Open Access

Role of the ERas gene in gastric cancer cells

  • Authors:
    • Yang Liu
    • Zhaoqian Wang
    • Huiming Li
    • Zhaoping Wu
    • Fang Wei
    • Huiping Wang
  • View Affiliations

  • Published online on: April 23, 2013     https://doi.org/10.3892/or.2013.2417
  • Pages: 50-56
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

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


Abstract

As a novel member of the Ras family, ERas, found in murine embryonic stem (ES) cells in 2003, was considered a pseudogene. To date, there are a few reports on the relationship between ERas and tumors. It was recently suggested that ERas could affect gastric carcinoma (GC) metastasis, but no significant relationship was found with tumor proliferation. Since ERas plays an important role in tumor-like growth of ES cells subcutaneously injected into nude mice, we hypothesized that ERas plays a role in tumor proliferation. In this experiment, we selected 7 GC strains from different sources with different differentiation degrees, we detected the expression of full length ERas transcript, and selected two ERas highly expressing GC strains, MKN‑28 and BGC‑823. After knocking down the ERas gene by siRNA, we observed that there was a significant decrease in proliferation, metastasis as well as clonality. Therefore, ERas is confirmed to be an important gene in affecting tumor proliferation and metastasis. Furthermore, the significance of the ERas mechanism and signaling pathway is shown.

Introduction

The ERas gene, which can support embryonic stem (ES) cell tumor-like growth (14), was first identified in murine ES cells by Takahashi et al(1). It encodes a protein of 227 amino acids with 43, 46 and 47% identity to conventional Ras oncogenes H-ras, K-ras and N-ras, respectively, defined as a new member of the Ras family. Unlike other Ras family members, the ERas product is a constitutively active protein without any mutation (1), while conventional ras oncogenes acquire activation of carcinogenesis by point mutation of several amino acids, which include Gly12, Ala59 or Glu63 (5,6). The ERas protein contains amino acid residues identical to those present in active mutants of the conventional Ras oncogenes K-ras, N-ras and H-ras (7). The conventional Ras members mainly function through activating either phosphatidylinositol-3-OH kinase (PI3K) or Raf pathway (8,9), while ERas interacts with PI3K (10) but not with Raf (11,12). The human ERas gene was initially erroneously recognized as a processed pseudogene HRasp (Ha-Ras2) (13,14) with deficiency or meaningless mutation, but it was more recently described as a gene potentially encoding a functional human ERas protein (1518). Therefore, a few studies on human ERas have previously been reported.

In 2002, Bjorklund et al(19) found that mouse ES cells developed into teratomas when transplanted into nude mice. In 2003, Takahashi et al(1) found that ERas is key in the tumor-like growth properties of ES cells. In 2009, Kaizaki et al(17) discovered ERas was actively expressed in gastric cancer (GC) and was closely related to its oncogenesis. These results indicate that the expression of ERas may be associated with cell proliferation and transformation.

However, in 2010, Kubota et al(20) detected 142 clinical samples of GC, which showed that ERas expression was strongly associated with liver and lymph node metastases. Meanwhile, there was no significant correlation between ERas expression and histological differentiation. Overexpression of ERas in GC cell lines promoted colony formation while it showed no significant effect on cell proliferation. These results indicated that ERas was only associated with the metastasis of GC. However, clonality is an indicator of proliferation capacity. Hence, considering the multiaspect influences of oncogenesis, we decided to knock down the ERas gene in order to study the impactions on proliferation and clonality of GC cells.

In addition, Kameda and Thomson (2) used RT-PCR to analyze expression of the ERas gene in human ES cells in 2005, and they could not detect a full-length ERas coding transcript. Instead, a truncated noncoding transcript was found, which was caused by a premature polyadenylation signal predicted through sequence analysis and confirmed by 3′RACE analysis. Except for the premature PloyA locus, humans and chimpanzees have typical-Alu-s-retrotransposon insertions, which also influence the expression of ERas at this specific locus. Moreover, the lack of ERas expression in human ES cells indicates that the oncogenesis is very different from that of murines (21). These findings indicate that further studies should be performed to ascertain whether or not a full-length ERas coding transcript is present in human GC cells.

In this study, 8 cell strains from different sources, GC lymph node metastasis, GC liver metastasis, GC ascites and GC tissues, with different differentiation degrees, were chosen to determine whether a full-length ERas mRNA exists and to elucidate the difference of degree of ERas expression among these GC strains by RT-PCR, real-time PCR and western blotting. Furthermore, we confirmed the effect of ERas knockdown on cell proliferation, metastasis and clonality in ERas highly expressed GC strains.

Materials and methods

Cell culture and cell lines

The cell lines GES-1, MKN-28, MKN-45, BGC-823, NCL-N87, SNU-16, SGC-7901 and AGS were cultured in RPMI-1640, supplemented with 10% fetal bovine serum (FBS; HyClone, Logan, UT, USA). They were cultured in an atmosphere of 5% CO2 at 37°C. BGC-823, NCL-N87 and AGS cells were obtained from Shanghai Institute of Cell Bank (Shanghai, China); GES-1, MKN-28, MKN-45, SNU-16 and SGC-7901 were purchased from the American Type Culture Collection (ATCC, Rockville, MD, USA).

Amplification of the ERas full-length transcripts and sequencing analysis

Total cellular RNA was extracted from each cell line with TRIzol® (Invitrogen Life Technologies, Carlsbad, CA, USA), and cDNA was synthesized with Reverse Transcription kit (Promega Corp., Madison, WI, USA), and both were performed according to the manufacturer’s protocol. The cDNA was synthesized by PCR with PrimeSTAR HS DNA polymerase (Takara Bio, Shiga, Japan). The primers used for the full-length ERas coding sequence were: forward, 5′-atggagctgccaacaaagcctggca-3′ and reverse, 5′-ttcaggccacagagcagccacagt-3′, which gave an amplified fragment of 702 bp. The reaction conditions were: 94°C for 15 sec, 62°C for 45 sec and 72°C for 30 sec, repeated for 35 cycles. The products were separated by 1.2% agarose gel electrophoresis, and the 702 bp bands were recycled using aqua-Spin gel extraction mini kit (Watson Biotechnologies, Inc., Shanghai, China). The identification of PCR products was confirmed by sequencing analysis (Songon Biotech Co., Shanghai, China).

Real-time quantitative PCR

The real-time quantitative PCR analyses were performed in triplicate using SYBR® Premix Ex Taq™II kit (Takara Bio). The GAPDH gene was chosen as an endogenous control. The primers used for ERas were: forward, 5′-cacatggagcccttccttc-3′ and reverse, 5′-tgtccagggtcaactccttc-3′; the primers used for GAPDH were: forward, 5′-ggacctgacctgccgtctag-3′ and reverse, 5′-gtagcccaggatgcccttga-3′; the PCR conditions were as follows: 94°C for 15 sec, 58°C for 45 sec, 72°C for 20 sec, repeated for 35 cycles. Amplified products were separated by 0.9% agarose gel electrophoresis.

Western blot analysis

Cells were lysed in a lysis buffer containing 2% sodium dodecyl sulfate (SDS) and 0.125 M Tris-HCl (pH 6.8) on ice for 30 min, followed by high-speed centrifugation, and the supernatant protein was finally collected. SDS-PAGE was performed using 10% polyacrylamide gels. PAGE separated proteins were electrophoretically transferred onto nitrocellulose membranes. The membrane filters were blocked with 5% powdered milk in TBST (0.1% Tween-20) for 2 h and then incubated in rabbit ERas antibody (Abgent, Suzhou, China) diluted 1:100 in TBST at 4°C overnight, and finally incubated with HRP anti-rabbit secondary antibody (Kangchen Biotech, Shanghai, China) diluted 1:2,000 for 1 h at room temperature. Antigens on the membrane were detected with enhanced chemiluminescense detection reagents (Roche, Basel, Switzerland).

Small interfering RNA transfection

Two ERas stealth siRNA, no. 30 forward, GCAACUAGCUUUGAGGGAC(dTdT) and reverse, GUCCCUCAAAGCUAGUUGC(dTdT); no. 32 forward, GUAACAUGGGAGUGCCUAA(dTdT) and reverse, UUAGGCACUCCCAUGUUAC(dTdT); one high GC% negative control siRNA forward, CCUACGCCACCAAUUUC GU(dTdT) and reverse, ACGAAAUUGGUGGCGUAGG (dTdT) were designed and synthesized (Bioneer, Daejon, Korea). siRNA was mixed with Lipofectamine™ 2000 (Invitrogen Life Technologies) in an OptiMEM serum-free medium (HyClone) for 30 min at room temperature and then added to each 24-well plate containing MKN-28 or BGC-823 cells. Cells were maintained in a humidified 5% CO2 incubator at 37°C for 6 h with the old medium being replaced by a fresh medium. After 24 h of transfection, cells were harvested for cell proliferation, migration and colony formation assays.

CCK-8 assay

siRNA-transfected MKN-28 and BGC-823 cells were seeded into 96-well plates at a density of 3×103 cells/well and maintained in culture medium for 5 days. Each well set five duplicates. We measured cell growth using cholecystokinin (CCK) assay by Cell Counting Kit (Dojindo, Tokyo, Japan) according to the manufacturer’s instructions.

Cell migration assay

For wound-healing experiments, MKN-28 and BGC-823 cells transfected with siRNA were cultured to 80% confluence after being seeded into 6-well plates, then scraped using a p10 tip (time 0), and suspended cells were washed with PBS three times. Cells were incubated for another 4 days and images were captured by microscope (Zeiss, Oberkochen, German) at the same time every day. Migration distance was measured from images (5 fields) at each indicated time point.

Transwell assay of MKN-28 and BGC-823 cells was assessed using 6.5 mm diameter inserts (Corning Costar Corp., Corning, NY, USA). A total of 3×104 cells were suspended in 100 μl serum-free RPMI-1640 medium and loaded into upper wells; lower chambers were filled with 600 μl of complete medium (RPMI-1640 supplemented with 10% FBS). Migration chambers were incubated in a humidified 5% CO2 incubator at 37°C for 24 and 48 h. Cells were then fixed with 600 μl of paraformaldehyde for 20 min. The inner surfaces of the upper chambers were wiped using cotton swabs to remove non-migrated cells in the migration assay. The chambers were then washed with PBS and stained with 500 μl crystal violet for 20 min at room temperature. Stained cells were counted using the ImageJ software, and 5 random fields were counted (Zeiss).

Colony formation assay

A total of 500 siRNA-transfected MKN-28 and BGC-823 cells were seeded in 6-well plates and incubated for 14 days respectively, with the medium replaced every 4 days. On the 15th day, the cells were stained with crystal violet for 20 min and washed with tap water for 10 min. For each dish, colonies in five random fields were counted using the ImageJ software.

Statistical analysis

Each measurement was performed in triplicate. Original real-time PCR data, CCK-8 data, migration/invasion data and colony formation data were recorded as continuous variables and analyzed using Student’s t-test or linear polynomial ANOVA with LSD post hoc examination. All statistical analyses were performed using SPSS 16.0 software. P-values <0.05 were considered to indicate statistically significant differences.

Results

ERas expressed in GC cells

The full-length ERas mRNA transcript was detected in all seven GC cell lines and GES-1 gastric mucosa cell line by RT-PCR. The forward and reverse primers were located in open reading frame (ORF) 1–25 and 680–702 bp separately, which give rise to an amplified fragment of 702 bp (Fig. 1A). To assess whether ERas expression was different with mutant H-ras, K-ras and N-ras, mutation analysis was performed by sequencing analysis, revealing no mutation of ERas in all the gastric cell lines that we selected (data not shown).

The expression levels of ERas mRNA were also determined by real-time PCR. All eight cell lines were divided into five groups by one-way ANOVA. The GC cell line BGC-823 expressed most highly (P<0.01), MKN-28 expressed highly (P<0.05), SNU-16, SGC-7901, MKN-45 expressed moderately (P<0.05), while AGS, NCL-N87 expressed poorly, similar to the normal gastric mucosal cell line, GES-1 (P>0.05) (Fig. 1B-D). ERas protein expression was also confirmed by western blotting (Fig. 1E).

ERas increases GC cell proliferation

To examine the role of ERas in cell proliferation, we measured BGC-823, MKN-28 cell growth by CCK-8 assay after transfecting with ERas siRNA30 and siRNA32.

We observed a significant decrease in cell proliferation when the expression of ERas was knocked down in MKN-28 and BGC-823 cells (Fig. 2).

On the third day, the OD value of MKN-28 cells decreased from 1.60±0.05 to 1.28±0.10 when transfected with siRNA30 (P<0.01), and it decreased to 1.35±0.10 when transfected with siRNA32 (P<0.01). Meanwhile, the OD value of BGC-823 cells decreased from 0.64±0.18 to 0.30±0.08 and 0.40±0.10 when transfected with siRNA30 (P<0.01) and siRNA32 (P<0.05) individually.

On the fourth day, the OD value decreased from 2.09±0.09 to 1.74±0.10 and 1.64±0.11 individually in MKN-28 cells when transfected with siRNA30 (P<0.01) and siRNA32 (P<0.01), respectively, while it decreased from 1.57±0.07 to 1.00±0.46 (P<0.05) and 1.20±0.19 (P<0.01) in BGC-823 cells. These data indicate that the proliferation of these GC cell lines is significantly promoted by the ERas gene.

ERas promotes GC cell migration

We also confirmed the effect of ERas on migration in MKN-28 and BGC-823 cells by Transwell and wound-healing assay.

As shown in Fig. 3A and B, knockdown of ERas by siRNA significantly impaired the ability of MKN-28 and BGC-823 cells to migrate through the membranes. Twenty-four hours later, the number of migratory cells decreased from 163.5±9.19 to 62.50±7.78 (P<0.05) and 71±11.31 (P<0.01) when transfected with siRNA30 and siRNA32 in MKN-28 cells, while it decreased from 155.50±9.19 to 50±4.24 (P<0.01) and 60.5±9.19 (P<0.05) in BGC-823 cells. Forty-eight hours later, the number of migratory MKN-28 cells decreased from 350.5±13.44 to 62.50±7.78 (P<0.01) and 71±11.31 (P<0.01) respectively, while the number of migratory BGC-823 cells decreased from 255±8.49 to 69.5±4.95 (P<0.01) and 86±8.49 (P<0.01).

Then, we further analyzed migration ability by using wound-healing for 4 days. As shown in Fig. 4, the speed of wound repair was markedly slower when ERas was silenced by siRNA in MKN-28 and BGC-823 cells at 24, 48, 72 and 96 h. The statistical significance was most notable at 48 h, when the wound repair percentage decreased from 61.35±3.54% to 25±6.25% (P<0.001) and 28.89±3.85% (P<0.001) after ERas was knocked down by siRNA30 and siRNA32 respectively in MKN-28 cells, while the percentage decreased from 58.13±3.61% to 19.61±3.78% (P<0.001) and 18±3.85% (P<0.001) respectively in BGC-823 cells.

ERas promotes GC cell colony formation

The colony formation was assessed two weeks later. As shown in Fig. 5, ERas knockdown by siRNA30 or siRNA32 reduced the number of colonies from 413±11.31 to 237±12.73 (P<0.01) and 254±9.90 (P<0.01) in MKN-28 cells, while the number of colonies in BGC-823 cells was reduced from 318.5±12.02 to 166±9.90 (P<0.01) and 180±12.73 (P<0.01).

Discussion

The ERas gene is strongly expressed in BGC-823 and MKN-28 cell strains among the eight gastric cell lines (Fig. 1B-D). To examine the effect of ERas on GC cell proliferation, these two highly expressing endogenous ERas cell strains were investigated after treatment with ERas siRNA by CCK-8 assay for 5 days. Knockdown of ERas inhibited the proliferation ability markedly compared to control in BGC-823 and MKN-28 cells on the third and fourth day (Fig. 2). Kubota et al(20) concluded that ERas could not promote the proliferation in GCIY cells transfected with an ERas-overexpressing vector by MTS assays for 6 days, whereas it could enhance colony formation as the cell clonality experiment results showed. They came to the same result that there was no relationship between proliferation and the ERas gene in GCIY and NUGC-4 cells with ERas knocked down by stealth siRNA using MTS assays for 2 days. However, whether or not knockdown of ERas inhibits cell colony formation was not further established. GCIY cell strain was used in both ERas overexpression and knockdown experiment. However, the expression level of ERas in this strain was relatively low, indicating it was not suitable for the knockdown experiment. On the other hand, the observation period in their study was relatively short. These reasons may lead to the disparity. Meanwhile, the results of Transwell test, scratch test and colony formation test (22,23) proved that ERas is able to increase GC cell migration and colony formation which were also two aspects that could reflect cell proliferation. Hence, there is sufficient evidence to prove that ERas enhances GC cell proliferation.

Furthermore, ERas was expressed most highly in poorly differentiated BGC-823 cells and well differentiated MKN-28 cells, less highly in poorly differentiated SNU-16 cells, moderately differentiated SGC-7901 cells and poorly differentiated MKN-45 cells, almost silently in poorly differentiated AGC cells, immortal GES-1 cells and poorly differentiated NCL-N87 cells (Fig. 1A-C). From these date, the conclusion that expression of ERas is not related to histological differentiation is the same as that of Kubota et al. However, from analyzing the following seven cell strains, SGC-7901 from GC lymph metastasis, NCL-N87 from GC liver metastasis, SNU-16 from GC ascites, GES-1 from fetal gastric mucosa and others from GC tissues, the connection between ERas and metastasis of gastric lymph and liver is not so strong, which is different from the result of Kubota et al. Therefore, to fully understand the connection between ERas and gastric lymph metastasis and liver metastasis, further research is required.

The full-length transcript of ERas (2,20) in those 7 GC cell strains were examined (Fig. 1A), suggesting that activated ERas is present universally in GC cells. Since a full-length transcript cannot be found in human ES cells, there should be some common activation factors which inhibit the prematuration of early polyadenylation signal and the insertion of Alu-S transposons to activate ERas expression.

To date, there are few studies on the relationship between ERas and cancer, and the function of ERas has yet to be determined. This study used CCK-8, Transwell, scratch test and clonality test to successfully prove that ERas has the ability to enhance GC cell proliferation, metastasis and clonality. Compared to Kubota et al(20), this study proved that the activation of the ERas gene in GC cells is common and the function of ERas in the processes of GC cell development and metastasis is important; there must be a significance of clarifying the correlative key signal pathways and the cytokines to activate the pathways.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (30672440). We thank Yufei Wang, Qiang Fu, Ronghua Wang, Ran Hu for their technical assistance. We also gratefully acknowledge the entire staff of the Experimental Center of Shanghai Jiaotong University Affiliated First People’s Hospital.

Abbreviations:

ES

embryonic stem

PI3K

phosphatidylinositol-3-OH kinase

GC

gastric carcinoma

References

1 

Takahashi K, Mitsui K and Yamanaka S: Role of ERas in promoting tumour-like properties in mouse embryonic stem cells. Nature. 423:541–545. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Kameda T and Thomson JA: Human ERas gene has an upstream premature polyadenylation signal that results in a truncated, noncoding transcript. Stem Cells. 23:1535–1540. 2005. View Article : Google Scholar

3 

Takahashi K, Murakami M and Yamanaka S: Role of the phosphoinositide 3-kinase pathway in mouse embryonic stem (ES) cells. Biochem Soc Trans. 33:1522–1525. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Takahashi K, Nakagawa M, Young SG and Yamanaka S: Differential membrane localization of ERas and Rheb, two Ras-related proteins involved in the phosphatidylinositol 3-kinase/mTOR pathway. J Biol Chem. 280:32768–32774. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Fasano O, Aldrich T, Tamanoi F, et al: Analysis of the transforming potential of the human H-ras gene by random mutagenesis. Proc Natl Acad Sci USA. 81:4008–4012. 1984. View Article : Google Scholar : PubMed/NCBI

6 

Schubbert S, Shannon K and Bollag G: Hyperactive Ras in developmental disorders and cancer. Nat Rev Cancer. 7:295–308. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Seeburg PH, Colby WW, Capon DJ, et al: Biological properties of human c-Ha-ras1 genes mutated at codon 12. Nature. 312:71–75. 1984. View Article : Google Scholar : PubMed/NCBI

8 

Cass LA and Meinkoth JL: Ras signaling through PI3K confers hormone-independent proliferation that is compatible with differentiation. Oncogene. 19:924–932. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Deora AA, Hajjar DP and Lander HM: Recruitment and activation of Raf-1 kinase by nitric oxide-activated Ras. Biochemistry. 39:9901–9908. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Rodriguez-Viciana P, Warne PH, Dhand R, et al: Phosphatidylinositol-3-OH kinase as a direct target of Ras. Nature. 370:527–532. 1994. View Article : Google Scholar : PubMed/NCBI

11 

Moodie SA, Willumsen BM, Weber MJ and Wolfman A: Complexes of Ras.GTP with Raf-1 and mitogen-activated protein kinase kinase. Science. 260:1658–1661. 1993. View Article : Google Scholar : PubMed/NCBI

12 

Zhang XF, Settleman J, Kyriakis JM, et al: Normal and oncogenic p21ras proteins bind to the amino-terminal regulatory domain of c-Raf-1. Nature. 364:308–313. 1993. View Article : Google Scholar : PubMed/NCBI

13 

Chang EH, Gonda MA, Ellis RW, et al: Human genome contains four genes homologous to transforming genes of Harvey and Kirsten murine sarcoma viruses. Proc Natl Acad Sci USA. 79:4848–4852. 1982. View Article : Google Scholar

14 

Miyoshi J, Kagimoto M, Soeda E and Sakaki Y: The human c-Ha-ras2 is a processed pseudogene inactivated by numerous base substitutions. Nucleic Acids Res. 12:1821–1828. 1984. View Article : Google Scholar : PubMed/NCBI

15 

Yasuda K, Yashiro M, Sawada T, et al: ERas oncogene expression and epigenetic regulation by histone acetylation in human cancer cells. Anticancer Res. 27:4071–4075. 2007.PubMed/NCBI

16 

Yashiro M, Yasuda K, Nishii T, et al: Epigenetic regulation of the embryonic oncogene ERas in gastric cancer cells. Int J Oncol. 35:997–1003. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Kaizaki R, Yashiro M, Shinto O, et al: Expression of ERas oncogene in gastric carcinoma. Anticancer Res. 29:2189–2193. 2009.PubMed/NCBI

18 

Kubota E, Kataoka H, Tanaka M, et al: ERas enhances resistance to CPT-11 in gastric cancer. Anticancer Res. 31:3353–3360. 2011.PubMed/NCBI

19 

Bjorklund LM, Sánchez-Pernaute R, Chung S, et al: Embryonic stem cells develop into functional dopaminergic neurons after transplantation in a Parkinson rat model. Proc Natl Acad Sci USA. 99:2344–2349. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Kubota E, Kataoka H, Aoyama M, et al: Role of ES cell-expressed Ras (ERas) in tumorigenicity of gastric cancer. Am J Pathol. 177:955–963. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Rao M: Conserved and divergent paths that regulate self-renewal in mouse and human embryonic stem cells. Dev Biol. 275:269–286. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Hsu RJ, Ho JY, Cha TL, et al: WNT10A plays an oncogenic role in renal cell carcinoma by activating WNT/β-catenin pathway. PLoS One. 7:e476492012.PubMed/NCBI

23 

Li WG, Yuan YZ, Qiao MM and Zhang YP: High dose glargine alters the expression profiles of microRNAs in pancreatic cancer cells. World J Gastroenterol. 18:2630–2639. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July 2013
Volume 30 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu Y, Wang Z, Li H, Wu Z, Wei F and Wang H: Role of the ERas gene in gastric cancer cells. Oncol Rep 30: 50-56, 2013
APA
Liu, Y., Wang, Z., Li, H., Wu, Z., Wei, F., & Wang, H. (2013). Role of the ERas gene in gastric cancer cells. Oncology Reports, 30, 50-56. https://doi.org/10.3892/or.2013.2417
MLA
Liu, Y., Wang, Z., Li, H., Wu, Z., Wei, F., Wang, H."Role of the ERas gene in gastric cancer cells". Oncology Reports 30.1 (2013): 50-56.
Chicago
Liu, Y., Wang, Z., Li, H., Wu, Z., Wei, F., Wang, H."Role of the ERas gene in gastric cancer cells". Oncology Reports 30, no. 1 (2013): 50-56. https://doi.org/10.3892/or.2013.2417