Ginsenoside 20(S)-Rh2 exerts anti-cancer activity through the Akt/GSK3β signaling pathway in human cervical cancer cells

  • Authors:
    • Xin Shi
    • Ji Yang
    • Gang Wei
  • View Affiliations

  • Published online on: January 18, 2018     https://doi.org/10.3892/mmr.2018.8454
  • Pages: 4811-4816
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ginsenoside 20(S)-Rh2 (GRh2) is a bioactive compound derived from ginseng that is believed to maintain health in traditional Chinese medicine. Emerging evidence has suggested that GRh2 exhibits anti‑cancer activity. The present study hypothesized that GRh2 has an anti‑cancer function in human cervical cancer cells. An MTS assay demonstrated that GRh2 attenuated proliferation of HeLa cells in a dose‑ and time‑dependent manner. In addition, GRh2 inhibited migration and invasion, as determined by wound healing and transwell invasion assays, respectively. Furthermore, GRh2 treatment reduced expression of mesenchymal markers N‑cadherin and vimentin as well as epithelial mesenchymal transition transcriptional factor zinc finger E‑box‑binding homeobox 1 and snail1, and increased the protein expression levels of epithelial marker E‑cadherin. In addition, the results revealed that GRh2 prevented activation of the protein kinase B (Akt)/glycogen synthase kinase (GSK)3β signaling pathway in HeLa cells. In conclusion, the results suggested that GRh2 inhibits cervical cancer cell proliferation by targeting the Akt pathway, and prevents cervical cancer cell migration and invasion by suppressing the Akt/GSK3β regulated EMT process, and therefore, GRh2 may have the potential to be a novel anti‑cancer agent for cervical cancer.

Introduction

Cervical cancer is one of the most common malignancies in woman worldwide, which occurs and develops on the surface of the cervix (1). A total of >510,000 patients are diagnosed with cervical cancer, and >288,000 patients die from the disease each year worldwide (1). The primary cause of cervical cancer is infection with human papillomavirus (HPV) (2). Although vaccines to high-risk human HPV (hrHPV) strains may prevent infection with hrHPV, cervical cancer remains a primary cause of cancer-associated mortality, particularly in under-developed countries (3,4).

Tumor metastasis is the primary cause of cancer-associated death. Numerous studies have suggested that epithelial mesenchymal transition (EMT) contributes to migration, invasion and metastasis of cancer (5,6). During the EMT process, epithelial cells undergo a cell morphology alteration from the polarized epithelial phenotype to mesenchymal phenotype, loss of cell-cell junction and epithelial markers including E-cadherin, acquirement of mesenchymal markers N-cadherin, vimentin and febronectin, and cytoskeleton rearrangement, which leads to gain of mesenchymal traits including cell migration and invasion (7,8). The EMT process is regulated by several transcription factors including zinc finger E-box-binding homeobox 1 (Zeb1), snail1, snail2 (Slug) and Twist, which repress the expression of E-cadherin and other cell adhesion and cytokeratin genes, and increase expression of mesenchymal markers vimentin, fibronectin and N-cadherin (9). Several signaling pathways are activated during EMT, including nuclear factor (NF)-κB, glycogen synthase kinase (GSK)3β, Notch, protein kinase B (Akt) and mitogen activated protein kinase (MAPK), which have been demonstrated to promote metastasis of lung, breast, ovarian and prostate cancers (1013). In addition, EMT is involved in anti-cancer drug resistance (14). Therefore, EMT is a target of cancer therapy, particularly in metastasized cancer types.

Currently, chemotherapy and radiotherapy are the principal therapeutic strategies for cervical cancer. However, these two therapeutic approaches have severe side effects including headache, abnormal respiration, cirrhosis and cardiac dysfunction. There is therefore increasing interest in the study of safe and effective anti-cancer compounds from natural plants (15). Ginseng is considered as a panacea and is frequently used in traditional Chinese medicine (16,17). Ginsenosides are one of the primary bioactive ingredients in ginseng, which have various pharmacological effects including anti-obesity, anti-diabetic, regulation of blood pressure, anti-aging, immune regulation and anti-tumor functions (1822). Ginsenosides are classified into three types, including protopanaxatriol-type, oleanolic acid-type and protopanaxadiol-type ginsenosides (23). Ginsenoside 20(S)-Rh2 (GRh2) belongs to the protopanaxadiol-type ginsenoside group. Several studies have reported that GRh2 exhibits anticancer activities in glioblastoma, skin squamous cell carcinoma, pancreatic cancer, prostatic cancer and leukemia (2428). A previous study used GRh2 to inhibit cancer cell proliferation and induce transformation to normal cells in patients with cervical cancer (29). However, the effect of GRh2 on cervical cancer, the EMT process and its underlying mechanism remains unclear.

In the present study, it was demonstrated that GRh2 effectively inhibited proliferation, migration, invasion and EMT by targeting the Akt/GSK3β signaling pathway in human cervical cancer cells. Therefore, the results suggested that GRh2 may have the potential to be a novel anti-cancer agent for cervical cancer.

Materials and methods

Reagents and antibodies

Commercial GRh2 was obtained from Sichuan Weike Biotechnology Co., Ltd. (Chengdu, China). GRh2 was prepared in a stock of 100 mg/ml and applied to cultured HeLa cells at 10, 25 and 50 µM. Antibodies for N-cadherin (cat. no. 13116), E-cadherin (cat. no. 3195), vimentin (cat. no. 5741), Zeb1 (cat. no. 3396), snail1 (cat. no. 3879), phosphorylated (p)-Akt (cat. no. 4060), Akt (cat. no. 4691), p-GSK3β (cat. no. 9327), GSK3β (cat. no. 5676) and β-actin (cat. no. 4970) were obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA).

Cell culture

Human cervical carcinoma HeLa cells were purchased from Shanghai Institute of Pharmaceutical Industry (Shanghai, China). Cells were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% streptomycin and penicillin at 37°C in an environment containing 5% CO2. DMEM and FBS were purchased from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA).

Cell proliferation assay

Cell proliferation was detected by 3-(4, 5-dimethylthiazol-2-yl)-5 3-carboxymethonyphenol)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay. 1×103/ml HeLa were seeded into each well of a 96-well microplate. Then, cells were incubated with media containing normal saline (control) or different concentrations of GRh2 (10, 25 and 50 µM) respectively. Cells were incubated for 6, 24, 48, 72, 96 and 120 h and then incubated with 10 µl MTS reagent (Promega Corporation, Madison, WI, USA) at 37°C for 4 h according to the manufacturer's protocol. The absorbance was measured at a wavelength of 490 nm using a microplate reader. Proliferation inhibition rates were determined following treatment for 120 h. % proliferation inhibition rate=(1-drug group/control group) ×100.

Transwell invasion assays

For the matrigel assay, 5×104 HeLa cells at the logarithmic growth phase were supplemented with 100 µl serum-free DMEM and seeded into the upper chamber of a transwell plate with 8-mm pore size (BD Biosciences, Franklin Lakes, NJ, USA). The upper side of the filter member of the chamber was coated with Matrigel (BD Biosciences) that was diluted with DMEM media (1:3). A total of 600 µl DMEM media supplemented with 1% FBS was added in the lower chamber, and GRh2 at a concentration of 0 µM (control) or 10, 25 and 50 µM was added into the upper chamber. Following incubation for 24 h at 37°C, the cells in the upper membrane were discarded and cells on the lower membrane were fixed using 4% paraformaldehyde for 30 min and stained with crystal violet (Beyotime Institute of Biotechnology, Haimen, China) for 10 min at room temperature. Next, five random fields were captured with a phase contrast video microscope (ELWD 0.3; Nikon Corporation, Tokyo, Japan; magnification, ×40) and then the cells were counted. Each experiment was performed in triplicate.

Wound healing assay

For wound healing assay, 3×105 HeLa cells at the logarithmic growth phase were seeded into 6-well plates. At 80% confluence, cells were incubated with GRh2 at a concentration of 0 µM (control), or 10, 25 and 50 µM, and the monolayer was disrupted with a cell scraper and captured at 0 and 48 h with a phase contrast video microscope (model ELWD 0.3; Nikon Corporation, Tokyo, Japan; magnification, ×100). Each experiment was performed in triplicate. The percentage of wound closure between the wound edges at different time points was measured with Image-Pro Plus version 6.0 software (Media Cybernetics, Inc., Rockville, MD, USA). % wound closure=(1-distance at 0 h/distance at 48 h) ×100.

Protein extraction and western blotting

For detection of N-cadherin, E-cadherin, vimentin, zeb1 and snail1 expression, HeLa cells were treated with GRh2 at a concentration of 0 µM (control), or 25, and 50 µM for 48 h at 37°C in an environment containing 5% CO2. To investigate the activity of the Akt and GSK3β signaling pathway, HeLa cells were treated with GRh2 at the concentration of 0 µM (control) or 25 and 50 µM for 12 h at 37°C in an environment containing 5% CO2. Cells were lysed using a radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology) supplemented with complete EDTA-free protease inhibitor cocktail tablets (Roche Diagnostics GmbH, Mannheim, Germany) according to the manufacturer's protocol. Total protein concentrations were determined with the bicinchoninic acid protein assay kit (Applygen Technologies Inc., Beijing, China). A total of 40 µg protein in each sample was loaded onto 8% SDS-PAGE gels, and transferred onto polyvinylidene difluoride membranes (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Following blocking with 5% nonfat milk for 30 min at room temperature, the membranes were incubated with primary antibodies including N-cadherin (1:1,000 dilution), E-cadherin (1:1,000 dilution), vimentin (1:1,000 dilution), Zeb1 (1:1,000 dilution), snail1 (1:1,000 dilution), phosphorylated (p)-Akt (1:2,000 dilution), Akt (1:1,000 dilution), p-GSK3β (1:1,000 dilution), GSK3β (1:1,000 dilution) and β-actin (1:1,000 dilution) at 4°C overnight. The membranes were next incubated with horseradish peroxidase-conjugated anti-rabbit IgG (cat. no. A0545; 1:5,000 dilution; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) for 1 h at room temperature. Signals were visualized using enhanced chemiluminescence reagent (EMD Millipore, Billerica, USA) and protein expression levels normalized to β-actin were calculated by using ImageJ software (version 1.48; National Institutes of Health, Bethesda, MD, USA). Detection of the protein of interest was repeated three times. The quantification of the western blots presented in the figures are representative of three independent experiments.

Statistical analysis

All tests were performed in triplicate and the experiments were repeated three times independently. Data was expressed as mean ± standard deviation. A one-way analysis of variance followed by Tukey's post hoc test was conducted to assess differences among multiple groups. All statistical calculations were carried out using SPSS software, version 19.0 (IBM Corp., Armonk, NY, USA) and P<0.05 was considered to indicate a statistically significant difference.

Results

GRh2 inhibits cell viability and proliferation of cervical cancer

To evaluate the function of GRh2 on cervical cancer cells, HeLa cells were exposed to various concentrations of GRh2 and the cell proliferation was assessed. Compared with the control group, GRh2 treatments inhibited HeLa cell proliferation in a dose- and time-dependent manner (P<0.001; Fig. 1A). Compared with the control group, proliferation inhibition rates were 40, 60 and 80% at GRh2 concentrations of 10, 25 and 50 µM, respectively, at 120 h (Fig. 1B). These results suggested that GRh2 inhibited the proliferation of cervical cancer cells.

GRh2 inhibits cell migration and invasion of cervical cancer

To ascertain the role of GRh2 in cervical cancer cell migration, wound healing and transwell invasion assays were performed in Hela cells treated with GRh2 at concentrations of 0 µM (control), or 10, 25 and 50 µM. The migration abilities were inhibited in HeLa cells treated with GRh2 in a dose-dependent manner (P<0.05 vs. control; Fig. 2A and B). Similar inhibitory effects were observed when detecting the invasion ability of HeLa cells by a Matrigel-coated transwell invasion assay (P<0.05 vs. control; Fig. 2C and D). These results suggested that GRh2 inhibited cell migration and invasion of cervical cancer.

GRh2 inhibits epithelial to mesenchymal transition of cervical cancer

To investigate whether GRh2 inhibits the EMT process, EMT markers were detected in HeLa cells treated with different concentrations of GRh2 (0, 25 and 50 µM) for 48 h. As expected, GRh2 treatment decreased the expression levels of N-cadherin and vimentin, and increased the protein expression levels of epithelial marker E-cadherin (Fig. 3A and B). The effects of GRh2 on the expression levels of EMT control transcription factors, Zeb1 and snail1, were also determined. The results revealed a downregulation in the expression levels of Zeb1 and snail1 in the GRh2-treated groups (Fig. 3A and B). These data suggested that GRh2 effectively inhibited EMT in a dose-dependent manner in cervical cancer cells.

GRh2 inhibits the Akt signaling pathway in cervical cancer cells

The effect of GRh2 on the Akt signaling pathway was investigated. HeLa cells were treated with different concentrations of GRh2 (0, 10, 25 and 50 µM) for 12 h. The phosphorylation and protein expression levels of Akt were then detected. GRh2 treatment significantly inhibited the phosphorylation of Akt compared with cells treated with 0 µM GRh2 (Fig. 4A and B), which suggested that GRh2 inhibited the activation of the Akt pathway.

GRh2 inhibits the GSK3β signaling pathway in cervical cancer cells

GSK3β is a downstream pathway of Akt. It has been reported that the Akt/GSK3β pathway regulates EMT (30). Therefore, the effect of GRh2 on the GSK3β pathway was investigated. HeLa cells were treated with different concentrations of GRh2 (0, 10, 25 and 50 µM) for 12 h. The activation of GSK3β was then detected. GRh2 treatment significantly suppressed the phosphorylation of GSK3β compared with cells treated with 0 µM GRh2 (Fig. 5A and B), which suggested that GRh2 inhibited the activation of the Akt/GSK3β pathway.

Discussion

Currently, cancer is one of the most common diseases that leads to mortality in humans (31). Chemotherapy and radiotherapy are the primary clinical therapeutic approaches. However, a limitation of these treatment strategies is toxicity to normal cells in addition to the tumor cells. The identification of safe and efficacious anticancer drugs is of primary concern (32). Natural products are the primary source for the development of anti-cancer agents (33). The anticancer activities of Panax ginseng have been demonstrated by several studies (34,35). Ginsenosides, a group of bioactive compounds of Panax ginseng, exhibit anticancer activities (36,37). GRh2 has been demonstrated to inhibit cancer cell proliferation, migration and invasion, and prevent tumor growth and metastasis (2428). However, only a small number of studies have explored the effects of GRh2 in human cervical cancer.

In the present study, the effect of GRh2 on cell proliferation of human cervical cancer cells was investigated. The results revealed that GRh2 impaired HeLa cell proliferation in a dose- and time-dependent manner. These findings are in accordance with previous reports that demonstrated that ginseng Rh2 prevents cell proliferation in colon, lung, liver, and gastric cancers, in addition to leukemia and glioblastoma (3842). In addition, it was observed that GRh2 inhibited the activation of the Akt pathway in HeLa cells. Akt mediates various biological processes, including glucose metabolism, cell survival, proliferation and differentiation (43,44). Collectively, the results of the present study, and previous studies, suggest that GRh2 may inhibit human cervical cancer cell proliferation via impairment of the Akt signaling pathway.

Furthermore, the present study revealed that GRh2 decreased the migration and invasion of human cervical cancer cells, which concurred with previous reports that Rh2 inhibits cell migration and invasion in C2C12 skeletal muscle cells (45,46). EMT is an important cellular process that promotes tumor progression and metastasis (47). To ascertain the underlying molecular mechanism of GRh2 inhibition of cervical cancer migration and invasion, the present study investigated the effects of GRh2 on EMT. The results demonstrated that Rh2 treatment increased the expression levels of epithelial marker E-cadherin and decreased the expression levels of mesenchymal N-cadherin and vimentin, in addition to EMT-control transcription factors Zeb1 and snail1. In addition, GRh2 treatment impaired the activation of Akt and the phosphorylation of GSK3β. Aberrant activation of the Akt signaling pathway was observed during tumor proliferation and metastasis as well as during the progression of EMT (45,48,49). GSK3β is activated when it is dephosphorylated. The inhibition of Akt pathway can activateGSK3β by preventing its phosphorylation (50,51). The activation of GSK3β induces the degradation of Snail. As a transcriptional factor, Snail inhibits the expression of epithelial marker E-cadherin and activates the transcription of mesenchymal N-cadherin and vimentin (47,5254). Thus, degradation of Snail increases the expression of epithelial marker E-cadherin and decreases the expression of mesenchymal N-cadherin and vimentin. Accordingly, the results of the present study demonstrated that GRh2 inhibited AKT pathway, dephosphorylated and activated GSK3β and inhibited the expression of Snail, which lead to upregulation of E-cadherin and downregulation of N-cadherin and vimentin. Taken together with the data from the present study and previous observations, it is suggested that GRh2 inhibited the expression of Snail and prevented EMT by inhibiting AKT and activating GSK3β.

In conclusion, the present study demonstrated that GRh2 inhibits cell proliferation of cervical cancer by suppressing the Akt signaling pathway. In addition, the data suggested that GRh2 prevents cell migration, invasion and EMT via inhibition of the Akt/GSK3β signaling pathway. Therefore, these findings suggested that GRh2 may be a potential treatment strategy for cervical cancer treatment.

References

1 

Saslow D, Castle PE, Cox JT, Davey DD, Einstein MH, Ferris DG, Goldie SJ, Harper DM, Kinney W, Moscicki AB, et al: American cancer society guideline for human papillomavirus (HPV) vaccine use to prevent cervical cancer and its precursors. CA Cancer J Clin. 57:7–28. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Steben M and Duarte-Franco E: Human papillomavirus infection: Epidemiology and pathophysiology. Gynecol Oncol. 107 2 Suppl 1:S2–S5. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Ginsburg O, Bray F, Coleman MP, Vanderpuye V, Eniu A, Kotha SR, Sarker M, Huong TT, Allemani C, Dvaladze A, et al: The global burden of women's cancers: A grand challenge in global health. Lancet. 389:847–860. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Yang A, Farmer E, Wu TC and Hung CF: Perspectives for therapeutic HPV vaccine development. J Biomed Sci. 23:752016. View Article : Google Scholar : PubMed/NCBI

5 

Cheng WY, Kandel JJ, Yamashiro DJ, Canoll P and Anastassiou D: A multi-cancer mesenchymal transition gene expression signature is associated with prolonged time to recurrence in glioblastoma. PLoS One. 7:e347052012. View Article : Google Scholar : PubMed/NCBI

6 

Tian L, Shen D, Li X, Shan X, Wang X, Yan Q and Liu J: Ginsenoside Rg3 inhibits epithelial-mesenchymal transition (EMT) and invasion of lung cancer by down-regulating FUT4. Oncotarget. 7:1619–1632. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Wijnhoven BP, Dinjens WN and Pignatelli M: E-cadherin-catenin cell-cell adhesion complex and human cancer. BR J Surg. 87:992–1005. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Nieto MA, Huang RY, Jackson RA and Thiery JP: Emt: 2016. Cell. 166:21–45. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Kaowinn S, Kim J, Lee J, Shin DH, Kang CD, Kim DK, Lee S, Kang MK, Koh SS, Kim SJ and Chung YH: Cancer upregulated gene 2 induces epithelial-mesenchymal transition of human lung cancer cells via TGF-β signaling. Oncotarget. 8:5092–5110. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Lee J, Byun HJ, Lee MS, Jin YJ, Jeoung D, Kim YM and Lee H: The metastasis suppressor CD82/KAI1 inhibits fibronectin adhesion-induced epithelial-to-mesenchymal transition in prostate cancer cells by repressing the associated integrin signaling. Oncotarget. 8:1641–1654. 2017.PubMed/NCBI

12 

Lu L, Wang J, Wu Y, Wan P and Yang G: Rap1A promotes ovarian cancer metastasis via activation of ERK/p38 and notch signaling. Cancer Med. 5:3544–3554. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Ci Y, Qiao J and Han M: Molecular mechanisms and metabolomics of natural polyphenols interfering with breast cancer metastasis. Molecules. 21:E16342016. View Article : Google Scholar : PubMed/NCBI

14 

Du B and Shim JS: Targeting epithelial-mesenchymal transition (EMT) to overcome drug resistance in cancer. Molecules. 21:E9652016. View Article : Google Scholar : PubMed/NCBI

15 

Lim HK, Bae W, Lee HS and Jung J: Anticancer activity of marine sponge Hyrtios sp. extract in human colorectal carcinoma RKO cells with different p53 status. BioMed Res Int. 2014:4135752014. View Article : Google Scholar : PubMed/NCBI

16 

Bent S and Ko R: Commonly used herbal medicines in the United States: A review. Am J Med. 116:478–485. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Nocerino E, Amato M and Izzo AA: The aphrodisiac and adaptogenic properties of ginseng. Fitoterapia. 71 Suppl 1:S1–S5. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Cheng Y, Shen LH and Zhang JT: Anti-amnestic and anti-aging effects of ginsenoside Rg1 and Rb1 and its mechanism of action. Acta Pharmacol Sin. 26:143–149. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Dong H, Bai LP, Wong VK, Zhou H, Wang JR, Liu Y, Jiang ZH and Liu L: The in vitro structure-related anti-cancer activity of ginsenosides and their derivatives. Molecules. 16:10619–10630. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Hwang JT, Kim SH, Lee MS, Kim SH, Yang HJ, Kim MJ, Kim HS, Ha J, Kim MS and Kwon DY: Anti-obesity effects of ginsenoside Rh2 are associated with the activation of AMPK signaling pathway in 3T3-L1 adipocyte. Biochemical Biophys Res Commun. 364:1002–1008. 2007. View Article : Google Scholar

21 

Jovanovski E, Bateman EA, Bhardwaj J, Fairgrieve C, Mucalo I, Jenkins AL and Vuksan V: Effect of Rg3-enriched Korean red ginseng (Panax ginseng) on arterial stiffness and blood pressure in healthy individuals: A randomized controlled trial. J Am Soc Hypertens. 8:537–541. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Kim J, Han BJ, Kim H, Lee JY, Joo I, Omer S, Kim YS and Han Y: Th1 immunity induction by ginsenoside Re involves in protection of mice against disseminated candidiasis due to Candida albicans. Int Immunopharmacol. 14:481–486. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Oh SJ, Lee S, Choi WY and Lim CJ: Skin anti-photoaging properties of ginsenoside Rh2 epimers in UV-B-irradiated human keratinocyte cells. J Biosci. 39:673–682. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Li S, Gao Y, Ma W, Guo W, Zhou G, Cheng T and Liu Y: EGFR signaling-dependent inhibition of glioblastoma growth by ginsenoside Rh2. Tumour Biol. 35:5593–5598. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Tang XP, Tang GD, Fang CY, Liang ZH and Zhang LY: Effects of ginsenoside Rh2 on growth and migration of pancreatic cancer cells. World J Gastroenterol. 19:1582–1592. 2013. View Article : Google Scholar : PubMed/NCBI

26 

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

27 

Zhang Q, Hong B, Wu S and Niu T: Inhibition of prostatic cancer growth by ginsenoside Rh2. Tumour Biol. 36:2377–2381. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Liu S, Chen M, Li P, Wu Y, Chang C, Qiu Y, Cao L, Liu Z and Jia C: Ginsenoside rh2 inhibits cancer stem-like cells in skin squamous cell carcinoma. Cell Physiol Biochem. 36:499–508. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Li Q, Li Y, Wang X, Fang X, He K, Guo X, Zhan Z, Sun C and Jin YH: Co-treatment with ginsenoside Rh2 and betulinic acid synergistically induces apoptosis in human cancer cells in association with enhanced capsase-8 activation, bax translocation and cytochrome c release. Mol Carcinog. 50:760–769. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Guo H, Luo H, Yuan H, Xia Y, Shu P, Huang X, Lu Y, Liu X, Keller ET, Sun D, et al: Litchi seed extracts diminish prostate cancer progression via induction of apoptosis and attenuation of EMT through Akt/GSK-3β signaling. Sci Rep. 7:416562017. View Article : Google Scholar : PubMed/NCBI

31 

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

32 

Toh DF, Patel DN, Chan EC, Teo A, Neo SY and Koh HL: Anti-proliferative effects of raw and steamed extracts of Panax notoginseng and its ginsenoside constituents on human liver cancer cells. Chin Med. 6:42011. View Article : Google Scholar : PubMed/NCBI

33 

Yang ZG, Sun HX and Ye YP: Ginsenoside Rd from Panax notoginseng is cytotoxic towards HeLa cancer cells and induces apoptosis. Chem Biodivers. 3:187–197. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Unlu A, Nayir E, Kirca O, Ay H and Ozdogan M: Ginseng and cancer. J BUON. 21:1383–1387. 2016.PubMed/NCBI

35 

Hafez MM, Hamed SS, El-Khadragy MF, Hassan ZK, Al Rejaie SS, Sayed-Ahmed MM, Al-Harbi NO, Al-Hosaini KA, Al-Harbi MM, Alhoshani AR, et al: Effect of ginseng extract on the TGFβ1 signaling pathway in CCl4-induced liver fibrosis in rats. BMC Complement Altern Med. 17:452017. View Article : Google Scholar : PubMed/NCBI

36 

Zheng X, Chen W, Hou H, Li J, Li H, Sun X, Zhao L and Li X: Ginsenoside 20(S)-Rg3 induced autophagy to inhibit migration and invasion of ovarian cancer. Biomed Pharmacother. 85:620–626. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Lahiani MH, Eassa S, Parnell C, Nima Z, Ghosh A, Biris AS and Khodakovskaya MV: Carbon nanotubes as carriers of Panax ginseng metabolites and enhancers of ginsenosides Rb1 and Rg1 anti-cancer activity. Nanotechnology. 28:0151012017. View Article : Google Scholar : PubMed/NCBI

38 

Yang J, Yuan D, Xing T, Su H, Zhang S, Wen J, Bai Q and Dang D: Ginsenoside Rh2 inhibiting HCT116 colon cancer cell proliferation through blocking PDZ-binding kinase/T-LAK cell-originated protein kinase. J Ginseng Res. 40:400–408. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Liu X, Sun Y, Yue L, Li S, Qi X, Zhao H, Yang Y, Zhang C and Yu H: JNK pathway and relative transcriptional factor were involved in ginsenoside Rh2-mediated G1 growth arrest and apoptosis in human lung adenocarcinoma A549 cells. Genet Mol Res. 15:2016. View Article : Google Scholar

40 

Shi Q, Shi X, Zuo G, Xiong W, Li H, Guo P, Wang F, Chen Y, Li J and Chen DL: Anticancer effect of 20(S)-ginsenoside Rh2 on HepG2 liver carcinoma cells: Activating GSK-3β and degrading beta-catenin. Oncol Rep. 36:2059–2070. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Qian J, Li J, Jia JG, Jin X, Yu DJ, Guo CX, Xie B and Qian LY: Ginsenoside-Rh2 inhibits proliferation and induces apoptosis of human gastric cancer SGC-7901 side population cells. Asian Pac J Cancer Prev. 17:1817–1821. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Wanderi C, Kim E, Chang S, Choi C and Choi K: Ginsenoside 20(S)-protopanaxadiol suppresses viability of human glioblastoma cells via down-regulation of cell adhesion proteins and cell-cycle arrest. Anticancer Res. 36:925–932. 2016.PubMed/NCBI

43 

Lim S, Yoon JW, Choi SH, Cho BJ, Kim JT, Chang HS, Park HS, Park KS, Lee HK, Kim YB and Jang HC: Effect of ginsam, a vinegar extract from Panax ginseng, on body weight and glucose homeostasis in an obese insulin-resistant rat model. Metabolism. 58:8–15. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Lee HM, Lee OH, Kim KJ and Lee BY: Ginsenoside Rg1 promotes glucose uptake through activated AMPK pathway in insulin-resistant muscle cells. Phytother Res. 26:1017–1022. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Manning BD and Cantley LC: AKT/PKB signaling: Navigating downstream. Cell. 129:1261–1274. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Ge Y and Chen J: Mammalian target of rapamycin (mTOR) signaling network in skeletal myogenesis. J Biol Chem. 287:43928–43935. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Kalluri R and Weinberg RA: The basics of epithelial-mesenchymal transition. J Clin Invest. 119:1420–1428. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Toker A and Yoeli-Lerner M: Akt signaling and cancer: Surviving but not moving on. Cancer Res. 66:3963–3966. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Larue L and Bellacosa A: Epithelial-mesenchymal transition in development and cancer: Role of phosphatidylinositol 3′ kinase/AKT pathways. Oncogene. 24:7443–7454. 2005. View Article : Google Scholar : PubMed/NCBI

50 

Frame S and Cohen P: GSK3 takes centre stage more than 20 years after its discovery. Biochem J. 359:1–16. 2001. View Article : Google Scholar : PubMed/NCBI

51 

Harwood AJ: Regulation of GSK-3: A cellular multiprocessor. Cell. 105:821–824. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Zhou BP, Deng J, Xia W, Xu J, Li YM, Gunduz M and Hung MC: Dual regulation of Snail by GSK-3beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat Cell Biol. 6:931–940. 2004. View Article : Google Scholar : PubMed/NCBI

53 

Peinado H, Olmeda D and Cano A: Snail, Zeb and bHLH factors in tumour progression: An alliance against the epithelial phenotype? Nat Rev Cancer. 7:415–428. 2007. View Article : Google Scholar : PubMed/NCBI

54 

Thiery JP: Epithelial-mesenchymal transitions in cancer onset and progression. Bull Acad Natl Med. 193:1969. 2009.(In French). PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 17 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shi X, Yang J and Wei G: Ginsenoside 20(S)-Rh2 exerts anti-cancer activity through the Akt/GSK3β signaling pathway in human cervical cancer cells. Mol Med Rep 17: 4811-4816, 2018
APA
Shi, X., Yang, J., & Wei, G. (2018). Ginsenoside 20(S)-Rh2 exerts anti-cancer activity through the Akt/GSK3β signaling pathway in human cervical cancer cells. Molecular Medicine Reports, 17, 4811-4816. https://doi.org/10.3892/mmr.2018.8454
MLA
Shi, X., Yang, J., Wei, G."Ginsenoside 20(S)-Rh2 exerts anti-cancer activity through the Akt/GSK3β signaling pathway in human cervical cancer cells". Molecular Medicine Reports 17.3 (2018): 4811-4816.
Chicago
Shi, X., Yang, J., Wei, G."Ginsenoside 20(S)-Rh2 exerts anti-cancer activity through the Akt/GSK3β signaling pathway in human cervical cancer cells". Molecular Medicine Reports 17, no. 3 (2018): 4811-4816. https://doi.org/10.3892/mmr.2018.8454