Open Access

Inhibition of Ep3 attenuates migration and promotes apoptosis of non‑small cell lung cancer cells via suppression of TGF‑β/Smad signaling

  • Authors:
    • Lei Li
    • Yanping Lv
    • Dengfeng Yan
  • View Affiliations

  • Published online on: September 4, 2018     https://doi.org/10.3892/ol.2018.9391
  • Pages: 5645-5654
  • Copyright: © Li 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

Non‑small cell lung cancer (NSCLC) is the most common cause of cancer‑associated mortality worldwide. Prostaglandin E2 (PGE2) regulates various biological processes, including invasion, proliferation and apoptosis. E‑prostanoid 3 (Ep3) is a PGE2 receptor, and the functional role of Ep3 in the progression of NSCLC remains unresolved. The present study investigated the effects of Ep3 in A549 cells and explored the underlying molecular mechanisms. The results revealed that the mRNA and protein expression levels of Ep3 were significantly upregulated in NSCLC tissues and A549 cells. Pharmacological inhibition of Ep3 or RNA interference against Ep3 attenuated the cell viability, migration and invasion, and promoted apoptosis in A549 cells. Ep3 deficiency also decreased the expression of transforming growth factor (TGF)‑β, phosphorylated (p)‑Smad2 and p‑Smad3. The transfection of TGF‑β overexpression plasmids reversed the effects of Ep3 deficiency on the cell viability and apoptosis in A549 cells. Finally, an in vivo experiment revealed that Ep3‑siRNA transfection strongly reduced the tumor growth and tumor volume. The Ep3‑siRNA transfection also inhibited tumor metastasis via suppression of the expression of metastasis‑associated proteins. Taken together, these findings indicate that inhibition of Ep3 attenuates the viability and migration, and promotes the apoptosis of NSCLC through suppression of the TGF‑β/Smad signaling pathway. Targeting of the Ep3/TGF‑β/Smad signaling pathway may be a novel therapeutic strategy for the prevention and treatment of NSCLC.

Introduction

Lung cancer is a leading cause of cancer-associated mortality worldwide (1,2). Among different types of lung cancer, non-small cell lung cancer (NSCLC) accounts for >80% of mortalities (2,3). Although several therapeutic strategies have been developed, including surgery, chemotherapy and radiation therapy, the prognosis of NSCLC patients remains poor, and the 5-year survival rate is <15% (4). NSCLC is difficult to treat due to malignant migration and metastasis, and the worsening physical conditions seriously affect the quality of life of patients (5,6). Therefore, understanding the underlying molecular mechanism of migration and metastasis in NSCLC may aid the development of more effective therapeutic strategies.

Prostaglandin E2 (PGE2) is the predominant product of arachidonic acid metabolism by cyclooxygenase-2 (COX-2). A number of studies have suggested that PGE2 serves functions in various biological processes, including inflammation, cell survival, migration and invasion (79). PGE2 has been demonstrated to bind to E prostanoid receptors on the surface of the cell membrane and thus regulate signaling pathways, including cAMP-PKA (10), PI3K/Akt and Ras/ERK (11), to exert their function. E-prostanoid 3 (Ep3) is a G protein-coupled receptor that serves an essential function in various diseases, including inflammation, hypertension and cancer (1215). Amano et al (16) suggested that Ep3 signaling on endothelial cells is essential for the matrix metalloproteinase (MMP)-9 upregulation that subsequently enhances tumor metastasis and angiogenesis. Fang et al (17) reported that Actinidia chinensis Planch root extract (acRoots) inhibits hepatocellular carcinoma cell invasion and metastasis via the inhibition of EP3 expression, resulting in decreased activation of vascular endothelial growth factor (VEGF), epidermal growth factor receptor, MMP2 and MMP9. Jiang et al (18) demonstrated that the inhibition of cell growth and induction of apoptosis by retinoic acid chalcone in colon cancer is mediated by inhibition of COX-2 expression, and subsequent inhibition of PGE2 and PGE2 receptors. The aforementioned studies suggest that the abnormal expression of Ep3 serves an important function in a number of cancer cells, and is associated with cell growth and metastasis. Yano et al (19) demonstrated that the expression of Ep3 may be a factor in the PGE2-mediated activation of the Ras signaling pathway in A549 cells. Yamaki et al (20) suggested that PGE2-dependent activation of Src signaling via Ep3 serves an important function in growth of A549 cells. These results suggest that Ep3 is involved in PGE2-mediated cellular processes in A549 cells. However, the functional effects and underlying molecular mechanisms of Ep3 in the development of NSCLC remain to be elucidated.

A number of studies have suggested that the regulation of Ep3 in cancer cells may be mediated by numerous signaling pathways, including extracellular signal-related kinase, phosphoinositide 3-kinase/protein kinase B and nuclear factor κ-light-chain-enhancer of activated B cells signaling (21,22). It has been reported that transforming growth factor (TGF)-β signaling serves a function in numerous types of cancer by regulating a variety of cellular events, including proliferation, migration and apoptosis (23,24). By binding to its receptor, TGF-β is able to activate Smad2 and Smad3, and initiate their translocation to the nucleus by forming a trimer with Smad4, to regulate the expression of TGF-β dependent genes (25,26). Several investigations have demonstrated that the activation of TGF-β, and the subsequent phosphorylation of Smad2 and Smad3 promote the invasion and migration of lung cancer cells (27,28), suggesting that TGF-β/Smad signaling is involved in the regulation of lung cancer cells. A previous study reported that the inhibition of Ep3 attenuates pulmonary hypertension through suppression of Rho/TGF-β1 signaling (29), suggesting that the regulation of Ep3 may be associated with TGF-β signaling. Thus, establishing whether TGF-β signaling is involved in the effects of Ep3 in lung cancer cells is of interest.

In the present study, the expression of Ep3 in NSCLC tissues and A549 cells was evaluated. The effects of Ep3 on the cell viability, migration, invasion and apoptosis of A549 cells were investigated, and the underlying molecular mechanisms of each were explored. It was hypothesized that the inhibition of Ep3 may suppress the cell viability, migration and invasion, and promote cell apoptosis of A549 cells.

Materials and methods

Tissue specimens

A total of 17 NSCLC tissues and corresponding adjacent normal lung tissues were obtained from patients from Zhoukou Central Hospital (Zhoukou, China) who underwent curative resection for NSCLC between August 2015 and October 2016. Among them, twelve were males, and five were females. Their ages were between 47 and 68 years, and the mean age was 61 years. All of the patients provided written informed consent, and the present study was approved by the Ethics Committee of Zhoukou Central Hospital.

Cell culture and L-798106 treatment

The A549 NSCLC cell line and HPAEpiC human alveolar epithelial cell line were purchased from the American Type Culture Collection (Manassas, VA, USA). The cells were cultured in RPMI-1640 (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and maintained at 37°C in a humidified atmosphere with 5% CO2. A549 cells were treated with 1 µM Ep3 antagonist L-798106 (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) for 48 h in the following experiments.

Cell transfection

Ep3 siRNA (si-Ep3) and negative control siRNA (si-control) were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). The TGF-β cDNA was cloned into pcDNA 3.1 vector (Invitrogen; Thermo Fisher Scientific, Inc.). The constructs were verified by DNA sequencing. The construction and verification of the pcDNA 3.1-TGF-β plasmids was performed by Generay Biotech Co., Ltd (Shanghai, China). Cells were seeded into 6-well, 24-well or 96-well plates according to experimental requirements. When the cells reached 70–80% confluence, Ep3 siRNA, negative control siRNA or plasmids were transfected into cells using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturers' protocol. Then, 6 h after transfection, the medium was changed to RPMI-1640 medium supplemented with 10% FBS.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total mRNA was extracted from tissues and cells using TRIzol® (Thermo Fisher Scientific, Inc.) according to the manufacturers' protocol. mRNA was reverse transcribed to cDNA using a PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China) according to the manufacturers' protocol. The mRNA levels of Ep3 and β-actin were measured using SYBR Premix Real-Time PCR reagent (Takara Biotechnology Co., Ltd.) according to the manufacturer's protocol. The primers used in the present study were as follows: Ep3 forward, 5′-TCCTTCCTAATCGCCGTTC-3′ and reverse, 5′-CTCCGCTTCAGGTTGTTCAT-3′; β-actin forward, 5′-CCTGGACTTCGAGCAAGAGA-3′ and reverse, 5′-ACTTGCGCTCAGGAGGAGCA-3′. The PCR conditions were as follows: 10 min at 95°C, followed by 40 cycles of 15 sec at 95°C, 30 sec at 60°C and 30 sec at 72°C, and a final extension for 5 min at 72°C. The relative expression level of Ep3 was normalized to β-actin levels. The relative expression levels were calculated using the 2−ΔΔCt method (30).

Western blot analysis

The protein was extracted from tissues and cells using radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Haimen, China) supplemented with a PMSF protease inhibitor (Sigma-Aldrich; Merck KGaA). Protein concentration was quantified using a bicinchoninic acid protein assay kit (Pierce; Thermo Fisher Scientific, Inc.). A total of 40 µg protein lysates were separated via 10–12% SDS-PAGE and transferred onto polyvinylidene difluoride membranes. Membranes were blocked in 5% bovine serum albumin (Sigma Aldrich; Merck KGaA) at room temperature for 1 h, and subsequently immunoblotted with the following primary antibodies according to the recommended dilution concentration: Anti-Ep3 (1:500; cat. no. P8372; Sigma-Aldrich; Merck KGaA), anti-caspase-3 (1:1,000; cat. no. 9662), anti-B cell lymphoma (Bcl)-2 (1:1,000; cat. no. 2872), anti-Bcl-associated × protein (Bax; 1:1,000; cat. no. 2772; all Cell Signaling Technology, Inc., Danvers, MA, USA), anti-MMP-9 (1:500; cat. no. ab58803; Abcam, Cambridge, UK), anti-VEGF (1:1,000; cat. no. V6627; Sigma-Aldrich; Meck KGaA), anti-TGF-β1 (1:1,000; cat. no. SAB4502954; Sigma-Aldrich; Merck KGaA), anti-Smad-2 (1:1,000; cat. no. ab63576), anti-Smad-3 (1:2,000; cat. no. ab40854), anti-phosphorylated (p)-Smad2 (1:800; cat. no. ab53100), anti-p-Smad3 (1:2,000; cat. no. ab52903) and anti-GAPDH (1:2,000; cat. no. ab8245; all Abcam) at 4°C overnight. Then, membranes were incubated with corresponding horseradish peroxidase (HRP)-conjugated secondary antibodies: Anti-rabbit IgG (1:10,000; cat. no. 7074), anti-mouse IgG (1:10,000; cat. no. 7076; both Cell Signaling Technology, Inc.) and donkey anti-goat IgG (1:10,000; cat. no. ab6885; Abcam) at room temperature for 1 h. GAPDH was used as the loading control. The blots were detected using an enhanced chemiluminescence solution (EMD Millpore, Billerica, MA, USA), followed by observing the signals under the Molecular Imager ChemiDoc XRS Gel Imagine System (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The relative intensities of protein bands were analyzed using Image lab analysis software (version 4.0; Bio-Rad Laboratories, Inc.).

MTT assay

Following transfection with Ep3 siRNA or treatment with L-798106 for 48 h, the cells were treated with 100 µg/ml MTT for 4 h at 37°C, followed by dissolving of the formazan crystals with 100 µl of dimethyl sulfoxide. The absorbance value was determined at a wavelength of 490 nm.

Wound-healing assay

Following transfection with Ep3 siRNA for 6 h, artificial wounds were created by scraping using a sterile 200-µl pipette tip. In a separate assay, following washing with phosphate-buffered saline (PBS), the cells were cultured in RPMI-1640 medium for 48 h. Artificial wounds were created followed by treatment with L-798106 for 48 h to measure the effects of L-798106. Images of cell migration were obtained at 0 h and 48 h using an inverted light microscope (magnification, ×40). The distance between the two edges of a wound was calculated using ImageJ software (version 1.8.0; National Institutes of Health, Bethesda, MD, USA). The width of wound at 0 h was regarded as 100% and the rate of wound healing=(0 h width of the wound-48 h width of the wound)/(0 h width of the wound).

Transwell assay

Following transfection with Ep3 siRNA or treatment with L-798106 for 48 h, the cells were harvested and washed twice with PBS. A total of 200 ml cell suspension in serum-free medium (1×105 cells) was seeded in the upper compartment of Transwell plates (with Matrigel-coated membrane; 8-µm pore size; BD Biosciences, Franklin Lakes, NJ, USA). In the lower chamber, 600 µl medium containing 10% FBS was added. After 24 h incubation, cells remaining on the upper membranes were removed by cotton swabs. The migrated cells were fixed with 4% paraformaldehyde for 30 min, stained with 0.4% crystal violet for 20 min at room temperature, and then counted under an inverted light microscope (magnification, ×200).

Flow cytometry assay

Following transfection with Ep-3 siRNA or treatment with L-798106 for 48 h, the cells were harvested and stained with Annexin V/propidium iodide double staining kit (BD Biosciences) according to the manufacturer's protocol. Apoptotic cells were assessed by flow cytometry on an FC500 flow cytometer (Beckman Coulter, Inc., Brea, CA, USA).

NSCLC xenografts

Specific pathogen-free (SPF) athymic nude mice (all male; age range, 6–8 weeks; weighing 20–25 g), purchased from the Experimental Animal Center of the Southwest Medical University (Sichuan, China), were housed and manipulated according to protocols approved by the Experimental Animal Center of the Southwest Medical University. They were maintained at a constant humidity (60±5%) and temperature (20±1°C) and kept on a 12-h light/dark cycle. All animals were provided with food and water ad libitum. In order to research tumorigenicity of Ep3 in vivo, A549 cells were transfected with or without Ep3 siRNA. Each mouse was subcutaneously inoculated with either 1×107 Ep3 siRNA transfected A549 cells or control cells (fluorescent-labeled) in 50% Matrigel (BD Biosciences) (Each group contained 10 mice). Following the development of a palpable tumor, the tumor volume was monitored every 5 days and assessed by measuring the two perpendicular dimensions using a caliper with the formula: (a × b2)/2, where a is the larger and b is the smaller dimension of the tumor. At 30 days after inoculation, the mice were sacrificed and tumor weights were assessed. Tumors from each mouse were randomly selected for immunohistochemical (IHC) analysis. All animal procedures were performed according to relevant national and international guidelines and were approved by the Animal Experimental Ethical Committee.

Immunohistochemistry

NSCLC tumor tissues were fixed in 10% formalin at 4°C for 24 h, and then embedded in paraffin for IHC analysis. Briefly, 5 µm-thick paraffin sections were deparaffinized in xylene, and gradually rehydrated in 100, 95 and 75% ethanol. In order to quench the activity of endogenous peroxidase, the tissue sections were incubated in 30% H2O2 for 30 min at room temperature. Following antigen retrieval in heated 10 mM citrate buffer for 10 min at room temperature, the tissue sections were immunostained with mouse anti-human MMP-9 primary antibody (cat. no. ab58803; 1:500; Abcam) overnight at 4°C. Tissues were incubated with the corresponding anti-mouse HRP-conjugated secondary antibody for 1 h at room temperature. Images were viewed under a light microscope (magnification, ×200).

Statistical analysis

Statistical analyses were performed using GraphPad Prism 5.0 (GraphPad Software, Inc., La Jolla, CA, USA). The data were analyzed by one-way analysis of variance followed by Bonferroni's post hoc test for multiple comparisons. The Student's t-test was used for comparisons between two groups. Each experiment was performed in triplicate and the values are presented as the mean ± standard error of the mean. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of Ep3 is increased in NSCLC tissues and A549 cells

The expression of Ep3 was measured by RT-qPCR and western blotting in NSCLC tissues, A549 cells and HPAEpiC cells. As presented in Fig. 1A and B, the mRNA and protein expression levels were significantly increased in the NSCLC tissues compared with the normal tissues (P<0.05). The same results were observed in the NSCLC cell line, whereby the expression of Ep3 was significantly upregulated in A549 cells compared with HPAEpiC cells (P<0.05; Fig. 1C and D).

Inhibition of Ep3 inhibits the cell viability of A549 cells

The elevated expression of Ep3 in NSCLC tissues and A549 cells suggest that Ep3 may serve an important function in the development of NSCLC. To explore the biological role of Ep3, A549 cells were transfected with Ep3 siRNA or its inhibitor L-798106 to suppress the expression of Ep3. As presented in Fig. 2A and B, the mRNA and protein expression levels of Ep3 were significantly downregulated in L-798106 and si-Ep3 groups as compared with the control A549 cells (P<0.05). Subsequently, it was determined whether Ep3 was involved in the regulation of the cell viability in A549 cells using an MTT assay. As presented in Fig. 2C, compared with control group, the cells treated with L-798106 or Ep3 siRNA exhibited significantly lower cell viability rates (P<0.05). These data suggest that Ep3 deficiency may be associated with the growth activity of A549 cells.

Ep3 deficiency promotes the cell apoptosis of A549 cells

To further investigate the effects of Ep3 on the apoptosis of A549 cells, a flow cytometry assay was performed. As presented in Fig. 3A, the proportion of apoptotic cells was significantly increased in L-798106 and si-Ep3 groups compared with control group (P<0.05). Additionally, western blotting was performed to assess the expression of apoptosis-associated proteins. As presented in Fig. 3B-D, compared with the control group, Ep3 deficiency by L-798106 or Ep3 siRNA significantly upregulated the expression of caspase-3 and Bax, and downregulated the expression of Bcl-2 (P<0.05).

Ep3 deficiency inhibits the cell migration and invasion of A549 cells

To determine whether Ep3 was involved in the migration and invasion of A549 cells, wound-healing and Transwell assays were performed. As presented in Fig. 4, the abilities of cell migration and invasion in L-798106 and si-Ep3 groups were significantly suppressed compared with that in control cells (P<0.05). Furthermore, the expression level of matrix metalloproteinase MMP-9 and VEGF was significantly reduced in L-798106 and si-Ep3 groups compared with control group (P<0.05). These results suggest that Ep3 deficiency may be a suppressor of cell motility of NSCLC. Together, these data suggest that inhibition of Ep3 may be an effective approach in the treatment of NSCLC due to its functions in the cell viability, migration, invasion and apoptosis in NSCLC cells.

Ep3 deficiency suppresses the growth of A549 cells via inhibition of the TGF-β/Smad signaling pathway

In the present study, it was demonstrated that Ep3 deficiency was involved in the regulation of cell viability, migration, invasion and apoptosis of A549 cells, and the underlying molecular mechanisms were investigated further. A number of studies have reported that TGF-β/Smad signaling is an important pathway in the progression of NSCLC (3133), therefore whether TGF-β/Smad signaling mediated the roles of Ep3 was further investigated in A549 cells. The expression of pathway-associated proteins was evaluated using western blotting. As presented in Fig. 5A-C, treatment with L-798106 or Ep3 siRNAs significantly inhibited the expression of TGF-β, p-Smad2 and p-Smad3 inA549 cells, suggesting that TGF-β/Smad signaling may mediate the functions of Ep3 in A549 cells (P<0.05). To address the direct involvement of TGF-β/Smad signaling in the regulation of Ep3 in A549 cells, pcDNA3.1-TGF-β plasmids were introduced into A549 cells prior to the treatment of L-798106 or Ep3 siRNA. As presented in Fig. 5A-C, compared with the L-798106 group, transfection of pcDNA3.1-TGF-β plasmids significantly elevated the levels of TGF-β, p-Smad2 and p-Smad3 (P<0.05). The same results were observed in the TGF-β+si-Ep3 group as compared with the si-Ep3 group (P<0.05). The MTT assay revealed that the reduction in cell viability caused by Ep3 knockdown was partially reversed in pcDNA3.1-TGF-β-transfected A549 cells (P<0.05; Fig. 5D). Furthermore, compared with the L-798106 or si-Ep3 group, cell apoptosis was reduced when TGF-β was overexpressed in these cells (P<0.05; Fig. 5E). These data suggest that cell viability inhibition and cell apoptosis induced by Ep3 knockdown were at least partially associated with the inhibition of TGF-β/Smad signaling.

Ep3 deficiency inhibits tumor growth and metastasis in vivo

To investigate the effects of Ep3 deficiency on NSCLC cells growth and invasion in vivo, a xenograft mouse model was established by subcutaneous injection of A549 cells to SPF nude mice. The mean tumor volume was significantly reduced in the si-Ep3 group compared with the control group after 25 days (P<0.05; Fig. 6A and B). Furthermore, the expression level of MMP-9 was markedly decreased in the si-Ep3 group compared with the control group using IHC analysis (Fig. 6C). In addition, the levels of Ep3, TGF-β, p-Smad2 and p-Smad3 were markedly reduced in the si-Ep3 group compared with the control group (Fig. 6D). Combined with the results of the aforementioned experiments, it is hypothesized that Ep3 deficiency inhibits tumor growth and metastasis in vivo.

Discussion

NSCLC remains a threat to public health worldwide. Despite advances in therapeutic modalities, little progress has been made in improving the quality of life and survival in patients with advanced and metastatic NSCLC. Therefore, an increased understanding of the underlying molecular mechanism of cancer proliferation and migration is essential for the development of novel effective therapeutic strategies against NSCLC. In the present study, it was identified that inhibition of Ep3 significantly attenuated the viability and migration, and promoted the apoptosis of A549 cells through suppression of the TGF-β/Smad signaling pathway, suggesting that inhibition of Ep3 may be a novel therapeutic strategy for the treatment of NSCLC.

Increased expression of COX-2 and the consequent upregulation of PGE2 have been implicated in the pathogenesis of several types of cancer, including colon, breast, and lung cancer due to their roles in regulating cell growth and invasion (3438). Ep3 is a receptor via which PGE2 exerts its cellular effects and has been reported to be involved in the progression of lung cancer (20,23,24,39). However, the effects and detailed molecular mechanisms of Ep3 in lung cancer remain unresolved. In the present study, it was identified that Ep3 expression was significantly upregulated in NSCLC lung tissues and A549 cells, suggesting that Ep3 may function as a tumor promoter in NSCLC. To investigate the effects of Ep3 in the development of NSCLC, A549 cells were transfected with Ep3 siRNA or treated with Ep3 inhibitor L-798106 to downregulate the expression of Ep3. MTT assays, wound-healing assays and Transwell assays revealed that pharmacological inhibition with L-798106 and Ep3 siRNA transfection significantly reduced the cell viability, and the migration and invasion abilities of A549 cells. Furthermore, flow cytometry analysis demonstrated that the proportion of apoptotic cells was significantly increased when A549 cells were treated with L-798106 and Ep3 siRNA. Additionally, the expression levels of several apoptosis-associated proteins were assessed using western blotting, and the results revealed that the protein levels of Caspase-3 and Bax had were significantly upregulated, whereas the expression of Bcl-2 was significantly downregulated when the expression of Ep3 was inhibited by L-798106 or Ep3 siRNA. These results demonstrated that decreased Ep3 expression may be a factor contributing to inhibition of the development of NSCLC, including suppressing proliferation and invasion, and promoting apoptosis of lung cancer cells.

The activation of TGF-β signaling, as well as the subsequent phosphorylation of Smad2 and Smad3 have been reported to serve an important function in the regulation of expression of numerous genes, and thus regulates cellular responses, including proliferation, migration and apoptosis, in various types of cancer, including lung cancer (40,41). This suggests that TGF-β/Smad signaling may be associated with the development of lung cancer. Therefore, approaches to harbor this signaling may be valuable for treating lung cancer. A previous study reported that inhibition of Ep3 attenuates pulmonary hypertension through suppression of Rho/TGF-β1 signaling (33), indicating that the regulation of Ep3 may be associated with TGF-β signaling. It was therefore investigated whether TGF-β signaling is involved in the effects of Ep3 in lung cancer cells. In the present study, treatment with L-798106 or Ep3 siRNA significantly inhibited the expression of TGF-β, p-Smad2 and p-Smad3 in A549 cells, suggesting that TGF-β/Smad signaling is involved in the regulation of Ep3 in A549 cells. To further confirm these results, pcDNA3.1-TGF-β plasmids were introduced into A549 cells prior to treatment with L-798106 or Ep3 siRNA. MTT and flow cytometric assays revealed that the inhibition of cell viability and the promotion of cell apoptosis induced by Ep3 knockdown were partially reversed by the overexpression of TGF-β. These data suggest that the Ep3-mediated biological effects in A549 cells are at least partially mediated by the TGF-β/Smad signaling pathway.

In conclusion, the results of the present study suggest that inhibition of Ep3 attenuates the viability and migration, and promotes the apoptosis of A549 cells, which was associated with the suppression of TGF-β/Smad signaling. The current study provides a novel insight into the underlying molecular mechanism associated with Ep3-mediated effects in NSCLC cells, and suggests that targeting the Ep3/TGF-β/Smad signaling pathway provides novel therapeutic strategies for the treatment of NSCLC. Further studies are required to verify these conclusions in vivo.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LL and YL made substantial contributions to the study conception and design. LL and DY acquired data and performed analysis and interpretation of the data. LL drafted the manuscript. YL revised the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All of the patients provided written informed consent and approval was provided by the Ethics Committee of Zhoukou Central Hospital for the human studies. The animal studies were approved by the Experimental Animal Center of the Southwest Medical University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Travis WD, Travis LB and Devesa SS: Lung cancer. Cancer. 75:191–202. 1995. View Article : Google Scholar : PubMed/NCBI

2 

Novello S, Barlesi F, Califano R, Cufer T, Ekman S, Levra MG, Kerr K, Popat S, Reck M, Senan S, et al: Metastatic non-small-cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 27:v1–v27. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Liu G, Pei F, Yang F, Li L, Amin AD, Liu S, Buchan JR and Cho WC: Role of autophagy and apoptosis in non-small-cell lung cancer. Int J Mol Sci. 18:E3672017. View Article : Google Scholar : PubMed/NCBI

4 

Payandeh M, Sadeghi M, Sadeghi E and Aeinfar M: The survival of lung cancer based on type of treatment in non-small-cell lung cancer: A review of literatureInternational Congress on Anti-Cancer Treatment. Paris, France: 2015

5 

Lim BJ, Jung SS, Choi SY and Lee CS: Expression of metastasis-associated molecules in non-small cell lung cancer and their prognostic significance. Mol Med Rep. 3:43–49. 2010.PubMed/NCBI

6 

Müllertidow C, Diederichs S, Bulk E, Pohle T, Steffen B, Schwäble J, Plewka S, Thomas M, Metzger R, Schneider PM, et al: Identification of metastasis-associated receptor tyrosine kinases in non-small cell lung cancer. Cancer Res. 65:1778–1782. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Schoenberger SD, Kim SJ, Sheng J, Rezaei KA, Lalezary M and Cherney E: Increased prostaglandin E2 (PGE2) levels in proliferative diabetic retinopathy, and correlation with VEGF and inflammatory cytokines. Invest Ophthalmol Vis Sci. 53:5906–5911. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Chell S, Kaidi A, Kadi A, Williams AC and Paraskeva C: Mediators of PGE2 synthesis and signalling downstream of COX-2 represent potential targets for the prevention/treatment of colorectal cancer. Biochim Biophys Acta. 1766:104–119. 2006.PubMed/NCBI

9 

Li Z, Zhang Y, Wanju K and Yehia D: PGE2 promotes renal carcinoma cell invasion through activated RalA. Oncogene. 32:1408–1415. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Kim CH, Park YG, Noh SH and Kim YK: PGE2 induces the gene expression of bone matrix metalloproteinase-1 in mouse osteoblasts by cAMP-PKA signaling pathway. Int J Biochem Cell Biol. 37:375–385. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Dajani OF, Meisdalen K, Guren TK, Aasrum M, Tveteraas IH, Lilleby P, Thoresen GH, Sandnes D and Christoffersen T: Prostaglandin E2 upregulates EGF-stimulated signaling in mitogenic pathways involving Akt and ERK in hepatocytes. J Cell Physiol. 214:371–380. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Goulet JL, Pace AJ, Key ML, Byrum RS, Nguyen M, Tilley SL, Morham SG, Langenbach R, Stock JL, McNeish JD, et al: E-prostanoid-3 receptors mediate the proinflammatory actions of prostaglandin E2 in acute cutaneous inflammation. J Immunol. 173:1321–1326. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Chen L, Miao Y, Zhang Y, Dou D, Liu L, Tian X, Yang G, Pu D, Zhang X, Kang J, et al: Inactivation of the E-prostanoid 3 receptor attenuates the angiotensin II pressor response via decreasing arterial contractility. Arterioscler Thromb Vasc Biol. 32:3024–3032. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Miyata Y, Ohba K, Matsuo T, Watanabe S, Hayashi T, Sakai H and Kanetake H: Tumor-associated stromal cells expressing E-prostanoid 2 or 3 receptors in prostate cancer: Correlation with tumor aggressiveness and outcome by angiogenesis and lymphangiogenesis. Urology. 81:136–142. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Reader J, Holt D and Fulton A: Prostaglandin E 2 EP receptors as therapeutic targets in breast cancer. Cancer Metastasis Rev. 30:449–463. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Amano H, Ito Y, Suzuki T, Kato S, Matsui Y, Ogawa F, Murata T, Sugimoto Y, Senior R, Kitasato H, et al: Roles of a prostaglandin E-type receptor, EP3, in upregulation of matrix metalloproteinase-9 and vascular endothelial growth factor during enhancement of tumor metastasis. Cancer Sci. 100:2318–2324. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Fang T, Hou J, He M, Wang L, Zheng M, Wang X and Xia J: Actinidia chinensis Planch root extract (acRoots) inhibits hepatocellular carcinoma progression by inhibiting EP3 expression. Cell Biol Toxicol. 32:499–511. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Jiang C, Wang Q, Xu Z, Li WS, Chen C, Yao XQ and Liu FK: Cyclooxygenase-2 knockdown using retinoic acid chalcone (RAC), a promising therapeutic strategy for colon cancer. Am J Cancer Res. 5:2012–2021. 2015.PubMed/NCBI

19 

Yano T, Zissel G, Muller-Qernheim J, Shin Jae S, Satoh H and Ichikawa T: Prostaglandin E-2 reinforces the activation of Ras signal pathway in lung adenocarcinoma cells via EP3. FEBS Lett. 518:154–158. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Yamaki T, Endoh KM, Miyahara M, Nagamine I, Thi Thu, Huong N, Sakurai H, Pokorny J and Yano T: Prostaglandin E2 activates Src signaling in lung adenocarcinoma cell via EP3. Cancer Lett. 214:115–120. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Israel DD and Regan JW: EP(3) prostanoid receptor isoforms utilize distinct mechanisms to regulate ERK 1/2 activation. Biochim Biophys Acta. 1791:238–245. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Wang P, Zhu F, Lee NH and Konstantopoulos K: Shear-induced interleukin-6 synthesis in chondrocytes: Roles of E Prostanoid (EP) 2 and EP3 in cAMP/protein KINASE A- and PI3-K/Akt-dependent NF-κB activation. J Biol Chem. 285:24793–24804. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Levy L and Hill CS: Alterations in components of the TGF-beta superfamily signaling pathways in human cancer. Cytokine Growth Factor Rev. 17:41–58. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Massagué J: TGFbeta in cancer. Cell. 134:215–230. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Massagué J and Wotton D: Transcriptional control by the TGF-beta/Smad signaling system. EMBO J. 19:1745–1754. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Massagué J: How cells read TGF-beta signals. Nat Rev Mol Cell Biol. 1:169–178. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Zu L, Xue Y and Wang J, Fu Y, Wang X, Xiao G, Hao M, Sun X, Wang Y, Fu G and Wang J: The feedback loop between miR-124 and TGF-β pathway plays a significant role on non-small cell lung cancer metastasis. Carcinogenesis. 37:333–343. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Wang Y, Yi J, Chen X, Zhang Y, XU M and Yang Z: The regulation of cancer cell migration by lung cancer cell-derived exosomes through TGF-β and IL-10. Oncol Lett. 11:1527–1530. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Lu A, Zuo C, He Y, Chen G, Piao L, Zhang J, Xiao B, Shen Y, Tang J, Kong D, et al: EP3 receptor deficiency attenuates pulmonary hypertension through suppression of Rho/TGF-β1 signaling. J Clin Invest. 125:1228–1242. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Zhang JX, Zhai JF, Yang XT and Wang J: MicroRNA-132 inhibits migration, invasion and epithelial-mesenchymal transition by regulating TGFβ1/Smad2 in human non-small cell lung cancer. Eur Rev Med Pharmacol Sci. 20:3793–3801. 2016.PubMed/NCBI

32 

Da C, Liu Y, Zhan Y, Liu K and Wang R: Nobiletin inhibits epithelial-mesenchymal transition of human non-small cell lung cancer cells by antagonizing the TGF-β1/Smad3 signaling pathway. Oncol Rep. 35:2767–2774. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Wang X, Li M, Hu M, Wei P and Zhu W: BAMBI overexpression together with β-sitosterol ameliorates NSCLC via inhibiting autophagy and inactivating TGF-β/Smad2/3 pathway. Oncol Rep. 37:3046–3054. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Prescott SM and Fitzpatrick FA: Cyclooxygenase-2 and carcinogenesis. Biochim Biophys Acta. 1470:M69–M78. 2000.PubMed/NCBI

35 

Dufour M, Faes S, Dormond-Meuwly A, Demartines N and Dormond O: PGE2-induced colon cancer growth is mediated by mTORC1. Biochem Biophys Res Commun. 451:587–591. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Bocca C, Ievolella M, Autelli R, Motta M, Mosso L, Torchio B, Bozzo F, Cannito S, Paternostro C, Colombatto S, et al: Expression of Cox-2 in human breast cancer cells as a critical determinant of epithelial-to-mesenchymal transition and invasiveness. Exp Opin Ther Targets. 18:121–135. 2014. View Article : Google Scholar

37 

Sharma S, Yang SC, Zhu L, Reckamp K, Gardner B, Baratelli F, Huang M, Batra RK and Dubinett SM: Tumor cyclooxygenase-2/prostaglandin E2-dependent promotion of FOXP3 expression and CD4+ CD25+ T regulatory cell activities in lung cancer. Cancer Res. 65:5211–5220. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Pan J, Yang Q, Shao J, Zhang L, Ma J, Wang Y, Jiang BH, Leng J and Bai X: Cyclooxygenase-2 induced β1-integrin expression in NSCLC and promoted cell invasion via the EP1/MAPK/E2F-1/FoxC2 signal pathway. Sci Rep. 6:338232016. View Article : Google Scholar : PubMed/NCBI

39 

Amano H, Hayashi I, Endo H, Kitasato H, Yamashina S, Maruyama T, Kobayashi M, Satoh K, Narita M, Sugimoto Y, et al: Host prostaglandin E2-EP3 signaling regulates tumor-associated angiogenesis and tumor growth. J Exp Med. 197:221–232. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Shen JL, Yan CH, Liu Y, Yan XQ, Zhang XL, Jin Y, Zhang KF, Sang ZF, Zhang GY, Li P and Fu SB: Studies of TGF-β/Smads expression in lung cancer. Yi Chuan Xue Ba. 30:681–686. 2003.(In Chinese).

41 

Liu RY, Zeng Y, Lei Z, Wang L, Yang H, Liu Z, Zhao J and Zhang HT: JAK/STAT3 signaling is required for TGF-β-induced epithelial-mesenchymal transition in lung cancer cells. Int J Oncol. 44:1643–1651. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 16 Issue 5

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
Li L, Lv Y and Yan D: Inhibition of Ep3 attenuates migration and promotes apoptosis of non‑small cell lung cancer cells via suppression of TGF‑β/Smad signaling. Oncol Lett 16: 5645-5654, 2018
APA
Li, L., Lv, Y., & Yan, D. (2018). Inhibition of Ep3 attenuates migration and promotes apoptosis of non‑small cell lung cancer cells via suppression of TGF‑β/Smad signaling. Oncology Letters, 16, 5645-5654. https://doi.org/10.3892/ol.2018.9391
MLA
Li, L., Lv, Y., Yan, D."Inhibition of Ep3 attenuates migration and promotes apoptosis of non‑small cell lung cancer cells via suppression of TGF‑β/Smad signaling". Oncology Letters 16.5 (2018): 5645-5654.
Chicago
Li, L., Lv, Y., Yan, D."Inhibition of Ep3 attenuates migration and promotes apoptosis of non‑small cell lung cancer cells via suppression of TGF‑β/Smad signaling". Oncology Letters 16, no. 5 (2018): 5645-5654. https://doi.org/10.3892/ol.2018.9391