MicroRNA-181a regulates epithelial-mesenchymal transition by targeting PTEN in drug-resistant lung adenocarcinoma cells

  • Authors:
    • Haihui Li
    • Pei Zhang
    • Xiaojin Sun
    • Yiming Sun
    • Chao Shi
    • Hao Liu
    • Xuegang Liu
  • View Affiliations

  • Published online on: August 31, 2015     https://doi.org/10.3892/ijo.2015.3144
  • Pages: 1379-1392
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chemoresistance is an inevitable occurrence in lung adenocarcinoma, which has been associated with decreased expression of the phosphatase and tensin homolog deleted on chromosome ten (PTEN). Therefore, it is important to identify novel molecular mechanisms to suppress chemoresistance in lung adenocarcinoma cells. Paclitaxel- and cisplatin-resistant A549 lung carcinoma cell derivatives were developed by long-term serial culture. The metastatic properties of the cells were assessed using wound-healing assays, migration assays, invasion assays, morphological examination, and western blot analysis/RT-PCR of genes associated with the epithelial-mesenchymal transition (EMT). To identify novel regulators of EMT in A549 cells, differentially expressed miRNAs in drug-resistant cells were identified by microarray analysis. The role of miR-181a was established by transfection with specific mimic and inhibitor followed by functional assays. Luciferase assays were performed to assess the ability of miR-181a to target the PTEN promoter, and regulation of PTEN expression by miR-181a was assessed by western blot analysis and RT-PCR. Paclitaxel- and cisplatin-resistant A549 cells acquired metastatic properties and EMT phenotype and had reduced PTEN expression as compared to sensitive cells. miR‑181a was identified as a differentially expressed miRNA in drug-resistant A549 cells, and miR-181a mimic and inhibitor were shown to affect migration, invasion, morphology and expression of EMT-associated genes. PTEN was identified as a direct target of miR-181a. Our findings demonstrate that miR-181a expression in lung adenocarcinoma is associated with EMT progression, potentially through targeting of PTEN. Regulation of miR-181a may provide a novel strategy for overcoming resistance to paclitaxel and cisplatin in lung adenocarcinoma.

Introduction

Lung adenocarcinoma is a highly malignant disease with aggressive clinical behavior. In 2013, lung cancer was predicted to account for 26% of all female cancer deaths and 28% of all male cancer deaths (1). Patients diagnosed with lung adenocarcinoma usually survive only a few months, even with the assistance of individualized treatment strategies, including surgery combined with radiation and chemotherapy. The high mortality is mainly accounted for by drug resistance during chemotherapy treatment. Thus, to achieve better treatment outcome in lung adenocarcinoma patients, it is of the utmost importance to explore the mechanisms of drug resistance and to identify novel methods to overcome drug resistance.

Paclitaxel and cisplatin, as first-line treatment for patients suffering from lung adenocarcinoma, improves survival with manageable toxicity (2,3). However, tumor cells acquire resistance to paclitaxel/cisplatin, which causes eventual failure to prolong the survival of lung adenocarcinoma patients. There is abundant evidence that chemoresistance is associated with the acquisition of EMT phenotypic of cancer cells (4). The serial process of EMT involves epithelial cells switching to mesenchymal phenotypic cells. The epithelial cell-cell junctions become disassembled, and the expression of epithelial markers, including E-cadherin and β-catenin, is reduced, while the motility and invasion potential is increased, and mesen-chymal properties, such as the high expression of molecular markers vimentin, Snail, Slug, Twist, Zinc-finger E-boxbinding homeobox1 (ZEB1) and ZEB2, become apparent (58). Additionally, EMT is often associated with the loss of expression of the phosphatase and tensin homolog deleted on chromosome ten (PTEN), an inhibitor of the phosphatidylinositol 3-kinase/Akt pathway (9). Mutation or reduced expression of PTEN is frequently observed in lung cancer, though the mechanism of PTEN loss is not well understood (10,11).

MicroRNAs (miRNAs) have been see as key post-transcriptional level regulators of gene expression (12). Involvement of miRNAs has been demonstrated in tumorigenesis, metastasis, embryonic development, metabolism and other pathological and normal physiological (13,14). PTEN has been shown to be a tumor suppressor gene in human hepatocellular carcinoma, and miRNA-21 has been shown to regulate the expression of PTEN (14). MicroRNA-492 expression promotes the progression of hepatic cancer by targeting PTEN (15). The PTEN gene is targeted by miRNA-221 and miRNA-222 to regulate gastric carcinoma cell proliferation and radioresistance by targeting (16). High-frequency miRNA dysfunction is also associated with lung adenocarcinoma development and progression (17). Therefore, it is of value to determine whether dysregulation of the miRNA-regulatory network is responsible chemoresistance in lung adenocarcinoma.

The dysregulation of miR-181a was identified in the present study on lung adenocarcinoma. We determined that miR-181a responds to chemotherapy by changing the levels of its target PTEN. The identification of miR-181a/PTEN as a novel regulatory circuit that mediates EMT and chemoresistance in lung adenocarcinoma provides a new molecular mechanism, which could be targeted as a novel therapy for chemoresistance in lung adenocarcinoma.

Materials and methods

Cell culture, reagents and antibodies

The cell line A549, which is a human lung adenocarcinoma cell line, and its drug-resistant strain were cultured for use in the present study. The culture conditions were 37°C in 5% CO2 in RPMI-1640 medium (Gibco, Gaithersburg, MD, USA) supplemented with 10% fetal bovine serum (FBS; Gibco). The cells were sub-cultured every 3–4 days. Cisplatin was purchased from Qilu Pharmaceutical Co., Ltd. (Jinan, China). Paclitaxel was purchased from Beijing SL Pharmaceutical Co., Ltd. (Beijing, China). MTT [3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide] was from Sigma (St. Louis, MO, USA). Primary antibodies against E-cadherin (cat. AF0131, dilution, 1:500–1:3,000), β-catenin (cat. AF0122, dilution, 1:500–1:3,000), MMP-2 (cat. AF0577, dilution, 1:500–1:2,000), MMP-9 (cat. AF0577, dilution, 1:500–1:2,000), Snail (cat. AF6032, dilution, 1:500–1:2,000), Slug (cat. AF4002, dilution, 1:500–1:2,000), vimentin (cat. AF7013, dilution, 1:500–1:2000), ZEB1 (cat. DF7414, dilution, 1:500–1:2,000), PTEN (cat. AF6351, dilution, 1:500–1:2,000) and β-actin (cat. AF0115, dilution, 1:500–1:3,000) were from Affinity Biosciences, Inc. (Cincinnati, OH, USA). Secondary antibodies, goat anti-rabbit IgG (H+L)-HRP (cat. S0001, dilution, 1:5,000–1:10,000), were also obtained from Affinity Biosciences.

Cell proliferation assays

Cells (A549, A549/PTX and A549/DDP) were seeded in 96-well plate at 7×103 cells/well and cultured overnight. The next day, cells were treated with different concentrations of PTX/DDP for up to 72 h. MTT assays were conducted as previously described (18). The IC50 (drug concentration causing 50% inhibition of viability) was calculated for each cell line, and the resistance index (RI) was obtained by dividing the IC50 value of the resistant cell lines by the IC50 value of the non-resistant cell lines.

Woundhealingassays

Cells (A549, A549/PTX and A549/DDP) were seeded in 6-well plates and cultured to 90–95% confluency. Scratch wounds were then generated on the surface of the plates using a pipette tip. Photographic images were taken immediately after the scratch wound and also 24 h later.

Transwell migration and invasion assays

A 24-well Transwell chamber (Corning Inc., Corning, NY, USA) with gelatin-coated polycarbonate membrane filters was used to assess the migration ability of A549, A549/PTX and A549/DDP cells. The invasive capacity of A549, A549/PTX and A549/DDP cells was assessed using Transwell inserts with Matrigel (BD Biosciences). After incubation for 24 h, the cells in the upper surfaces of the Transwell chambers were removed with cotton swabs, and the migrated and invaded cells were fixed with 4% paraformaldehyde, and then stained with Giemsa solution. The stained cells were photographed and counted under a light microscope in four randomly selected fields.

RNA extraction and mRNA expression analyzed by reverse transcription-PCR analysis

TRIzol (Invitrogen, Grand Island, NY, USA) was used to isolate the total RNA from A549, A549/PTX and A549/DDP cells. The obtained RNA was purified with RNeasy Mini kit and RNase-free DNase Set (Qiagen) according to the protocols suggested by the manufacturer. Table I shows the primers used in the PCR reactions. GAPDH expression was used as an internal control. The kit manufacturer's protocol was used for RT-PCR amplifications.

Table I

Primer sequences and amplification lengths of qRT-PCR products.

Table I

Primer sequences and amplification lengths of qRT-PCR products.

GenePrimer sequence (5′-3′)Size of product (bp)
E-cadherinF: CATTTCCCAACTCCTCTCCTGGC
R: ATGGGCCTTTTTCATTTTCTGGG
90
β-cateninF: CACAAGCAGAGTGCTGAAGGTG
R: GATTCCTGAGAGTCCAAAGACAG
146
VimentinF: AGATGGCCCTTGACATTGAG
R: TGGAAGAGGCAGAGAAATTC
80
FibronectinF: CCCACCGTCTCAACATGCTTAG
R: CTCGGCTTCCTCCATAACAAGTAC
264
MMP-2F: GATAACCTGGATGCCGTCGTG
R: CTTCACGCTCTTCAGACTTTGGTTC
105
MMP-9F: CGGAGTGAGTTGAACCAG
R: GTCCCAGTGGGGATTTAC
118
SnailF: CCAGCTCTCTGAGGCCAAGGATC
R: TGGCTTCGGATGTGCATCTTGAG
108
SlugF: CCCTGAAGATGCATATTCGGAC
R: CTTCTCCCCCGTTGTAGTTCTA
116
TwistF: TGCGGAAGATCATCCCCA
R: TCCATCCTCCAGACCGAGAA
187
ZEB1F: GCACAACCAAGTGCAGAAGA
R: GCCTGGTTCAGGAGAAGATG
141
GAPDHF: AAGGTGAAGGTCGGAGTCAAC
R: CTTGATTTTGGAGGGATCTCG
252

[i] F, forward; R, reverse.

miRNA microarray and quantitative miRNA analysis

Cells (A549/PTX or A549/DDP) were cultured for one week without treatment before the experiments. Total RNA from drug-resistant and drug-sensitive cells was isolated with miRNeasy Mini kit (Qiagen; cat. 217004) following the manufacturer's instructions and RNA concentration was determined by NanoVue™ Plus (Thermo Fisher Scientific, Inc., Waltham, MA, USA). For miRNA analysis, the Human Cancer PathwayFinder miRNA PCR Array (Qiagen; cat. MIHS-102Z) allows the simultaneous detection of 84 miRNAs previously identified in human cancers. The fold change for each miRNA was calculated by plugging the Crossing point (Cp) values into the manufacturer's web-based software, and so microarray images were acquired. For each array, a multiple controls were used; i.e. RT negative and positive controls, and genomic DNA contamination controls and endogenous controls. miScript PCR primer (Qiagen) was used for validation of the miRNA samples. The expression of hsa-miR181a (Qiagen; cat. MS00008827) in drug-resistant and drug-sensitive cells was performed with a similar approach. The cells were transfected as described above. U6 was used as endogenous control to normalize Ct values obtained for each gene. The changes in the expression were calculated using the 2−ΔΔCt method.

Protein extraction and western blotting

Cells were plated at a density of 4×105 cells/well in 6-well culture plates (Corning Life Sciences). RIPA buffer (1X PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS and protease inhibitor cocktail) was used to harvested and lyse cells. A bicinchoninic acid (BCA) assay (Beyotime Institute of Biotechnology, Beijing, China) was used to measure protein concentrations. The resulting proteins (40 μg) were separated with sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Proteins were then transferred onto polyvinylidene difluoride (PVDF) membranes. The membranes were blocked with 5% skim milk in TPBS, and probed with primary antibodies overnight at 4°C. HRP-conjugated secondary antibodies were incubated with the membranes after they were washed. Chemiluminescent ECL reagent (Millipore, Millipore, MA, USA) was used for visualization. Finally, gel imaging equipment (Bio-Rad Laboratories, Hercules, CA, USA) was used to image the membranes. β-actin was the loading control. In fact, antibodies were dilution as follows: E-cadherin (1:800), β-catenin (1:800), MMP-2 (1:1,000), MMP-9 (1:1,000), Snail (1:1,000), Slug (1:1,000), vimentin (1:1,000), ZEB1 (1:1,000), PTEN (1:1,000), β-actin (1:3,000) and goat anti-rabbit IgG (H+L)-HRP (1:5,000).

Cell transfection

Six-well plates were used for cell seeding and cells were transfected with miR-181a mimic, miR-181a inhibitor or their negative controls. These miRNAs were synthesized by Shanghai GenePharma Co. (Shanghai, China) (Table II) and cells were transfected with the Lipofectamine 2000 (Invitrogen) using the manufacturer's protocol. After the indicated incubation period, the cells were subjected to further analysis by functional assays as indicated.

Table II

The sequence of the miR-181a mimic and inhibitor and their negative controls.

Table II

The sequence of the miR-181a mimic and inhibitor and their negative controls.

miRNASequence
hsa-miR-181a mimic5′-3′ Sense AACAUUCAACGCUGUCGGUGAGU
Antisense UCACCGACAGCGUUGAAUGUUUU
hsa-miR negative control5′-3′ Sense UUCUCCGAACGUGUCACGUTT
Antisense ACGUGACACGUUCGGAGAATT
hsa-miR-181a inhibitor5′-3′ Sense ACUCACCGACAGCGUUGAAUGUU
Antisense AACAUUCAACGCUGUCGGUGAGU
hsa-miR inhibitor negative control5′-3′ Sense CAGUACUUUUGUGUAGUACAA
Antisense UUGUACUACACAAAAGUACUG
Luciferase reporter assay

PCR was used to amplify the full-length 3′-UTR segments of PTEN mRNA that contained the miR-181a binding site. These segments were then inserted into the Xba1-site of the pGL3 vector (Promega, Madison, WI, USA) and the vector pGL3-PTEN was produced. A site-directed mutagenesis kit (Stratagene, La Jolla, CA, USA) was used to construct the pGL3-PTEN-mut reporter construct with point mutations in the seed sequence. A total of 1×106 cells were cotransfected with 50 pmol of miR-181a inhibitor (or control inhibitor), 1 μg of pGL3-PTEN (or pGL3-PTEN-mut) plasmid, and 1 μg of a Renilla luciferase expression construct pRL-TK (Promega) to assess the endogenous inhibitory activity of miR-181a, using Lipofectamine 2000. Cells were cultured for 36 h after the transfection, and then luciferase activity was assessed using a dual luciferase assay system (Promega). Results were normalized to Renilla luciferase activity.

Statistical analysis

Results were analyzed using GraphPad Prism 4.0 (Graphpad Software, La Jolla, CA, USA) and the data are shown as means ± SEM. A Student's t-test was used for comparisons between different groups. P<0.05 was considered to be statistically significant.

Results

Establishment of paclitaxel- or cisplatin-resistant A549 cells

To develop A549 lung carcinoma cells that are resistant to paclitaxel or cisplatin, A549 cells were exposed to increasing concentrations of paclitaxel or cisplatin for more than 12 months. After each round, the surviving cells that reached >70% confluency were passaged by trypsinization, and the concentration of paclitaxel or cisplatin was increased. The procedure was performed repeatedly until the cells showed resistance to the inhibitory activities of 200 μg/ml paclitaxel and 1,000 μg/ml cisplatin. The resulting cells (A549/PTX and A549/DDP cells) were cultured for an additional 3 months in RPMI-1640 medium containing 200 μg/ml paclitaxel or 1,000 μg/ml cisplatin.

To assess the resistance properties of A549 cells and their derivatives, we exposed the cells to increasing amounts of paclitaxel or cisplatin for 24, 48 or 72 h. The viability was decreased by drug exposure for all cell lines in a dose- and time-dependent manner; however, A549/PTX and A549/DDP cells were significantly more resistant than A549 cells (Fig. 1). The IC50 was greater for A540/PTX and A549/DDP cells than for the parental A549 cells at all times of drug treatment, and the RI ranged from 6 to 10.2 for A549/PTX cells and from 3.3 to 9.4 for A549/DDP cells (Tables III and IV). These observations demonstrate that A549/PTX and A549/DDP cells acquired chemoresistance.

Table III

The inhibitory 50% concentration (IC50) and resistance index (RI) of A549 and A549/PTX cells treated with paclitaxel for 24, 48 and 72 h.

Table III

The inhibitory 50% concentration (IC50) and resistance index (RI) of A549 and A549/PTX cells treated with paclitaxel for 24, 48 and 72 h.

IC50 (μmol·l−1)

Time (h)A549A549/PTXRI
241.22±0.467.3±0.446
480.52±0.083.22±0.266.2
720.25±0.172.55±0.3110.2

Table IV

The resistance index (RI) and inhibitory 50% concentration (IC50) of A549 and A549/DDP cells treated with cisplatin for 24 h, 48 h and 72 h.

Table IV

The resistance index (RI) and inhibitory 50% concentration (IC50) of A549 and A549/DDP cells treated with cisplatin for 24 h, 48 h and 72 h.

IC50 (μmol·l−1)

Time (h)A549A549/DDPRI
2475.83±0.53250.24±0.943.3
4817.62±0.1266.96±0.593.8
725.84±0.3254.90±0.639.4
A549/PTX and A549/DDP cells have increased motility and invasion activity

The acquisition of metastatic properties by cancer cells is characterized by increased migration and invasion abilities. Wound healing, migration and invasion assays were performed to assess the migratory potential of A549/PTX and A549/DDP cells in relation to A549 cells. The results show that significantly increased numbers of cells migrated across the ‘wound’ in a scratch assay, suggesting that A549/PTX and A549/DDP cells acquired enhanced migration capacity (Fig. 2A). Transwell chamber assays were performed to further compare the migration and invasion capacity of A549/DDP and A549/PTX cells to A549 cells (Fig. 2B). A549/PTX had ~1.8-fold increased migration and 2.9-fold increased invasion levels relative to A549 cells, and A549/DDP cells had ~1.5-fold increased migration and 2.2-fold increased invasion levels. Thus, A549/PTX and A549/DDP cells have increased metastatic properties as compared with the parental A549 cells.

A549/DDP and A549/PTX cells showed molecular and morphological alterations that were consistent with EMT

To determine whether EMT progression is detected in A549/PTX and A549/DDP cells, we assessed the morphological changes of the cells. Our results show that each of the resistant A549 cell lines had marked morphologic changes compared with the parental cell lines (Fig. 3A). Whereas A549 cells displayed a rounded shape and little formation of pseudopodia, A549/PTX and A549/DDP cells displayed phenotypic changes, including a loss of cell polarity and increased formation of pseudopodia, leading to elongated, irregular fibroblastoid appearance.

To further determine whether A549/PTX and A549/DDP cells have specific molecular changes consistent with EMT, we measured the expression of epithelial and mesenchymal phenotype markers with western blotting (Fig. 3B) and RT-PCR analysis (Fig. 3C). Consistent with EMT progression in A549/PTX and A549/DDP cells, the expression of epithelial adhesion molecules E-cadherin and β-catenin were significantly reduced in A549/PTX and A549/DDP cells. The expression of the mesenchymal markers (vimentin, MMP-2, MMP-9, Snail, Slug and ZEB1) was elevated in A549/PTX and A549/DDP cells, although the expression of Twist showed no significant change. These results showed the expression levels of genes that are known to play a critical role in EMT are modulated in lung adenocarcinoma cells upon acquisition of either paclitaxel- or cisplatin-resistance.

miR-181a is differentially expressed in A549/PTX, A549/DDP, and A549 parental A549 cells

To identify miRNAs that are potentially involved in the underlying mechanisms of drug-resistant cells and induction of EMT-like properties, we used microarray analysis to assess the differential expression of miRNA in A549/PTX and parental A549 cells. Ten miRNAs were significantly upregulated and 5 were significantly down-regulated in A549/PTX cells relative to A549 cells (absolute log fold-change |logFC|>4; Table V). Among the differentially expressed miRNAs, the miR-181 family (miR-181a, b and c) was significantly upregulated, and among the miR-181 family members, miR-181a was the most significantly dysregulated. qRT-PCR analysis verified that miR-181a was upregulated 16-fold in A549/PTX compared with A549 cells. Furthermore, miR-181a was also upregulated 12-fold in A549/DDP compared to parental A549 cells (Fig. 4A). Therefore, we hypothesized that miR-181a may represent a primary regulator in lung adenocarcinoma cells.

Table V

Differentially expressed miRNAs in A549/PTX compared with A549 cells (|LogFC|>4).

Table V

Differentially expressed miRNAs in A549/PTX compared with A549 cells (|LogFC|>4).

miRNAUp/downFold change (log2)
hsa-miR-135b-5pUp16.2407
hsa-miR-155-5pUp15.7135
hsa-miR-181a-5pUp8.0039
hsa-miR-301a-3pUp7.9692
hsa-miR-19a-3pUp7.9104
hsa-miR-205-5pUp5.0039
hsa-miR-181c-5pUp4.9968
hsa-miR-181b-5pUp4.0564
hsa-miR-23b-3pUp4.0254
hsa-miR-96-5pUp4.006
hsa-let-7g-5pDown−4.0231
hsa-miR-128-5pDown−4.0284
hsa-miR-191-5pDown−4.1711
hsa-miR-200c-3pDown−5.0792
hsa-miR-25-3pDown−6.0224

[i] Up, upregulation; down, downregulation.

miR-181a regulates metastatic properties and EMT in human lung adenocarcinoma cells

To explore the potential role of miR-181a in regulating metastasis and EMT in lung adenocarcinoma cells, we used miR-181a mimic and inhibitor to modulate miR-181 expression (Fig. 4B). Our results demonstrate that miR-181a mimic increases the ability of A549 cells to migrate and invade (Fig. 5A). Conversely, miR-181a inhibitor decreases the migration and invasion abilities in both A549/PTX and A549/DDP cells (Fig. 5B). These results suggest that miR-181a is an upstream regulator of migration and invasion in lung adenocarcinoma cells.

To further assess the role of miR-181a in EMT, we examined the morphological and molecular characteristics of cells that were transfected with miR-181a inhibitor or miR-181a mimic. Morphological study showed that the A549 cells transfected with miR-181a mimic had a more mesenchymal appearance (Fig. 6A). Furthermore, at both the protein (Fig. 6B) and mRNA (Fig. 6C) levels, miR-181a mimic caused a reduction in the expression of the epithelial adhesion molecules (E-cadherin and β-cadherin) and an increase in the expression of the mesen-chymal markers (vimentin, MMP-2, MMP-9, Snail, ZEB1 and Slug), which was similar to the pattern observed for A549/PTX and A549/DDP cells (Fig. 3). Conversely, transfection of A549/PTX cells with miR-181a inhibitor may reverse drug resistance, as indicated by MTT. These cells also showed a modest but clearly visible change in morphological characteristics, changing from mesenchymal-like spindle-cell shape to epithelial-like shapes (Fig. 7A and B), with opposite effects on the expression of epithelial and mesenchymal markers (Fig. 7C and D). Similar results were observed for A549/DDP cells (Fig. 8). These results directly demonstrate that overexpression of miR-181a promotes the acquisition of EMT phenotype in parental A549 cells and that inhibition of miR-181a expression reverses the EMT phenotype in A549/PTX or A549/DDP cells. Taken together, miR-181a plays an important role in the regulation of the EMT in human lung adenocarcinoma cells.

miR-181a regulates the protein expression of PTEN by directly targeting 3′UTR of PTEN

To further explore the molecular mechanism of miR-181a in promoting EMT progression in lung adenocarcinoma cells, miRNA target prediction databases (miRNA.org and TargetScan) available online were used to identify potential targets of miR-181a in humans. This analysis identified PTEN, an inhibitor of the PI3 kinase/Akt pathway that is reported to play an important role in the pathogenesis and drug resistance, as a potential target of miR-181a (Fig. 9A). Consistent with a potential role for PTEN inhibition in the development of drug resistance, the protein levels of PTEN were reduced in the paclitaxel- or cisplatin-resistant A549 cells compared with the parental A549 cells (Fig. 9B). We cloned the wild-type 3′UTR of PTEN and a mutated version of the 3′UTR into the pGL3-report plasmid to investigate whether the predicted binding site of miR-181a to the 3′UTR of the PTEN gene was responsible for this regulation. The activity of the reporter containing the wild-type sequence, but not the mutant sequence, was decreased in A549 cells by co-transfection with miR-181a mimic as demonstrated by luciferase assays. Conversely, miR-181a inhibitor increased the activity of wild-type, but mutant reporter activity did not change in A549/PTX and A549/DDP cells (Fig. 9C). These results suggest that miR-181a may target the PTEN promoter to inhibit its expression.

Figure 9

miR-181a directly targets PTEN by binding to its 3′UTR. (A) The predicted miR-181a binding site within PTEN 3′UTR and its mutant version resulting from site mutagenesis are presented. Red indicates the nucleotides that were mutated to create a mismatch. (B) PTEN expression in lung adenocar-cinoma cell lines was assessed by western blotting. (C) Luciferase assays were performed for the indicated cell lines transfected with a luciferase reported containing either the wild-type or mutant PTEN 3′UTR sequence; together with a blank control, a negative control, or miR-181 mimic/inhibitor. Results were determined relative to a co-transfected Renilla construct and were standardized to 1.0 in the blank control cells (compared to blank or mimic/inhibitor-NC, mean ± SEM, n=3, *P<0.05). (D) Western blots of PTEN are shown for A549, A549/PTX or A549/DDP cells that were non-transfected (blank) or that were transfected with negative control or miR-181a mimic/inhibitor. β-actin was also tested as a loading control. Shown are results that are representative of three independent experiments. (E) A proposed model for miR-181a/PTEN signaling pathway in A549/PTX and A549/DDP EMT-type cells, prolonged exposure of A549 cells to either paclitaxel or cisplatin caused elevated expression of miR-181a. miR-181a targets the promoter of PTEN to inhibit its expression. Because PTEN is an inhibitor of the PI3 kinase pathway, which regulates metastatic processes, inhibition of PTEN expression leads to the enhancement of migration, invasion, and EMT progression.

To verify these findings with endogenous PTEN, we investigated whether miR-181a modulates the expression of PTEN protein in A549 cells and its drug-resistant derivatives. Western blotting results demonstrated that PTEN protein levels are reduced in A549 cells transfected with miR-181a mimic, but that conversely, the protein expression of PTEN is increased when miR-181a inhibitor is transfected into A549/PTX and A549/DDP cells (Fig. 9D). Overall, these results suggest that miR-181a inhibits the protein expression of PTEN by directly targeting 3′UTR of PTEN.

Discussion

Drug resistance is achieved by the sequential acquisition of genetic alterations that re-route crucial pathways promoting tumor cell invasive/aggressive phenotypes. Although the causes of drug resistance have been explored for many years, there remains no remedy to overcome it to improve clinical outcomes in resistant cancers. There is growing evidence suggesting that drug-resistance in cancer cells is associated with the EMT process in human cancers, including lung adenocarcinoma (19). For example, gefitinib resistance of cancer cells is correlated with TM4SF5-mediated EMT (20). Similarly, activation of the PI3 kinase/Akt/HIF-1α pathway contributes to hypoxia-induced EMT and chemoresistance in hepatocellular carcinoma (21). Moreover, paclitaxel-resistant ovarian cancer cells, cisplatin resistant colorectal cancer cells, gemcitabine-resistant pancreatic cancer cells and tamoxifen-resistant breast cancer cells have an EMT phenotype which includes downregulation of E-cadherin and upregulation of vimentin (22). Similarly, we have observed that paclitaxel or cisplatin-resistant lung adenocarcinoma cells acquire EMT features. Consistently, in the present study, we found that multi-resistant lung adenocarcinoma cells demonstrated altered morphological characteristics of cells similar to EMT with decreased E-cadherin and increased vimentin, Snail and Slug, suggesting that there is a link between chemoresistance and EMT in lung adenocarcinoma.

Recently, miRNAs have emerged as crucial mediators in regulating the cellular responses of cancer cells to therapy (23). Patient response to chemotherapy has been shown to be closely correlated to the functional status of miRNAs. Although the mechanisms of miRNA-regulated drug resistance are still largely unknown, current evidence suggest several roles for miRNAs, including influencing therapy-induced cell death, altering drug targets, regulating multiple drug resistance (MDR)-related proteins, modifying bioavailable drug concentrations and promoting angiogenesis (2428). It is also becoming increasingly evident that miRNAs are key modulators of EMT, which is an important process that drives cancer metastasis. Recent studies have shown that miRNAs can play a role as important modulators of EMT through the regulation of E-cadherin and other molecules such as vimentin and ZEB (29). For example, the members of the miR-429 family inhibit cells growth and invasion and regulate EMT-related marker genes which happens throngh the targeting of Onecut2 in colorectal carcinoma (30). Moreover, miR-205 is downregulated during EMT with the associated downregulation of ZEB1 and ZEB2 expression in a panel of epithelial breast cancer cells (31). In the present study, we identified 10 miRNAs that are upregulated and 5 miRNAs are downregulated in A549/PTX cells after compared with A549 cells. To determine a potential relation with drug resistance in lung cancer cells, we selected miR-181a, the most highly differentially expressed miRNA among the miR-181a, miR-181b, and miR-181c genes. This miRNA was also upregulated in A549/DDP cells, which supports its general association with the multi-drug resistant phenotype.

Our results indicated that overexpression of miR-181a promotes invasion and migration and the acquisition of EMT in lung adenocarcinoma cells, but that inhibition of miR-181a expression partly reverses EMT. Target prediction tools identified PTEN as a putative target gene of miR-181a. The PTEN signaling pathway has been reported to play a role in the control of various cellular processes, such as cell proliferation, apoptosis, invasion, metastases and EMT in human cancer (32,33); therefore, a role for miR-181 in targeting PTEN is consistent with its known biological effect. In fact, some other miRNAs are capable of conferring drug resistance by targeting PTEN in other types of cancer. For example, miR-21 induces cell survival and chemoresistance, by binding the 3′UTR of PTEN mRNA (34). Loss of PTEN is a very frequent genetic aberration in malignant tumors such as breast cancer, gastric cancer and glioblastoma (3436), and PTEN loss is significantly associated with cytotoxic drug resistance (37). In this study, we found that miR-181a negatively regulates PTEN expression in lung adenocarcinoma cell lines. As a result, these cells became more aggressive and invasive after transfection with an miR-181a expression construct. This finding is consistent with clinical observations, which have revealed that more advanced stage patients expressed higher levels of miR-181a (38). There is growing evidence suggesting that dysfunction of PTEN has prognostic importance in several malignancies, including lung adenocarcinoma. Our findings reveal that targeting PTEN at the post-transcriptional level by miRNAs such as miR-181a mediates PTEN inactivation (Fig. 9E). These results suggest a mechanism whereby increased miR-181a expression may be associated with reduced survival in lung adenocarcinoma patients, and also support the development of miR-181a inhibitor as a potential target for reversing the effects of drug resistance.

In conclusion, we demonstrated that miR-181a responds to chemotherapy in lung adenocarcinoma cells by changing the levels of both itself and its target PTEN. Downregulation of miR-181a could successfully sensitize cancer cells to chemotherapy. In the future, therapeutic strategies could be developed based on the predictive levels of miR-181a. Additionally, miR-181a repression could potentially be combined with chemotherapy to prolong drug sensitivity in lung adenocarcinoma, and potentially other types of cancer.

Acknowledgements

The present study was supported by grants from the National Natural Science Foundation of China (nos. 81372899 and 81000992), the Twelfth Five-year Science and Technology Research Program of Anhui Provincial Scientific Committee (no. 1301042200), the Key Project of Natural Science Research of the Educational Department of Anhui Province, China (no. 1208085MH130), and the Natural Science Foundation of Anhui province (no. 1508085MH166).

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Brown T, Pilkington G, Bagust A, Boland A, Oyee J, Tudur-Smith C, Blundell M, Lai M, Martin Saborido C, Greenhalgh J, et al: Clinical effectiveness and cost-effectiveness of first-line chemotherapy for adult patients with locally advanced or metastatic non-small cell lung cancer: A systematic review and economic evaluation. Health Technol Assess. 17:1–278. 2013.

3 

Macedo-Pérez EO, Morales-Oyarvide V, Mendoza-García VO, Dorantes-Gallareta Y, Flores-Estrada D and Arrieta O: Long progression-free survival with first-line paclitaxel plus platinum is associated with improved response and progression-free survival with second-line docetaxel in advanced non-small-cell lung cancer. Cancer Chemother Pharmacol. 74:681–690. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Hugo H, Ackland ML, Blick T, Lawrence MG, Clements JA, Williams ED and Thompson EW: Epithelial-mesenchymal and mesenchymal-epithelial transitions in carcinoma progression. J Cell Physiol. 213:374–383. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, Bruyneel E, Mareel M, Huylebroeck D and van Roy F: The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell. 7:1267–1278. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Wang X, Ling MT, Guan XY, Tsao SW, Cheung HW, Lee DT and Wong YC: Identification of a novel function of TWIST, a bHLH protein, in the development of acquired taxol resistance in human cancer cells. Oncogene. 23:474–482. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Haslehurst AM, Koti M, Dharsee M, Nuin P, Evans K, Geraci J, Childs T, Chen J, Li J, Weberpals J, et al: EMT transcription factors snail and slug directly contribute to cisplatin resistance in ovarian cancer. BMC Cancer. 12:912012. View Article : Google Scholar : PubMed/NCBI

8 

Zhao W, Zhou Y, Xu H, Cheng Y and Kong B: Snail family proteins in cervical squamous carcinoma: Expression and significance. Clin Invest Med. 36:E223–E233. 2013.PubMed/NCBI

9 

Bowen KA, Doan HQ, Zhou BP, Wang Q, Zhou Y, Rychahou PG and Evers BM: PTEN loss induces epithelial - mesenchymal transition in human colon cancer cells. Anticancer Res. 29:4439–4449. 2009.PubMed/NCBI

10 

Soria JC, Lee HY, Lee JI, Wang L, Issa JP, Kemp BL, Liu DD, Kurie JM, Mao L and Khuri FR: Lack of PTEN expression in non-small cell lung cancer could be related to promoter methylation. Clin Cancer Res. 8:1178–1184. 2002.PubMed/NCBI

11 

Jin G, Kim MJ, Jeon HS, Choi JE, Kim DS, Lee EB, Cha SI, Yoon GS, Kim CH and Jung TH: PTEN mutations and relationship to EGFR, ERBB2, KRAS, and TP53 mutations in non-small cell lung cancers. Lung Cancer. 69:279–283. 2010. View Article : Google Scholar

12 

De Craene B and Berx G: Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 13:97–110. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Moustakas A and Heldin CH: Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 98:1512–1520. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST and Patel T: MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 133:647–658. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Jiang J, Zhang Y, Yu C, Li Z, Pan Y and Sun C: MicroRNA-492 expression promotes the progression of hepatic cancer by targeting PTEN. Cancer Cell Int. 14:952014. View Article : Google Scholar : PubMed/NCBI

16 

Chun-Zhi Z, Lei H, An-Ling Z, Yan-Chao F, Xiao Y, Guang-Xiu W, Zhi-Fan J, Pei-Yu P, Qing-Yu Z and Chun-Sheng K: MicroRNA-221 and microRNA-222 regulate gastric carcinoma cell proliferation and radioresistance by targeting PTEN. BMC Cancer. 10:3672010. View Article : Google Scholar : PubMed/NCBI

17 

Gibbons DL, Lin W, Creighton CJ, Rizvi ZH, Gregory PA, Goodall GJ, Thilaganathan N, Du L, Zhang Y, Pertsemlidis A, et al: Contextual extracellular cues promote tumor cell EMT and metastasis by regulating miR-200 family expression. Genes Dev. 23:2140–2151. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Zhang P, Liu H, Xia F, Zhang QW, Zhang YY, Zhao Q, Chao ZH, Jiang ZW and Jiang CC: Epithelial-mesenchymal transition is necessary for acquired resistance to cisplatin and increases the metastatic potential of nasopharyngeal carcinoma cells. Int J Mol Med. 33:151–159. 2014.

19 

Reka AK, Chen G, Jones RC, Amunugama R, Kim S, Karnovsky A, Standiford TJ, Beer DG, Omenn GS and Keshamouni VG: Epithelial-mesenchymal transition-associated secretory phenotype predicts survival in lung cancer patients. Carcinogenesis. 35:1292–1300. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Lee MS, Kim HP, Kim TY and Lee JW: Gefitinib resistance of cancer cells correlated with TM4SF5-mediated epithelial-mesenchymal transition. Biochim Biophys Acta. 1823:514–523. 2012. View Article : Google Scholar

21 

Jiao M and Nan KJ: Activation of PI3 kinase/Akt/HIF-1α pathway contributes to hypoxia-induced epithelial-mesenchymal transition and chemoresistance in hepatocellular carcinoma. Int J Oncol. 40:461–468. 2012.

22 

Li Y, VandenBoom TG II, Kong D, Wang Z, Ali S, Philip PA and Sarkar FH: Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 69:6704–6712. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Guo H, Ingolia NT, Weissman JS and Bartel DP: Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 466:835–840. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A and Lund AH: Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem. 283:1026–1033. 2008. View Article : Google Scholar

25 

Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, Wojcik SE, Aqeilan RI, Zupo S, Dono M, et al: miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA. 102:13944–13949. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Dews M, Homayouni A, Yu D, Murphy D, Sevignani C, Wentzel E, Furth EE, Lee WM, Enders GH, Mendell JT, et al: Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. Nat Genet. 38:1060–1065. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Jiang K: Biotech comes to its ‘antisenses’ after hard-won drug approval. Nat Med. 19:2522013. View Article : Google Scholar

28 

Bockhorn J, Dalton R, Nwachukwu C, Huang S, Prat A, Yee K, Chang YF, Huo D, Wen Y, Swanson KE, et al: MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat Commun. 4:13932013. View Article : Google Scholar : PubMed/NCBI

29 

Ren J, Chen Y, Song H, Chen L and Wang R: Inhibition of ZEB1 reverses EMT and chemoresistance in docetaxel-resistant human lung adenocarcinoma cell line. J Cell Biochem. 114:1395–1403. 2013. View Article : Google Scholar

30 

Sun Y, Shen S, Liu X, Tang H, Wang Z, Yu Z, Li X and Wu M: MiR-429 inhibits cells growth and invasion and regulates EMT-related marker genes by targeting Onecut2 in colorectal carcinoma. Mol Cell Biochem. 390:19–30. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Lee JY, Park MK, Park JH, Lee HJ, Shin DH, Kang Y, Lee CH and Kong G: Loss of the polycomb protein Mel-18 enhances the epithelial-mesenchymal transition by ZEB1 and ZEB2 expression through the downregulation of miR-205 in breast cancer. Oncogene. 33:1325–1335. 2014. View Article : Google Scholar

32 

Alimonti A, Carracedo A, Clohessy JG, Trotman LC, Nardella C, Egia A, Salmena L, Sampieri K, Haveman WJ, Brogi E, et al: Subtle variations in Pten dose determine cancer susceptibility. Nat Genet. 42:454–458. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Aguissa-Touré AH and Li G: Genetic alterations of PTEN in human melanoma. Cell Mol Life Sci. 69:1475–1491. 2012. View Article : Google Scholar

34 

Liu ZL, Wang H, Liu J and Wang ZX: MicroRNA-21 (miR-21) expression promotes growth, metastasis, and chemo- or radioresistance in non-small cell lung cancer cells by targeting PTEN. Mol Cell Biochem. 372:35–45. 2013. View Article : Google Scholar

35 

Wu Z, He B, He J and Mao X: Upregulation of miR-153 promotes cell proliferation via downregulation of the PTEN tumor suppressor gene in human prostate cancer. Prostate. 73:596–604. 2013. View Article : Google Scholar

36 

Yang TS, Yang XH, Wang XD, Wang YL, Zhou B and Song ZS: MiR-214 regulate gastric cancer cell proliferation, migration and invasion by targeting PTEN. Cancer Cell Int. 13:682013. View Article : Google Scholar : PubMed/NCBI

37 

Wang Q, Li SH, Wang H, Xiao Y, Sahin O, Brady SW, Li P, Ge H, Jaffee EM, Muller WJ, et al: Concomitant targeting of tumor cells and induction of T-cell response synergizes to effectively inhibit trastuzumab-resistant breast cancer. Cancer Res. 72:4417–4428. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Schwind S, Maharry K, Radmacher MD, Mrózek K, Holland KB, Margeson D, Whitman SP, Hickey C, Becker H, Metzeler KH, et al: Prognostic significance of expression of a single microRNA, miR-181a, in cytogenetically normal acute myeloid leukemia: A Cancer and Leukemia Group B study. J Clin Oncol. 28:5257–5264. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2015
Volume 47 Issue 4

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li H, Zhang P, Sun X, Sun Y, Shi C, Liu H and Liu X: MicroRNA-181a regulates epithelial-mesenchymal transition by targeting PTEN in drug-resistant lung adenocarcinoma cells. Int J Oncol 47: 1379-1392, 2015
APA
Li, H., Zhang, P., Sun, X., Sun, Y., Shi, C., Liu, H., & Liu, X. (2015). MicroRNA-181a regulates epithelial-mesenchymal transition by targeting PTEN in drug-resistant lung adenocarcinoma cells. International Journal of Oncology, 47, 1379-1392. https://doi.org/10.3892/ijo.2015.3144
MLA
Li, H., Zhang, P., Sun, X., Sun, Y., Shi, C., Liu, H., Liu, X."MicroRNA-181a regulates epithelial-mesenchymal transition by targeting PTEN in drug-resistant lung adenocarcinoma cells". International Journal of Oncology 47.4 (2015): 1379-1392.
Chicago
Li, H., Zhang, P., Sun, X., Sun, Y., Shi, C., Liu, H., Liu, X."MicroRNA-181a regulates epithelial-mesenchymal transition by targeting PTEN in drug-resistant lung adenocarcinoma cells". International Journal of Oncology 47, no. 4 (2015): 1379-1392. https://doi.org/10.3892/ijo.2015.3144