Inhibition of multiple myeloma cell proliferation by ginsenoside Rg3 via reduction in the secretion of IGF-1

  • Authors:
    • Yan Li
    • Tao Yang
    • Jing Li
    • Hong‑Ling Hao
    • Su‑Yun Wang
    • Jie Yang
    • Jian‑Min Luo
  • View Affiliations

  • Published online on: July 7, 2016     https://doi.org/10.3892/mmr.2016.5475
  • Pages: 2222-2230
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ginsenoside Rg3 (Rg3) is one of the primary constituents isolated from ginseng, and has been found to exhibit cytotoxic effects against cancer cells. The present study aimed to investigate the effects of Rg3 on human multiple myeloma cell proliferation and apoptosis, and to examine its underlying molecular mechanisms. Cell viability was detected using a Cell Counting kit‑8 assay, and cell cycle arrest and cell apoptosis were analyzed using flow cytometry. In addition, the expression levels of cell cycle‑associated markers and apoptosis‑associated proteins, and the release of cytochrome C were determined using western blot analysis. The effects of Rg3 on the insulin‑like growth factor (IGF)-1/AKT/mammalian target of rapamycin (mTOR) and mitogen-activated protein kinase signaling pathways were also investigated using western blot analysis. The results showed that Rg3 inhibited cell viability in U266, RPMI8226 and SKO‑007 cells in a time‑ and dose‑dependent manner, and caused cell cycle arrest in the G1 phase by regulating the cyclin‑dependent kinase pathway. Furthermore, Rg3 induced multiple myeloma cell apoptosis, and was involved in B cell lymphoma-2 (Bcl2)/Bcl2-associated X protein imbalance, caspase activation and the release of cytochrome C from the mitochondria into the cytoplasm. Mechanistically, it was found that the inhibitory effects of Rg3 on multiple myeloma cell proliferation were essential for secretion of IGF‑1 and inactivation of the Akt/mTOR pathway. Collectively, these findings demonstrated that Rg3 effectively inhibited cell proliferation and induced apoptosis of multiple myeloma cells. These data broaden the clinical investigation of Rg3 in the treatment of multiple myeloma, associated with the inactivation of IGF-1/AKT/mTOR signaling.

Introduction

Multiple myeloma is a clonal malignancy of plasma cells characterized by bone destruction, monoclonal proteins, hypercalcemia, excess bone marrow plasma, renal damage and immunodeficiency (1,2). The incidence of multiple myeloma varies globally, from 1/100,00 individuals in China to ~4/100,000 individuals in developed countries (3). Patients with multiple myeloma will often develop recurrence or an increased susceptibility to fungal, viral and bacterial infections, which are the major cause of multiple myeloma-associated mortality (4,5). The survival rates of patients with multiple myeloma can now exceed 10 years as a result of therapy comprising hematopoietic stem cell transplantation in combination with novel chemotherapeutic agents, including thalidomide, lenalidomide and bortezomib (68). However, chemotherapy often produces drug resistance and high levels of toxicity, therefore, developing a more effective agent remains a priority in the treatment of multiple myeloma.

Over previous decades, several natural products derived from plants have shown promising structures for the development of novel agents for use in cancer treatment. Ginsenosides, a traditional Chinese medicine, have been reported to exhibit antitumor properties (9,10). Ginsenoside Rg3 (Rg3), a monomer derived from heat-processed ginseng, has been found to have potent antitumor effects (11,12). Although Rg3 has been reported to inhibit cancer cell proliferation and induce cell death in melanoma (13), breast cancer (14), acute leukemia (15), glioma (16) and hepatocellular carcinoma (17), the activity of Rg3 against cell growth in multiple myeloma and its functional targets remain to be fully elucidated.

The objectives of the present study were to investigate the activity of Rg3 in inhibiting the growth of multiple myeloma cell lines, and to elucidate the underlying mechanisms. The results demonstrated the antiproliferative effect of Rg3 against multiple myeloma cells. In addition, the mechanism underlying the action of Rg3 was correlated with the inhibition of secretion of insulin-like growth factor (IGF)-1 and inactivation of the AKT/mammalian target of rapamycin (mTOR) signaling pathway. The present study indicates that Rg3 may be a potential clinical therapeutic agent for multiple myeloma.

Materials and methods

Materials and reagents

Ginsenoside Rg3 (Rg3) was purchased from Yatai Pharmaceuticals Co., Ltd (Jilin, China) with 98% purity, assayed using high-performance liquid chromatography (HPLC; Fig. 1A). The powder was dissolved in dimethyl sulfoxide (DMSO) in a stock concentration of 100 mg/ml. The final concentration of DMSO in the culture medium was ≤0.1%. Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), penicillin, streptomycin and phosphate-buffered saline (PBS) were purchased from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Antibodies obtained from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA) were as follows, all used at 1:500 dilution: Mouse anti-cyclin D1 monoclonal antibody (cat. no. sc-8396), mouse anti-p27 monoclonal antibody (cat. no. sc-393380), mouse anti-phosphorylated (phospho)-extracellular signal regulated kinase (Erk)1/2 monoclonal antibody (cat. no. sc-7383), rabbit anti-Erk1/2 polyclonal antibody (cat. no. sc-292838), mouse anti-phospho-c-Jun N-terminal kinase (JNK) monoclonal antibody (cat. no. sc-6254), mouse anti-JNK monoclonal antibody (cat. no. sc-7345), rabbit anti-phophos-p38 polyclonal antibody (cat. no. sc-17852-R), mouse anti-p38 monoclonal antibody (cat. no. sc-81621) and rabbit anti-IGF-1 polyclonal antibody (cat. no. sc-9013). Antibodies obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA) were as follows, all used at 1:1,000 dilution: Mouse anti-B cell lymphoma-2 (Bcl-2) monoclonal antibody (cat. no. 15071), rabbit anti-Bcl-2-associated X protein (Bax) polyclonal antibody (cat. no. 5023), mouse anti-cytochrome C monoclonal antibody (cat. no. 12963), rabbit anti-cytochrome c oxidase (Cox) IV polyclonal antibody (cat. no. 4850), mouse anti-caspase-9 monoclonal antibody (cat. no. 9508), mouse anti-caspase-8 monoclonal antibody (cat. no. 9746), rabbit anti-caspase-3 polyclonal antibody (cat. no. 9665), rabbit anti-phospho-AKT polyclonal antibody (cat. no. 5012), mouse anti-AKT monoclonal antibody (cat. no. 2920), rabbit anti-phospho-mTOR polyclonal antibody (cat. no. 2976) and mouse anti-mTOR monoclonal antibody (cat. no. 2983). IGF-1, rabbit polyclonal anti-retinoblastoma (Rb; cat. no. SAB4502589), mouse monoclonal anti-phospho-Rb (cat. no. R6878) and rabbit polyclonal anti-GAPDH (cat. no. G8795) were purchased from Sigma-Aldrich (St. Louis, MO, USA) and used at 1:1,000 dilution. The Cell Counting kit-8 assay (CCK-8), radioimmunoprecipitation assay (RIPA) lysis buffer, bicinchoninic acid (BCA) kit, enhanced chemiluminescence (ECL) system and Fluorescein isothiocyanate (FITC)-Annexin V Apoptosis Detection kit were obtained from Beyotime Institute of Biotechnology (Jiangsu, China).

Cell culture

The U266, RPMI8226 and SKO-007 human multiple myeloma cell lines, were obtained from America Type Culture Collection (Rockville, MD, USA) and maintained in DMEM supplemented with 10% FBS, 100 U/ml penicillin and 100 U/ml streptomycin at 37°C in a 5% CO2 atmosphere.

Cell viability assay

Multiple myeloma cells were seeded in 96-well plates at a density of 5×103 cells/well overnight and then treated with Rg3 at different concentrations (0, 20, 40, 60, 80 and 100 µg/ml), and for different durations (6, 12, 24 and 48 h), as indicated. Subsequently, fresh medium containing 10 µl CCK-8 reagent was added, followed by incubation at 37°C for 2 h. The absorbance of each well was measured at 450 nm on a plate reader (Bio-Tek Instruments, Inc., Winooski, VT, USA).

Western blot analysis

The cells were washed with cold PBS and lysed with lysis buffer. The protein concentration was determined using the BCA kit. Equal quantities (40 µg) of protein were separated on 12% SDS-PAGE gels (GenScript, Piscataway, NJ, USA) and then transferred onto nitrocellulose membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked with 5% non-fat milk for 1 h, and then probed with appropriate primary antibodies overnight at 4°C, followed by blotting with horseradish peroxidase (HRP)-labeled goat anti-mouse IgG (cat. no. A0216) or HRP-labeled goat anti-rabbit IgG (cat. no. A0208) secondary antibodies (1:1,000; Beyotime Institute of Biotechnology) at room temperature for 1 h. The target bands were detected using the ECL system, and the band intensity was determined using ImageJ software (version 1.41; NIH, Bethesda, MD, USA).

Cell cycle analysis

The cells were harvested by centrifugation and processed for cell cycle analysis, as described previously (3). Briefly, the cells were digested with 0.25 g/l trypsin and harvested by centrifugation at 1,000 × g for 10 min at room temperature. Following incubation with 70% ethanol at −20°C for 15 min, the cells were stained with 20 mg/ml propidium iodide (PI) and incubated for 30 min at room temperature. The cells were analyzed for DNA content using FACScalibur flow cytometry (BD Biosciences, San Jose, CA, USA). The percentages of cells containing different DNA contents were quantified using CellQuest software (version 5.1; BD Biosciences).

Apoptosis detection

The cellular apoptotic ratios were detected with the FITC-Annexin V Apoptosis Detection kit using flow cytometry. Briefly, the cells were trypsinized and harvested by centrifugation. The cell pellets were re-suspended in a binding buffer of Annexin V-FITC and PI at room temperature in the dark for 15 min. The apoptotic cells were counted using flow cytometry (BD Biosciences), with the percentage of apoptotic cells expressed as the FITC/PI ratio.

Isolation of mitochondria

The isolation of mitochondrial and cytoplasmic proteins was performed using a Mitochondria Isolation kit (Thermo Fisher Scientific Inc.), according to the manufacturer's protocol. The cytosolic and mitochondrial fractions were analyzed using western blot analysis. Cox IV was used as an internal control for the mitochondrial fraction.

ELISA assay

The concentrations of IGF-1 were determined using a Human IGF-I Quantikine ELISA kit (R&D Systems, Inc., Minneapolis, MN, USA), according to the manufacturer's protocol. The absorbance was measured at a wavelength of 450 nm using a microplate reader (Bio-Tek Instruments, Inc.).

Statistical analysis

All data are presented as the mean ± standard error of the mean, and the n value indicates the number of independent experiments. Data were statistically analyzed using Student's t-test on GraphPad Prism 5.0 software (GraphPad Software, Inc., La Jolla CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of Rg3 on the viability of multiple myeloma cell lines

The uncontrolled cell proliferation of cancer cells is vital in the progression of cancer, therefore, the present study evaluated the effects of Rg3 on the viability of multiple myeloma cells. As shown in Fig. 1B, treatment with Rg3 inhibited the viability of the U266 cells in a time- and dose-dependent manner. The inhibition of cell viability in the U266 cells following treatment with Rg3 for 48 h reached the maximal level at 80 µg/ml, and the half maximal inhibitory concentration (IC50) value was 47.5 mg/l. In addition to U266, two other multiple myeloma cell lines, RPMI8226 and SKO-007, were also included in the present study to examine these effects. Rg3 had a more potent effect in inhibiting the viability of the RPMI8226 (IC50=36.8 µg/ml) and SKO-007 (IC50=31.5 µg/ml) cells, compared with the U266 cells (Fig. 1C and D).

Rg3 arrests the cell cycle of multiple myeloma cells

Flow cytometric cell cycle analysis was performed using PI staining to determine the effect of Rg3 on cell cycle progression. Treatment of the U266 cells with 20, 40 and 80 µg/ml Rg3 increased the percentage of the cell population in the G1 phase and decreased the percentage in the S phase. However, Rg3 had no effect on the G2/M phase (Fig. 2A and B). The effect of Rg3 on cell cycle progression was also examined in the SKO-007 cells (Fig. 2C and D). Similarly, the cell cycle of the SKO-007 cells was arrested in the G1 phase by Rg3, indicating that Rg3 may inhibit multiple myeloma cell proliferation through suppressing the cell cycle transition from the G1 to the S phase. The cell cycle transition between the G1 and S phase is strictly regulated by the balance of cyclins and cyclin-dependent kinase inhibitors (18,19). Therefore, to further investigate the mechanisms underlying how Rg3 arrested G1/S transition, the expression levels of proteins regulating cell cycle progression in U266 cells, including cyclin D1, p27 and phospho-Rb, were determined. The results of the western blot analysis showed that Rg3 decreased the protein expression of cyclin D1 and phosphorylation of Rb, and increased the expression of p27 in a dose-dependent manner (Fig. 2E and F).

Rg3 induces multiple myeloma cells apoptosis via the mitochondria-dependent pathway

To examine the effect of Rg3 on cell survival, its effects on cell apoptosis were determined. Annexin V-FITC/PI staining followed by flow cytometric analysis revealed that treatment of the U266 cells with 20, 40 and 80 µg/ml Rg3 for 48 h significantly increased the percentage of apoptotic cells in the in U266 cell population (Fig. 3A and B). Similar results were observed in the RPMI8226 and SKO-007 cells (data not shown). Bcl-2 and Bax are anti-apoptotic and pro-apoptotic proteins, and the ratio of Bcl-2/Bax appears to be a determinant of cell survival and death. To understand the mechanism by which Rg3 induces cell apoptosis, the expression levels of Bcl-2 and Bax, and the ratio of Bcl-2 to Bax were measured. The results of the western blot analysis showed that Rg3 treatment markedly decreased the protein expression of Bcl-2 and increased the protein expression of Bax in the U266 cells, resulting in a further decrease in the Bcl-2/Bax ratio (Fig. 3C and D). The mitochondria-dependent signaling pathway is critical for cell apoptosis. The release of cytochrome C from the mitochondria into the cytoplasm triggers the downstream apoptotic signal, consequently resulting in cell apoptosis (20). Therefore, the present study examined the release of cytochrome C. As expected, Rg3 treatment caused a significant decrease in the protein expression of cytochrome C in the mitochondria, and an increase in the cytoplasm (Fig. 3E and F). Cytochrome C release sequentially activates downstream apoptosis-associated proteins, including caspases. Western blot analysis demonstrated that Rg3 increased the protein expression levels of cleaved caspase-9, caspase-8 and caspase-3 (Fig. 3G and H). Collectively, these data suggested that mitochondrial dysfunction may underlie, at least partially, the enhanced effect of Rg3 on myeloma cell apoptosis.

AKT/mTOR, but not MAP kinase, signaling is involved in the action of Rg3 on multiple myeloma cell proliferation and survival

The phosphorylation of AKT and downstream mTOR has been reported to be involved in the progression of several types of malignancy, including multiple myeloma (3,21). To clarify the signal transduction pathways by which Rg3 exerts its antitumor effects, the present study first examined the activation of the AKT/mTOR pathway. As shown in Fig. 4A and B, the phosphorylation of AKT and its downstream protein, mTOR, was attenuated by Rg3 treatment in a dose-dependent manner. MAP kinase signaling is also a regulator of cell cycle progression and tumorigenesis (22,23). However, Rg3 treatment did not alter the phosphorylation of Erk1/2, JNK or p38 (Fig. 4C and D). These data excluded the possibility that MAP kinase signaling was involved in the effects of Rg3 on multiple myeloma cell proliferation and survival.

Rg3 attenuates AKT/mTOR activation via inhibiting the secretion of IGF-1

IGF-1 is an important pathway of AKT/mTOR (24). Therefore, the present study investigated whether Rg3 affects this pathway. The results of the western blot analysis showed that Rg3 had no effect on the protein expression of IGF-1 (Fig. 5A). However, the secretion of IGF-1 was markedly decreased following Rg3 treatment (Fig. 5B). In addition, treatment with IGF-1 reversed the Rg3-induced inactivation of the AKT/mTOR pathway, as evidenced by the significant restoration in AKT and mTOR phosphorylation (Fig. 5C and D). The present study further examined whether IGF-1 was involved in the effects of Rg3 on cell proliferation and survival. The results of the CCK-8 assay revealed that the reduction in cell viability induced following Rg3 treatment was gradually inhibited by IGF-1 in a dose-dependent manner (Fig. 5E). As expected, Rg3-induced cell apoptosis was almost eliminated by the addition of IGF-1 (Fig. 5F).

Discussion

Several experimental and clinical studies have been performed on the effects of Rg3 on cancer. These studies have identified Rg3 as being effective in the treatment of certain types of cancer. Shen Yi capsule has been approved by the China Food and Drug Administration, with Rg3 as the active pharmaceutical ingredient, for clinical use in cancer treatment (25). However, the specific functions of Rg3 in the treatment of multiple myeloma remain to be fully elucidated. The present study provided evidence of the anti-multiple myeloma activity of Rg3 in cultured cells. The data revealed that Rg3 inhibited the proliferation of the U266, RPMI8226 and SKO-007 human multiple myeloma cell lines, and induced cellular apoptosis, which were consistent with previous studies (26,27).

The data obtained in the present study revealed that the anti-proliferative effect of Rg3 in multiple myeloma cells was due to inhibition of the cell cycle transition between the G1 and S phases. Cell cycle progression is strictly regulated by cyclin-dependent kinases 4 and 6, which are activated by cyclin D1, but attenuated by p27 (28). In addition, the activation of cyclin-dependent kinases 4 and 6 can lead to the phosphorylation of Rb and consequently promote the transition between the G1 and S phase (3). In the present study, it was found that the expression of cyclin D1 and the phosphorylation of Rb were attenuated by Rg3, whereas the expression of p27 was elevated. The apoptotic process is usually associated with the imbalance between the levels of Bcl-2 and Bax (29). During apoptotic stimulation, the increased expression of Bax enhances membrane permeability, which results in the release of cytochrome C from the mitochondria into cytoplasm, and activates a family of proteases, including caspase-9, caspase-8 and caspase-3, driving the cell toward apoptosis (30,31). The present study showed that Rg3 treatment induced the release of mitochondrial cytochrome C into the cytoplasm, increased the protein expression levels of cleaved caspase-9, caspase-8 and caspase-3, and decreased the Bcl-2/Bax ratio. These data indicated that Rg3-induced apoptosis in multiple myeloma cells was mitochondria-dependent.

A previous study demonstrated that Rg3 induced U266 human multiple myeloma cell apoptosis through activation of the Bax protein (26). In addition, Song et al (27) reported that inhibition of the secretion of vascular endothelial growth factor may contribute to the anti-proliferative effects of Rg3. However, apart from these reports, there is no more information regarding the mechanisms underlying the functions of Rg3 in inhibiting the growth of multiple myeloma cells. Deregulation of the Akt/mTOR and MAPK pathway is a common event in human cancer, and is crucial for tumor cell proliferation, cell cycle transition and apoptosis (32,33). Notably, the results of the present study provided the first evidence, to the best of our knowledge, that Rg3 affected cell proliferation and survival, predominantly via Akt/mTOR pathway, and less via the MAPK pathway. The detailed mechanisms to explain why Rg3 is linked less with MAPK and more with Akt/mTOR remain to be elucidated, and further investigations are required. In addition, the abnormal secretion of IGF-1 has been documented to be a tumorigenic factor (21). In the present study, it was found that Rg3 inhibited the secretion of IGF-1, but did not alter its expression. IFG-1 is essential for the inhibitory effect of Rg3 on activation of the AKT/mTOR pathway, suggesting that Rg3 mediated cell proliferation and survival through IGF-1/AKT/mTOR signaling.

In conclusion, the present study demonstrated that Rg3 acted on the IGF-1/AKT/mTOR signal transduction pathway to inhibit multiple myeloma cell proliferation. These results provided evidence to support further investigation for the development of Rg3 as a clinical drug candidate in the treatment of multiple myeloma.

References

1 

Raab MS, Podar K, Breitkreutz I, Richardson PG and Anderson KC: Multiple myeloma. Lancet. 374:324–339. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Di Martino MT, Gullà A, Cantafio ME, Lionetti M, Leone E, Amodio N, Guzzi PH, Foresta U, Conforti F, Cannataro M, et al: In vitro and in vivo anti-tumor activity of miR-221/222 inhibitors in multiple myeloma. Oncotarget. 4:242–255. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Huang EW, Xue SJ, Li XY, Xu SW, Cheng JD, Zheng JX, Shi H, Lv GL, Li ZG, Li Y, et al: EEN regulates the proliferation and survival of multiple myeloma cells by potentiating IGF-1 secretion. Biochem Biophys Res Commun. 447:271–277. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Kumar S and Rajkumar SV: Thalidomide and lenalidomide in the treatment of multiple myeloma. Eur J Cancer. 42:1612–1622. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Kyle RA and Rajkumar SV: Multiple myeloma. Blood. 111:2962–2972. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Richardson PG, Mitsiades C, Schlossman R, Ghobrial I, Hideshima T, Munshi N and Anderson KC: Bortezomib in the front-line treatment of multiple myeloma. Expert Rev Anticancer Ther. 8:1053–1072. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Hideshima T, Chauhan D, Shima Y, Raje N, Davies FE, Tai YT, Treon SP, Lin B, Schlossman RL, Richardson P, et al: Thalidomide and its analogs overcome drug resistance of human multiple myeloma cells to conventional therapy. Blood. 96:2943–2950. 2000.PubMed/NCBI

8 

Brown RE, Stern S, Dhanasiri S and Schey S: Lenalidomide for multiple myeloma: Cost-effectiveness in patients with one prior therapy in England and Wales. Eur J Health Econ. 14:507–514. 2013. View Article : Google Scholar

9 

Lee JY, Jung KH, Morgan MJ, Kang YR, Lee HS, Koo GB, Hong SS, Kwon SW and Kim YS: Sensitization of TRAIL-induced cell death by 20(S)-ginsenoside Rg3 via CHOP-mediated DR5 upregulation in human hepatocellular carcinoma cells. Mol Cancer Ther. 12:274–285. 2013. View Article : Google Scholar

10 

Park EH, Kim YJ, Yamabe N, Park SH, Kim HK, Jang HJ, Kim JH, Cheon GJ, Ham J and Kang KS: Stereospecific anticancer effects of ginsenoside Rg3 epimers isolated from heat-processed American ginseng on human gastric cancer cell. J Ginseng Res. 38:22–27. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Yuan HD, Quan HY, Zhang Y, Kim SH and Chung SH: 20(S)-Ginsenoside Rg3-induced apoptosis in HT-29 colon cancer cells is associated with AMPK signaling pathway. Mol Med Rep. 3:825–831. 2010.

12 

Min JK, Kim JH, Cho YL, Maeng YS, Lee SJ, Pyun BJ, Kim YM, Park JH and Kwon YG: 20(S)-Ginsenoside Rg3 prevents endothelial cell apoptosis via inhibition of a mitochondrial caspase pathway. Biochem Biophys Res Commun. 349:987–994. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Shan X, Fu YS, Aziz F, Wang XQ, Yan Q and Liu JW: Ginsenoside Rg3 inhibits melanoma cell proliferation through down-regulation of histone deacetylase 3 (HDAC3) and increase of p53 acetylation. PLoS One. 9:e1154012014. View Article : Google Scholar : PubMed/NCBI

14 

Kim BM, Kim DH, Park JH, Surh YJ and Na HK: Ginsenoside Rg3 inhibits constitutive activation of NF-kB signaling in human breast cancer (MDA-MB-231) cells: ERK and Akt as potential upstream targets. J Cancer Prev. 19:23–30. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Zeng D, Wang J, Kong P, Chang C and Li J and Li J: Ginsenoside Rg3 inhibits HIF-1α and VEGF expression in patient with acute leukemia via inhibiting the activation of PI3K/Akt and ERK1/2 pathways. Int J Clin Exp Pathol. 7:2172–2178. 2014.

16 

Sin S, Kim SY and Kim SS: Chronic treatment with ginsenoside Rg3 induces Akt-dependent senescence in human glioma cells. Int J Oncol. 41:1669–1674. 2012.PubMed/NCBI

17 

Jiang JW, Chen XM, Chen XH and Zheng SS: Ginsenoside Rg3 inhibit hepatocellular carcinoma growth via intrinsic apoptotic pathway. World J Gastroenterol. 17:3605–3613. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Sedding DG, Hermsen J, Seay U, Eickelberg O, Kummer W, Schwencke C, Strasser RH, Tillmanns H and Braun-Dullaeus RC: Caveolin-1 facilitates mechanosensitive protein kinase B (Akt) signaling in vitro and in vivo. Circ Res. 96:635–642. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Héron-Milhavet L, Franckhauser C, Rana V, Berthenet C, Fisher D, Hemmings BA, Fernandez A and Lamb NJ: Only Akt1 is required for proliferation, while Akt2 promotes cell cycle exit through p21 binding. Mol Cell Biol. 26:8267–8280. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Kuwabara I, Kuwabara Y, Yang RY, Schuler M, Green DR, Zuraw BL, Hsu DK and Liu FT: Galectin-7 (PIG1) exhibits pro-apoptotic function through JNK activation and mitochondrial cytochrome c release. J Biol Chem. 277:3487–3497. 2002. View Article : Google Scholar

21 

Cea M, Cagnetta A, Fulciniti M, Tai YT, Hideshima T, Chauhan D, Roccaro A, Sacco A, Calimeri T, Cottini F, et al: Targeting NAD+ salvage pathway induces autophagy in multiple myeloma cells via mTORC1 and extracellular signal-regulated kinase (ERK1/2) inhibition. Blood. 120:3519–3529. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Su F, Viros A, Milagre C, Trunzer K, Bollag G, Spleiss O, Reis-Filho JS, Kong X, Koya RC, Flaherty KT, et al: RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors. N Engl J Med. 366:207–215. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Wajapeyee N, Serra RW, Zhu X, Mahalingam M and Green MR: Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell. 132:363–374. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Palazzolo I, Stack C, Kong L, Musaro A, Adachi H, Katsuno M, Sobue G, Taylor JP, Sumner CJ, Fischbeck KH and Pennuto M: Overexpression of IGF-1 in muscle attenuates disease in a mouse model of spinal and bulbar muscular atrophy. Neuron. 63:316–328. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Sun Y, Lin H, Zhu Y, Feng J, Chen Z, Li G, Zhang X, Zhang Z, Tang J, Shi M, et al: A randomized, prospective, multi-centre clinical trial of NP regimen (vinorelbine+cisplatin) plus Gensing Rg3 in the treatment of advanced non-small cell lung cancer patients. Zhongguo Fei Ai Za Zhi. 9:254–258. 2006.In Chinese. PubMed/NCBI

26 

Luo Y, Zhang P, Zeng HQ, Lou SF and Wang DX: Ginsenoside Rg3 induces apoptosis in human multiple myeloma cells via the activation of Bcl-2-associated X protein. Mol Med Rep. 12:3557–3562. 2015.PubMed/NCBI

27 

Song Y, Hou J, Kang L and Gao S: Effect of 20 (S)-ginsenoside Rg3 on the proliferation inhibition and secretion of vascular endothelial growth factor of multiple myeloma cell line U266. Zhonghua Xue Ye Xue Za Zhi. 35:519–523. 2014.In Chinese. PubMed/NCBI

28 

Woo RA and Poon RY: Cyclin-dependent kinases and S phase control in mammalian cells. Cell Cycle. 2:316–324. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Misiti F, Orsini F, Clementi ME, Lattanzi W, Giardina B and Michetti F: Mitochondrial oxygen consumption inhibition importance for TMT-dependent cell death in undifferentiated PC12 cells. Neurochem Int. 52:1092–1099. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Ott M, Gogvadze V, Orrenius S and Zhivotovsky B: Mitochondria, oxidative stress and cell death. Apoptosis. 12:913–922. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Mignotte B and Vayssiere JL: Mitochondria and apoptosis. Eur J Biochem. 252:1–15. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Sharp ZD and Bartke A: Evidence for down-regulation of phosphoinositide 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR)-dependent translation regulatory signaling pathways in Ames dwarf mice. J Gerontol A Biol Sci Med Sci. 60:293–300. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Morgensztern D and McLeod HL: PI3K/Akt/mTOR pathway as a target for cancer therapy. Anticancer Drugs. 16:797–803. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 14 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
Li Y, Yang T, Li J, Hao HL, Wang SY, Yang J and Luo JM: Inhibition of multiple myeloma cell proliferation by ginsenoside Rg3 via reduction in the secretion of IGF-1. Mol Med Rep 14: 2222-2230, 2016
APA
Li, Y., Yang, T., Li, J., Hao, H., Wang, S., Yang, J., & Luo, J. (2016). Inhibition of multiple myeloma cell proliferation by ginsenoside Rg3 via reduction in the secretion of IGF-1. Molecular Medicine Reports, 14, 2222-2230. https://doi.org/10.3892/mmr.2016.5475
MLA
Li, Y., Yang, T., Li, J., Hao, H., Wang, S., Yang, J., Luo, J."Inhibition of multiple myeloma cell proliferation by ginsenoside Rg3 via reduction in the secretion of IGF-1". Molecular Medicine Reports 14.3 (2016): 2222-2230.
Chicago
Li, Y., Yang, T., Li, J., Hao, H., Wang, S., Yang, J., Luo, J."Inhibition of multiple myeloma cell proliferation by ginsenoside Rg3 via reduction in the secretion of IGF-1". Molecular Medicine Reports 14, no. 3 (2016): 2222-2230. https://doi.org/10.3892/mmr.2016.5475