblot assay. In addition, the roles of p53 and NF-κB on SN-induced autophagy were also investigated. These experimental results will be helpful to determine the mechanism by which SN induces human liver tumor cell death and its potential effects on chemoprevention and/or cancer therapy.

Materials and methods

Chemicals and reagents

SN and MDC stain were purchased from Sigma Chemical Co. (St Louis, MO, USA). Antiserum against p53, DRAM, phosphorylated (p)-IκB, IκB or NF-κB (P65) were obtained from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Antiserum against TIGAR was obtained from Abcam, Inc. (Cambridge, MA, USA). Antiserum against PI3K-I, PI3K-III, LC3-II, Beclin-1 and mTOR were purchased from Cell Signaling Technology (Beverly, MA, USA).

Cell culture and SN treatment

The human hepatocellular carcinoma HepJ5 (J5) cell line was obtained from the Food Industry Research and Development Institute (Hsinchu, Taiwan). J5 cells (2.5×106) were plated on 60-mm plastic tissue culture dishes (Becton-Dickinson Labware, Franklin Lakes, NJ, USA) and incubated at 37°C (in humidified 5% CO2 and 95% air at 1 atm) in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum, 1% penicillin and streptomycin (100 U/ml), and 1% L-glutamine (0.33%, w/v).

After primary plating for 24 h, cells were treated with 100, 250 or 500 μM of SN for 24 h. All SN was diluted in dimethyl sulfoxide (DMSO). Cells treated with DMSO alone were regarded as the control group.

Cell viability and morphology examination

Cell viability was measured using an MTT (3-(4, 5-dimethyl-2-yl)-2, 5-diphenyl tetrazolium bromide) assay and morphological examination. The MTT assay was performed as described by Debizot and Lang (1986) (25). Briefly, J5 cells were incubated in 3-cm plates (1×106 cells) for 24 h and treated with 100, 250 or 500 μM of SN for 24 h. Then, MTT reagent (5 mg/ml) was added, and cell viability was determined by measuring the optical density (OD) at 570 nm. A phase-contrast microscope was used to examine morphological changes (Olympus IX 51, Olympus, Tokyo, Japan).

Monodansylcadaverine staining

To determine whether J5 cell death induced by SN was attributed to autophagy, the autophagic vacuole staining dye MDC was used as an autophagy indicator (26). Cells were grown on cover slips and treated with 100, 250 or 500 μM of SN for 24 h. After SN treatment, cells were incubated with 0.1 mM MDC for 15 min. Micrographs were acquired at ×400 magnification on an inverted fluorescence microscope (Olympus IX 51, Olympus). MDC stained cells were measured fluorometrically at an excitation wavelength of 360 nm and an emission wavelength of 530 nm.

Immunoblot analysis of autophagy-associated protein expression

To elucidate the signal transduction pathways by which autophagy was induced by SN in J5 cells, we analyzed the protein expression of autophagy regulators of pre-autophagosome and autophagosome formation. J5 cells were treated with 100, 250 or 500 μM of SN for 24 h. Total protein was collected and prepared from cells. Nuclear extracts were obtained from cell pellets using an NE-PER extraction kit (Thermo Scientific, Rockford, IL, USA) as per the manufacturer's instructions.

Cellular protein concentration of cells was assayed by the method of Lowry et al (27). Cytoplasmic PI3K-I, PI3K-III, Akt, mTOR, Beclin-1, LC3-II, p53, DRAM and TIGAR as well as nuclear p53 expression were determined using sodium dodecylsulfate polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblot assay (28,29). Bands were visualized using hydrogen peroxide/tetrahydrochloride diaminobenzidine or an enhanced chemiluminescent detection kit (Amersham Life Science, Buckinghamshire, UK) and were quantitated with an AlphaImager 2000 (Alpha Innotech, San Leandro, CA, USA).

Analysis of NF-κB activation and NF-κB-DNA binding activation

To determine whether autophagy-related molecules are regulated by the NF-κB pathway after SN treatment, J5 cells were treated with 100, 250 or 500 μM of SN for 24 h. Total protein was collected and prepared from cells. Cytosolic p-IκB, IκB, and NF-κB as well as nuclear NF-κB (p65) expression were determined using SDS-PAGE and immunoblot assay (28,29). Bands were visualized using hydrogen peroxide/ tetrahydrochloride diaminobenzidine or an enhanced chemiluminescent detection kit (Amersham Life Science) and were quantitated with an AlphaImager 2000 (Alpha Innotech).

For the NF-κB-DNA binding activity assay, nuclear extracts were obtained from cell pellets using an NE-PER extraction kit (Thermo Scientific) as per the manufacturer's instructions. NF-κB-DNA binding activity within the nuclear fraction was determined using an NF-κB (p65) transcription factor activity assay kit (Cayman Chemical Co., Ann Arbor, MI, USA) as per the manufacturer's instructions.

Statistical analysis

Statistical analyses were performed using SAS software (SAS Institute). Analysis of variance (ANOVA) and Duncan's multiple-range test were used to identify significant differences among the means (p<0.05).

Results

SN induces cytotoxicity in human liver tumor cells

According to the results of the MTT assay (Fig. 1), when J5 cells were treated with 100, 250 or 500 μM SN for 24 h, the cell viability measurements were 101±8, 96±7 and 71±7%, respectively. The cell viability of the 500 μM SN group was significantly reduced compared with the control group (100%) (p<0.05). From the microscopic observation results, no significant changes in the morphology of J5 cells treated with 100 or 250 μM SN were noted compared with the controls (data not shown). However, significant changes in J5 cell morphology were observed upon treatment with 500 μM SN after 24-h incubation compared with controls.

SN induces autophagy in human liver tumor cells

As shown in Fig. 2A, J5 cells were stained by MDC after various concentrations of SN treatment for 24 h, MDC-stained cell levels significantly increased (p<0.05), as measured fluorometrically. After J5 cells were treated with 100, 250 or 500 μM SN for 24 h, MDC fluorescence (35±5, 46±8 and 83±9%, respectively) also significantly increased in a dose-dependent manner compared with the control group (6±3%) (Fig. 2B). These results demonstrate that SN induces autophagy in J5 cells.

Effects of SN on protein levels of autophagy regulators

Fig. 3 indicates that in J5 cells treated with 250 or 500 μM SN for 24 h, PI3K-I protein levels (89±7 and 78±4%, respectively) significantly decreased following 24 h incubation compared with the control group (100%) (p<0.05). After J5 cell treatment with 250 or 500 μM SN for 24 h, the Akt protein levels were decreased (86±5 and 62±3%, respectively) compared with the control group (100%) (p<0.05). mTOR protein levels of the 100, 250 or 500 μM SN-incubated groups were significantly decreased (75±4, 65±3 and 53±4%, respectively) compared with the control group (100%) (p<0.05). These results demonstrate that SN suppresses the PI3K/Akt/mTOR pathways. Thus, this compound may play an important role in pre-autophagosome formation.

Conversely, Fig. 4 indicates that when J5 cells are treated with 500 μM SN for 24 h, Beclin-1 protein levels were increased (144±14%) compared with the control group (100%) (p<0.05). When J5 cells were treated with 250 or 500 μM SN, PI3K-III (124±12 and 164±18%, respectively) and LC3-II (167±13 and 185±15%, respectively) protein levels were also significantly increased compared with the control group (100%) (p<0.05). These results suggest that SN regulates the PI3K-I/Akt/mTOR and Beclin-1/PI3K-III/LC3-II pathways that induce autophagy in J5 cells.

Immunoblot analysis also demonstrated that nuclear p53 protein levels of the 500 μM SN group were significantly increased (183±17%) compared with the control group (100%) (p<0.05) (Fig. 5). DRAM protein levels were significantly increased (168±14%) after 500 μM SN treatment compared with the control group (100%) (p<0.05). However, cytoplasmic p53 significantly decreased (45±13%) after J5 cells were treated with 500 μM SN for 24 h compared with the control group (100%) (p<0.05). When J5 cells were treated with 500 μM SN, TIGAR protein levels (56±13%) were significantly reduced compared with the control group (100%) (p<0.05). These results indicate that SN affects autophagosome formation via p53 signaling.

SN induces NF-κB activation

To examine whether the suppression of Beclin-1, LC3-II and DRAM expression by SN is dependent on the inhibition of NF-κB activation, both immunoblot and NF-κB-DNA binding activity assays were performed. Fig. 6A and B demonstrate that p-IκB protein was significantly increased by 256 and 278% after 250 and 500 μM SN treatment, respectively (p<0.05). Nuclear p65 protein levels were also significantly decreased by 334 and 345% after 250 and 500 μM SN treatment, respectively (p<0.05). The changes in p65 protein levels in the nuclear fraction indicate that SN could significantly increase translocation of NF-κB from the cytoplasm to the nucleus in J5 cells. Furthermore, Fig. 6C demonstrates that NF-κB nuclear protein DNA-binding activity was significantly increased after 250 and 500 μM SN treatment (p<0.05). These results suggest that SN regulates NF-κB activation in J5 cells.

Discussion

SN is one of the major flavor compounds from celery seed oil (30). Our study demonstrates that SN suppresses human tumor cell viability through the induction of autophagy. This finding is novel in the anticancer research of SN. Autophagy plays an important role in tumor inhibition because autophagy defects are associated with metabolic stress, genomic damage, and tumorigenesis (3). By genetically or pharmacologically triggering autophagy, these autophagy inducers could act as potential therapeutic candidates to kill tumor cells (5,31). Various clinical anticancer agents, such as polyoxomolybdates, platonin, phenethyl isothiocyanate, tamoxifen, rapamycin, temozolomide, histone deacetylase inhibitors and camptothecin, involve the induction of autophagy to enhance chemotherapeutic efficacy (3236). In this study, SN significantly reduced human liver cell viability and induced autophagy. Therefore, SN may act as a potential cancer preventative or therapeutic.

SN is a phthalide-like compound. Phthalide structures contain a lactone core with a variety of complex chemical compound substituents typically found in herbs or vegetables, such as celery, Dong quai (Angelica sinensis) and Chuanxiong (Rhizoma Chuanxiong) (37,38). SN has a similar chemical structure to butylphthalide (3-n-butylphthalide) in celery seed oil, and both compounds are primarily responsible for the aroma and taste of celery (30). Previous studies demonstrate that Angelica sinensis phthalides, including n-butylidene-phthalide, senkyunolide A and z-ligustilide, significantly inhibit cell proliferation of human colon cancer HT-29 cells (39). Noscapine, a phthalide isoquinoline alkaloid derived from opium, inhibits cell proliferation by inducing apoptosis in colon cancer cells (40). Furthermore, 4-(3′,3′-dimethyl-allyloxy)-5-methyl-6-methoxyphthalide, which is isolated from Podocarpus macrophyllus, inhibits proliferation of HeLa tumor cell lines by inducing G1 cell cycle arrest and apoptosis (41). No study has investigated the effects of SN, a phthalide-like structure, on cancer cell death. Our results demonstrate that SN induces autophagy in human liver tumor cells, and this specific physiological effect may be attributed to its phthalide-like structure.

The PI3K pathway, a critical signal transduction system linking oncogenes and multiple receptor classes to many essential cellular functions, is perhaps the most commonly activated signaling pathway in human cancer (42). PI3K-I is highly correlated with Akt activation. Constitutive activation of the PI3K/ AKT signaling pathway often causes cells to proliferate in an uncontrolled manner (10). PI3K-III is an important regulator of autophagy, a cellular response to nutrient starvation (43). Among the signaling pathways that promote autophagy induction, the PI3K-I/Akt/mTOR and Beclin-1/PI3K-III/LC3-II signaling pathways are the major transduction pathways that regulate autophagy (44). These pathways negatively and positively regulate autophagy, respectively (45,46). In this study, SN significantly suppresses PI3K-I, Akt and mTOR protein levels in J5 cells, indicating that SN is a critical suppressor of PI3K-I/Akt/mTOR signaling that leads to autophagy. In contrast, when J5 cells were treated with SN, Beclin-1 and PI3K-III expression increased, thus triggering LC-3 II activation and inducing autophagy. Clinical anticancer drugs, such as polyoxomolybdates, platonin, phenethyl isothiocyanate and rapamycin, regulate PI3K, LC-3 or mTOR expression and lead to decreased cancer cell viability (3234,47). Previous studies demonstrate that caffeine suppresses HeLa cell growth and enhances autophagy by inhibiting the PI3K/Akt/mTOR signaling pathway (48). Wang et al also report that quercetin activates Akt-mTOR signaling in human gastric cancer cells, leading to increased LC3-II and Beclin-1 expression and subsequent induction of autophagy (49). Phytochemicals or chemicals that affect autophagy induction by suppressing PI3K-I/Akt/mTOR signaling and/or enhancing Beclin-1/ PI3K-III/LC3-II signaling show chemopreventative or chemotherapy potential.

p53 is a tumor suppressor that regulates numerous responses, such as cell cycle arrest, apoptosis, and senescence, each of which may lead to tumor inhibition (50). Recent studies indicate a new role for p53 in autophagy induction. Two p53-associated signaling pathways are involved in autophagy induction and based on p53 cellular localization. p53 promotes autophagy as a transcription factor in the nucleus by activating genes involved in apoptosis, cell cycle arrest and autophagy. However, autophagy is suppressed when cytosolic p53 expression is inhibited (15,51,52). In addition, p53 may induce autophagy by binding to the promoter region of multiple genes that code for pro-autophagic modulators, such as DRAM and TIGAR (53,54). Cytosolic p53 inhibits autophagy by inducing TIGAR, which indirectly affects reactive oxygen species (ROS) through modulation of the glycolytic pathway (53,55). To understand the effect of SN on p53 localization and its downstream regulation of autophagy, our study demonstrated that SN induces J5 cell autophagy by upregulating nuclear p53 and DARM and downregulating cytoplasmic p53 and TIGAR in J5 cells. Furthermore, SN not only regulates cytosolic and nuclear p53 levels but also modifies the expression of mTOR and DRAM in our study. Clearly, p53 plays an important role in SN-induced J5 cell autophagy. Previous studies demonstrate a similar biochemical regulation phenomenon wherein T-47D breast cancer cells treated with sulphathiazole, a common antibacterial drug, enhanced expression of p53/DRAM and downregulated the Akt/mTOR pathway, resulting in autophagy (56). Plumbagin (5-hydroxy-2-methyl-1, 4-naphthoquinone; PLB), a naturally occurring naphthoquinone isolated from the roots of Plumbaginaceae plants, induces autophagy in tongue squamous cell carcinoma (TSCC) through regulation of p53 and PI3K/Akt/mTOR-mediated pathways (57).

In this study, SN was significantly affected by transcript-specific autophagy genes, such as Beclin-1, LC3-II and DRAM, through upregulation of NF-κB activation, which results in increased Beclin-1, LC3-II and DRAM protein levels. Previous studies have demonstrated that the NF-κB pathway is activated in response to cellular starvation. When wild-type mouse embryonic fibroblasts are starved in medium, IκB phosphorylation and degradation is triggered. Then, NF-κB activation and increased LC3 expression is noted following starvation. These results also demonstrate that NF-κB-DNA binding activity and target gene expression were significantly increased under starvation conditions (19).

In conclusion, 250 and 500 μM SN induce autophagy in J5 cells, leading to human liver cancer cell death. The molecular mechanism involves regulation of PI3K, p53 and NF-κB expression during autophagy induction.

Acknowledgements

This study was supported by a grant (NSC101-2815-C-309-010-B) from the National Science Council, Taiwan, R.O.C..

Abbreviations:

SN

sedanolide

MDC

monodansylcadaverine

PI3K

phosphoinositide 3-kinase

mTOR

mammalian target of rapamycin

DRAM

damage-regulated autophagy modulator

TIGAR

Tp53-induced glycolysis and apoptosis regulator

IκB

inhibitor of kappa B

NF-κB

nuclear factor-kappa B

HCC

human hepatocellular carcinoma

References

1 

Ferenci P, Fried M, Labrecque D, Bruix J, Sherman M, Omata M, Heathcote J, Piratsivuth T, Kew M, Otegbayo JA, et al; World Gastroenterology Organisation Guidelines and Publications Committee. World Gastroenterology Organisation Guideline. Hepatocellular carcinoma (HCC): A global perspective. J Gastrointestin Liver Dis. 19:311–317. 2010.PubMed/NCBI

2 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Eskelinen EL, Mizushima N, Ohsumi Y, et al: Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest. 112:1809–1820. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Gozuacik D and Kimchi A: Autophagy as a cell death and tumor suppressor mechanism. Oncogene. 23:2891–2906. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Ding WX, Ni HM, Gao W, Chen X, Kang JH, Stolz DB, Liu J and Yin XM: Oncogenic transformation confers a selective susceptibility to the combined suppression of the proteasome and autophagy. Mol Cancer Ther. 8:2036–2045. 2009. View Article : Google Scholar : PubMed/NCBI

6 

White E and DiPaola RS: The double-edged sword of autophagy modulation in cancer. Clin Cancer Res. 15:5308–5316. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Mizushima N: The pleiotropic role of autophagy: From protein metabolism to bactericide. Cell Death Differ. 12(Suppl 2): 1535–1541. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Engelman JA, Luo J and Cantley LC: The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat Rev Genet. 7:606–619. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Bader AG, Kang S, Zhao L and Vogt PK: Oncogenic PI3K deregulates transcription and translation. Nat Rev Cancer. 5:921–929. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Vivanco I and Sawyers CL: The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat Rev Cancer. 2:489–501. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Klionsky DJ and Emr SD: Autophagy as a regulated pathway of cellular degradation. Science. 290:1717–1721. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Arico S, Petiot A, Bauvy C, Dubbelhuis PF, Meijer AJ, Codogno P and Ogier-Denis E: The tumor suppressor PTEN positively regulates macroautophagy by inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway. J Biol Chem. 276:35243–35246. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Brazil DP and Hemmings BA: Ten years of protein kinase B signalling: A hard Akt to follow. Trends Biochem Sci. 26:657–664. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Kondapaka SB, Singh SS, Dasmahapatra GP, Sausville EA and Roy KK: Perifosine, a novel alkylphospholipid, inhibits protein kinase B activation. Mol Cancer Ther. 2:1093–1103. 2003.PubMed/NCBI

15 

Sui X, Jin L, Huang X, Geng S, He C and Hu X: p53 signaling and autophagy in cancer: A revolutionary strategy could be developed for cancer treatment. Autophagy. 7:565–571. 2011. View Article : Google Scholar

16 

Feng Z, Zhang H, Levine AJ and Jin S: The coordinate regulation of the p53 and mTOR pathways in cells. Proc Natl Acad Sci USA. 102:8204–8209. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Trocoli A and Djavaheri-Mergny M: The complex interplay between autophagy and NF-κB signaling pathways in cancer cells. Am J Cancer Res. 1:629–649. 2011.

18 

Criollo A, Chereau F, Malik SA, Niso-Santano M, Mariño G, Galluzzi L, Maiuri MC, Baud V and Kroemer G: Autophagy is required for the activation of NFκB. Cell Cycle. 11:194–199. 2012. View Article : Google Scholar

19 

Comb WC, Cogswell P, Sitcheran R and Baldwin AS: IKK-dependent, NF-κB-independent control of autophagic gene expression. Oncogene. 30:1727–1732. 2011. View Article : Google Scholar

20 

Kang R, Zeh HJ, Lotze MT and Tang D: The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ. 18:571–580. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Zheng GQ, Kenney PM, Zhang J and Lam LK: Chemoprevention of benzo[a]pyrene-induced forestomach cancer in mice by natural phthalides from celery seed oil. Nutr Cancer. 19:77–86. 1993. View Article : Google Scholar

22 

Woods JA, Jewell C and O'Brien NM: Sedanolide, a natural phthalide from celery seed oil: Effect on hydrogen peroxide and tert-butyl hydroperoxide-induced toxicity in HepG2 and CaCo-2 human cell lines. In Vitr Mol Toxicol. 14:233–240. 2001. View Article : Google Scholar

23 

Sowbhagya HB: Chemistry, technology, and nutraceutical functions of celery (Apium graveolens L.): An overview. Crit Rev Food Sci Nutr. 54:389–398. 2014. View Article : Google Scholar

24 

Momin RA and Nair MG: Antioxidant, cyclooxygenase and topoisomerase inhibitory compounds from Apium graveolens Linn. seeds. Phytomedicine. 9:312–318. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Denizot F and Lang R: Rapid colorimetric assay for cell growth and survival. Modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. J Immunol Methods. 89:271–277. 1986. View Article : Google Scholar : PubMed/NCBI

26 

Itoh T, Ohguchi K, Nozawa Y and Akao Y: Intracellular glutathione regulates sesquiterpene lactone-induced conversion of autophagy to apoptosis in human leukemia HL60 cells. Anticancer Res. 29:1449–1457. 2009.PubMed/NCBI

27 

Lowry OH, Rosebrough NJ, Farr AL and Randall RJ: Protein measurement with the Folin phenol reagent. J Biol Chem. 193:265–275. 1951.PubMed/NCBI

28 

Laemmli UK: Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature. 227:680–685. 1970. View Article : Google Scholar : PubMed/NCBI

29 

Towbin H, Staehelin T and Gordon J: Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: Procedure and some applications. Proc Natl Acad Sci USA. 76:4350–4354. 1979. View Article : Google Scholar : PubMed/NCBI

30 

Gold HJ and Wilson CW III: The volatile flavor substances of celery. J Food Sci. 28:484–488. 1963. View Article : Google Scholar

31 

Amaravadi RK, Lippincott-Schwartz J, Yin XM, Weiss WA, Takebe N, Timmer W, DiPaola RS, Lotze MT and White E: Principles and current strategies for targeting autophagy for cancer treatment. Clin Cancer Res. 17:654–666. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Ogata A, Yanagie H, Ishikawa E, Morishita Y, Mitsui S, Yamashita A, Hasumi K, Takamoto S, Yamase T and Eriguchi M: Antitumour effect of polyoxomolybdates: Induction of apoptotic cell death and autophagy in in vitro and in vivo models. Br J Cancer. 98:399–409. 2008. View Article : Google Scholar

33 

Bommareddy A, Hahm ER, Xiao D, Powolny AA, Fisher AL, Jiang Y and Singh SV: Atg5 regulates phenethyl isothiocyanate-induced autophagic and apoptotic cell death in human prostate cancer cells. Cancer Res. 69:3704–3712. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Chen YJ, Huang WP, Yang YC, Lin CP, Chen SH, Hsu ML, Tseng YJ, Shieh HR, Chen YY and Lee JJ: Platonin induces autophagy-associated cell death in human leukemia cells. Autophagy. 5:173–183. 2009. View Article : Google Scholar

35 

Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, Thomas-Tikhonenko A and Thompson CB: Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest. 117:326–336. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Fleming A, Noda T, Yoshimori T and Rubinsztein DC: Chemical modulators of autophagy as biological probes and potential therapeutics. Nat Chem Biol. 7:9–17. 2011. View Article : Google Scholar

37 

Uhlig JW, Chang A and Jen JJ: Effect of phthalides on celery flavor. J Food Sci. 52:658–660. 1987. View Article : Google Scholar

38 

Zhang X, Xiao H, Xu Q, Li X, Wang J and Liang X: Characterization of phthalides in Ligusticum chuanxiong by liquid chromatographic-atmospheric pressure chemical ionization-mass spectrometry. J Chromatogr Sci. 41:428–433. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Kan WL, Cho CH, Rudd JA and Lin G: Study of the anti-proliferative effects and synergy of phthalides from Angelica sinensis on colon cancer cells. J Ethnopharmacol. 120:36–43. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Yang ZR, Liu M, Peng XL, Lei XF, Zhang JX and Dong WG: Noscapine induces mitochondria-mediated apoptosis in human colon cancer cells in vivo and in vitro. Biochem Biophys Res Commun. 421:627–633. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Chen C and Yang RL: A phthalide derivative isolated from endophytic fungi Pestalotiopsis photiniae induces G1 cell cycle arrest and apoptosis in human HeLa cells. Braz J Med Biol Res. 46:643–649. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Liu P, Cheng H, Roberts TM and Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 8:627–644. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Backer JM: The regulation and function of Class III PI3Ks: Novel roles for Vps34. Biochem J. 410:1–17. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Meijer AJ and Codogno P: Regulation and role of autophagy in mammalian cells. Int J Biochem Cell Biol. 36:2445–2462. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Shinojima N, Yokoyama T, Kondo Y and Kondo S: Roles of the Akt/mTOR/p70S6K and ERK1/2 signaling pathways in curcumin-induced autophagy. Autophagy. 3:635–637. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Pyo JO, Nah J and Jung YK: Molecules and their functions in autophagy. Exp Mol Med. 44:73–80. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Georgakis GV and Younes A: From Rapa Nui to rapamycin: Targeting PI3K/Akt/mTOR for cancer therapy. Expert Rev Anticancer Ther. 6:131–140. 2006. View Article : Google Scholar

48 

Saiki S, Sasazawa Y, Imamichi Y, Kawajiri S, Fujimaki T, Tanida I, Kobayashi H, Sato F, Sato S, Ishikawa K, et al: Caffeine induces apoptosis by enhancement of autophagy via PI3K/Akt/ mTOR/p70S6K inhibition. Autophagy. 7:176–187. 2011. View Article : Google Scholar :

49 

Wang K, Liu R, Li J, Mao J, Lei Y, Wu J, Zeng J, Zhang T, Wu H, Chen L, et al: Quercetin induces protective autophagy in gastric cancer cells: Involvement of Akt-mTOR- and hypoxia-induced factor 1α-mediated signaling. Autophagy. 7:966–978. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Murray-Zmijewski F, Slee EA and Lu X: A complex barcode underlies the heterogeneous response of p53 to stress. Nat Rev Mol Cell Biol. 9:702–712. 2008. View Article : Google Scholar : PubMed/NCBI

51 

Zilfou JT and Lowe SW: Tumor suppressive functions of p53. Cold Spring Harb Perspect Biol. 1:a0018832009. View Article : Google Scholar :

52 

Maiuri MC, Galluzzi L, Morselli E, Kepp O, Malik SA and Kroemer G: Autophagy regulation by p53. Curr Opin Cell Biol. 22:181–185. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, Gottlieb E and Vousden KH: TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell. 126:107–120. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Crighton D, Wilkinson S, O'Prey J, Syed N, Smith P, Harrison PR, Gasco M, Garrone O, Crook T and Ryan KM: DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell. 126:121–134. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Bensaad K, Cheung EC and Vousden KH: Modulation of intracellular ROS levels by TIGAR controls autophagy. EMBO J. 28:3015–3026. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Mohammadpour R, Safarian S, Sheibani N, Norouzi S and Razazan A: Death inducing and cytoprotective autophagy in T-47D cells by two common antibacterial drugs: Sulphathiazole and sulphacetamide. Cell Biol Int. 37:348–358. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Pan ST, Qin Y, Zhou ZW, He ZX, Zhang X, Yang T, Yang YX, Wang D, Qiu JX and Zhou SF: Plumbagin induces G2/M arrest, apoptosis, and autophagy via p38 MAPK- and PI3K/Akt/mTOR-mediated pathways in human tongue squamous cell carcinoma cells. Drug Des Devel Ther. 9:1601–1626. 2015.PubMed/NCBI

Related Articles

Journal Cover

December-2015
Volume 47 Issue 6

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
Hsieh S, Chen C, Wang J, Kuo Y, Li C, Hsieh L and Wu C: Sedanolide induces autophagy through the PI3K, p53 and NF-κB signaling pathways in human liver cancer cells. Int J Oncol 47: 2240-2246, 2015
APA
Hsieh, S., Chen, C., Wang, J., Kuo, Y., Li, C., Hsieh, L., & Wu, C. (2015). Sedanolide induces autophagy through the PI3K, p53 and NF-κB signaling pathways in human liver cancer cells. International Journal of Oncology, 47, 2240-2246. https://doi.org/10.3892/ijo.2015.3206
MLA
Hsieh, S., Chen, C., Wang, J., Kuo, Y., Li, C., Hsieh, L., Wu, C."Sedanolide induces autophagy through the PI3K, p53 and NF-κB signaling pathways in human liver cancer cells". International Journal of Oncology 47.6 (2015): 2240-2246.
Chicago
Hsieh, S., Chen, C., Wang, J., Kuo, Y., Li, C., Hsieh, L., Wu, C."Sedanolide induces autophagy through the PI3K, p53 and NF-κB signaling pathways in human liver cancer cells". International Journal of Oncology 47, no. 6 (2015): 2240-2246. https://doi.org/10.3892/ijo.2015.3206