Sinomenine inhibits proliferation of SGC-7901 gastric adenocarcinoma cells via suppression of cyclooxygenase-2 expression

  • Authors:
    • Yifei Lv
    • Changshun Li
    • Shuang Li
    • Zhiming Hao
  • View Affiliations

  • Published online on: May 13, 2011     https://doi.org/10.3892/ol.2011.305
  • Pages: 741-745
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sinomenine (SIN) is a bioactive alkaloid extracted from the Chinese medicinal plant Sinomenium acutum. Results of studies have shown that the anti-inflammatory, immunosuppressive and anti-arthritic effects of SIN are partially attributed to the inhibition of cyclooxygenase-2 (COX-2) expression. COX-2 overexpression is associated with enhanced proliferation and angiogenesis of gastric cancer (GC). SGC-7901 cells were treated with different concentrations of SIN in order to observe its effect on the proliferation of human gastric adenocarcinoma cells and to explore the potential underlying molecular mechanism via the detection of COX-2 expression. Celecoxib was used as the positive control. Morphological alterations of the cells were observed microscopically. Cell proliferation was evaluated using MTT assay. COX-2 expression was detected using semi-quantitative RT-PCR and Western blotting. The results showed that SIN inhibited the proliferation of SGC-7901 cells in a time- and dose-dependent manner. In the presence of SIN or celecoxib, SGC-7901 cells became round and detached morphologically, indicating cell apoptosis. The expression of COX-2 was inhibi-ted by SIN in a dose-dependent manner at both the mRNA and protein levels. Our findings indicate that the protective effects of SIN are mediated through the inhibition of COX-2 expression. These findings suggest a novel therapy to treat inflammation-mediated gastric adenocarcinomata.

Introduction

Sinomenine (SIN; 7,8-didehydro-4-hydroxy-3,7-dimethoxy-17-methylmorphinane-6-one) is a biomonomer alkali derived from the Chinese medicinal plant Sinomenium acutum. Traditionally, SIN has been used in the treatment of rheumatoid arthritis due to its anti-inflammatory effect (1). Previous studies demonstrated that SIN has cardioprotective (2) and immunosuppressive effects (3,4). In vitro studies indicated that the suppression of cyclooxygenase-2 (COX-2) expression is one of the possible mechanisms for the anti-inflammatory characteristic of SIN (5). Furthermore, in the pioneer experiment conducted by Zhang et al SIN was found to inhibit the proliferation of HeLa cells, possibly by inhibiting the expression of COX-2 (6).

COX is a key enzyme mediating the conversion of arachidonic acid to prostaglandins. Two distinct COX enzymes have been identified: COX-1, a constitutive enzyme, and COX-2, an inducible form (7). COX-1 is a housekeeping molecule that can be detected in most cells and tissues under normal conditions and is involved in maintaining homeostasis by regulating normal physiological functions, such as immune response, acid secretion and blood supply. The expression of COX-2 is rapidly induced by growth factors, oncogenes, carcinogens, mitogens and lipopolysaccharides (8). The majority of the data from animal and human studies indicate that COX-2 is crucial to inflammation and oncogenesis. COX-2 is up-regulated in transformed cells and in a variety of solid tumors such as lung, colorectal, pancreatic and breast cancers (912). COX-2 inhibitors induce apoptosis in various cancer cells both in vitro and in vivo (13). COX-2 is considered to be a potential preventive and therapeutic target for malignancies (14).

Gastric cancer is one of the most common causes of cancer-related mortality in China and other Asian countries (15). At present, surgery and chemotherapy are the standard treatment modalities utilized in gastric cancer (16). However, the 5-year survival of gastric cancer patients is estimated to be only 30%. To improve the prognosis of GC, the development of novel strategies based on its molecular alterations is required. The majority of gastric adenocarcinomas have a high-level expression of COX-2 (1719). Both angiogenesis and Helicobacter pylori infection have been reported to be associated with the COX-2 expression in gastric cancer patients (20). The knockdown of COX-2 in a SGC-7901 gastric adenocarcinoma cell line by RNA interference inhibits proliferation and induces apoptosis (21), indicating that suppression of COX-2 may be developed into an effective approach for the treatment of gastric cancer. The majority of selective COX-2 inhibitors have pronounced side effects that limit the administration of these drugs. In the present study, the inhibitory effect of SIN on the proliferation of SGC-7901 gastric adenocarcinoma cells was observed. Additionally, the question of whether the suppression of COX-2 expression is a potential mechanism for SIN on the proliferation of SGC-7901 cells was investigated.

Materials and methods

Cell cultures and reagents

SGC-7901 gastric adenocarcinoma cells were cultured with Dulbecco’s modified Eagle’s medium (DMEM; Gibco, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS; Gibco), 100 U/ml penicillin and 100 μg/ml streptomycin. Cultures were maintained at 37°C in a humidified incubator in an atmosphere of 5% CO2. Cells were passaged at 1:3 every 3 days. SIN and celecoxib (Sino-American Biotech, Henan, China) were dissolved in dimethylsulfoxide (DMSO; Sigma, St. Louis, MO, USA), stored at −20°C and diluted in DMEM in different proportions (DMSO density of <0.1%). The morphological and growth patterns of the cells were dynamically observed under an inverted microscope (Olympus IX-50; Olympus Optical, Tokyo, Japan).

MTT assay

Following the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, 5×103 cells were seeded in 96-well plates and cultured for 24 h at 37°C and 5% CO2. Media containing various concentrations of SIN were added to the wells 24 h later to reach final concentrations of 125, 250, 500 and 1,000 μmol/l. Celecoxib at a final concentration of 50 μmol/l was used as a positive control. For the DMSO control, DMSO was added to a final concentration of 1‰ to exclude the possible effect of DMSO on cell proliferation. For the blank control, no reagent was added. Drug treatment was continued for another 24, 48, 72 or 96 h, and 5 mg/ml MTT (Sigma) was added to the wells. All of the groups were incubated for 4 h at 37°C. The supernatant was removed and crystals were dissolved in 200 μl DMSO. The absorbance was examined with an automated microplate reader (Bio-Tek, Winooski, VT, USA) at an absorption wavelength of 490 nm. Only the medium was added to the negative control well, which was used to zero the absorbance. Three wells were set up for each group and three independent experiments were conducted.

Reverse transcription-polymerase chain reaction (RT-PCR)

The relative expression of COX-2 mRNA was evaluated using a semi-quantitative reverse transcriptase PCR kit (Takara, Otsu, Shiga, Japan). Total RNA was isolated from SGC-7901 cells using a TRIzol reagent (Promega, Madison, WI, USA). Reverse transcription of total RNA (2 μg) was performed in 20 μl volume according to the manufacturer’s instructions. The primers used for COX-2 were: 5′-CGAGGTGTATGTATGAGTGTG-3′ (forward) and 5′-TCTAGCCAGAGTTTCACCGTA-3′ (reverse). β-actin was amplified as an internal control using the primers: 5′-GTAA AGACCTCTATGCCATCA-3′ (forward) and 5′-GGACTCAT CGTACTCCTGCT-3′ (reverse), resulting in products of 550 and 227 bp, respectively. Each PCR product was visualized by staining with ethidium bromide after electrophoresis on 2% agarose gels under ultraviolet light. The gel images were photographed (Olympus) and relative densities were analyzed using the Bandscan software.

Western blotting

All groups of SGC-7901 cells were collected in 1.5-ml Eppendorf tubes when the cells were treated with drugs for 48 h. The total protein was extracted with RIPA lysis buffer containing proteinase inhibitors. Protein concentration was determined using the Bradford assay. The protein (100 μg) of each sample was separated on 10% sodium dodecyl sulfate (SDS) polyacrylamide gel and transferred to nitrocellulose membranes. Non-specific binding was blocked by 5% skimmed milk for 2 h at room temperature. The membranes were incubated with primary antibody against COX-2 and β-actin (1:1,000 dilution; Sigma) for 4 h at room temperature or overnight at 4°C. After washing with PBST followed by incubation with peroxidase-conjugated goat anti-mouse IgG as secondary antibody (1:2,000 dilution; Sigma) for 1 h at room temperature, protein was detected using enhanced chemiluminescence solution, and by exposing membranes to Kodak X-ray film. The expression of β-actin was detected as an internal control.

Statistical analysis

Statistical analysis was performed using SPSS software (SPSS13.0). Statistical analyses of the data were performed using one-way analysis of variance (ANOVA) followed by a post hoc test. Data were shown as the mean ± standard deviation. P<0.05 was considered to be statistically significant.

Results

Cell morphology

After SGC-7901 cells were treated with different concentrations of drugs, proliferation of SGC-7901 cells was inhibited, the number of cells decreased significantly and cell growth was retarded. Morphologically, the cells detached from the bottle and became round. Achromatolysis, deflation and pyknosis of the nucleus was observed. This phenomenon was most obvious in the 1,000 μmol/l SIN and celecoxib-positive groups. SGC-7901 cells grew more rapidly in the DMSO control group (Fig. 1).

Cell proliferation

We found that the proliferation of SGC-7901 cells was inhibited to various extents in all of the experimental groups and the celecoxib-positive control group (Fig. 2). The DMSO control group was not depressed. SIN inhibited the growth of SGC-7901 cells in a dose-dependent manner and the number of cells decreased following the increased concentration of SIN. Compared to that of the blank control group, the growth of cells treated with SIN decreased significantly (P<0.05 by ANOVA and Tukey’s post hoc test to detect significantly different means). A significant difference was also observed between the celecoxib-positive control group and the blank control and SIN groups (P<0.05). The DMSO control group showed no effects on SGC-7901 cells; in a group comparison between the various densities of SIN, the high-dose group resulted in a markedly reduced growth of SGC-7901 cells as compared to that of the low-dose treated group (P<0.05). Concomitantly, SGC-7901 cells treated with SIN for 24–96 h resulted in an obviously increased inhibitory rate of cell growth. We observed that the highest inhibitory rate among the SIN groups was 93.89% in the 1,000 μmol/l SIN group at 96 h. Moreover, the inhibitory action of SIN on SGC-7901 cells occurred in a time-dependent manner (P<0.05).

SIN inhibits COX-2 expression in human gastric adenocarcinoma cells

To determine COX-2 expression in response to SIN treatment, RT-PCR was performed and SGC-7901 cells were examined (Fig. 3). SIN at a concentration of 125 μmol/l caused a decrease in the expression of COX-2 mRNA, which began 48 h after the initial treatment was administered and occurred in a dose-dependent manner in SGC-7901 cells compared to the blank control group (P<0.05). The DMSO control group exhibited no effects on the expression of COX-2 mRNA in SGC-7901 cells. The celecoxib-positive control group was significantly different from the blank control group (P<0.05).

Western blotting verified the expression of COX-2

Western blot analysis revealed that COX-2 protein was expressed in gastric cancer cells (Fig. 4). No significant difference was observed between the blank and DMSO control groups. Compared to the blank control group, the expression of COX-2 was decreased in various densities of the SIN group in a dose-dependent manner (P<0.05). In contrast to the blank control group, the expression of COX-2 was decreased in the celecoxib-positive control group (p<0.05).

Discussion

Gastric cancer is the most common cause of cancer-related mortality worldwide. Numerous molecular studies have been performed to investigate the developmental mechanism of gastric cancer and COX-2 expression in the pathogenesis of gastric cancer. COX-2 was found to play a significant role in gastric cancer by various pathways. Additionally, the correlation between COX-2 and clinicopathological characteristics, such as tumor size, stage, invasion and lymph node metastasis, of gastric cancer have been identified. COX-2 overexpression protects cancer cells against various apoptotic stimuli (22). The up-regulation of COX-2 is closely related to gastric cancer metastasis through the promotion of lymphangiogenesis and the angiogenesis of gastric cancer (23). Findings of studies have demonstrated that COX-2 is constitutively overexpressed in gastric cancer (24). The relationship between Helicobacter pylori infection and gastric cancer has also been demonstrated. Thus, Helicobacter pylori infection is thought to contribute to the development of gastric cancer via COX-2, which may be due to the stimulation of tumor growth and angiogenesis (25). Several molecular pathways have been hypothesized in the development of gastric cancer. Previous studies indicated that the COX-2-PGI2-PPARδ pathway was also involved in tumorigenesis (26). VEGF is one of the most significant mediators of the COX-2 pathway (27). COX-2 produced by cancer cells is correlated with the elevation of Bcl-2 protein and inhibition of apoptosis in gastric cancer tissue.

In the present study, we observed that COX-2 was highly expressed in gastric cancer cells, a result that is consistent with findings of other studies. COX-2 selective inhibitors have been shown to induce apoptosis in gastric cancer (28). Our study found that SIN was suppressed COX-2 expression in SGC-7901 cells, which grew slowly and became round. In their study, Zhang et al found that SIN inhibited the proliferation of HeLa cells as a COX-2 selective inhibitor (6). This inhibition may relate to SIN blockage of the cell cycle and induction of apoptosis, the mechanism of which may constitute the inhibition of COX-2 expression in a dose- dependent manner. Studies have also shown that SIN mediated the down-regulation of COX-2 expression and the production of induced PGE2 in PC-12 cells by suppressing the activity of NF-κB (5). To assess whether the inhibition of COX-2 expression is involved in gastric cancer cells, MTT assay, RT-PCR analysis and Western blotting were performed to test cell viability, COX-2 mRNA and protein expression, respectively.

The results of this study suggest that SIN has an inhibitory effect on the growth of gastric cancer. Based on our observation of cell morphology, we found that SIN effectively inhibited the growth of SGC-7901 cells. Compared to the control group, the number of cells decreased significantly in the SIN groups and the proliferation of SGC-7901 cells was inhibited. The highest inhibitory rate was 93.89% in the 1,000 μmol/l SIN group at 96 h. The preliminary inhibitory effect of SIN on gastric cancer cells was demonstrated by this result. We showed that SIN was capable of reducing up-regulated mRNA and the protein levels of COX-2. COX-2 mRNA was significantly decreased compared to the blank control group. SIN down-regulated the COX-2 protein expression in a dose-dependent manner in gastric cancer cells. The present results indicate that the inhibitory effect of SIN on gastric cancer cells may be activated by the COX-2 pathway. COX-2 is a key enzyme in prostaglandin synthesis. PGE2 may promote the growth of gastric cancer cells and induce Foxp3 expression independently of TGF-β and IL-10 in the gastric cancer microenvironment (29). SIN may also inhibit PGE2 synthesis by suppressing the expression of COX-2. Further investigation is required to identify the signal transduction pathway of COX-2. Blocking this pathway using SIN may facilitate tumor therapeutics.

In conclusion, the present study suggests that SIN is involved in inhibiting the proliferation of gastric cancer cells in vitro and that its therapeutic mechanism is related to the inhibition of COX-2 expression. The findings of this study suggest that SIN has a preliminarily therapeutic effect on gastric cancer, indicating that SIN is an effective candidate drug for treating gastric cancer.

Acknowledgements

The authors are indebted to Professor Hongxia Li (The First Affiliated Hospital of Xi’an Jiao Tong University, College of Medicine, China) for the kind assistance with cell cultures, and wish to thank Professor Xinyang Wang (The First Affiliated Hospital of Xi’an Jiaotong University, College of Medicine, China) for directing the experimental work.

References

1 

Ke XY, Yu MX and Jiang M: Clinical observation on treatment of rheumatoid arthritis with sinomenine. Bei Jing Yi Xue. 8:186–188. 1986.

2 

Satoh H: Electropharmacology of sinomeni caulis et rhizome and its constituents in cardiomyocytes. Am J Chin Med. 33:967–979. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Vieregge B, Resch K and Kaever V: Synergistic effects of the alkaloid sinomenine in combination with the immunosuppressive drugs tacrolimus and mycophenolic acid. Planta Medica. 65:80–82. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Dai YB, Huang X and Luo ZG: Immunosuppressive effect of sinomenine on ICAM-1 expression in rat renal allograft rejection. Mod J Integr Trad Chin West Med. 12:1358–1363. 2003.

5 

Chen W, Shen YD and Zhao GS: Inhibitory effect of sinomenine on expression of cyclooxygenase-2 in lipopolysaccharide-induced PC-12 cells. China J Chin Mat Medica. 29:900–903. 2004.PubMed/NCBI

6 

Zhang Y, Wu M and Li XG: Experimental study on the effect of selective COX-2 inhibitor sinomenine on HeLa cells. Prog Mod Biomed. 6:38–40. 2006.

7 

Hla T, Bishop-Bailey D, Liu CH, Schaefers HJ and Trifan OC: Cyclooxygenase-1 and -2 isoenzymes. Int J Biochem Cell Biol. 31:551–557. 1999. View Article : Google Scholar

8 

Morita I: Distinct functions of COX-1 and COX-2. Prostaglandins Other Lipid Mediat. 68:165–175. 2002. View Article : Google Scholar

9 

Su JL, Shih JY, Yen ML, Jeng YM, Chang CC and Hsieh CY: Cyclooxygenase-2 induces EP1-and HER-2/Neu-dependent vascular endothelialgrowth factor-C up-regulation: a novel mechanism of lymphangiogenesis in lung adenocarcinoma. Cancer Res. 64:554–564. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Soumaoro LT, Uetake H, Higuchi T, Takagi Y, Enomoto M and Sugihara K: Cyclooxygenase-2 expression: a significant prognostic indicator for patients with colorectal cancer. Clin Cancer Res. 10:8465–8471. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Tucker ON, Danneberg AJ and Tang EK: Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer. Cancer Res. 59:987–990. 1999.PubMed/NCBI

12 

Timoshenko AV, Chakraborty C, Wagner GF and Lala PK: COX-2-mediated stimulation of the lymphangiogenic factor VEGF-C in human breast cancer. Br J Cancer. 94:1154–1163. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Meric JB, Rottey S, Olaussen K, Soria JC, Khayat D and Rixe O: Cyclooxygenase-2 as a target for anticancer drug development. Crit Rev Oncol Hematol. 59:51–64. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Dannenberg AJ and Subbaramaiah K: Targeting cyclooxygenase-2 in human neoplasia: rationale and promise. Cancer Cell. 4:431–436. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics. CA Cancer J Clin. 59:225–249. 2009.

16 

Wagner AD, Grothe W, Haerting J, et al: Combination chemotherapies in advanced gastric cancer: an updated systematic review and meta-analysis. American Society of Clinical Oncology. 25:45552007.

17 

Chen CN, Sung CT, Lin MT, Lee PH and Chang KJ: Clinicopathologic association of cyclooxygenase-1 and cyclooxygenase-2 expression in gastric adenocarcinoma. Ann Surg. 233:183–188. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Yamaca D, Ayyildiz T, Coskun U, et al: Cyclooxygenase-2 expression and its association with angiogenesis, Helicobacter pylori, and clinicopathologic characteristics of gastric carcinoma. Pathol Res Pract. 204:527–536. 2008. View Article : Google Scholar

19 

Tatsuguchi A, Matsui K and Shinji Y: Cyclooxygenase-2 expression correlates with angiogenesis and apoptosis in gastric cancer tissue. Hum Pathol. 35:488–495. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Sun WH, Yu Q, Shen H, et al: Roles of Helicobacter pylori infection and cyclooxygenase-2 expression in gastric carcinogenesis. World J Gastroenterol. 10:2809–2813. 2004.PubMed/NCBI

21 

Wang BH, Qian W and Gao YJ: Effects of inhibition of cyclooxygenase-2 by RNA interference on proliferation and apoptosis of human gastric cancer cells: an experimental study with human gastric cancer cells and mice. Nat Med J China. 86:266–271. 2006.

22 

Tjiu JW, Liao YH, Lin SJ, et al: Cyclooxygenase-2 over-expression in human basal cell carcinoma cell line increases antiapoptosis, angiogenesis, and tumorigenesis. J Invest Dermatol. 126:1143–1151. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Fosien E: Biochemistry of cyclooxygenase (COX-2) inhibitors and molecular pathology of COX-2 in neoplasia. Crit Rev Las Sci. 37:431–502. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Willams CS and DuBois RN: Prostaglandin endoperoxide synthase: why two isoforms? Am J Physiol. 270:393–400. 1996.PubMed/NCBI

25 

Konturek PC, Hartwich A, Zuchowicz M, et al: Helicobacter pylori gastrin and cyclooxygenases in gastric cancer. J Physiol Pharmacol. 51:737–749. 2000.

26 

Yu J, Leung WK, Chen J, Ebert MPA, Malfertheiner P and Sung JJY: Expression of peroxisome proliferators-activated receptor δ in human gastric cancer and its response to specific COX-2 inhibitor. Cancer Lett. 223:11–17. 2005.

27 

Da MX, Wu XT, Wang J, et al: Expression of cyclooxygenase-2 and vascular endothelial growth factor-c correlates with lymph-angiogenesis and lymphatic invasion in human gastric cancer. Arch Med Res. 39:92–99. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Tegeder I, Pfeilschifter J and Geisslinger G: Cyclooxygenase-independent actions of cyclooxygenase inhibitors. FASEB J. 15:2057–2072. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Yuan XL, Chen L, Li MX, et al: Elevated expression of Foxp3 in tumor-infiltrating Treg cells suppresses T-cell proliferation and contributes to gastric cancer progression in a COX-2-dependent manner. Clin Immunol. 134:277–288. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-August 2011
Volume 2 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
Lv Y, Li C, Li S and Hao Z: Sinomenine inhibits proliferation of SGC-7901 gastric adenocarcinoma cells via suppression of cyclooxygenase-2 expression. Oncol Lett 2: 741-745, 2011
APA
Lv, Y., Li, C., Li, S., & Hao, Z. (2011). Sinomenine inhibits proliferation of SGC-7901 gastric adenocarcinoma cells via suppression of cyclooxygenase-2 expression. Oncology Letters, 2, 741-745. https://doi.org/10.3892/ol.2011.305
MLA
Lv, Y., Li, C., Li, S., Hao, Z."Sinomenine inhibits proliferation of SGC-7901 gastric adenocarcinoma cells via suppression of cyclooxygenase-2 expression". Oncology Letters 2.4 (2011): 741-745.
Chicago
Lv, Y., Li, C., Li, S., Hao, Z."Sinomenine inhibits proliferation of SGC-7901 gastric adenocarcinoma cells via suppression of cyclooxygenase-2 expression". Oncology Letters 2, no. 4 (2011): 741-745. https://doi.org/10.3892/ol.2011.305