Dammarane-type triterpene ginsenoside-Rg18 inhibits human non-small cell lung cancer A549 cell proliferation via G1 phase arrest

  • Authors:
    • Dong‑Gyu Leem
    • Ji‑Sun Shin
    • Kyung‑Tack Kim
    • Sang Yoon Choi
    • Myung‑Hee Lee
    • Kyung‑Tae Lee
  • View Affiliations

  • Published online on: February 15, 2018     https://doi.org/10.3892/ol.2018.8057
  • Pages: 6043-6049
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

A previous study reported that a novel dammarane-type triterpene saponin, ginsenoside-Rg18, derived from the root of Panax ginseng, displayed hydroxyl radical scavenging, anti‑bacterial and cytotoxic activities. However, the underlying molecular mechanisms of its anti‑proliferative effect on non‑small cell lung cancer (NSCLC) A549 cells remains unclear. In the present study, it was determined that Rg18 inhibited the proliferation of A549 cells with a half‑maximal inhibitory concentration of 150 µM. Flow cytometry analysis indicated that cell cycle progression was blocked by Rg18 at G1 phase in A549 cells, which was accompanied by downregulation of cyclin‑dependent kinase 2 (CDK2), CDK4, CDK6, cyclin D1, cyclin D2, cyclin E and phosphorylated retinoblastoma protein expression at the protein level. In addition, the CDK inhibitors (CDKNs), CDKN1A and CDKN1B, were upregulated following Rg18 treatment. Furthermore, Rg18 treatment resulted in the intracellular accumulation of reactive oxygen species (ROS), and a dose‑dependent inhibition of p38 mitogen activated protein kinase (p38), c‑Jun N‑terminal kinase (JNK) and nuclear factor‑κB (NF‑κB)/p65 phosphorylation. Taken together, Rg18‑mediated G1 phase arrest was closely associated with the generation of intracellular ROS, and p38, JNK and NF‑κB/p65 inhibition in A549 human NSCLC cells.

Introduction

Lung cancer is one of the leading causes of cancer-associated mortality worldwide (1). Non-small-cell lung cancer (NSCLC) accounts for ~80% all cases of lung cancer and has a 5-year survival rate of 15% (2). Resistance can be developed to contemporary chemotherapy regimens for NSCLC and various adverse side effects can be invoked (3). Therefore, the identification and development of more effective, less toxic antitumor NSCLC therapies is urgently required.

The cell cycle is a highly regulated mechanism of controlling cell growth, proliferation and survival. Cancer cells exhibit dysregulated cell cycles, with the overexpression of positive regulators and inhibition of negative regulators, resulting in unlimited replication potential (4). Therefore, the development of agents targeting the dysregulated cell cycle has been considered to be a suitable strategy for cancer therapy (3). Cell cycle progression depends on cyclin-dependent kinase (CDK) activation status, which act consecutively in G1 phase to initiate S phase and, in G2 phase, to initiate mitosis. With mitogenic stimulation, cyclin D is activated, in turn activating CDK4 and 6. Cyclin D-dependent kinases phosphorylate retinoblastoma proteins (Rb), which relieves transcription factor E2F1 from inhibition and allows for the expression of specific E2F1-target genes (5). The cyclin E-CDK2 complex concludes Rb phosphorylation and permits the activation of E2F1-responsive genes (6). Cyclin-CDK complexes are inhibited by two CDK inhibitor (CDKN) families: Inhibitors of CDKN4 (INK4) (p15INK4B, p16INK4A, p18INK4C and p19INK4D) and CDKN1A (p21CIP1/WAF1), 1B (p27KIP1) and 1C (p57KIP2) (7). The CDKNs also perform central cell cycle-regulating functions, coordinating internal and external signals that modulate cell cycle progression (7). Thus, the cell cycle is tightly controlled in response to external and internal factors. In abnormal conditions, including DNA damage, cell cycle progression is inhibited to prevent harmful progression.

Reactive oxygen species (ROS) are chemically reactive molecules, including the superoxide anion, hydrogen peroxide and the hydroxyl radical. ROS are endogenously produced during metabolic activities of the cell, including the oxidative phosphorylation of mitochondria (8). ROS may also arise from reactions with exogenous sources, including xenobiotic compounds (9). Cellular function requires moderate ROS levels for normal cell signaling. However, ROS overproduction can be toxic to cells owing to their peroxidative activity (10). One proposed anticancer strategy is the use of exogenous ROS-stressing agents or inhibition of endogenous antioxidants to elevate intracellular ROS levels to toxic amounts, triggering cell cycle arrest in the target cancer cell (11).

Ginsenosides are triterpene saponins and the major pharmacologically active components extracted from the roots and rhizomes of various Panax species. Ginsenosides are responsible for the majority of functions of ginseng, including angiogenesis, vasorelaxation and antioxidation (8). These molecules also possess anti-inflammatory and anticancer properties (1214). In a previous study, four dammarane-type triterpene saponins isolated from the Panax ginseng root exhibited effective hydroxyl radical scavenging ability, as well as antibacterial and cytotoxic activities (15). However, to the best of our knowledge, there has been no research into the anti-proliferative effects of these compounds or their underlying molecular mechanisms. As part of an ongoing screening program for the evaluation of the anti-proliferative potential of natural compounds, the mechanisms underlying the cell cycle-arresting activities of Rg18 in NSCLC A549 cells were investigated in the present study.

Materials and methods

Chemicals and reagents

Rg18 and Rs11 (Fig. 1A) were kindly provided by Dr. Kyung-Tack Kim (Korea Food Research Institute, Wanju-gun, South Korea), and its purity of >96% was determined by high-performance liquid chromatography-mass spectrometry analyses (15). RPMI-1640 medium, fetal bovine serum (FBS), penicillin and streptomycin were all obtained from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). MTT, phenylmethylsulfonyl fluoride (PMSF), N-acetylcysteine (NAC), 2′,7′-dichlorofluorescin diacetate (DCFH-DA), bisacrylamide and sodium dodecyl sulfate were purchased from Sigma-Aldrich; Merck KGaA (Darmstadt, Germany). CDK2 (sc-163), CDK4 (sc-260), CDK6 (sc-7961), cyclin D1 (sc-8396), cyclin D2 (sc-593), cyclin E (sc-198), p21CIP1/WAF1 (sc-397), p27KIP1 (sc-1641), Rb (sc-102), p38 mitogen activated protein kinase (p38, sc-535), c-Jun N-terminal kinase (JNK, sc-7345), extracellular signal-regulated kinase (ERK, sc-94), p65 (sc-372), p-JNK (sc-12882), p-ERK (sc-7383) and β-actin (sc-81178) primary antibodies were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Phosphorylated (p)-Rb (no. 9307), p-p38 (no. 9215), p-p65 (Ser536, no. 3031) primary antibodies were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Proteinase K, ribonuclease A, and TEMED were purchased from Bio-Rad Laboratories, Inc. (Hercules, CA, USA).

Cell culture

The human lung adenocarcinoma A549 cell line was obtained from the Korean Cell Line Bank (Seoul, Korea). A549 cells were grown at 37°C in RPMI-1640, supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 µg/ml streptomycin sulfate, in a humidified atmosphere of 5% CO2.

MTT assay

Cell viability was assessed by MTT assay, as described previously (16). A total of 5×104 cells/ml were seeded in wells containing 100 µl complete medium in a 96-well plate. After 24 h, Rg18 or Rs11 was added at concentrations of 0, 6.25, 12.5, 25, 50, 100 and 200 µM and cells were incubated for 48 h. A total of 50 µl MTT (stock solution, 5 mg/ml in PBS) was added, and the plates were incubated for an additional 4 h. The medium was disposed and the formazan blue was dissolved using 100 µl DMSO/well. The optical density was measured at 540 nm.

Cell cycle analysis

The cell cycle distribution was analyzed using propidium iodide staining as described previously (17). A total of 1×104 cells were collected from each experimental group, washed twice with PBS (4°C), fixed and permeabilized with 70% ethanol at 4°C for 1 h. The cells were washed once more with PBS and resuspended in a solution containing propidium iodide (50 µg/ml) and RNase A (250 µg/ml) for 30 min at room temperature. Fluorescence-activated cell sorting (FACS) was then performed to determine the cell cycle stage of each cell, using the Cytomics™ FC 500 flow cytometry and CXP cytometer software (version 2.0; (Beckman Coulter, Inc, Indianapolis, IN, USA).

Protein extraction and western blot analysis

Cells were collected by centrifugation at 200 × g for 10 min at 4°C, washed twice with PBS at 4°C, and centrifuged at 200 × g for 5 min. The resulting cell pellet was resuspended in 1X protein lysis buffer (Intron Biotechnology, Inc., Seongnam, Korea). The protein concentration was determined using the Bio-Rad protein assay reagent (Bio-Rad Laboratories, Inc.) and bovine serum albumin as a standard solution according to the manufacturer's instruction. Equal amounts of cell lysates (5–15 µl) were separated by 8–12% SDS-PAGE and transferred into nitrocellulose membranes. The membranes were incubated with the aforementioned primary antibodies. The membranes were then incubated with a 1:2,000 dilution of horseradish peroxidase-conjugated Goat anti-mouse IgG (no. 31430), Goat anti-rabbit IgG (no. 31460), or Rabbit anti-Goat IgG (no. 81-1620) (Thermo Fisher Scientific Inc.) and visualized using an enhanced chemiluminescence detection system (Santa Cruz Biotechnology, Inc., Dallas, TX, USA), according to the manufacturer's protocol. β-actin was used as a loading control and densitometric analysis was performed using Quantity One® Software (version 4.6.3; Bio-Rad Laboratories, Inc.).

ROS detection (DCFH-DA assay)

Intracellular ROS levels were detected using a DCFH-DA assay, as described previously (18). Cells (5×104 cells/ml) were pretreated with and without 10 mM NAC for 1 h and then treated with 150 µM Rg18 for 24 h. The cells were collected and suspended in pre-warmed PBS (37°C) prior to the addition of 20 µM DCFH-DA and incubated for 30 min at 37°C. When transported into the cells, DCFH-DA forms DCFH by deacetylation, and upon oxidation, fluorescent 2′,7′-dichlorofluorescin is formed. Fluorescence intensity was analyzed by Cytomics™ FC 500 flow cytometry and CXP cytometer software (version 2.0; Beckman Coulter, Inc., Indianapolis, IN, USA).

Statistical analysis

All data are presented as the mean ± standard deviation of the results from three independent experiments. One-way analysis of variance followed by Dunnett's post hoc test was performed to identify differences between groups using GraphPad Prism software (version 5.01; GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Rg18 inhibits cell proliferation and induces cell cycle arrest in A549 cells

The anti-proliferative effects of Rg18 and Rs11 were examined in A549 cells using an MTT assay (Fig. 1B) and half-maximal inhibitory concentration (IC50) values. The A549 cells were treated with different concentrations of Rg18 and Rs11 for 48 h, and it was found that Rg18 significantly decreases the viability of A549 cells, and these decreases are concentration-dependent (IC50=140.09 µM). However, Rs11 did not show obvious cytotoxic effects up to 100 µM and it decreased cell viability at 200 µM (cell viability: 51.78 %). The following experiments in A549 cells were conducted using Rg18 owing to their greater sensitivity to this ginsenoside. To investigate the mechanism of the anti-proliferative effect of Rg18, flow cytometry was used to identify changes in cell cycle distribution following treatment with Rg18. A549 cells were treated with various concentrations of Rg18 (0, 50, 100 and 150 µM) for 48 h, and the percentage of A549 cells in G1 phase increased from 63.3±1.5% (control group) to 68.6±2.1, 69.8±1.4 and 71.1±2.5% upon treatment with 50, 100 and 150 µM Rg18, respectively (Fig. 1C).

Effect of Rg18 on protein expression levels of various G1 phase cell cycle regulators in A549 cells

Rg18-induced G1 arrest in A549 cells (Fig. 1C), therefore, the protein expression levels of cell cycle regulatory molecules were investigated following 50, 100 or 150 µM Rg18 treatment. Rg18 treatment significantly reduced the protein expression level of CDK2, CDK4, CDK6, cyclin D1, cyclin D2 and cyclin E in A549 cells. Rg18 treatment also increased the protein expression levels of p21CIP1/WAF1 and p27KIP1 after a 48-h treatment. These proteins modulate CDK activity in various phases of the cell cycle, including G1. As Rb phosphorylation is influential in the transit from G1 to S phase (19), the phosphorylation levels of Rb were also examined. Following treatment with 150 µM Rg18 for 48 h, the degree of phosphorylation and expression levels of Rb significantly decreased (Fig. 2).

Rg18 treatment results in upregulation of intracellular ROS levels and downregulates p38, JNK and nuclear factor-κB (NF-κB) activation in A549 cells

ROS generation is an early sign of cell cycle arrest (20). To analyze ROS generation induced by Rg18 treatment, a DCFH-DA assay was performed. Following treatment with 150 µM Rg18 for 2 h, the generation of ROS increased significantly, an effect that was blocked by pretreatment with the antioxidant NAC (Fig. 3A). To investigate the molecular mechanism of Rg18-induced G1 arrest, p38, ERK, JNK, and NF-κB (p65) phosphorylation were analyzed in Rg18-treated A549 cells. These molecules serve notable roles in cell cycle regulation (21,22). Rg18 treatment resulted in a concentration-dependent decrease in p38, JNK and NF-κB/p65 phosphorylation, but there was no change to ERK phosphorylation levels (Fig. 3B).

Discussion

Previous studies have demonstrated that ginsenosides isolated from the root of P. ginseng C. A. Meyer could inhibit cancer cell growth in vitro and in vivo via cell cycle arrest (14,2325). In a previous study, it was demonstrated that four novel ginsenosides isolated from the P. ginseng root exhibited hydroxyl radical scavenging, anti-bacterial and cytotoxic activities (15). The aim of the present study was to determine whether Rg18 exerted an anti-proliferative effect on A549 cells and to characterize the molecular mechanism involved. The results demonstrated that Rg18 inhibited the proliferation of A549 cells and flow cytometric assays indicated that treatment with Rg18 lead to G1 arrest in A549 cells.

Cell cycle progression is highly controlled by interactions of various regulators, including the cyclins and their catalytic partners, CDKs (6). CDK complexes are formed and activated at specific cell cycle phases; their activities are necessary for progression through distinct cell cycle phases (7). Progressing through the G1 phase requires either CDK4 or CDK6 activity, followed by the activation of CDK2. The cyclin-CDK complex formed during G1 phase catalyzes the phosphorylation of the dominant inhibitors of G1/S phase cell cycle progression, the Rb family of tumor suppressor proteins, thereby allowing progression to S phase (26,27). Cyclin-CDK complexes can bind p21CIP1/WAF1 and p27KIP1, which inhibit kinase activities and prevent cell cycle progression (28). Western blot analysis demonstrated that Rg18 decreased the expression levels of cyclin D1, cyclin D2, cyclin E, CDK4, CDK6 and CDK2 in A549 cells. Furthermore, decreased CDK expression has been demonstrated to be associated with Rb under-phosphorylation, which is known to result in the sequestering of E2F, and thereby inhibition of the cell cycle progression (29).

The results indicate that Rg18 influences cell cycle progression via the upregulation of p21CIP1/WAF1 and p27KIP1 protein expression in A549 cells. It was apparent that strong CKI upregulation mediated Rg18-induced G1 phase arrest and the inhibition of cell growth. Overall, the G1 phase blockade in A549 cells appeared to be mediated by the downregulation of CDK activity associated with CKI induction, such as by p21CIP1/WAF1 and p27KIP1.

ROS are involved in multiple types of chemically induced cell cycle arrest; evidence indicates that increased oxidative stress is associated with cell cycle arrest induced by certain anticancer agents (11,30). Among the protopanaxadiols, ginsenoside-Rb2 has been demonstrated to significantly increase the expression of genes encoding antioxidant enzymes, including superoxide dismutase and catalase in vitro (31). The present study demonstrated that Rg18 treatment increased intracellular ROS levels, which led to cell cycle arrest.

The mitogen-activated protein kinases (MAPKs) are also involved in cell cycle regulation (21), and three pathways, ERK, JNK and p38, are closely associated with the progression of a number of malignant types of cancer, including breast and ovarian cancer, and NSCLC (32,33). JNK and p38 function in stress reactions and the induction of cell cycle arrest (34). The anticancer activity of 20(S)-protopanaxadiol in colon cancer cells is mediated by downregulation of the ERK, JNK and NF-κB signaling pathways (35). Additionally, compound K significantly inhibited phorbol 12-myristate 13-acetate-induced matrix metallopeptidase 9 protein expression and secretion via suppression of DNA-binding and activator protein-1 transcriptional activities, downstream of the p38, ERK and JNK pathways (36). However, it has been established that selenite-induced ROS arrest the cell cycle of NB4 cells at the G1 phase by inhibiting the JNK/activating transcription factor 2 axis in vitro and in vivo (37). In the present study, it was demonstrated that Rg18 treatment suppressed the phosphorylation of JNK and p38 in A549 cells.

Data from previous studies indicated that blocking the activation of NF-κB could be a critical target for the regulation of cell proliferation and antioxidant behaviors (3840). Ginsenoside Rg3 has been reported to inhibit NF-κB, induce G1 arrest and enhance susceptibility to docetaxel and other chemicals in prostate cancer cells (41). Furthermore, the ginsenoside Rd has been demonstrated to elevate intracellular glutathione levels by increasing γ-glutamyl cysteine ligase activation in rat hepatocyte H4IIE cells through NF-κB-DNA binding (42). This result indicates that NF-κB serves as a cellular marker for cell cycle arrest in HL-60 cells. In the present study, Rg18 treatment inhibited the phosphorylation of NF-κB/p65 in A549 cells. However, the exact mechanism of this effect, and whether it took place at the transcriptional and/or translational levels, requires further investigation.

In summary, Rg18 was found to inhibit the proliferation of A549 cells by arresting the cell cycle at the G1 phase by downregulating CDK2, CDK4 and CDK6 expression, in association with the induction of p21CIP1/WAF1 and p27KIP1. The results indicated that this was, at least in part, due to intracellular ROS production and the downregulation of multiple signaling pathways, including JNK, p38 and NF-κB/p65. Further research is required to dissect the underlying mechanisms of these pathway changes. However, these results illustrate the potential use of Rg18 in cancer treatment, either alone or in combination with other anticancer agents.

Acknowledgements

Not applicable.

Funding

This study was supported by a research grant from the Korea Food Research Institute (E0145202).

Availability of data and materials

All data generated and/or analyzed during this study are included in this published article.

Authors' contributions

DGL and JSS designed experiments, analyzed the data and statistics, and drafted the manuscript. KTK, SYC, and MHL. did isolation, structural analysis, and a purity analysis of Rg18 and Rs11. KTL designed the study, coordinated the project and gave final approval of publication. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Torre LA, Siegel RL and Jemal A: Lung cancer statistics. Adv Exp Med Biol. 893:1–19. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Molina JR, Yang P, Cassivi SD, Schild SE and Adjei AA: Non-small cell lung cancer: Epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 83:pp. 584–594. 2008; View Article : Google Scholar : PubMed/NCBI

3 

Huang CY, Ju DT, Chang CF, Muralidhar Reddy P and Velmurugan BK: A review on the effects of current chemotherapy drugs and natural agents in treating non-small cell lung cancer. Biomedicine (Taipei). 7:232017. View Article : Google Scholar : PubMed/NCBI

4 

Chen HS, Liu Y, Lin LQ, Zhao JL, Zhang CP, Jin JC, Wang L, Bai MH, Wang YC, Liu M and Shen BZ: Anti-proliferative effect of an extract of the root of Polygonum multiflorum Thunb. On MCF-7 human breast cancer cells and the possible mechanisms. Mol Med Rep. 4:1313–1319. 2011.PubMed/NCBI

5 

Dickson MA and Schwartz GK: Development of cell-cycle inhibitors for cancer therapy. Curr Oncol. 16:36–43. 2009.PubMed/NCBI

6 

Giacinti C and Giordano A: RB and cell cycle progression. Oncogene. 25:5220–5227. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Foster DA, Yellen P, Xu L and Saqcena M: Regulation of G1 cell cycle progression: Distinguishing the restriction point from a nutrient-sensing cell growth checkpoint(s). Genes Cancer. 1:1124–1131. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Nita M and Grzybowski A: The role of the reactive oxygen species and oxidative stress in the pathomechanism of the age-related ocular diseases and other pathologies of the anterior and posterior eye segments in adults. Oxid Med Cell Longev. 2016:31647342016. View Article : Google Scholar : PubMed/NCBI

9 

Ray PD, Huang BW and Tsuji Y: Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 24:981–990. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Wang Z, Zhao X and Gong X: Costunolide induces lung adenocarcinoma cell line A549 cells apoptosis through ROS (reactive oxygen species)-mediated endoplasmic reticulum stress. Cell Biol Int. 40:289–297. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Xiao W, Jiang Y, Men Q, Yuan L, Huang Z, Liu T, Li W and Liu X: Tetrandrine induces G1/S cell cycle arrest through the ROS/Akt pathway in EOMA cells and inhibits angiogenesis in vivo. Int J Oncol. 46:360–368. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Qi B, Zhang L, Zhang Z, Ouyang J and Huang H: Effects of ginsenosides-Rb1 on exercise-induced oxidative stress in forced swimming mice. Pharmacogn Mag. 10:458–463. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Li J, Zhong W, Wang W, Hu S, Yuan J, Zhang B, Hu T and Song G: Ginsenoside metabolite compound K promotes recovery of dextran sulfate sodium-induced colitis and inhibits inflammatory responses by suppressing NF-κB activation. PLoS One. 9:e878102014. View Article : Google Scholar : PubMed/NCBI

14 

Chung KS, Cho SH, Shin JS, Kim DH, Choi JH, Choi SY, Rhee YK, Hong HD and Lee KT: Ginsenoside Rh2 induces cell cycle arrest and differentiation in human leukemia cells by upregulating TGF-β expression. Carcinogenesis. 34:331–340. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Lee DG, Lee AY, Kim KT, Cho EJ and Lee S: Novel dammarane-type triterpene saponins from Panax ginseng root. Chem Pharm Bull (Tokyo). 63:927–934. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Plumb JA, Milroy R and Kaye SB: Effects of the pH dependence of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide-formazan absorption on chemosensitivity determined by a novel tetrazolium-based assay. Cancer Res. 49:4435–4440. 1989.PubMed/NCBI

17 

Choi JH and Lee KT: Costunolide-induced apoptosis in human leukemia cells: Involvement of c-jun N-terminal kinase activation. Biol Pharm Bull. 32:1803–1808. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Park EY, Kim JI, Leem DG, Shin JS, Kim KT, Choi SY, Lee MH, Choi JH, Lee YS and Lee KT: Resveratrol analogue (E)-8-acetoxy-2-[2-(3,4-diacetoxyphenyl)ethenyl]-quinazoline induces apoptosis via Fas-mediated pathway in HL-60 human leukemia cells. Oncol Rep. 36:3577–3587. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Taya Y: RB kinases and RB-binding proteins: New points of view. Trends Biochem Sci. 22:14–17. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Verbon EH, Post JA and Boonstra J: The influence of reactive oxygen species on cell cycle progression in mammalian cells. Gene. 511:1–6. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Zhang W and Liu HT: MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 12:9–18. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Joyce D, Albanese C, Steer J, Fu M, Bouzahzah B and Pestell RG: NF-kappaB and cell-cycle regulation: The cyclin connection. Cytokine Growth Factor Rev. 12:73–90. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Abe H, Arichi S, Hayashi T and Odashima S: Ultrastructural studies of Morris hepatoma cells reversely transformed by ginsenosides. Experientia. 35:1647–1649. 1979. View Article : Google Scholar : PubMed/NCBI

24 

Kim HS, Lee EH, Ko SR, Choi KJ, Park JH and Im DS: Effects of ginsenosides Rg3 and Rh2 on the proliferation of prostate cancer cells. Arch Pharm Res. 27:429–435. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Cheng CC, Yang SM, Huang CY, Chen JC, Chang WM and Hsu SL: Molecular mechanisms of ginsenoside Rh2-mediated G1 growth arrest and apoptosis in human lung adenocarcinoma A549 cells. Cancer Chemother Pharmacol. 55:531–540. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Hwang HC and Clurman BE: Cyclin E in normal and neoplastic cell cycles. Oncogene. 24:2776–2786. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Connell-Crowley L, Harper JW and Goodrich DW: Cyclin D1/Cdk4 regulates retinoblastoma protein-mediated cell cycle arrest by site-specific phosphorylation. Mol Biol Cell. 8:287–301. 1997. View Article : Google Scholar : PubMed/NCBI

28 

Schmidt M, Lu Y, Parant JM, Lozano G, Bacher G, Beckers T and Fan Z: Differential roles of p21(Waf1) and p27(Kip1) in modulating chemosensitivity and their possible application in drug discovery studies. Mol Pharmacol. 60:900–906. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Lee KW, Kim HJ, Lee YS, Park HJ, Choi JW, Ha J and Lee KT: Acteoside inhibits human promyelocytic HL-60 leukemia cell proliferation via inducing cell cycle arrest at G0/G1 phase and differentiation into monocyte. Carcinogenesis. 28:1928–1936. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Wang R, Zhang Q, Peng X, Zhou C, Zhong Y, Chen X, Qiu Y, Jin M, Gong M and Kong D: Stellettin B induces G1 arrest, apoptosis and autophagy in human non-small cell lung cancer A549 cells via blocking PI3K/Akt/mTOR pathway. Sci Rep. 6:270712016. View Article : Google Scholar : PubMed/NCBI

31 

Chang MS, Lee SG and Rho HM: Transcriptional activation of Cu/Zn superoxide dismutase and catalase genes by panaxadiol ginsenosides extracted from Panax ginseng. Phytother Res. 13:641–644. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Dhillon AS, Hagan S, Rath O and Kolch W: MAP kinase signalling pathways in cancer. Oncogene. 26:3279–3290. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Roberts PJ and Der CJ: Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 26:3291–3310. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Wagner EF and Nebreda AR: Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 9:537–549. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Gao JL, Lv GY, He BC, Zhang BQ, Zhang H, Wang N, Wang CZ, Du W, Yuan CS and He TC: Ginseng saponin metabolite 20(S)-protopanaxadiol inhibits tumor growth by targeting multiple cancer signaling pathways. Oncol Rep. 30:292–298. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Jung SH, Woo MS, Kim SY, Kim WK, Hyun JW, Kim EJ, Kim DH and Kim HS: Ginseng saponin metabolite suppresses phorbol ester-induced matrix metalloproteinase-9 expression through inhibition of activator protein-1 and mitogen-activated protein kinase signaling pathways in human astroglioma cells. Int J Cancer. 118:490–497. 2006. View Article : Google Scholar : PubMed/NCBI

37 

An JJ, Shi KJ, Wei W, Hua FY, Ci YL, Jiang Q, Li F, Wu P, Hui KY, Yang Y and Xu CM: The ROS/JNK/ATF2 pathway mediates selenite-induced leukemia NB4 cell cycle arrest and apoptosis in vitro and in vivo. Cell Death Dis. 4:e9732013. View Article : Google Scholar : PubMed/NCBI

38 

Kaltschmidt B, Kaltschmidt C, Hehner SP, Dröge W and Schmitz ML: Repression of NF-kappaB impairs HeLa cell proliferation by functional interference with cell cycle checkpoint regulators. Oncogene. 18:3213–3225. 1999. View Article : Google Scholar : PubMed/NCBI

39 

Rajitha B, Belalcazar A, Nagaraju GP, Shaib WL, Snyder JP, Shoji M, Pattnaik S, Alam A and El-Rayes BF: Inhibition of NF-κB translocation by curcumin analogs induces G0/G1 arrest and downregulates thymidylate synthase in colorectal cancer. Cancer Lett. 373:227–233. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Tran KQ, Tin AS and Firestone GL: Artemisinin triggers a G1 cell cycle arrest of human Ishikawa endometrial cancer cells and inhibits cyclin-dependent kinase-4 promoter activity and expression by disrupting nuclear factor-κB transcriptional signaling. Anticancer Drugs. 25:270–281. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Kim SM, Lee SY, Cho JS, Son SM, Choi SS, Yun YP, Yoo HS, Yoon DY, Oh KW, Han SB and Hong JT: Combination of ginsenoside Rg3 with docetaxel enhances the susceptibility of prostate cancer cells via inhibition of NF-kappaB. Eur J Pharmacol. 631:1–9. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Kim ND, Pokharel YR and Kang KW: Ginsenoside Rd enhances glutathione levels in H4IIE cells via NF-kappaB-dependent gamma-glutamylcysteine ligase induction. Pharmazie. 62:933–936. 2007.PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 15 Issue 4

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
Leem DG, Shin JS, Kim KT, Choi SY, Lee MH and Lee KT: Dammarane-type triterpene ginsenoside-Rg18 inhibits human non-small cell lung cancer A549 cell proliferation via G1 phase arrest. Oncol Lett 15: 6043-6049, 2018
APA
Leem, D., Shin, J., Kim, K., Choi, S.Y., Lee, M., & Lee, K. (2018). Dammarane-type triterpene ginsenoside-Rg18 inhibits human non-small cell lung cancer A549 cell proliferation via G1 phase arrest. Oncology Letters, 15, 6043-6049. https://doi.org/10.3892/ol.2018.8057
MLA
Leem, D., Shin, J., Kim, K., Choi, S. Y., Lee, M., Lee, K."Dammarane-type triterpene ginsenoside-Rg18 inhibits human non-small cell lung cancer A549 cell proliferation via G1 phase arrest". Oncology Letters 15.4 (2018): 6043-6049.
Chicago
Leem, D., Shin, J., Kim, K., Choi, S. Y., Lee, M., Lee, K."Dammarane-type triterpene ginsenoside-Rg18 inhibits human non-small cell lung cancer A549 cell proliferation via G1 phase arrest". Oncology Letters 15, no. 4 (2018): 6043-6049. https://doi.org/10.3892/ol.2018.8057