Pomolic acid induces apoptosis in SK-OV-3 human ovarian adenocarcinoma cells through the mitochondrial-mediated intrinsic and death receptor-induced extrinsic pathways

  • Authors:
    • Ki Hyun Yoo
    • Jong-Hwa Park
    • Do Kyung Lee
    • Yuan Yuan Fu
    • Nam In Baek
    • In Sik Chung
  • View Affiliations

  • Published online on: October 22, 2012     https://doi.org/10.3892/ol.2012.985
  • Pages: 386-390
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The cytotoxic effect of pomolic acid (PA), a pentacyclic triterpene isolated from flowers of Osmanthus fragrans var. aurantiacus Makino, was investigated in SK-OV-3 human ovarian adenocarcinoma cells. PA dose-dependently inhibited the viability of SK-OV-3 cells. PA-induced apoptosis was further characterized by detection of cell surface annexin V and sub-G1 apoptotic cell populations. The number of cells immunostained with annexin V-fluorescein isothiocyanate (FITC) increased following treatment with PA. The sub-G1 cell populations also increased in PA-treated SK-OV-3 cells. PA induced the activation of caspase-8, -9 and -3, critical mediators of apoptosis signaling. PA decreased the mitochondrial transmembrane potential (∆Ψm), resulting in the activation of caspase-9. In addition, PA increased the expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis signaling-related death receptor 5 (DR5), mediating caspase-8-involved extrinsic pathway. Taken together, our results indicate that PA induces apoptosis in SK-OV-3 cells, which is mediated by the mitochondrial-mediated intrinsic and death receptor-induced extrinsic pathways.

Introduction

Terpenes are widespread natural compounds involved in various biological activities, playing a significant role in human medicine. They include triterpenes, saponines and other triterpenoids, representing one of numerous classes of natural compounds. The main groups of triterpenes and saponines are represented by tetracyclic derivatives of protostane, cycloartane, dammarane, euphane and pentacyclic derivatives of ursane, gammacerane, lupane and hopane (1). Pentacyclic triterpenes are secondary metabolites that are based on a 30-carbon skeleton and synthesized in many plants by the cyclization of squalene (2). These are widely distributed in fruit peel, leaves and stem bark (3). It has been demonstrated that several triterpenes have the ability to induce cancer cell apoptosis and prevent oxidative stress, inflammation and hypertension (4,5).

Pomolic acid (PA) is a pentacyclic triterpene isolated from the flowers of Osmanthus fragrans var. aurantiacus Makino that are found in China, Japan and the southern portion of Korea. The flowers are used by the Chinese to give an aroma to tea or wine and in cosmetics for hair and skin (6,7). Also, dried flowers have neuroprotective, free radical scavenging and anti-oxidative effects (8). PA inhibited the growth of leukemia cell line HL-60 and induced mitochondria-dependent apoptotic cell death (9). PA has been shown to induce apoptosis by activating the caspase cascade through loss of the mitochondrial transmembrane potential (ΔΨm), independently of anti-apoptotic Bcl-2 expression in leukemia cells (9,10). PA is suggested to overcome multidrug resistance mediated by overexpression of anti-apoptotic Bcl-2 proteins (10,11). PA also showed anti-proliferative activities against human gastric adenocarcinoma (MK-1), human uterine carcinoma (HeLa) and murine melanoma (B16F10) cells (12). However, the apoptotic mechanisms of PA in human carcinoma cells were not investigated in detail. In our preliminary experiment on cytotoxic effects, PA reduced the viabilities of human ovarian adenocarcinoma SK-OV-3, human colon carcinoma HCT-116, human breast adenocarcinoma MCF-7, SK-BR-3 and human melanoma SK-MEL-5 cells. The greatest inhibition of PA was observed in SK-OV-3 cells (data not shown). To the best of our knowledge, the apoptotic effect of PA on SK-OV-3 cells has not yet been examined. Thus, in this study, we report the PA-induced apoptosis and its molecular mechanism in human ovarian adenocarcinoma SK-OV-3 cells.

Materials and methods

Ethical considerations

The study was approved by the Kyung Hee University Institutional Animal Care and Use Committee (Yongin, Korea).

Extraction and isolation of pomolic acid

The dried and powdered flowers (800 g) of Osmanthus fragrans var. aurantiacus Makino, were extracted with 80% aqueous methanol (MeOH) (3 liters x3). Extracts were partitioned with ethyl acetate (EtOAc; 3 liters x3) and H2O (3 liters). The soluble fraction of EtOAc extract (30 g) was subjected to SiO2 column (8x15 cm) chromatography (c.c.), eluted with n-hexane-EtOAc (5:1 → 1:1, 5 liters of each) and monitored using thin-layer chromatography (TLC) to produce 30 fractions (OSE-1 to OSE-30). Non-soluble precipitate (OSE-P, 2.69 g) of EtOAc extracts was applied to the octadecylsilica gel (ODS) c.c. and eluted with MeOH-H2O (4:1, 18 liters) to produce 8 fractions (OSE-P-1 to OSE-P-8). OSE-P-4 [Ve/Vt (elution volume/total volume) 0.27–0.38, 224 mg] was subjected to SiO2 c.c. and eluted with CHCl3-MeOH (25:1, 2.1 liters) to give 14 fractions (OSE-P-4-1 to OSE-P-4-14). The subfraction OSE-P-4-4 yielded 20 mg PA (Ve/Vt 0.21–0.28, ODS TLC Rf 0.21, MeOH-H2O=13:1).

Cell culture

Human ovarian adenocarcinoma SK-OV-3 cells were obtained from the Korea Cell Line Bank (KCLB, Seoul, Korea). Cells were grown at 37°C with 5% CO2 in RPMI-1640 medium with 10% (v/v) fetal bovine serum (FBS) and 1% (v/v) penicillin-streptomycin. All cell culture media and reagents were purchased from Thermo Scientific Hyclone (Waltham, MA, USA).

Cytotoxic assay

The cytotoxicity of PA was measured using an MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, Sigma, St. Louis, MO, USA] colorimetric assay. Cells were seeded onto 96-well plates at a density of 1x104 cells/well in 100 μl RPMI-1640 supplemented with 10% (v/v) FBS. Following 24 h incubation at 37°, the cells were treated with serum-free RPMI-1640, containing various concentrations of PA. Following a further 24h incubation, 50 μl MTT [5 mg/ml in phosphate-buffered saline, (PBS)] was added to each well. The cells were incubated at 37°C for 2 h. Following removal of the medium, the cells were treated with 100 μl dimethyl sulfoxide (DMSO) for 5 min and then the optical density was measured using a microplate reader (Bio-Tek, Winooski, VT, USA) at 550 nm. Cell viability was calculated as a percentage of viable cells in the PA-treated group (5, 15, 25 and 50 μM) vs. a control group using the following equation: Cell viability (%) = [(ODCompound - ODBlank)/(ODContol - ODBlank)] × 100.

Annexin V assay

Modulation of phosphatidylserine externalization during apoptosis was identified using annexin V conjugated with the fluorescent dye FITC. SK-OV-3 cells were seeded onto 6-well plates at a density of 3x105 cells/well. Following treatment with 25 μM PA for 6 h, the cells were stained with annexin V-FITC conjugate and then imaged under x40 objective magnification using a confocal laser scanning microscope (LSM 510 Meta, Carl Zeiss, Oberkochen, Germany).

Cell cycle analysis

SK-OV-3 cells were seeded onto 6-well plates at a density of 3x105 cells/well. Following dose-dependent treatment with PA for various times up to 12 h, cells were collected and washed twice with ice-cold PBS. Cell pellets were fixed in 70% (v/v) cold ethanol overnight at −20°C. Fixed cells were centrifuged, washed and resuspended in 100 μl PBS, then mixed with 100 μl RNase A (1 mg/ml, Sigma) and incubated for 30 min at 37°. The cells were stained by adding 400 μl propidium iodide (PI; 50 μg/ml, Sigma). After filtering through a nylon mesh (40 μm), the DNA content of stained cells was analyzed using the FACSVantage SE and CellQuest program (BD Biosciences, San Jose, CA, USA).

Mitochondrial membrane potential (ΔΨm) assay

SK-OV-3 cells treated with 25 μM PA for 3 or 6 h were labeled for 30 min with 0.1 μM tetramethylrhodamine ethyl ester (TMRE; Sigma), a cationic, lipophilic dye that accumulates in the negatively charged mitochondrial matrix and collected by trypsinization. The fluorescence intensity was monitored at 582 nm (FL2 channel) by FACSVantage SE (BD Biosciences).

Real-time polymerase chain reaction (PCR) analysis

Total RNA was isolated using an RNeasy Mini kit (Qiagen, Hilden, Germany) according to the manufacturer’s protocol. DNase I (Takara Bio Inc., Shiga, Japan)-treated total RNA was transcribed into cDNA using an ImProm-II Reverse Transcription System (Promega, Madison, WI, USA). Quantitative real-time PCR was performed using the ABI prism 7000 sequence detection system (Applied Biosystems, Foster City, CA, USA). The reaction was carried out in a total volume of 20 μl containing 10 μl 2X SYBR Premix EX Taq™ (Takara Bio Inc.), 1 μl each oligonucleotide primer (10 μM) and 1 μl DNA. The sequences of the sense and antisense primers for death receptor (DR) 5, DR4 and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) were as follows: DR5 sense: 5′-TGC ACC ACG ACC AGA AAC AC-3′; DR5 antisense: 5′-ATC ACC GAC CTT GAC CAT CC-3′; DR4 sense: 5′-AGG GTC TCA GAG GAG GAG GC-3′; DR4 antisense: 5′-GGA GTC AAA GGG CAC GAT GT-3′; GAPDH sense: 5′-AGG AGG CAT TGC TGA TGA TC-3′; GAPDH antisense: 5′-AGT GAG GGT CTC TCT CTT CC-3′. Following completion of PCR, the data were analyzed using ABI prism 7000 sequence detection system software. The expression of mRNA was normalized with GAPDH.

Western blot analysis

After seeding onto 6-well plates at a density of 3x105 cells/well, cells were treated with 25 μM PA for various time periods up to 12 h, then lysed with RIPA buffer (Thermo Fisher Scientific Inc., Rockford, IL, USA) supplemented with a protease inhibitor cocktail (Roche, Basel, Switzerland). Protein concentrations were determined using an RC/DC Bio-Rad assay kit (Bio-Rad, Hercules, CA, USA) following the manufacturer’s instructions. Protein samples were separated using electrophoresis on 10–15% polyacrylamide-SDS gel. The electrophoresized proteins on the gel were transferred onto a polyvinylidene fluoride (PVDF) membrane (Pall Life Sciences, Port Washington, NY, USA), blocked with 5% (w/v) skimmed milk (BD Biosciences), incubated with anti-human cleaved caspase-3 and -8, anti-human caspase-9 (Cell Signaling Technology Inc., Danvers, MA, USA), anti-human DR4 and DR5 (Imgenex, San Diego, CA, USA), anti-human PARP and anti-human α-tubulin (Santa Cruz Biotechnology, Santa Cruz, CA, USA), then probed with horseradish peroxidase-conjugated anti-rabbit or mouse IgG antibody (GE Healthcare Life Sciences, Stockholm, Sweden). The membrane was detected using an enhanced chemiluminescent western blotting detection system (GE Healthcare Life Sciences).

Statistical analysis

All data are presented as mean ± standard deviation (SD). Student’s t-test was used to compare different data groups. P<0.05, P<0.01 and P<0.001 were considered to indicate statistically significant differences.

Results

PA demonstrates dose-dependent cytotoxic effects

PA was isolated from flowers of Osmanthus fragrans var. aurantiacus Makino (Fig. 1A). Identification of the structure was confirmed on the basis of several spectroscopic analyses, including infrared (IR), 1H- and 13C-nuclear magnetic resonance (NMR) and 2D-NMR (correlation spectroscopy, COSY; heteronuclear single quantum coherence, HSQC and heteronuclear multiple-bond correlation spectroscopy, HMBC) (data not shown). To evaluate the cytotoxic effect of PA on SK-OV-3 cells, cells were treated with different concentrations (5, 15, 25 and 50 μM) of PA for 24 h and cell viabilities were measured using an MTT assay. PA dose-dependently inhibited the viability of SK-OV-3 cells (Fig. 1B). When 50 μM PA was treated for 24 h, the viability of SK-OV-3 cells decreased to 10.3% of that for untreated control cells. IC50 (50% inhibitory concentration) value occurred between 15 and 25 μM.

PA induces apoptosis in SK-OV-3 cells

To determine whether the cytotoxic effect of PA was caused by apoptosis, PA-treated SK-OV-3 cells were immunostained with annexin V-FITC conjugate and monitored under a confocal microscope (Fig. 2A). Annexin V-FITC-stained cells were observed in PA-treated cells, but not in an untreated control group. Cell cycle analysis was performed to determine the sub-G1 apoptotic population of PA-treated SK-OV-3 cells. Cells were treated with different concentrations (5, 15, 25 and 50 μM) of PA for 6 or 12 h and their DNA contents were analyzed using flow cytometry following propidium iodide (PI) staining. As shown in Fig. 2B and C, the number of cells in the sub-G1 population increased in a time- and dose-dependent manner. Following 6 h incubation with 5, 15, 25 and 50 μM PA, the sub-G1 populations increased to 0.6, 1.8, 9.8 and 19.8%, respectively. The sub-G1 populations of cells treated with 5, 15, 25 and 50 μM PA for 12 h increased to 1.01, 7.41, 13.01 and 33.93%, respectively. Taken together, these results indicate that PA induces apoptosis in SK-OV-3 cells.

PA induces the activation of the caspase cascade

The activation of the caspase cascade was determined in SK-OV-3 cells treated with 25 μM PA for indicated times (3, 6, 9 and 12 h). Cleaved caspase-8 (41 and 43 kDa) and caspase-3 (17 and 19 kDa) were increased by the treatment of PA (Fig. 3A). PA induced the reduction of procaspase-9 (49 kDa) and increased the cleaved caspase-9 (35 and 37 kDa). PA also induced the cleavage of poly ADP-ribose polymerase (PARP). These results indicate that PA induces apoptosis in SK-OV-3 cells via the activation of the caspase cascade.

PA reduces the mitochondrial transmembrane potential (ΔΨm) and increased the expression of DR5 in SK-OV-3 cells

To determine the effect of PA on mitochondrial transmembrane potential (ΔΨm), SK-OV-3 cells were treated with 25 μM PA for 3 or 6 h and labeled with TMRE. The fluorescence intensity was measured using flow cytometry. The number of TMRE-stained cells showing fluorescence intensities up to 10 were 80.7 and 77.5% following 3 and 6 h incubation of 25 μM PA, respectively (Fig. 3B). This means that the treatment of 25 μM PA for 3 and 6 h reduced the ΔΨm by 15 and 19.5%, respectively, compared to PA-nontreated cells. This indicates that PA induces the loss of mitochondrial transmembrane potential, resulting in the activation of caspase-9.

To investigate the effect of PA on death receptor-induced extrinsic apoptosis pathway, the expression levels of DR4 and DR5 were analyzed using real-time PCR and western blot analysis. As shown in Fig. 3C, DR5 transcript was time-dependently increased in PA-treated SK-OV-3 cells, resulting in 1.3, 1.7, 2.3 and 2.5-fold increases following 3, 6, 9 and 12 h incubation with 25 μM PA, respectively. The protein levels of DR5, based on densitometry (Fig. 3D), increased 3.5, 5.1, 6 and 6.1-fold, respectively, following 3, 6, 9 and 12 h incubation with 25 μM PA. However, DR4 transcript and protein levels decreased in PA-treated SK-OV-3 cells. This indicates that PA increases expression of DR5, resulting in the activation of caspase-8, a caspase cascade mediator of the death receptor-induced extrinsic apoptosis pathway.

Discussion

PA inhibited the growth of leukemia HL-60 cells (9) and showed anti-proliferative activities in several human carcinoma cells including human gastric adenocarcinoma (MK-1), human uterine carcinoma (HeLa) and murine melanoma (B16F10) cells (12). Although mitochondria-dependent apoptotic cell death was evaluated in leukemia HL-60 cells, PA-induced apoptotic mechanisms of human carcinoma cells were not investigated in detail. In a preliminary experiment, we determined the cytotoxic effect of PA on the viabilities of human carcinoma cells. The greatest inhibition of PA was observed in SK-OV-3 cells (data not shown). We chose SK-OV-3 cells to investigate PA-induced apoptotic mechanisms of human carcinoma cells. In this study, we examined the effect of PA on the viability of human ovarian adenocarcinoma SK-OV-3 cells and investigated the mechanisms involved in PA-induced apoptosis. PA dose-dependently inhibited the viability of SK-OV-3 cells (Fig. 1B) and induced apoptosis, which was characterized by detection of cell surface annexin V and sub-G1 apoptotic cell populations (Fig. 2).

The caspase-3 activation cascade played a central role in apoptotic mechanisms (13) as determined in PA-treated SK-OV-3 cells. Cleaved caspase-8, -9 and -3 levels increased following treatment with PA (Fig. 3A). Our findings indicate that PA activates two major apoptotic pathways, intrinsic and extrinsic, as is the case for other reported triterpenoids, to induce apoptosis through mitochondria-mediated intrinsic and death receptor-induced extrinsic pathways (14,15). PA has been reported to induce apoptosis by activation of caspase-9 and -3 through the loss of mitochondrial transmembrane potential (ΔΨm) in leukemia cells (9). In SK-OV-3 cells, PA was also found to reduce the mitochondrial transmembrane potential (ΔΨm) (Fig. 3B). This implies that PA induces apoptosis in SK-OV-3 cells via a mitochondria-mediated intrinsic pathway.

In addition, PA induced activation of caspase-8, a caspase cascade mediator of the death receptor-induced extrinsic pathway. This indicates that PA induces apoptosis through the death receptor-induced extrinsic pathway. Recently, triterpenoids such as celastrol and ursolic acid have been suggested to potentiate tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through upregulation of DR4 and/or DR5 (16,17). In our previous study, 3-O-acetyloleanolic acid was also shown to induce apoptosis in HCT-116 cells, which is mediated by upregulation of DR5 (18). TRAIL is a TNF family member showing anti-tumor activity in various cancer cell types as well as minimal cytotoxicity to normal cells and tissues (19). The binding between TRAIL and the two death receptors DR4 (TRAILR1/APO-2) and DR5 (TRAILR2/KILLER) leads to oligomerization of the death receptors and activation of the death receptor-induced extrinsic pathway (20). DR5 and DR4 signaling induces apoptosis via caspase-8-mediated activation of the caspase cascade (21,22). PA increased the expression of DR5, increasing induction of the caspase-8-mediated extrinsic pathway. To the best of our knowledge, this is the first report showing that PA induces apoptosis in SK-OV-3 cells as well as upregulation of TRAIL signaling-related DR5 in PA-mediated apoptosis.

In conclusion, our results demonstrated for the first time PA-induced apoptotic mechanisms in human ovarian adenocarcinoma SK-OV-3 cells. Our findings suggest that PA increases expression of TRAIL signaling-related death receptor DR5 and induces apoptosis in SK-OV-3 cells via the mitochondria-mediated intrinsic and the death receptor-induced extrinsic pathway.

Acknowledgements

This study was supported by grants from the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (2012001575) and from Kyung Hee University in 2010 (KHU-20110257).

References

1 

Patocka J: Biologically active pentacyclic triterpenes and their current medicine signification. J Appl Biomed. 1:7–12. 2003.

2 

Phillips DR, Rasbery JM, Bartel B and Matsuda SP: Biosynthetic diversity in plant triterpene cyclization. Curr Opin Plant Biol. 9:305–314. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Jager S, Trojan H, Kopp T, Laszczyk MN and Scheffler A: Pentacyclic triterpene distribution in various plants - rich sources for a new group of multi-potent plant extracts. Molecules. 14:2016–2031. 2009.PubMed/NCBI

4 

Baek JH, Lee YS, Kang CM, Kim JA, Kwon KS, Son HC and Kim KW: Intracellular Ca2+ release mediates ursolic acid-induced apoptosis in human leukemic HL-60 cells. Int J Cancer. 73:725–728. 1997.

5 

Yamaguchi Y, Yamada K, Yoshikawa N, Nakamura K, Haginaka J and Kunitomo M: Corosolic acid prevents oxidative stress, inflammation and hypertension in SHR/NDmcr-cp rats, a model of metabolic syndrome. Life Sci. 79:2474–2479. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Duke JA and Ayensu ES: Medicinal Plants of China. 2. Reference Publications Inc; Algonac, MI, USA: pp. 3811985

7 

Hedrick UP: Sturtevant’s Edible Plants of the World. Dover Publications; Mineola, NY, USA: pp. 6861972

8 

Lee HH, Lin CT and Yang LL: Neuroprotection and free radical scavenging effects of Osmanthus fragrans. J Biomed Sci. 14:819–827. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Fernandes J, Weinlich R, Castilho RO, Kaplan MAC, Amarante-Mendes GP and Gattass CR: Pomolic acid triggers mitochondria dependent apoptotic cell death in leukemia cell line. Cancer Lett. 219:49–55. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Fernandes J, Weinlich R, Castilho RO, Amarante-Mendes P and Gattass CR: Pomolic acid may overcome multidrug resistance mediated by overexpression of anti-apoptotic Bcl-2 proteins. Cancer Lett. 245:315–320. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Vasconcelos FC, Gattass RR, Rumjanek VM and Maia RC: Pomolic acid-induced apoptosis in cells from patients with chronic myeloid leukemia exhibiting different drug resistance profile. Invest New Drugs. 25:525–533. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Yoshida M, Fuchigami M, Nagao TT, et al: Antiproliferative constituents from umbelliferase plants VII. Active triterpenes and rosmarinic acid from Centella asiatica. Biol Pharm Bull. 28:173–175. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Zheng TS, Schlosser SF, Dao T, Hingorani R, Crispe IN, Boyer JL and Flavell RA: Caspase-3 controls both cytoplasmic and nuclear events associated with Fas-mediated apoptosis in vivo. Proc Natl Acad Sci USA. 95:13618–13623. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Hengartner MO: The biochemistry of apoptosis. Nature. 407:770–776. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Lee B, Lee DY, Yoo KH, Baek NI, Park JH and Chung IS: Calenduloside E 6’-methyl ester induces apoptosis in CT-26 mouse colon carcinoma cells and inhibits tumor growth in a CT-26 xenograft animal model. Oncol Lett. 4:22–28. 2012.

16 

Sung B, Park B, Yadav VR and Aggarwal BB: Celastrol, a triterpene, enhances TRAIL-induced apoptosis through the down-regulation of cell survival proteins and up-regulation of death receptors. J Biol Chem. 285:11498–11507. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Prasad S, Yadav VR, Kannappan R and Aggarwal BB: Ursolic acid, a pentacyclin triterpene, potentiates TRAIL-induced apoptosis through p53-independnet up-regulation of death receptors. J Biol Chem. 286:5546–5557. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Yoo KH, Park JH, Cui EJ, et al: 3-O-Acetyloleanolic acid induces apoptosis in human colon carcinoma HCT-116 cells. Phytother Res. View Article : Google Scholar : 2012.PubMed/NCBI

19 

Pitti RM, Marsters SA, Ruppert S, Donahue CJ, Moore A and Ashkenazi A: Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family. J Biol Chem. 271:12687–12690. 1996. View Article : Google Scholar : PubMed/NCBI

20 

Zhang L and Fang B: Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Therapy. 12:228–237. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Ashkenazi A and Dixit VM: Death receptors: Signaling and modulation. Science. 281:1305–1308. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Wajant H, Gerspach J and Pfizenmaier K: Tumor therapeutics by design: targeting and activation of death receptors. Cytokine Growth Factor Rev. 16:55–76. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January 2013
Volume 5 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yoo KH, Park J, Lee DK, Fu YY, Baek NI and Chung IS: Pomolic acid induces apoptosis in SK-OV-3 human ovarian adenocarcinoma cells through the mitochondrial-mediated intrinsic and death receptor-induced extrinsic pathways. Oncol Lett 5: 386-390, 2013
APA
Yoo, K.H., Park, J., Lee, D.K., Fu, Y.Y., Baek, N.I., & Chung, I.S. (2013). Pomolic acid induces apoptosis in SK-OV-3 human ovarian adenocarcinoma cells through the mitochondrial-mediated intrinsic and death receptor-induced extrinsic pathways. Oncology Letters, 5, 386-390. https://doi.org/10.3892/ol.2012.985
MLA
Yoo, K. H., Park, J., Lee, D. K., Fu, Y. Y., Baek, N. I., Chung, I. S."Pomolic acid induces apoptosis in SK-OV-3 human ovarian adenocarcinoma cells through the mitochondrial-mediated intrinsic and death receptor-induced extrinsic pathways". Oncology Letters 5.1 (2013): 386-390.
Chicago
Yoo, K. H., Park, J., Lee, D. K., Fu, Y. Y., Baek, N. I., Chung, I. S."Pomolic acid induces apoptosis in SK-OV-3 human ovarian adenocarcinoma cells through the mitochondrial-mediated intrinsic and death receptor-induced extrinsic pathways". Oncology Letters 5, no. 1 (2013): 386-390. https://doi.org/10.3892/ol.2012.985