Open Access

Anti-metastatic activity of fangchinoline in human gastric cancer AGS cells

  • Authors:
    • Zhengrong Chen
    • Tengfei He
    • Kui Zhao
    • Chungen Xing
  • View Affiliations

  • Published online on: December 2, 2016     https://doi.org/10.3892/ol.2016.5457
  • Pages: 655-660
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Fangchinoline (FCL) is an active component isolated from the traditional medicinal plant Stephania tetrandra S. Moore, and has been reported to possess anti‑cancer functions in several types of cancers; however, the effect of FCL on gastric cancer metastasis and its underlying molecular mechanisms remain unknown. The current study aimed to investigate the effect of FCL on the cell migration and invasion of human metastatic gastric cancer AGS cells and its mechanisms. Our study demonstrates that FCL dosage dependently suppressed the adhesion, migration and invasion capacities of human gastric cancer AGS cells without obvious cytotoxic effects. Reverse transcription‑polymerase chain reaction and western blot assays demonstrated that FCL greatly inhibited the expression of matrix metalloproteinase (MMP)‑2 and MMP‑9 at both the mRNA and protein levels, while it significantly increased the expression of tissue inhibitor of metalloproteinase (TIMP) 1 and TIMP2 messenger RNAs. Our results also indicated that FCL repressed the phosphorylation of AKT in gastric cancer AGS cells. In summary, FCL may exert its anti‑metastatic property in human gastric cancer cells in vitro by suppression of MMP‑2 and MMP‑9, increase of TIMP1 and TIMP2 genes, and inhibition of AKT phosphorylation. FCL may be a drug candidate for the treatment of gastric cancer metastasis.

Introduction

Gastric cancer is the fourth most common neoplasia and the second highest cause of mortality associated with cancer worldwide (1). It is reported that gastric cancer is also the third most common type of cancer and the second highest cause of mortality in patients with cancer in China (1). Complete excision of the gastric tumor is the best option of curative treatment (2). In patients diagnosed at advanced stages, although chemotherapy effectively improves the survival quality and time, the median survival time of patients with advanced gastric cancer remains poor (1,2). The metastatic ability of gastric cancer is the leading cause of poor prognosis and high mortality (3). Therefore, cancer metastasis is still a big challenge in the clinic for oncologists.

Accumulating data have demonstrated that degradation of extracellular matrix (ECM) proteins is the antecedent condition of malignant tumor metastasis (1). Matrix metalloproteinases (MMPs) are part of the endopeptidase family and can cleave the majority of components of the ECM, including fibronectin, collagen, elastin, proteoglycan and laminin, thus playing critical roles in the development, progression and metastasis of malignant tumors (3,4). Upregulation of MMPs and altered expression of their tissue inhibitors [known as tissue inhibitors of metalloproteinases (TIMPs)] are associated with the invasiveness and metastasis of cancer cells (4). Among them, MMP-2 and MMP-9 are the main enzymes for the degradation of basement membranes in cancer cells and/or stromal cells (4,5). TIMPs are natural inhibitors of MMPs, and TIMP2 specifically inhibits MMP-2, while TIMP1 inhibits MMP-9 (3).

Fangchinoline (FCL) is a bisbenzylisoquinoline alkaloid in Stephania tetrandra S. Moore (Fen fang ji) (6). It is reported that FCL can inhibit histamine release (7), lower blood pressure as a non-specific calcium channel antagonist (8) and inhibit glutamate release from rat cortical synaptosomes (9). In addition, FCL exerts anti-cancer activities in several types of malignant tumors, including breast cancer (10), prostate carcinoma (11), hepatocellular carcinoma (12) and lung cancer (13). However, the effects of FCL on the metastasis of gastric cancer and its underlying mechanisms remain poorly understood.

In the present study, the anti-metastatic activity of FCL (Fig. 1) in gastric cancer cells and its molecular mechanism of action were explored. Our data revealed that FCL inhibited the phosphorylation of AKT and upregulated TIMP2/1, leading to reduced expression of MMP-2/9 and inhibition of gastric cancer cell invasion in vitro.

Materials and methods

Chemicals and antibodies

FCL (purity >98%) was purchased from Shanghai Standard Technology Co., Ltd. (Shanghai, China). Cell culture materials were purchased from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Specific antibodies against GAPDH (1:1,000, sc-25778), MMP-2 (1:500, sc-53630), MMP-9 (1:500, sc-21733), phosphorylated (p)-AKT (1:500, sc-135650) and AKT (1:500, sc-5298) were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA).

Cell culture

Human gastric cancer AGS cells (Type Culture Collection of the Chinese Academy of Sciences, Shanghai, China) were cultured in high glucose-Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS; both HyClone; GE Healthcare Life Sciences, Chalfont, UK) and 200 mM glutamine at 37°C under a humidified 95% air/5% CO2 mixture (v/v).

Cell viability assay

Cells were seeded at a density of 5×103 cells/well in a 96-well microplate containing DMEM with 10% FBS, and incubated for 24 h. Next, cells were incubated with different concentrations of FCL for 24 h. The proliferation rate was determined with an MTT assay kit (Beyotime Institute of Biotechnology, Shanghai, China). The results were expressed as the absorbance at 570 nm, as previously reported (11).

Cell adhesion assay

After pre-treating the cells with or without FCL (2, 4 and 8 µM) for 24 h, AGS cells were trypsinized and suspended at a final concentration of 2×105 cells/ml in serum-free medium. A total of 100 µl cell suspension was seeded to each well in a 96-well plate coated with 20 µg/ml fibronectin overnight. The cells were incubated for 30 min at 37°C, and the non-adherent cells were removed by washing with PBS. MTT assay was used to determine the number of remaining adherent cells.

Wound healing assay

AGS cells were seeded on a 6-well plate and incubated in serum-free medium for 24 h. A transverse scratch wound on each monolayer of cells was created using a sterilized 200-µl pipette tip. The scratch wounded cell monolayers were then stimulated with various concentrations of FCL (0, 2, 4 and 8 µM) in 5% FBS for an additional 36 h, at which point, the cells that had migrated into the wound were photographed and analyzed.

Transwell chamber invasion assay

Transwell chamber invasion assay was conducted as described by Magee et al (14). In brief, the transwell chambers (EMD Millipore, Billerica, MA, USA) were pre-coated with Matrigel (BD Biosciences, Franklin Lakes, NJ, USA) for 1 h, and the upper chambers were washed using serum-free medium. Following incubation of AGS cells with FCL (0, 2, 4 and 8 µM) for 24 h, the cells were trypsinized and suspended at a density of 2×105 cells/ml in serum-free medium. A total of 200 µl cells were placed in the upper chambers, and medium with 10% FBS was placed in the lower chambers. After 24-h incubation at 37°C, the non-invaded cells were removed using a cotton swab, while the invaded cells were fixed with 100% methanol and stained with hematoxylin and eosin (Nanjing Jiancheng Biotechnology Institute Co., Ltd., Nanjing, China). The invaded cells on the lower surface of the membrane filter were counted under an inverted microscope. The data are presented as the mean number of cells attached to the bottom surface from six randomly selected fields.

RNA extraction and reverse transcription-polymerase chain reaction (RT-PCR)

Total RNA was isolated from AGS cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Synthesis of complementary DNA was performed using the PrimeScript RT Reagent kit (Takara Biotechnology Co., Ltd., Dalian, China), and the PCR conditions were as follows: Initial denaturation at 95°C for 5 min, followed by 24–33 cycles of denaturation at 94°C for 30 sec, annealing at 54°C for 30 sec and extension at 72°C for 45 sec. The primer sequences were as follows: MMP-2 (forward 5′-TGGATGATGCCTTTGCTCGT-3′ and reverse 5′-AAACTTGCAGGGCTGTCCTT-3′); MMP-9 (forward 5′-GGACAAGCTCTTCGGCTTCT-3′ and reverse 5′-TTCAGGGCGAGGACCATAGA-3′); TIMP1 (forward 5′-CTCGTCATCAGGGCCAAGTT-3′ and reverse 5′-GTAGGTCTTGGTGAAGCCCC-3′); TIMP2 (forward 5′-TAGTGATCAGGGCCAAAGCG-3′ and reverse 5′-CAGGCTCTTCTTCTGGGTGG-3′); and GAPDH (forward 5′-GAGAAGGCTGGGGCTCATTT-3′ and reverse 5′-GTCAGGTCCACCACTGACAC-3′). The expression levels of MMP-2, MMP-9, TIMP1 and TIMP2 were normalized to those of GAPDH, which served as an internal control.

Western blot analysis

To analyze the level of protein expression, cell lysates were prepared, as described previously (15), and separated by 10% SDS-PAGE and then transferred to polyvinylidene difluoride membranes for 2 h. The membrane was treated with blocking solution (5% bovine serum albumin in TBS) at room temperature for 1 h, and then incubated overnight at 4°C with antibodies against MMP-2, MMP-9, AKT, p-AKT or GAPDH. The membranes were washed with Tris-buffered saline containing 0.5% Tween-20 (Beyotime Institute of Biotechnology), and then incubated for 1 h at room temperature with an anti-immunoglobulin G secondary antibody conjugated to horseradish peroxidase (1:1,000; A0208; Beyotime Institute of Biotechnology). The expression levels of the proteins were analyzed via chemiluminescence (Beyotime Institute of Biotechnology) and quantified using Quantity One software, version 4.6.2 (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The expression levels of MMP-2, MMP-9, p-AKT and AKT were normalized to those of the internal control GAPDH.

Statistical analysis

The results are expressed as the mean ± standard deviation from ≥3 independent experiments. The statistical differences between the experimental groups were assessed using a Student's t-test in SPSS version 13.0 (SPSS Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

FCL decreases cell proliferation in gastric cancer AGS cells

To determine the inhibitory effect of FCL (Fig. 1A) on gastric cancer cell proliferation, an MTT assay was used. AGS cells were pre-treated with FCL (0–64 µM) for 24 h. As shown in Fig. 1B, gastric cancer cell proliferation was inhibited by FCL in a dose-dependent manner, while FCL at doses <8 µM exhibited little effect on the cell viability of gastric cancer AGS cells (Fig. 1B). Therefore, these doses (0, 2, 4 and 8 µM) were used in the following experiments.

FCL inhibits the attachment of gastric cancer cells to fibronectin

To determine the effect of FCL on cell adhesion to the ECM, an adhesion assay was conducted in gastric cancer AGS cells. As shown in Fig. 2, FCL greatly inhibited cell adhesion ability in a dose-dependent manner, as demonstrated by cell attachment assay. Cell adhesion was inhibited by 41.53±4.85% for the 4-µM dose of FCL and by 85.63±3.51% for 8 µM FCL (Fig. 2).

FCL reduces the migration of gastric cancer cells

Next, the inhibitory effect of FCL on cell migration was assessed in the artificial scratch wound of gastric cancer AGS cells. As shown in Fig. 3A, FCL obviously reduced cell numbers in the scratch wound area in gastric cancer AGS cells in a dose-dependent manner. The inhibitory rates of cell migration were 29.55±5.74, 64.65±4.99 and 76.68±4.82% for 2, 4 and 8 µM FCL, respectively (Fig. 3B).

FCL suppresses cell invasion in gastric cancer cells

A Boyden chamber assay was used to examine whether FCL could suppress the cell invasion of gastric cancer cells. As shown in Fig. 4A, exposure to FCL greatly decreased the invasive ability of gastric cancer cells in a concentration-dependent manner. The inhibitory rates of FCL on gastric cancer cells invasion were 13.47±7.30, 51.93±3.56 and 75.97±4.30% for 2, 4 and 8 µM FCL, respectively (Fig. 4B).

Effects of FCL on the expression of MMP-2, MMP-9, TIMP2 and TIMP1 genes

Accumulating evidence has demonstrated that MMPs, particularly MMP-2/MMP-9, and their inhibitors TIMP2/TIMP1 participated in the invasiveness and metastasis of malignant tumors via stimulating the degradation of ECM and cell migration (3,5). To verify the possible anti-metastatic molecular mechanism of FCL on gastric cancer cells, the expression of MMP-2/MMP-9 and TIMP2/TIMP1 genes was detected in FCL-treated gastric cancer AGS cells. The expression of MMP-2 and MMP-9 messenger (m) RNAs was obviously decreased in a concentration-dependent manner, with a maximum decrease of 31.23±4.16 and 63.83±4.64% for MMP-2 and MMP-9, respectively, following exposure to 8 µM FCL (Fig. 5). In addition, the mRNA levels of their specific inhibitors TIMP2 and TIMP1 were significantly increased in a dose-dependent manner, with a maximum elevated rate of 1.96±0.08-fold and 1.50±0.06-fold for TIMP2 and TIMP1, respectively, upon exposure to 8 µM FCL (Fig. 5). Additionally, the expression levels of MMP-2 and MMP-9 were also confirmed by western blot assay, and the results demonstrated that FCL also significantly inhibited the expression of MMP-2 and MMP-9 proteins in a similar pattern (Fig. 6). The above data suggested that FCL inhibited the invasiveness of gastric cancer AGS cells via downregulation of MMP-2/MMP-9 and upregulation of TIMP2/TIMP1.

FCL reduces the phosphorylation of AKT in gastric cancer cells

Since our data revealed that FCL exhibited inhibitory effects on cell adhesion, migration, invasion and proteinases, the effects of FCL on the expression of the components of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT pathway were explored by western blotting to elucidate the underlying molecular mechanisms. As shown in Fig. 6, the phosphorylation of AKT was significantly reduced in a dose-dependent manner to 79.6±4.2 and 31.8±3.5% upon treatment with 4 and 8 µM FCL, respectively, while the total AKT levels were not significantly affected upon exposure to FCL.

Discussion

As an active compound of Stephania tetrandra S. Moore, FCL has been reported to exhibit anti-cancer effects, which are associated with cell apoptosis promotion, autophagic death induction, cell cycle arrest and multidrug resistance (68); however, the anti-metastatic activity of FCL has not been clarified yet, particularly in gastric cancer. The present study attempted to identify novel functions of FCL, and our data revealed that FCL inhibited cell adhesion, migration and invasion in human gastric cancer AGS cells without obvious cytotoxicity. In addition, the anti-metastatic function of FCL was associated with downregulation of MMP-2/9, upregulation of TIMP2/1 and decreased phosphorylation of AKT.

The invasion and metastasis of malignant tumors are the leading causes of poor prognosis in patients with gastric cancer, and these processes are usually accompanied by the destruction of basement membrane components in the ECM, which is mainly controlled by a number of proteolytic enzymes such as MMPs and their endogenous inhibitors TIMP2/1 (1,5). Immunohistochemistry analysis revealed that expression of MMP-2 and MMP-9 was detected in 94 and 70% of gastric cancer specimens, respectively, and it was associated with poor overall survival in patients with gastric cancer (16,17). Overexpression of MMP-2/9 was observed to accelerate the invasion of tumor cells in vitro and in vivo (18,19), and metastasis in MMP-2 and MMP-9-null mice was greatly suppressed compared with that in wild-type mice, suggesting that the elevated expression of MMP-2/9 promotes the metastatic potential of malignant tumor cells (3,5). Therefore, suppression of cancer migration and invasion mediated by MMP-2/9 provides a new direction for drug research in cancer metastasis. In the current study, exposure of human gastric cancer AGS cells to non-toxic doses of FCL inhibited cell adhesion, migration and invasion, compared with control cells, in a concentration-dependent manner. In addition, FCL was observed to inhibit the expression of MMP-2/9 mRNAs as well as their protein levels, while it increased the expression of TIMP2/1 genes, implying that FCL exhibited an ability to inhibit matrix degradation proteases, thus limiting the adhesive and invasive capabilities of gastric cancer cells.

A considerable number of studies have shown that PI3K/AKT signaling serves a critical role in regulating the expression of MMPs; therefore, suppression of PI3K/AKT signaling may inhibit proliferation, invasion, angiogenesis and metastasis in various malignant tumors (15). In addition, inhibition of PI3K/AKT signaling could result in decreased expression of MMP-2/9 (2022). Considering that FCL at non-toxic doses (0–8 µM) could greatly suppress cell adhesion, migration and invasion via inhibition of MMP-2/9 mRNAs and proteins, the present authors hypothesized whether PI3K/AKT signaling was involved in this process. Our results demonstrated that FCL suppressed the phosphorylation of AKT in gastric cancer cells, suggesting that the inhibition of cell adhesion, migration and invasion by FCL could be mediated by suppression of MMP-2/9 and increase of TIMP2/1, in part, via decreasing the phosphorylation of AKT.

In summary, our current study has shown that FCL prevented the metastatic potential of human gastric cancer AGS cells via inhibition of MMP-2/9 and increase of TIMP2/1 through downregulation of PI3K/AKT signaling, thus highlighting the potential role of FCL in the therapy of gastric cancer metastasis.

Acknowledgements

The present study was supported by a Youth Development Fund from the Health Authorities of Suzhou (Suzhou, China; grant no. 201017).

References

1 

Qiu MZ and Xu RH: The progress of targeted therapy in advanced gastric cancer. Biomark Res. 1:322013. View Article : Google Scholar : PubMed/NCBI

2 

Wong H and Yau T: Targeted therapy in the management of advanced gastric cancer: Are we making progress in the era of personalized medicine. Oncologist. 17:346–358. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Murray GI, Duncan ME, Arbuckle E, Melvin WT and Fothergill JE: Matrix metalloproteinases and their inhibitors in gastric cancer. Gut. 43:791–797. 1998. View Article : Google Scholar : PubMed/NCBI

4 

Pereira AL, Veras SS, Silveira EJ, Seabra FR, Pinto LP, Souza LB and Freitas RA: The role of matrix extracellular proteins and metalloproteinases in head and neck carcinomas: An updated review. Braz J Otorhinolaryngol. 71:81–86. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Zhang X, Wang Y, Yamamoto G and Tachikawa T: Expression of matrix metalloproteinases MMP-2, MMP-9 and their tissue inhibitors TIMP-1 and TIMP-2 in the epithelium and stroma of salivary gland pleomorphic adenomas. Histopathology. 55:250–260. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Desgrouas C, Taudon N, Bun SS, Baghdikian B, Bory S, Parzy D and Ollivier E: Ethnobotany, phytochemistry and pharmacology of Stephania rotunda Lour. J Ethnopharmacol. 154:537–563. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Fang LH, Zhang YH and Ku BS: Fangchinoline inhibited the antinociceptive effect of morphine in mice. Phytomedicine. 12:183–188. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Kim HS, Zhang YH, Oh KW and Ahn HY: Vasodilating and hypotensive effects of fangchinoline and tetrandrine on the rat aorta and the stroke-prone spontaneously hypertensive rat. J Ethnopharmacol. 58:117–123. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Lin TY, Lu CW, Tien LT, Chuang SH, Wang YR, Chang WH and Wang SJ: Fangchinoline inhibits glutamate release from rat cerebral cortex nerve terminals (synaptosomes). Neurochem Int. 54:506–512. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Xing Z, Zhang Y, Zhang X, Yang Y, Ma Y and Pang D: Fangchinoline induces G1 arrest in breast cancer cells through cell-cycle regulation. Phytother Res. 27:1790–1794. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Wang CD, Huang JG, Gao X, Li Y, Zhou SY, Yan X, Zou A, Chang JL, Wang YS, Yang GX and He GY: Fangchinoline induced G1/S arrest by modulating expression of p27, PCNA, and cyclin D in human prostate carcinoma cancer PC3 cells and tumor xenograft. Biosci Biotechnol Biochem. 74:488–493. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Wang N, Pan W, Zhu M, Zhang M, Hao X, Liang G and Feng Y: Fangchinoline induces autophagic cell death via p53/sestrin2/AMPK signalling in human hepatocellular carcinoma cells. Br J Pharmacol. 164:731–742. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Guo B, Su J, Zhang T, Wang K and Li X: Fangchinoline as a kinase inhibitor targets FAK and suppresses FAK-mediated signaling pathway in A549. J Drug Target. 23:266–274. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Magee PJ, Allsopp P, Samaletdin A and Rowland IR: Daidzein, R-(+)equol and S-(−)equol inhibit the invasion of MDA-MB-231 breast cancer cells potentially via the down-regulation of matrix metalloproteinase-2. Eur J Nutr. 53:345–350. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Chen PN, Hsieh YS, Chiou HL and Chu SC: Silibinin inhibits cell invasion through inactivation of both PI3K-Akt and MAPK signaling pathways. Chem Biol Interact. 156:141–150. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Sampieri CL, León-Córdoba K and Remes-Troche JM: Matrix metalloproteinases and their tissue inhibitors in gastric cancer as molecular markers. J Cancer Res Ther. 9:356–363. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Sier CF, Kubben FJ, Ganesh S, Heerding MM, Griffioen G, Hanemaaijer R, van Krieken JH, Lamers CB and Verspaget HW: Tissue levels of matrix metalloproteinases MMP-2 and MMP-9 are related to the overall survival of patients with gastric carcinoma. Br J Cancer. 74:413–417. 1996. View Article : Google Scholar : PubMed/NCBI

18 

Beliën AT, Paganetti PA and Schwab ME: Membrane-type 1 matrix metalloprotease (MT1-MMP) enables invasive migration of glioma cells in central nervous system white matter. J Cell Biol. 144:373–384. 1999. View Article : Google Scholar : PubMed/NCBI

19 

Deryugina EI, Luo GX, Reisfeld RA, Bourdon MA and Strongin A: Tumor cell invasion through matrigel is regulated by activated matrix metalloproteinase-2. Anticancer Res. 17:3201–3210. 1997.PubMed/NCBI

20 

Chen HJ, Lin CM, Lee CY, Shih NC, Amagaya S, Lin YC and Yang JS: Phenethyl isothiocyanate suppresses EGF-stimulated SAS human oral squamous carcinoma cell invasion by targeting EGF receptor signaling. Int J Oncol. 43:629–637. 2013.PubMed/NCBI

21 

Lu KH, Yang HW, Su CW, Lue KH, Yang SF and Hsieh YS: Phyllanthus urinaria suppresses human osteosarcoma cell invasion and migration by transcriptionally inhibiting u-PA via ERK and Akt signaling pathways. Food Chem Toxicol. 52:193–199. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Yang SF, Chen MK, Hsieh YS, Yang JS, Zavras AI, Hsieh YH, Su SC, Kao TY, Chen PN and Chu SC: Antimetastatic effects of Terminalia catappa L. on oral cancer via a down-regulation of metastasis-associated proteases. Food Chem Toxicol. 48:1052–1058. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2017
Volume 13 Issue 2

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
Chen Z, He T, Zhao K and Xing C: Anti-metastatic activity of fangchinoline in human gastric cancer AGS cells. Oncol Lett 13: 655-660, 2017
APA
Chen, Z., He, T., Zhao, K., & Xing, C. (2017). Anti-metastatic activity of fangchinoline in human gastric cancer AGS cells. Oncology Letters, 13, 655-660. https://doi.org/10.3892/ol.2016.5457
MLA
Chen, Z., He, T., Zhao, K., Xing, C."Anti-metastatic activity of fangchinoline in human gastric cancer AGS cells". Oncology Letters 13.2 (2017): 655-660.
Chicago
Chen, Z., He, T., Zhao, K., Xing, C."Anti-metastatic activity of fangchinoline in human gastric cancer AGS cells". Oncology Letters 13, no. 2 (2017): 655-660. https://doi.org/10.3892/ol.2016.5457