Open Access

Effects of 17β-estradiol and tamoxifen on gastric cancer cell proliferation and apoptosis and ER-α36 expression

  • Authors:
    • Xuming Wang
    • Qiuyue Chen
    • Xuan Huang
    • Feng Zou
    • Zhengqi Fu
    • Ying Chen
    • Yan Li
    • Zhaoyi Wang
    • Lijiang Liu
  • View Affiliations

  • Published online on: November 23, 2016     https://doi.org/10.3892/ol.2016.5424
  • Pages: 57-62
  • Copyright: © Wang 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

The present study aimed to investigate the effects of 17β-estradiol and tamoxifen, an agonist and inhibitor of the estrogen receptor (ER), respectively, on the proliferation and apoptosis of gastric cancer cells, as well as the messenger (m)RNA expression levels of ER‑α36. Nested reverse transcription‑polymerase chain reaction (RT-PCR) confirmed that ER‑α36 was expressed in the BGC823, MKN45 and SGC7901 human gastric cancer cell lines. Subsequently, the BGC823 cell line was stimulated with various concentrations of 17β‑estradiol or tamoxifen for 24 or 48 h, and the proliferation, apoptosis and mRNA expression levels of ER‑α36 were determined by water‑soluble tetrazolium (WST)-1 assay, flow cytometry and RT‑quantitative PCR, respectively. The activity of BGC823 cells was significantly increased following treatment with 10‑12 mol/l 17β‑estradiol for 24 h (P=0.013), as compared with the control, and reached a peak at 48 h (P=0.002). Notably, the activity of BGC823 cells was decreased with increasing concentrations of 17β‑estradiol, although it remained higher compared with that of the control. In the tamoxifen‑treated groups, the cell activity decreased as the drug concentration increased. The apoptosis rate was markedly reduced in the 17β‑estradiol group after 24 h (10-12 mol/l, P=0.013; 10‑11 mol/l, P=0.023; and 10‑10 mol/l, P=0.017) and after 48 h (10‑12 mol/l, P=0.002; 10-11 mol/l, P=0.011; and 10‑10 mol/l, P=0.033), whereas the rate of apoptosis increased as the tamoxifen concentration increased (24 h: 5x10‑6 mol/l, P=0.002; and 10‑5 mol/l, P=0.001; and 48 h: 5x10‑6 mol/l, P=0.014 and 10‑5 mol/l, P=0.0021), as compared with the control group. The mRNA expression levels of ER‑α36 were significantly increased after 24 h of treatment with 10‑12 mol/l (P=0.024), 10‑11 mol/l (P=0.0113) and 10‑10 mol/l (P=0.0037) 17β‑estradiol compared with the control group when the concentration of 17β‑estradiol was low, and the same was observed after 48 h of treatment 10‑12 mol/l (P=0.0164), 10‑11 mol/l (P=0.0342) and 10‑10 mol/l (P=0.0198) 17β‑estradiol. The mRNA expression levels of ER‑α36 were significantly decreased with increasing concentrations of tamoxifen after 24 h (5x10‑6 mol/l, P=0.0233; and 10‑5 mol/l, P=0.007) and after 48 h (5x10‑6 mol/l, P=0.001; and 10‑5 mol/l, P=0.0153). In addition, the ability of tamoxifen to inhibit the growth of gastric cancer cells was concentration‑dependent. The results of the present study suggested that gastric cancer cells were sensitive to the effects of 17β‑estradiol and tamoxifen, and that tamoxifen is able to induce gastric cancer cell apoptosis. The expression levels of ER‑α36 were upregulated, and the growth of gastric cancer cells was increased, following treatment with 17β‑estradiol, thus suggesting that gastric cancer tumors are stimulated by estrogen.

Introduction

Epidemiological studies have suggested that gastric cancer is predominant in males, and that the ratio of morbidity of gastric cancer between male and female patients is 2:1–3:1 (13). The differences between the genders become negligible when female patients reach the menopause, and the morbidity associated with gastric cancer was reported to decrease in men who had been treated with estrogen for prostate carcinoma (13). These findings suggested that estrogen has a positive association with gastric cancer, although the underlying reasons are unclear. Estrogen acts by binding to its ligand (4,5). Previous studies reported that estrogen receptors (ERs) were expressed in the tumors of estrogen-independent organs, including the stomach, which indicates that, in these organs, the occurrence and development of a tumor is associated with estrogen (610).

ER-α36 is a novel ER variant identified by Professor Zhaoyi Wang (4), and whose molecular weight is 35.7 kDa. The difference between ER-α36 and the traditional ER is that ER-α36 lacks intrinsic transcriptional activity due to the loss of the activation function (AF) 1 and AF2 domains (4,5). Previous studies have demonstrated that ER-α36 is located at the cytomembrane, where is involved in various biological processes, including cell differentiation, proliferation and apoptosis, by mediating rapid signal transduction (5). There are few reports regarding the function, mechanism and clinical significance of ER-α36 in gastric cancer, although previous studies suggested that ER-α36 has a central role in balancing the proliferation and apoptosis of gastric cancer cells (1114). 17β-estradiol, which is a type of agonist of the ER, has a critical role in physiological processes by binding to its ligand to mediate the expression of various genes (1517). Conversely, tamoxifen, which is a non-steroidal triphenylethylene, affects the proliferation and apoptosis of cells by selectively competing with estrogen for the ER binding site, thus altering the expression levels of various cytokines (1820). The present study aimed to investigate the effects of 17β-estradiol and tamoxifen on the proliferation and apoptosis of gastric cancer cell lines cultured in vitro. In addition, the role of ER-α36 in the proliferation and apoptosis of gastric cancer cells was evaluated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR).

Materials and methods

Reagents

The BGC823 human gastric cancer and MCF-7 human breast adenocarcinoma cell lines were purchased from the Institute of Basic Medical Sciences at the Chinese Academy of Medical Sciences (Beijing, China). The MKN45 and SGC7901 human gastric cancer cell lines were donated by the Department of Immunology at Huazhong University of Science and Technology (Wuhan, China). Gibco® RPMI-1640 medium and fetal bovine serum (FBS) were purchased from Thermo Fisher Scientific, Inc., (Waltham, MA, USA). Trypsin was purchased from Sangon Biotech, Co., Ltd., (Shanghai, China) and glutamine was obtained from Ameresco, LLC (Solon, OH, USA). 17β-estradiol and tamoxifen were purchased from Sigma-Aldrich (Merck Millipore, Darmstadt, Germany). The water-soluble tetrazolium (WST)-1 kit was purchased from Beyotime Institute of Biotechnology (Haimen, China). The RT-PCR kit and PCR primers were purchased from Sangon Biotech, Co., Ltd. The THUNDERBIRD® SYBR® qPCR Mix was purchased from Toyobo Co., Ltd. (Osaka, Japan). The Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) Apoptosis Detection kit was purchased from Nanjing KeyGen Biotech Co., Ltd. (Nanjing, China).

Cell culture

BGC823, SGC7901, MKN45 and MCF-7 cells were cultured in RPMI-1640 medium supplemented with 10% FBS at 5% CO2 and 37°C. The cells were digested using trypsin and passaged upon reaching 70–80% confluence, followed by passaging every 2–3 days. BGC823 cells were digested and plated at a density of 1×105 cells/well onto Costar® 6-well plates in RPMI-1640 medium containing 10% FBS at 37°C. After 24 h, the culture medium was removed, and the cells were washed twice with phosphate-buffered saline, followed by culturing in phenol red-free RPMI-1640 medium containing 1% charcoal-stripped FBS for 6 h at 37°C. Subsequently, BGC823 cells were cultured in the same medium containing various concentrations of 17β-estradiol or tamoxifen for 24 or 48 h at 37°C. The MCF-7 control cells were cultured in medium containing 1:1,000 absolute ethyl alcohol at 37°C. 17β-estradiol and tamoxifen, which were dissolved in absolute ethyl alcohol and stored at a concentration of 10−5 and 10−2 mol/l, respectively, at −20°C, were diluted prior to use. BGC823 cells were treated with 10−12, 10−11 or 10−10 mol/l 17β-estradiol, or with 5×10−6 or 1×10−5 mol/l tamoxifen, for 24 or 48 h.

Nested RT-PCR

Total RNA was extracted from BGC823 cells upon reaching 80% confluence, using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol, and was reverse transcribed into complementary (c)DNA using an RT-PCR kit. The RT system (25 µl) consisted of the forward and reverse glyceraldehyde 3-phosphate dehydrogenase (GAPDH) primers, and the forward and reverse ER-α36 first and second nested primers. The primer sequences were as follows: GAPDH (452 bp) forward, 5′-ACCACAGTCCATGCCATCAC-3′ and reverse, 5′-TCCACCACCCTGTTGCTGTA-3; ER-α36 first nested primer (290 bp) forward, 5′-CAAGTGGTTTCCTCGTGTCTAAAG-3′ and reverse, 5′-TGTTGAGTGTTGGTTGCCAGG-3′; and ER-α36 second nested primer (219 bp) forward, 5′-TGGTTTCCTCGTGTCTAA-3′ and reverse, 5′-CAAAGTTTGTGGGTAGCT-3′. The first nested PCR system consisted of 2 µl cDNA, 2 µl first nested primer, 1.0 µl MgCl2, 2 µl 10X PCR buffer, 0.2 µl Taq polymerase (5 U/µl) and 13.2 µl double distilled (dd)H2O in a total volume of 20 µl. The cycling conditions were an initial denaturation for 1 min at 94°C, followed by 20 cycles consisting of denaturation at 94°C for 45 sec, an annealing step at 53°C for 45 sec and an extension step at 72°C for 60 sec. This was followed by a final extension at 72°C for 10 min. The second nested PCR system consisted of 2 µl cDNA, 2 µl each of the forward and reverse GAPDH primers, 1.5 µl MgCl2, 2 µl 10X PCR buffer (15 mmol/l), 0.1 µl Taq polymerase (5 U/µl) and 12.4 µl sterile ddH2O. The cycling conditions for this reaction system were an initial denaturation for 1 min at 94°C, followed by 15 cycles consisting of denaturation at 94°C for 45 sec, an annealing step at 55°C for 45 sec and an extension step at 72°C for 60 sec. This was followed by a final extension at 72°C for 10 min. PCR products were separated by 1.5% agarose gel electrophoresis and visualized by ethidium bromide staining under ultraviolet illumination.

WST-1 assay for assessment of cell proliferation

Exponential phase BGC823 cells were digested using trypsin and plated at a density of 3×103 cells/well onto 96-well plates. Each group included five parallel wells. To each well, 20 µl WST-1 solution was added and, after 24 or 48 h, the absorbance at 450 nm was measured using a microplate reader after culturing for a further 2 h in an incubator at 5% CO2 and 37°C.

RNA extraction and quantitative (q)PCR

Total RNA was extracted from gastric cancer cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Total RNA was reverse transcribed into cDNA using an RT-PCR kit. qPCR was performed on a StepOnePlus Real-Time PCR System (Thermo Fisher Scientific, Inc.) using the THUNDERBIRD® SYBR® qPCR Mix. The qPCR consisted of 35 cycles of 94°C for 30 sec, 56°C for 30 sec and 72°C for 30 sec. ER-α36 and β-actin primers were designed using Primer Premier 5.0 software (Premier Biosoft International, Palo Alto, CA, USA), and had the following sequences: ER-α36 forward, 5′-ACAAGTGGTTTCCTCGTGTCTAA-3′ and reverse, 5′-GGGTGTTGAGTGTTGGTTGC-3′; and β-actin forward, 5′-ATGATGATATCGCCGCGCTC-3′; and reverse, 5′-GTACATGGCTGGGGTGTTGA-3′. β-actin was used as an internal invariant endogenous control for qPCR. Expression levels were determined using the relative 2(−∆∆C(T)) method. All experiments were performed at least three times to ensure the reproducibility of the results.

Apoptosis assay

The cells were stained using the Annexin V-FITC/PI Apoptosis Detection kit, according to the manufacturer's protocol, and apoptotic cells, including early apoptotic cells (Annexin V+/PI) and necrotic or late apoptotic cells (Annexin V+/PI+), were measured by flow cytometry. Briefly, BGC823 cells were treated with 17β-estradiol or tamoxifen for 24 or 48 h, and subsequently, the cells were collected and resuspended in phenol red-free RPMI-1640 medium with 1% charcoal-stripped FBS at a density of 1×106 cells/ml. Next, the cells were stained with 5 µl Annexin V-FITC and 5 µl PI in 300 µl binding buffer (10 mmol/l 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid, pH 7.4, 140 mmol/l NaOH and 2.5 mmol/l CaCl2) for 15 min at room temperature in the dark. Quantification of apoptotic cells was performed using a flow cytometer (FACScan; Beckman Coulter, Inc., Brea, CA, USA).

Statistical analysis

Statistical analyses were performed using SPSS 12.0 software (SPSS, Inc., Chicago, IL, USA). Data are presented as the mean ± standard deviation of three replicate samples, and differences were compared using the Student's t-test or one-way analysis of variance. P<0.05 was considered to indicate a statistically significant difference. All experiments were performed at least three times to ensure the reproducibility of the results.

Results

Expression of ER-α36 in various human gastric cancer cell lines

The expression of ER-α36 messenger (m)RNA in BGC823, MKN45 and SGC7901 human gastric cancer cell lines was determined by semiquantitative nested RT-PCR, using the MCF-7 cell line as a positive control. Notably, ER-α36 mRNA expression was positive in the gastric cancer cell lines, and there was no detection of gene amplification (Fig. 1). These results suggest that ER-α36 is highly expressed in gastric cancer cells.

WST-1-based detection of cell proliferation in gastric cancer cells treated with an ER-α36 agonist or inhibitor

BGC823 cells were stimulated with various concentrations of 17β-estradiol or tamoxifen for 24 or 48 h, and WST-1 assays were then performed to assess cell proliferation. In the cells treated with 10−12 mol/l 17β-estradiol for 24 h, proliferation was increased by 13.2% compared with the control group, and the difference was significant (P=0.013; Fig. 2A). Conversely, the proliferation rates of the cells treated with 10−11 mol/l (P=0.0841) or 10−10 mol/l (P=0.0735) 17β-estradiol for 24 h were not significantly different from those exhibited by the control (P>0.05; Fig. 2A). After 48 h, the cell growth activity was increased by 30.5, 23.9 and 13.8% in the 10−12 mol/l (P=0.0015), 10−11 mol/l (P=0.0178) and 10−10 mol/l (P=0.0245) 17β-estradiol-treated groups, respectively, as compared with the control group, and the difference was significant (Fig. 2A). These results suggest that 17β-estradiol promotes the proliferation of gastric cancer cells in vitro.

There was no significant difference in the cell proliferation rate between BGC823 cells treated with 5×10−6 mol/l tamoxifen and the control group after 24 h (P=0.0724; Fig. 2B). After 24 h, the activity of the BGC823 cells treated with 1×10−5 mol/l tamoxifen was 56.4% of that displayed by the control group, which was significantly different (P=0.0233; Fig. 2B). The activity of the BGC823 cells treated with 5×10−6 mol/l was 61.5% of that of the control group after 48 h (P=0.0021), and the cell activity of the 1×10−5 mol/l group was 29.9% of that of the control group after 48 h (P=0.0059), which were significantly different (Fig. 2B). These results suggest that tamoxifen inhibits the growth of gastric cancer cells in vitro.

Gastric cancer cell apoptosis following treatment with an ER-α36 agonist or inhibitor

The apoptosis rate was significantly reduced in the BGC823 cells treated with 17β-estradiol for 24 h (10−12 mol/l, P=0.013; 10−11 mol/l, P=0.023; and 10−10 mol/l, P=0.017) and for 48 h (10−12 mol/l, P=0.002; 10−11 mol/l, P=0.011 and 10−10 mol/l, P=0.033) (Fig.3). Conversely, the rates of the apoptosis were significantly increased in the BGC823 cells as the tamoxifen concentration increased after 24 h (5×10−6 mol/l, P=0.002; and 5×10−5 mol/l, P=0.001) and after 48 h (5×10−6 mol/l, P=0.014; and 10−5 mol/l, P=0.0021), as compared with the control group. These results indicate that tamoxifen inhibits the growth of BGC823 cells, potentially by promoting gastric cancer cell apoptosis.

Alterations in the expression levels of ER-α36 following treatment of gastric cancer cells with 17β-estradiol or tamoxifen

BGC823 cells were treated with various concentrations of 17β-estradiol or tamoxifen for 24 or 48 h, and subsequently, the mRNA expression levels of ER-α36 were determined by RT-qPCR. After 24 h, the mRNA expression levels of ER-α36 in the 10−12 mol/l (P=0.024), 10−11 mol/l (P=0.0113) and 10−10 mol/l (P=0.0037) 17β-estradiol-treated groups exhibited a fold-change of 1.78, 1.44 and 1.15, respectively, as compared with those in the control (Fig. 4). After 48 h, the mRNA expression levels of ER-α36 in the 10−12 mol/l (P=0.0164), 10−11 mol/l (P=0.0342) and 10−10 mol/l (P=0.0198) 17β-estradiol-treated groups displayed a fold-change of 2.15, 1.56 and 1.26, respectively, as compared with those in the control (Fig. 4). Conversely, the mRNA expression levels of ER-α36 were decreased by 29.6% after 24 h in the 5×10−6 mol/l tamoxifen-treated group (P=0.0233), as compared with the mRNA levels observed in the control group, while those in the 1×10−5 mol/l tamoxifen-treated group (P=0.007) were decreased by 53.7%, as compared with the mRNA levels detected in the control group (Fig. 4). In the 5×10−6 mol/l tamoxifen-treated group (P=0.001), the mRNA expression levels of ER-α36 were decreased by 40.7% after 48 h, as compared with the levels measured in the control group, and those in the 1×10−5 mol/l tamoxifen-treated group (P=0.0153) were decreased by 64.8%, as compared with the levels displayed by the control group (Fig. 4). These results suggest that tamoxifen downregulates the expression of ER-α36 in gastric cancer cells.

Discussion

Gastric cancer is a type of gastrointestinal cancer and, as compared to other gastrointestinal cancers, there is no fundamental difference in its diagnosis and treatment (21). Recurrence is commonly observed in patients with advanced gastric cancer who have missed the opportunity for surgical resection and have instead undergone non-radical surgery. Although novel methods for the targeted treatment of gastric cancer are abundant, reports regarding the effects of these treatments have been less than satisfactory (21). It is not a coincidence that the incidence of gastric cancer is higher in males than in females (13). The present study aimed to investigate the role of estrogen in gastric cancer, in order to aid the development of better prophylactic and therapeutic strategies for gastric cancer and to improve the understanding of its ontogenesis.

Estrogen performs its biological functions by binding to the ER, which belongs to a family of receptors consisting of α and β subtypes. The ER exerts its function via the estrogen response element (ERE) and activating protein-1 in its target genes. It is considered that the ER is a ligand-dependent transcriptional activator (4,5). It has been demonstrated that membrane-bound ER quickly activates an intracellular second messenger to exert its biological functions in numerous cell types (5,2224).

ER-α36 is a novel subtype of ER that was discovered and cloned by Professor Zhaoyi Wang, and whose molecular weight is 35.7 kDa. As compared with the traditional ER, ER-α36 does not participate in the activation of the ERE due to loss of the AF1 and AF2 domains (4,5). However, ER-α36 possesses three myristoylation sites, including amino acids 25–30 [glycine (Gly)-valine-tryptophan-serine-cysteine-glutamate (Glu)], 76–81 [Gly-methionine (Met)-Met-lysine-Gly-Gly] and 171–176 [Glu-leucine (Leu)-Leu-threonine-asparagine-Leu], which are associated with the receptor's location at the membrane (5). Wang et al (4) reported that the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 was increased following stimulation of HEK-293 cells overexpressing ER-α36 with 17β-estradiol for 5 min. The level of ERK1/2 phosphorylation peaked at 30 min, and then commenced to decline, which may suggest that ER-α36 promotes the proliferation of cells by activating the mitogen-activated protein kinase (MAPK)/ERK signaling pathway (15).

The present study demonstrated that a low concentration of 17β-estradiol was able to promote the proliferation of gastric cancer cells in vitro, and that the proliferation of these cells was negatively correlated with the concentration of 17β-estradiol. Low-dose 17β-estradiol displayed an enhanced ability to promote the proliferation of gastric cancer cells, as compared with high concentrations of 17β-estradiol. These results were consistent with the findings of previous epidemiological studies, in which lower levels of estrogen in males were associated with higher morbidity of gastric cancer in males compared with females (1). In addition, in the present study, the proliferation of gastric cancer cells was inhibited by tamoxifen in a concentration- and time-dependent manner. These results suggested that gastric cancer cells were sensitive to estrogen, and that gastric cancer tumors are estrogen-responsive. Furthermore, the rate of apoptosis was increased in gastric cancer cells treated with tamoxifen, thus indicating that tamoxifen induces gastric cancer cell apoptosis in vitro.

The present study demonstrated that the proliferation of gastric cancer cells was increased to a greater extent following stimulation with lower concentrations of 17β-estradiol than using higher concentrations of this molecules. In addition, the current study demonstrated that the mRNA expression levels of ER-α36 were increased in the 17β-estradiol-treated group compared with the control group at all times, particularly when 17β-estradiol was obviously promoting cell proliferation. Tamoxifen was observed to induce gastric cancer cell apoptosis in vitro, and its concentration was negatively correlated with the expression of ER-α36. The apoptosis of gastric cancer cells was more obvious, and their mRNA expression levels of ER-α36 were decreased to a greater extent, which indicated that ER-α36 is important in the balance between proliferation and apoptosis of gastric cancer cells.

Since the ER is located at the cell membrane, ER-α36 may activate members of the MAPK family. MAPK is the main transducer of information from the cell surface to the nucleus (4). In eukaryotic cells, at least four types of MAPK signal transducers have been reported, including ERK, c-Jun N-terminal kinase (JNK), P38 and ERK5 (24). It has been hypothesized that JNK mediates the apoptosis of cells in the emergency response, thus inhibiting apoptosis and promoting proliferation when the ERK cascade is dominant, while initiating apoptosis when the JNK cascade is dominant (2533).

In conclusion, the present study demonstrated that low concentrations of 17β-estradiol were able to promote ER-α36 expression in gastric cancer cells, which in turn led to their increased proliferation, potentially via activation of the MAPK signaling pathway. Conversely, high concentrations of tamoxifen downregulated ER-α36 expression, which led to the apoptosis of gastric cancer cells. The aforementioned results indicated that a high concentration of tamoxifen could be important in the curative treatment of stomach cancer. Further studies are required to validate the results of the present study.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (Beijing, China; grant nos. 81470110, 81272754 and 30870981), the National Natural Science foundation of Guangxi (grant no. 0848014) and the Science Foundation of the Health Office of Hubei Province (Wuhan, China; grant no. WJ2015Z059).

References

1 

Chandanos E and Lagergren J: The mystery of male dominance in oesophageal cancer and the potential protective role of oestrogen. Eur J Cancer. 45:3149–3155. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Chung HW, Noh SH and Lim JB: Analysis of demographic characteristics in 3242 young age gastric cancer patients in Korea. World J Gastroenterol. 16:256–263. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Chandanos E and Lagergren J: Oestrogen and the enigmatic male predominance of gastric cancer. Eur J Cancer. 44:2397–2403. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Wang Z, Zhang X, Shen P, Loggie BW, Chang Y and Deuel TF: Identification, cloning, and expression of human estrogen receptor-alpha36, a novel variant of human estrogen receptor-alpha66. Biochem Biophys Res Commun. 336:1023–1027. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Wang Z, Zhang X, Shen P, Loggie BW, Chang Y and Deuel TF: A variant of estrogen receptor-{alpha}, hER-{alpha}36: Transduction of estrogen- and antiestrogen-dependent membrane-initiated mitogenic signaling. Proc Natl Acad Sci USA. 103:9063–9068. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Saqui-Salces M, Neri-Gomez T, Gamboa-Dominguez A, Ruiz-Palacios G and Camacho-Arroyo I: Estrogen and progesterone receptor isoforms expression in the stomach of Mongolian gerbils. World J Gastroenterol. 14:5701–5706. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Matsuyama S, Ohkura Y, Eguchi H, Kobayashi Y, Akagi K, Uchida K, Nakachi K, Gustafsson JA and Hayashi S: Estrogen receptor beta is expressed in human stomach adenocarcinoma. J Cancer Res Clin Oncol. 128:319–324. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Wang M, Pan JY, Song GR, Chen HB, An LJ and Qu SX: Altered expression of estrogen receptor alpha and beta in advanced gastric adenocarcinoma: Correlation with prothymosin alpha and clinicopathological parameters. Eur J Surg Oncol. 33:195–201. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Kameda C, Nakamura M, Tanaka H, Yamasaki A, Kubo M, Tanaka M, Onishi H and Katano M: Oestrogen receptor-alpha contributes to the regulation of the hedgehog signalling pathway in ERalpha-positive gastric cancer. Br J Cancer. 102:738–747. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Motohashi M, Wakui S, Muto T, Suzuki Y, Shirai M, Takahashi H and Hano H: Cyclin D1/cdk4, estrogen receptors alpha and β, in N-methyl-N'-nitro-N-nitrosoguanidine-induced rat gastric carcinogenesis: Immunohistochemical study. J Toxicol Sci. 36:373–378. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Wang X, Deng H, Zou F, Fu Z, Chen Y, Wang Z and Liu L: ER-α36-mediated gastric cancer cell proliferation via the c-Src pathway. Oncol Lett. 6:329–335. 2013.PubMed/NCBI

12 

Fu Z, Deng H, Wang X, Yang X, Wang Z and Liu L: Involvement of ER-α36 in the malignant growth of gastric carcinoma cells is associated with GRP94 overexpression. Histopathology. 63:325–333. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Wang X, Huang X, Fu Z, Zou F, Li Y, Wang Z and Liu L: Biphasic ER-α36-mediated estrogen signaling regulates growth of gastric cancer cells. Int J Oncol. 45:2325–2330. 2014.PubMed/NCBI

14 

Deng H, Huang X, Fan J, Wang L, Xia Q, Yang X, Wang Z and Liu L: A variant of estrogen receptor-alpha, ER-alpha36 is expressed in human gastric cancer and is highly correlated with lymph node metastasis. Oncol Rep. 24:171–176. 2010.PubMed/NCBI

15 

Nethrapalli IS, Tinnikov AA, Krishnan V, Lei CD and Toran-Allerand CD: Estrogen activates mitogen-activated protein kinase in native, nontransfected CHO-K1, COS-7, and RAT2 fibroblast cell lines. Endocrinology. 146:56–63. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Zappa L, Savady R and Sugarbaker PH: Gastric perforation following cytoreductive surgery with perioperative intraperitoneal chemotherapy. J Surg Oncol. 101:634–636. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Acconcia F and Marino M: The Effects of 17 β-estradiol in cancer are mediated by estrogen receptor signaling at the plasma membrane. Front Physiol. 2:302011. View Article : Google Scholar : PubMed/NCBI

18 

Zhou L, Cai B, Bao W, He YY, Chen XY, Yang YX, Liu XL and Wan XP: Crosstalk between estrogen receptor and mitogen-activated protein kinase signaling in the development and progression of endometrial cancer. Int J Gynecol Cancer. 21:1357–1365. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Anbalagan M, Carrier L, Glodowski S, Hangauer D, Shan B and Rowan BG: KX-01, a novel Src kinase inhibitor directed toward the peptide substrate site, synergizes with tamoxifen in estrogen receptor α positive breast cancer. Breast Cancer Res Treat. 132:391–409. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Mabuchi S, Ohmichi M, Kimura A, Ikebuchi Y, Hisamoto K, Arimoto-Ishida E, Nishio Y, Takahashi K, Tasaka K and Murata Y: Tamoxifen inhibits cell proliferation via mitogen-activated protein kinase cascades in human ovarian cancer cell lines in a manner not dependent on the expression of estrogen receptor or the sensitivity to cisplatin. Endocrinology. 145:1302–1313. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Lee JH, Kim JG, Jung HK, Kim JH, Jeong WK, Jeon TJ, Kim JM, Kim YI, Ryu KW, Kong SH, et al: Clinical practice guidelines for gastric cancer in Korea: An evidence-based approach. J Gastric Cancer. 14:87–104. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Fox EM, Andrade J and Shupnik MA: Novel actions of estrogen to promote proliferation: Integration of cytoplasmic and nuclear pathways. Steroids. 74:622–627. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Wong CW, McNally C, Nickbarg E, Komm BS and Cheskis BJ: Estrogen receptor-interacting protein that modulates its nongenomic activity-crosstalk with Src/Erk phosphorylation cascade. Proc Natl Acad Sci USA. 99:14783–14788. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Lei YY, Wang WJ, Mei JH and Wang CL: Mitogen-activated protein kinase signal transduction in solid tumors. Asian Pac J Cancer Prev. 15:8539–8548. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Shlevkov E and Morata G: A dp53/JNK-dependant feedback amplification loop is essential for the apoptotic response to stress in Drosophila. Cell Death Differ. 19:451–460. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Wu HC, Yang CY, Hung DZ, Su CC, Chen KL, Yen CC, Ho TJ, Su YC, Huang CF, Chen CH, et al: Nickel(II) induced JNK activation-regulated mitochondria-dependent apoptotic pathway leading to cultured rat pancreatic β-cell death. Toxicology. 289:103–111. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Pedram A, Razandi M, Sainson RC, Kim JK, Hughes CC and Levin ER: A conserved mechanism for steroid receptor translocation to the plasma membrane. J Biol Chem. 282:22278–22288. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Qu JL, Qu XJ, Zhao MF, Teng YE, Zhang Y, Hou KZ, Jiang YH, Yang XH and Liu YP: Gastric cancer exosomes promote tumour cell proliferation through PI3K/Akt and MAPK/ERK activation. Dig Liver Dis. 41:875–880. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Razandi M, Pedram A, Merchenthaler I, Greene GL and Levin ER: Plasma membrane estrogen receptors exist and functions as dimers. Mol Endocrinol. 18:2854–2865. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Lau WS, Chen WF, Chan RY, Guo DA and Wong MS: Mitogen-activated protein kinase (MAPK) pathway mediates the oestrogen-like activities of ginsenoside Rg1 in human breast cancer (MCF-7) cells. Br J Pharmacol. 156:1136–1146. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Wu RW, Yow CM, Wong CK and Lam YH: Photodynamic therapy (PDT)-Initiation of apoptosis via activation of stress-activated p38 MAPK and JNK signal pathway in H460 cell lines. Photodiagnosis Photodyn Ther. 8:254–263. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Wu WK, Cho CH, Lee CW, Wu YC, Yu L, Li ZJ, Wong CC, Li HT, Zhang L, Ren SX, et al: Macroautophagy and ERK phosphorylation counteract the antiproliferative effect of proteasome inhibitor in gastric cancer cells. Autophagy. 6:228–238. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Chuang SM, Wang IC and Yang JL: Roles of JNK, p38 and ERK mitogen-activated protein kinases in the growth inhibition and apoptosis induced by cadmium. Carcinogenesis. 21:1423–1432. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2017
Volume 13 Issue 1

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
Wang X, Chen Q, Huang X, Zou F, Fu Z, Chen Y, Li Y, Wang Z and Liu L: Effects of 17β-estradiol and tamoxifen on gastric cancer cell proliferation and apoptosis and ER-α36 expression. Oncol Lett 13: 57-62, 2017
APA
Wang, X., Chen, Q., Huang, X., Zou, F., Fu, Z., Chen, Y. ... Liu, L. (2017). Effects of 17β-estradiol and tamoxifen on gastric cancer cell proliferation and apoptosis and ER-α36 expression. Oncology Letters, 13, 57-62. https://doi.org/10.3892/ol.2016.5424
MLA
Wang, X., Chen, Q., Huang, X., Zou, F., Fu, Z., Chen, Y., Li, Y., Wang, Z., Liu, L."Effects of 17β-estradiol and tamoxifen on gastric cancer cell proliferation and apoptosis and ER-α36 expression". Oncology Letters 13.1 (2017): 57-62.
Chicago
Wang, X., Chen, Q., Huang, X., Zou, F., Fu, Z., Chen, Y., Li, Y., Wang, Z., Liu, L."Effects of 17β-estradiol and tamoxifen on gastric cancer cell proliferation and apoptosis and ER-α36 expression". Oncology Letters 13, no. 1 (2017): 57-62. https://doi.org/10.3892/ol.2016.5424