Emerging role of microRNAs in modulating endothelin-1 expression in gastric cancer

  • Authors:
    • Kuo-Wang Tsai
    • Ling-Yueh Hu
    • Ting-Wen Chen
    • Sung-Chou Li
    • Meng‑Ru Ho
    • Shou-Yu Yu
    • Ya-Ting Tu
    • Wei-Shone Chen
    • Hing-Chung Lam
  • View Affiliations

  • Published online on: November 11, 2014     https://doi.org/10.3892/or.2014.3598
  • Pages: 485-493
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endothelin-1 (ET-1) is a small 21-amino acid peptide that is known to exert diverse biological effects on a wide variety of tissues and cell types through its own receptors. The ET-1-ETRA axis is frequently dysfunctional in numerous types of carcinomas, and contributes to the promotion of cell growth and migration. microRNAs (miRNAs) are small non-coding RNAs that play a critical role in carcinogenesis through mRNA degradation or the translational inhibition of cancer-associated protein-coding genes. However, the role of ET-1 and the relationship between ET-1 and miRNAs in gastric cancer remain unknown. Results of the analysis of the database of The Cancer Genome Atlas (TCGA) revealed that ET-1 is significantly overexpressed in gastric cancer cells when compared with its expression in adjacent normal cells. Exogenous ET-1 significantly enhanced gastric cancer cell proliferation, implying that ET-1 plays an oncogenic role in gastric cancer carcinogenesis. Using a luciferase reporter assay we showed that 18 miRNA candidates had a significant silencing effect on ET-1 expression by up to 20% in HEK293T cells. Among them, 5 miRNAs (miR-1, miR-101, miR-125A, miR-144 and let-7c) were shown to be involved in ET-1 silencing through post-transcriptional modulation in gastric cancer. Our data also revealed that DNA hypermethylation contributes to the silenced miR-1 expression in gastric cancer cells. The ectopic expression of miR-1 significantly inhibited AGS cell proliferation by suppressing ET-1 expression. Overall, our study revealed that ET-1 overexpression may be due to DNA hypermethylation resulting in the silencing of miR-1 expression in gastric cancer cells. In addition, we identified several miRNAs as potential modulators for ET-1 in gastric cancer, which may be used as targets for gastric cancer therapy.

Introduction

Endothelin (ET) was first identified as a vasoconstrictive 21-amino acid S-peptide derived from vascular endothelial cells. It is currently known to exert diverse biological effects on a wide variety of tissues and cell types through its own receptors (1,2). Three ETs (ET-1, ET-2 and ET-3), 2 G-protein-coupled receptors (ETRA and ETRB) and 2 ET-converting enzymes (ECE-1 and ECE-2) comprise the ET axis (2). The ET-1 gene contains 5 exons located on chromosome 6, and expresses a 212-amino acid precursor, preproendothelin-1 (PPET-1). PPET-1 is converted by protease to form an intermediate peptide consisting of 38 amino acids, big-ET-1, which is further cleaved by ECE-1 or ECE-2 to release the mature form of ET-1 (21 amino acids) (3,4).

An increase in ET-1 expression has been detected in various malignancies including lung, prostate, colorectal, liver, breast and ovarian cancers, as well as melanomas, and may contribute to cell growth, metastasis and angiogenesis, and suppression of apoptosis (411). ET-1 typically induces proliferation through the ETRA receptor, and decreased ETRB expression has frequently been implicated in the pathogenesis of esophageal squamous cell carcinoma and nasopharyngeal carcinoma, as well as prostate and ovarian cancers (12). The promoter regulatory region of the ETRB gene contains a 5′ CpG island which is commonly (70%) methylated, suggesting that ETRB down-regulation occurs through DNA hypermethylation in human carcinomas (1317). In gastric cancer, a high level of ET-1 has been detected in plasma, which may be a reliable marker for predicting recurrence (18,19). Stow et al (20) summarized previous studies and revealed that ET-1 transcription can be enhanced using numerous physical and chemical stimuli such as phorbol esters, thrombin, angiotensin II and insulin, by activating signaling transduction pathways including the protein kinase C (PKC) and JUN-terminal kinase (JNK) pathways. In addition, GATA family transcription factors, β-catenin, transforming growth factor β (TGFβ) and hypoxia-inducible factor-1 (HIF-1) can enhance ET-1 transcriptional activity by modulating its promoter (8,12). Therefore, unbalanced stimulators and impaired signaling that frequently lead to ET-1 overexpression trigger the activation of the ET axis, and promote cancer cell progression through autocrine and paracrine signaling. However, the detailed functions and regulatory mechanisms of ET-1 in gastric cancer remain unknown. The number of studies has recently been increasing regarding small non-coding short RNA microRNAs (miRNAs) in cancer development through post-transcriptional regulation of the expression of oncogenes or tumor-suppressor genes. miRNAs are deregulated in numerous disease states, particularly in cancer, making them novel molecular indicators that are critical for early diagnosis, prognosis, and therapy. Previous studies have demonstrated several instances of abnormal miRNA expression in gastric cancer cells compared with adjacent normal cells (21,22). These dysfunctional miRNAs may disrupt the balance of their target genes, resulting in the downregulation of tumor-suppressor genes and oncogene overexpression in gastric cancer.

Previous studies have revealed that miRNAs play a critical role in modulating ET-1 expression by targeting the 3′untranslated region (3′UTR), including miR-1, miR-125a, miR-125b, miR-155 and miR-199 (2328). To date, the biological function and regulating mechanisms of ET-1 by miRNAs in gastric cancer remain unknown. In the present study, we demonstrated that ET-1 plays an oncogenic role in gastric cancer growth, and identified several potential target sites for miRNA candidates harbored within the 3′UTR of ET-1 using a bioinformatics approach. We identified 5 dysfunctional miRNAs that may contribute to ET-1 overexpression in gastric cancer, to provide a potential approach and novel targets for therapy.

Materials and methods

Cell line and nuclear acid extraction

Five human gastric cancer cell lines, AGS, AZ-521, HR, NUGC and TSGH, were obtained from the American Type Culture Collection, and maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% inactivated fetal bovine serum (FBS; Invitrogen, Carlsbad, CA, USA). After 5-Aza-dC (10 μM) treatment for 4 days, RNA was extracted using TRIzol reagent (Invitrogen), according to the manufacturer’s protocol. After RNA extraction, the interface was subjected to DNA extraction with a back extraction buffer (4 M guanidine thiocyanate, 50 mM sodium citrate and 1 M Tris). Genomic DNA was then precipitated with EtOH.

Constructed reporter of ET-1 and the miRNA expression vector

ET-1 3′UTR was amplified from AGS cDNA by using the specific primer. The amplified ET-1 3′UTR was cloned into the pmiR-RB-Report vector, between the XhoI and NotI restriction sites. The primary miRNA transcripts were amplified from the genomic DNA of AGS cells by using miR-specific forward and reverse primers. The amplified PCR products were then cloned into the pLKO vector, between the AgeI and EcoRI restriction sites. All constructs were further confirmed through direct sequencing. The individual primers used in the present study are shown in Table I.

Table I

Primer sequences.

Table I

Primer sequences.

Constructed primersSequences
ET-1-3′UTR-F 5′-CCGCTCGAGCAGACCTTCGGGGCCTGTCTGAAG-3′
ET-1-3′UTR-R 5′-GAATGCGGCCGCACAGTAAGGAAAAAAATATTTATTTTC-3′
miR-1-1-F 5′-TCTACCGGTGGACACCAGGCAGCAGTGGC-3′
miR-1-1-R 5′-TCTGAATTCACAATGCTGGCGGGGACACG-3′
let-7b-F 5′-TCTACCGGTGGGCCTCTGCCTGTGGAGGA-3′
let-7b-R 5′-TCTGAATTCTCACTGAGGTAGGGGGCGGC-3′
let-7c-F 5′-TCTACCGGTTGGCAGGTTAGATGGTCAGAAGACA-3′
let-7c-R 5′-ACCTTCTTGCACACAAATTGGCTCA-3′
miR-33a-F 5′-TCTACCGGTCCCATAGCCTCTGTAAGCCC-3′
miR-33a-R 5′-TCTGAATTCGCTAAGGACATGTTCCCCGT-3′
miR-101-F 5′-TCTACCGGTTGCCTCCTCACGTCTCCAACCA-3′
miR-101-R 5′-TCTGAATTCTGGCTGCACCAACAACTACCCC-3′
miR-125a-F 5′-TCTACCGGTTTCTGTCCTTGTCCCTGCATC-3′
miR-125a-R 5′-TCTGAATTCCCATCGTGTGGGTCTCAAGG-3′
miR-125b-F 5′-TCTACCGGTTGCGCCCCCAGATACTGCGT-3′
miR-125b-R 5′-TCTGAATTCGGGGCATGCTGGGACTTCAGC-3′
miR-134-F 5′-TCTACCGGTCGGGCCATGGACAATGCGCT-3′
miR-134-R 5′-TCTGAATTCAGGGGCTGCCAAGGCTGACT-3′
miR-135a-F 5′-TCTACCGGTTCTTGTTTCCCGGTCCTTGT-3′
miR-135a-R 5′-TCTGAATTCACCCGACTGGTGGGCTATTA-3′
miR-135b-F 5′-TCTACCGGTTTTATGGCCAGGAAGCCACC-3′
miR-135b-R 5′-TCTGAATTCCAGCACACCCTGAAGGTCTC-3′
miR-141-F 5′-TCTACCGGTGGACACAATGGGCCCCAGCC-3′
miR-141-R 5′-TCTGAATTCAAAGCAGACGTCGCAGCCCC-3′
miR-144-F 5′-TCTACCGGTACACTGGCCCTGGGTCCCTA-3′
miR-144-R 5′-TCTGAATTCTGCCCTGGCAGTCAGTAGGTT-3′
miR-203-F 5′-TCTACCGGTGTCGGGGGCTCCTCTCTCCG-3′
miR-203-R 5′-TCTGAATTCCACCGCCAGTTCCTCGCTGG-3′
miR-224-F 5′-TCTACCGGTGCAGAGGGCTGGGCTACCTT-3′
miR-224-R 5′-TCTGAATTCCCCAGGGCCCAACTGGAAGAGT-3′
miR-340-F 5′-TCTACCGGTTCCAGCTTGAGTCTTCAAGAG-3′
miR-340-R 5′-TCTGAATTCAGAGTTGTGATCAGTAAATTAGA-3′
miR-379-F 5′-TCTACCGGTGCCTGCTTCCAATGCCAAAT-3′
miR-379-R 5′-TCTGAATTCGCCCAAGTTGCATCACTTCC-3′
miR-410-F 5′-TCTACCGGTGCCCTTTTGAGGGTAGGAGC-3′
miR-410-R 5′-TCTGAATTCAATGATTCAATGGCGGGGGT-3′
miR-425-F 5′-TCTACCGGTGTGCCCCTGACCCCCAGACA-3′
miR-425-R 5′-TCTGAATTCAGCAGGGAAACCCAGGGGCA-3′
miR-494-F 5′-TCTACCGGTACCGTCAGGAAAGCTCCAAT-3′
miR-494-R 5′-TCTGAATTCTCAGGAACAGGAAGTGCCTC-3′
miR-495-F 5′-TCTACCGGTCGCCTCTGCTCAGTGTCAGCC-3′
miR-495-R 5′-TCTGAATTCTCAGGGTCCCGTCGGGGATG-3′
miR-590-F 5′-TCTACCGGTGAACGTCAGCACCCTCCCCCCA-3′
miR-590-R 5′-TCTGAATTCTTGAGCGCCAGTGGCCAAGC-3′
miR-873-F 5′-TCTACCGGTGTGACCAGTGTCTGGGATGC-3′
miR-873-R 5′-TCTGAATTCCCTTGGTGGGATTCAACACCT-3′
Gene-specific primer
miR-1-RT 5′-CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGATACATAC-3′
miR-1-GSF 5′-CGGCGGTGGAATGTAAAGAAGT-3′
Luciferase reporter assay

HEK293T cells (2.5×104 cells/well) were seeded in a 24-well plate and cotransfected with 0.3 μg of either the control vector or various miRNA constructs in conjunction with 0.1 μg of the ET-1-3′UTR reporter construct (pmiR-RB-REPORT™; RiboBio) by using the Lipofectamine 2000 reagent (Invitrogen). After 48 h, the luciferase activity was measured using the Dual-Glo Luciferase Assay System kit (Promega, Madison, WI, USA). Luciferase readings were corrected for background, and firefly luciferase values were normalized to Renilla values to control for transfection efficiency. All of the assays were performed in triplicate in 3 independent experiments.

Methylation status of CpG islands

The genomic DNA of 5 gastric cancer cell lines, and normal and tumor tissue pairs from 50 gastric cancer patients were subjected to bisulfite conversion using the EZ DNA Methylation-Gold kit (Zymo Research Corporation, Orange, CA, USA). The bisulfite conversion reaction was conducted at 98°C for 10 min, and then at 64°C for 2.5 h, with a final incubation at 4°C for up to 20 h in a PCR thermocycler. Then, the modified genomic DNA was used for the methylation analysis of CpG25 and CpG81 of the mir-1-1 upstream using the specific methylation primers. The PCR conditions were as follows: 94°C for 10 min, followed by 35 cycles of 94°C for 1 min, 60°C for 1 min, and 72°C for 30 sec, with a final extension at 72°C for 10 min using a PCR thermocycler and HotStart Taq DNA polymerase (Qiagen, Hilden, Germany). The methylation status of the genomic DNA of individual samples was also examined with the BstUI digestion (New England Biolabs, Beverly, MA, USA). The digested PCR fragments were then separated on 2% agarose gel. Detailed information is provided in a previous study (29). The individual primers used in the present study were as follows: CpG25-F, 5′-GGAGGGGTAGGATAGTAGTTTGAGT-3′ and CpG25-R, 5′-AAAAAAACCTAACCTAAAAAACCAAAA-3′; CpG81-F, 5 ′-G G T G AG T T T T G T T TAG T T TAT TAT TAT-3 ′ and CpG81-R, 5′-ATCAAAATTCCTACCTCCCAACTA-3′.

Stem-loop RT-PCR of miR-1

Total RNA (2 μg) was reverse-transcribed with miR-1-specific stem-loop RT primers using SuperScript III Reverse Transcriptase (Invitrogen). The reaction was performed under the following incubation conditions: 30 min at 16°C, followed by 20°C for 30 sec, 42°C for 30 sec and 50°C for 1 sec, totaling 50 cycles. The enzyme was subsequently inactivated by incubation at 85°C for 5 min. Afterward, cDNA was diluted 10X for subsequent real-time PCR analysis using the miR-1-GSF primer. Gene expression was detected using a SYBR-Green I assay (Applied Biosystems, Foster City, CA, USA). The U6 expression was used as the internal control, and the expression levels of miR-1 were normalized to that of U6 (ΔCt = CtmiR-1 - CtU6).

Ectopic expression of miRNA candidates

Stable or transient gastric cell cultures expressing miR-1 candidates were generated using the transfected gastric cells with pLKO-pre-miRNA by Lipofectamine 2000. After 24 h of transfection, the stably expressed miRNA cells were generated by puromycin selection for 14 days. The shLuc targeting the luciferase gene provided puromycin resistance as the control. The expression levels of miRNAs were confirmed using real-time PCR.

Expression levels of ET axis genes and miRNAs were analyzed using The Cancer Genome Atlas (TCGA) data

The TCGA project collects RNA transcriptome and small RNA transcriptome data, which are obtained using the next-generation sequencing approach from various types of human cancers. For the present study, we downloaded all level-3 expression data for gastric cancer from the TCGA data portal (https://tcga-data.nci.nih.gov/tcga/dataAccessMatrix.htm). We used 29 tumor samples and 29 normal samples to analyze the expression levels of each miRNA and ET axis-related gene.

Cell proliferation assay

For cell proliferation analysis, we seeded 3,000 living AGS cells or AGS cells stably expressing miR-1 in 96-well plates. After exogenous ET-1 stimulation, we determined cell growth at 0, 1, 2, 3 and 4 days using WST-1 (Roche, Mannhein, Germany). All experiments were repeated 3 times.

Results

ET-1 is upregulated in gastric cancer

We first assessed the expression pattern of the ET axis genes (ET-1, ET-2, ET-2, ETRA, ETRB, ECE-1 and ECE-2) in gastric cancer by analyzing data from the database of TCGA. We obtained RNA transcriptome data on cancer tissues and the corresponding adjacent normal tissues from information on 29 gastric cancer patients. As shown in Fig. 1A, the expression levels of ET-1, ECE-1 and ECE-2 were significantly increased in gastric cancer compared to corresponding normal tissues (P<0.001). Conversely, ET-2 expression was decreased significantly in gastric cancer (P<0.05). We further examined the expression levels of ET-1 in 3 gastric cancer cell lines using the RT-PCR approach. As shown in Fig. 1B, we detected a greater abundance of ET-1 RNA in the AZ-521 and HR cells, and low expression in AGS cells. To investigate the putative function of ET-1 on gastric cancer, we further treated AGS cells with ET-1 (10 nM) for 4 days. We evaluated the potential influence of ET-1 on gastric cancer cell proliferation through WST-1 for 96 h. As shown in Fig. 1C, cell proliferation in the ET-1 treatment groups was significantly increased compared with that in the control groups. These results demonstrate that ET-1 plays an oncogenic role in promoting the proliferation of gastric cancer cells. However, the mechanism that leads to ET-1 dysregulation in gastric cancer remains unclear. To further investigate the underlying mechanisms of ET-1 dysfunction in gastric cancer, we focused on the post-transcriptional regulation of ET-1 expression by miRNAs during the progression of gastric cancer.

Discovery of putative miRNAs that regulate ET-1 genes

Using a prediction program available on microRNA.org, we predicted that several miRNA candidates could suppress ET-1 expression by targeting its 3′UTR sequence. Among them, we selected 22 miRNA candidates, and constructed their pre-miRNA sequence into a pKLO expression vector (Fig. 2A). In addition, we cloned the full length of the 3′UTR (i.e., 1,126 bp) of ET-1 into the pmiR-RB-Report vector. Due to the fact that the HEK293T cells have a high transfection efficiency, we conducted a luciferase assay using HEK293T cells. As shown in Fig. 2B, miR-1, let-7c and miR-125b substantially inhibited luciferase activity by >50% compared to the control group after transfection for 48 h. Fifteen miRNA candidates slightly suppressed the luciferase activity, including let-7b, miR-33a, miR-101, miR-125a, miR-134, miR-135a/b, miR-141, miR-144, miR-379, miR-410, miR-425, miR-494, miR-495 and miR-590. Consistent with previous studies, our data indicated that miR-1 and miR-125a/b both suppressed ET-1 expression through post-transcriptional regulation. After using a reporter assay, we found that 18 miRNA candidates may be involved in ET-1 post-translational regulation through the targeting of its 3′UTR sequence.

Expression level of miRNA candidates from the TCGA database

After completing the reporter assay, we selected 18 miRNA candidates to further examine their expression levels in gastric cancer by analyzing data from the TCGA database. As shown in Fig. 3A, we identified 6 miRNAs (miR-134, miR-135b, miR-141, miR-425, miR-494 and miR-590) and 5 miRNAs (miR-1, let-7c, miR-101, miR-125a and miR-144) that were significantly upregulated and downregulated, respectively, in gastric cancer. Since the expression level of ET-1 was upregulated in the gastric cancer cells compared with the corresponding normal cells, miRNA candidate expression should decrease in gastric cancer. According to their expression levels in gastric cancer, we selected 6 miRNAs (miR-1, miR-101, miR-125a/b, miR-144 and let-7c) for further confirmation in the AGS cells. These miRNA candidates were ectopically expressed in the AGS cells, and let-7c, miR-1, miR-101, miR-125a/b and miR-144 were found to significantly suppress luciferase activity (Fig. 3B). In addition, we found that the endogenous protein levels of ET-1 were inhibited by let-7c, miR-1, miR-101, miR-125a and miR-144 overexpression (Fig. 3C).

DNA hypermethylation silences miR-1 expression in gastric cancer

Among the listed miRNAs, we selected miR-1 to further examine the biological function and the regulatory mechanisms in gastric cancer. The expression of miR-1 originates from 2 paralogous genes, mir-1-1 and mir-1-2, located on chromosomes 20 and 18, respectively. As shown in Fig. 4A, 4 CpG islands are located at the putative transcription start sites of the miR-1-1 loci, according to the UCSC database. We analyzed the methylation statuses of the CpG25 and CpG81 regions in 5 human gastric cancer cell lines by using a COBRA approach. As shown in Fig. 4B, the CpG81 regions were completely methylated in the AGS and AZ-521 cells, yet unmethylated in the HR cells. After treatment for 4 days with 5-Aza-dC, the expression levels of miR-1 were restored in the AGS, AZ-521 and HR cells (Fig. 4C). These data indicate that DNA methylation contributes to the modulation of miR-1 transcriptional activity in gastric cancer cell lines.

By analyzing TCGA data, we found that the expression levels of miR-1 were significantly decreased in the gastric cancer cells compared with the corresponding adjacent normal cells (Fig. 3A). Therefore, we further examined the methylation status of the CpG81 regions in 50 primary gastric cancers and the corresponding adjacent control, and observed a high frequence of DNA hypermethylation in gastric cancer (26 of 50). These results indicated that DNA hypermethylation leads to low expression levels of miR-1 in gastric cancer (Fig. 4D).

miR-1 suppresses gastric cancer cell growth

The ectopic expression of miR-1 in AGS cells increased mature miR-1 expression by ~250-fold (Fig. 5A). The growth of AGS cells with miR-1 overexpression was substantially inhibited compared with those transfected with the vector (Fig. 5B). Furthermore, the clonogenic survival of AGS cells also decreased after ectopic expression of miR-1 (Fig. 5C). Overall, our results imply that ET-1 overexpression may be due to the silencing of miR-1 expression by DNA hypermethylation in gastric cancer. In addition, we identified several miRNA candidates that could modulate the ET-1 oncogenic function, and could also act as potential therapeutic targets for therapy in the future.

Discussion

Dysfunction of the endothelin (ET) axis is known to be a feature of human cancer, including lung, prostate, colorectal, liver, breast, ovarian and oral cancer as well as melanomas. Abnormal activation of the ET axis leads to promotion of cell growth, metastasis and angiogenesis, and suppression of apoptosis (411). By analyzing TCGA data, we found that the expression levels of ET-1, ECE-1 and ECE-2 were significant increased in the gastric cancer compared to the corresponding normal tissues (Fig. 1). A previous study reported that the plasma big-ET-1 level in gastric cancer was significantly higher than that in the control (30). These findings imply that the expression level of ET-1 is increased in gastric cancer. Although ECE-1 (31) and ECE-2 were found to be upregulated in a number of cancers, including prostate, ovarian and oral cancer, thyroid carcinoma, and breast cancer (3235), the expression of ECE-1 and ECE-2 was not reported in gastric cancer. Previous studies found that, in human cancer, ET-2 and ET-3 expression levels are silenced by DNA hypermethylation of their promoter regions (36,37). In the present study, we observed that the expression level of ET-2 was significant decreased in gastric cancer. However, what mechanism is involved in the loss of expression in gastric cancer remains unclear. These studies support our finding that the ET-1 axis is overexpressed, and that ET-2 is downregulated in gastric cancer.

In the past decade, miRNAs have emerged as playing important roles via modulation of the cell cycle, cell growth and cell motility in gastric cancer progression (21,22). In the present study, we identified 15 miRNAs that may modulate ET-1 expression by targeting 3′UTR. Among them, 6 miRNAs (miR-134, miR-135b, miR-141, miR-425, miR-494 and miR-590) and 5 miRNAs (miR-1, let-7c, miR-101, miR-125a and miR-144) showed significant upregulation and downregulation, respectively, in gastric cancer. Consistent with our findings, previous studies have revealed that miR-135b and miR-425 act as oncomiRs involved in the tumorigenesis of gastric cancer (3840). Furthermore, miR-125a/b, miR-144 and miR-101 have been reported to be significantly decreased and display a tumor-suppressive function by silencing their oncongenic targets in gastric cancer. Here, we demonstrated that miR-125a/b, miR-101 and miR-144 suppressed ET1 expression by targeting the 3′UTR in gastric cancer. In addition, previous studies have reported that miR-1, miR-125a/b, miR-155 and miR-199 suppress ET-1 expression by targeting 3′UTR (2328). In numerous cancers, miR-1 performs a tumor-suppressive function by suppressing tumor cell growth and migration in prostate cancer, hepatocellular, bladder and esophageal squamous cell carcinoma, and lung cancer (29,4148). Furthermore, hypermethylation of the promoter region of mir-1-1 resulted in low expression in hepatocellular carcinoma and colorectal cancer (29,4850). In the present study, we also demonstrated that miR-1 expression levels were suppressed by DNA methylation and that ectopic miR-1 expression led to the suppression of gastric cancer cell growth. Collectively, our results imply that ET-1 overexpression may be due to DNA hypermethylation resulting in the silencing of miR-1 expression in gastric cancer.

In solid tumors, the ET-axis pathway plays a critical oncogenic role in facilitating cancer growth and metastasis. Various investigations have demonstrated that the ET-1-ETRA axis is a potential therapeutic target for cancer therapy. To date, several drugs have been designed as ETRA antagonists for anticancer growth and metastasis. The ET-1 is a short intercellular peptide derived from numerous tumor cells, and has emerged as a potential therapeutic target or biomarker for human cancer. In the present study, our identification of the post-translational regulation of ET-1 may lead to novel insights into the function of ET-1 in gastric cancer, and may provide a potential therapeutic target.

Acknowledgements

This study was supported by grants from the Kaohsiung Veterans General Hospital (VGHKS 102-020, VGHKS 102-074, VGHKS 103-066 and VGHKS 103-108).

References

1 

Yanagisawa M, Kurihara H, Kimura S, et al: A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature. 332:411–415. 1988. View Article : Google Scholar : PubMed/NCBI

2 

Kedzierski RM and Yanagisawa M: Endothelin system: the double-edged sword in health and disease. Annu Rev Pharmacol Toxicol. 41:851–876. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Rubanyi GM and Polokoff MA: Endothelins: molecular biology, biochemistry, pharmacology, physiology, and pathophysiology. Pharmacol Rev. 46:325–415. 1994.PubMed/NCBI

4 

Grimshaw MJ: Endothelins in breast tumour cell invasion. Cancer Lett. 222:129–138. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Asham E, Shankar A, Loizidou M, et al: Increased endothelin-1 in colorectal cancer and reduction of tumour growth by ETA receptor antagonism. Br J Cancer. 85:1759–1763. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Eberle J, Fecker LF, Orfanos CE and Geilen CC: Endothelin-1 decreases basic apoptotic rates in human melanoma cell lines. J Invest Dermatol. 119:549–555. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Douglas ML, Richardson MM and Nicol DL: Endothelin axis expression is markedly different in the two main subtypes of renal cell carcinoma. Cancer. 100:2118–2124. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Kim TH, Xiong H, Zhang Z and Ren B: β-Catenin activates the growth factor endothelin-1 in colon cancer cells. Oncogene. 24:597–604. 2005. View Article : Google Scholar

9 

Abdel-Gawad IA, Hassanein HM, Bahgat NA, et al: Study of endothelin-1 and vascular endothelial growth factor in patients with cancer colon. J Egypt Natl Canc Inst. 20:216–223. 2008.PubMed/NCBI

10 

Said N, Smith S, Sanchez-Carbayo M and Theodorescu D: Tumor endothelin-1 enhances metastatic colonization of the lung in mouse xenograft models of bladder cancer. J Clin Invest. 121:132–147. 2011. View Article : Google Scholar :

11 

Nelson JB, Udan MS, Guruli G and Pflug BR: Endothelin-1 inhibits apoptosis in prostate cancer. Neoplasia. 7:631–637. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Rosano L, Spinella F and Bagnato A: Endothelin 1 in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 13:637–651. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Nelson JB, Lee WH, Nguyen SH, et al: Methylation of the 5′ CpG island of the endothelin B receptor gene is common in human prostate cancer. Cancer Res. 57:35–37. 1997.PubMed/NCBI

14 

Pao MM, Tsutsumi M, Liang G, Uzvolgyi E, Gonzales FA and Jones PA: The endothelin receptor B (EDNRB) promoter displays heterogeneous, site specific methylation patterns in normal and tumor cells. Hum Mol Genet. 10:903–910. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Cohen AJ, Belinsky S, Franklin W and Beard S: Molecular and physiologic evidence for 5′CpG island methylation of the endothelin B receptor gene in lung cancer. Chest. 121:27S–28S. 2002. View Article : Google Scholar

16 

Lo KW, Tsang YS, Kwong J, To KF, Teo PM and Huang DP: Promoter hypermethylation of the EDNRB gene in nasopharyngeal carcinoma. Int J Cancer. 98:651–655. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Zhao BJ, Sun DG, Zhang M, Tan SN and Ma X: Identification of aberrant promoter methylation of EDNRB gene in esophageal squamous cell carcinoma. Dis Esophagus. 22:55–61. 2009. View Article : Google Scholar

18 

Mathieu MN and Chevillard C: Endothelin-1 and ETA receptor subtype are expressed in the gastric HGT-1 cell line. J Cardiovasc Pharmacol. 26(Suppl 3): S508–S509. 1995. View Article : Google Scholar : PubMed/NCBI

19 

Ferrari-Bravo A, Franciosi C, Lissoni P, Fumagalli L and Uggeri F: Effects of oncological surgery on endothelin-1 secretion in patients with operable gastric cancer. Int J Biol Markers. 15:56–57. 2000.PubMed/NCBI

20 

Stow LR, Jacobs ME, Wingo CS and Cain BD: Endothelin-1 gene regulation. FASEB J. 25:16–28. 2011. View Article : Google Scholar :

21 

Pan HW, Li SC and Tsai KW: MicroRNA dysregulation in gastric cancer. Curr Pharm Des. 19:1273–1284. 2013.

22 

Wu HH, Lin WC and Tsai KW: Advances in molecular biomarkers for gastric cancer: miRNAs as emerging novel cancer markers. Expert Rev Mol Med. 16:e12014. View Article : Google Scholar : PubMed/NCBI

23 

Jacobs ME, Wingo CS and Cain BD: An emerging role for microRNA in the regulation of endothelin-1. Front Physiol. 4:222013. View Article : Google Scholar : PubMed/NCBI

24 

Yeligar S, Tsukamoto H and Kalra VK: Ethanol-induced expression of ET-1 and ET-BR in liver sinusoidal endothelial cells and human endothelial cells involves hypoxia-inducible factor-1α and microRNA-199. J Immunol. 183:5232–5243. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Li D, Yang P, Xiong Q, et al: MicroRNA-125a/b-5p inhibits endothelin-1 expression in vascular endothelial cells. J Hypertens. 28:1646–1654. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Li D, Yang P, Li H, et al: MicroRNA-1 inhibits proliferation of hepatocarcinoma cells by targeting endothelin-1. Life Sci. 91:440–447. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Li D, He B, Zhang H, et al: The inhibitory effect of miRNA-1 on ET-1 gene expression. FEBS Lett. 586:1014–1021. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Feng B, Cao Y, Chen S, Ruiz M and Chakrabarti S: miRNA-1 regulates endothelin-1 in diabetes. Life Sci. 98:18–23. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Chen WS, Leung CM, Pan HW, et al: Silencing of miR-1-1 and miR-133a-2 cluster expression by DNA hypermethylation in colorectal cancer. Oncol Rep. 28:1069–1076. 2012.PubMed/NCBI

30 

Teng XJ, Shen ZX, Xiang JJ, et al: Pre- and post-operative plasma big endothelin-1 levels in patients with gastric carcinoma undergoing radical gastrectomy. Anticancer Res. 26:2503–2507. 2006.PubMed/NCBI

31 

Whyteside AR, Hinsley EE, Lambert LA, McDermott PJ and Turner AJ: ECE-1 influences prostate cancer cell invasion via ET-1-mediated FAK phosphorylation and ET-1-independent mechanisms. Can J Physiol Pharmacol. 88:850–854. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Smollich M, Götte M, Yip GW, et al: On the role of endothelin-converting enzyme-1 (ECE-1) and neprilysin in human breast cancer. Breast Cancer Res Treat. 106:361–369. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Dawson LA, Maitland NJ, Berry P, Turner AJ and Usmani BA: Expression and localization of endothelin-converting enzyme-1 in human prostate cancer. Exp Biol Med. 231:1106–1110. 2006.

34 

Awano S, Dawson LA, Hunter AR, Turner AJ and Usmani BA: Endothelin system in oral squamous carcinoma cells: specific siRNA targeting of ECE-1 blocks cell proliferation. Int J Cancer. 118:1645–1652. 2006. View Article : Google Scholar

35 

Van Beneden R, Michel L, Havaux X, Delos M and Donckier J: Increased expression of endothelin-1 converting enzyme in human thyroid carcinoma. Clin Endocrinol. 60:146–147. 2004. View Article : Google Scholar

36 

Wiesmann F, Veeck J, Galm O, et al: Frequent loss of endothelin-3 (EDN3) expression due to epigenetic inactivation in human breast cancer. Breast Cancer Res. 11:R342009. View Article : Google Scholar : PubMed/NCBI

37 

Wang R, Lohr CV, Fischer K, et al: Epigenetic inactivation of endothelin-2 and endothelin-3 in colon cancer. Int J Cancer. 132:1004–1012. 2013. View Article : Google Scholar

38 

Lim JY, Yoon SO, Seol SY, et al: Overexpression of miR-196b and HOXA10 characterize a poor-prognosis gastric cancer subtype. World J Gastroenterol. 19:7078–7088. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Ma J, Liu J, Wang Z, et al: NF-kappaB-dependent microRNA-425 upregulation promotes gastric cancer cell growth by targeting PTEN upon IL-1β induction. Mol Cancer. 13:402014. View Article : Google Scholar

40 

Peng WZ, Ma R, Wang F, Yu J and Liu ZB: Role of miR-191/425 cluster in tumorigenesis and diagnosis of gastric cancer. Int J Mol Sci. 15:4031–4048. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Yoshino H, Chiyomaru T, Enokida H, et al: The tumour-suppressive function of miR-1 and miR-133a targeting TAGLN2 in bladder cancer. Br J Cancer. 104:808–818. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Wu CD, Kuo YS, Wu HC and Lin CT: MicroRNA-1 induces apoptosis by targeting prothymosin alpha in nasopharyngeal carcinoma cells. J Biomed Sci. 18:802011. View Article : Google Scholar : PubMed/NCBI

43 

Wang F, Song G, Liu M, Li X and Tang H: miRNA-1 targets fibronectin1 and suppresses the migration and invasion of the HEp2 laryngeal squamous carcinoma cell line. FEBS Lett. 585:3263–3269. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Nohata N, Sone Y, Hanazawa T, et al: miR-1 as a tumor suppressive microRNA targeting TAGLN2 in head and neck squamous cell carcinoma. Oncotarget. 2:29–42. 2011.PubMed/NCBI

45 

Nasser MW, Datta J, Nuovo G, et al: Down-regulation of micro-RNA-1 (miR-1) in lung cancer. Suppression of tumorigenic property of lung cancer cells and their sensitization to doxorubicin-induced apoptosis by miR-1. J Biol Chem. 283:33394–33405. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Kinoshita T, Nohata N, Fuse M, et al: Tumor suppressive microRNA-133a regulates novel targets: moesin contributes to cancer cell proliferation and invasion in head and neck squamous cell carcinoma. Biochem Biophys Res Commun. 418:378–383. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Hudson RS, Yi M, Esposito D, et al: MicroRNA-1 is a candidate tumor suppressor and prognostic marker in human prostate cancer. Nucleic Acids Res. 40:3689–3703. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Datta J, Kutay H, Nasser MW, et al: Methylation mediated silencing of microRNA-1 gene and its role in hepatocellular carcinogenesis. Cancer Res. 68:5049–5058. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Migliore C, Martin V, Leoni VP, et al: MiR-1 downregulation cooperates with MACC1 in promoting MET overexpression in human colon cancer. Clin Cancer Res. 18:737–747. 2012. View Article : Google Scholar

50 

Suzuki H, Takatsuka S, Akashi H, et al: Genome-wide profiling of chromatin signatures reveals epigenetic regulation of microRNA genes in colorectal cancer. Cancer Res. 71:5646–5658. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2015
Volume 33 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
Tsai K, Hu L, Chen T, Li S, Ho MR, Yu S, Tu Y, Chen W and Lam H: Emerging role of microRNAs in modulating endothelin-1 expression in gastric cancer. Oncol Rep 33: 485-493, 2015
APA
Tsai, K., Hu, L., Chen, T., Li, S., Ho, M., Yu, S. ... Lam, H. (2015). Emerging role of microRNAs in modulating endothelin-1 expression in gastric cancer. Oncology Reports, 33, 485-493. https://doi.org/10.3892/or.2014.3598
MLA
Tsai, K., Hu, L., Chen, T., Li, S., Ho, M., Yu, S., Tu, Y., Chen, W., Lam, H."Emerging role of microRNAs in modulating endothelin-1 expression in gastric cancer". Oncology Reports 33.1 (2015): 485-493.
Chicago
Tsai, K., Hu, L., Chen, T., Li, S., Ho, M., Yu, S., Tu, Y., Chen, W., Lam, H."Emerging role of microRNAs in modulating endothelin-1 expression in gastric cancer". Oncology Reports 33, no. 1 (2015): 485-493. https://doi.org/10.3892/or.2014.3598