15-Deoxy-Δ12,14-prostaglandin J2 induces growth inhibition, cell cycle arrest and apoptosis in human endometrial cancer cell lines

  • Authors:
    • Haili Li
    • Hisashi Narahara
  • View Affiliations

  • Published online on: February 5, 2013     https://doi.org/10.3892/ijmm.2013.1268
  • Pages: 778-788
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

15-Deoxy-∆12,14-prostaglandin J2 (15d-PGJ2), a peroxisome proliferator-activated receptor γ ligand, has been reported to have antiproliferative activity in certain types of cancer. The purpose of this study was to elucidate the effect of 15d-PGJ2 on endometrial cancer cells, as well as the mechanism of action. Endometrial cancer-derived cells (HHUA, Ishikawa and HEC-59) were treated with various concentrations of 15d-PGJ2, and its effects on cell growth, the cell cycle and apoptosis were investigated in vitro. Using cDNA microarrays, some potential targets of this drug were identified. All endometrial cancer cell lines were sensitive to the growth-inhibitory effect of 15d-PGJ2. Cell cycle arrest at the G2/M phase of the cell cycle and induction of apoptosis were observed. Concerning the gene expression changes induced by 15d-PGJ2 treatment, the upregulation of aldo-keto reductase family 1 member C3 (AKR1C3) and the downregulation of anterior gradient homolog 3 (AGR3) and nitric oxide synthase 2A (NOS2A) were confirmed using western blot analysis in all the cell lines examined. These results suggest that 15d-PGJ2 may be a novel therapeutic option for the treatment of endometrial cancer.

Introduction

Endometrial cancer is the most common malignant tumor of the female genital tract, and its incidence has increased in recent years (1,2). Furthermore, the search for agents effective in the treatment of either advanced or recurrent endometrial cancer has proved to be disappointing (2,3). Therefore, innovative approaches are required for the treatment of endometrial cancer.

Peroxisome proliferator-activated receptor (PPAR)γ is a nuclear hormone receptor and its ligands, troglitazone and pioglitazone, have been shown to induce apoptosis in several types of cancer cells, including endometrial cancer cells (46). 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) is a PPARγ ligand that activates PPARγ at micromolar concentrations in humans in vivo (79). Recently, 15d-PGJ2 was reported to have antiproliferative activity in certain types of cancer (4,1012). However, the effect of 15d-PGJ2 on endometrial cancer cells has not yet been investigated.

The present study aimed to investigate the biological and therapeutic effects of 15d-PGJ2 on endometrial cancer. We examined whether this compound can mediate cell growth inhibition, cell cycle arrest and apoptosis in endometrial cancer cell lines (HHUA, Ishikawa and HEC-59). Furthermore, to identify potential and novel target genes responsive to the anticancer effect in 15d-PGJ2-treated endometrial cancer cells, we analyzed the global changes in gene expression in HHUA cells following treatment with 15d-PGJ2 using cDNA microarrays. The expression of candidate proteins was confirmed by western blot analysis in the 3 endometrial cancer cell lines.

Materials and methods

Cell lines

The HHUA human endometrial cancer cell line was obtained from Riken (Ibaraki, Japan). The Ishikawa human endometrial cancer cell line was kindly provided by Dr Masato Nishida (Tsukuba University, Ibaraki, Japan). The HEC-59 human endometrial cancer cell line was obtained from the American Type Culture Collection (Manassas, VA, USA). The cells were maintained as monolayers at 37°C in 5% CO2/air in Dulbecco’s modified Eagle’s medium (DMEM; Gibco, Rockville, MD, USA) containing 10% heat-inactivated fetal bovine serum (FBS; Omega, Tarzana, CA, USA).

Chemicals

15d-PGJ2 was obtained from Enzo Life Sciences (Plymouth Meeting, Montgomery County, PA, USA), and prepared as a 20 mg/ml stock solution in dimethyl sulfoxide (DMSO). The stock solution was stored in aliquots at −20°C.

Assessment of cell proliferation and cell viability

The cell proliferation and cell viability were determined in 96-well plates by a modified methylthiazol tetrazolium (MTT) assay using WST-1 (Roche Diagnostics, Penzberg, Germany) following the manufacturer’s instructions. We distributed 5×103 cells in DMEM supplemented with 10% FBS into each well of a 96-well flat-bottomed microplate (Corning, Inc., New York, NY, USA) and incubated them overnight. The medium was then removed, and the cells were incubated for 48 h with 100 μl of experimental medium containing various concentrations of 15d-PGJ2. Thereafter, 10 μl of WST-1 dye was added to each well, and the cells were further incubated for 4 h. All experiments were performed in the presence of 10% FBS. Cell proliferation was evaluated by measuring the absorbance at 540 nm. Data were calculated as the ratio of the values obtained for the 15d-PGJ2-treated cells to those for the untreated controls.

Cell cycle analysis by flow cytometry

The cell cycle was analyzed by flow cytometry after 2 days of culturing. Cells (5×104) were exposed to 15d-PGJ2 in 6-well flat-bottomed plates for 48 h. Analysis was performed immediately after staining using the CellFIT program (Becton-Dickinson, San Jose, CA, USA), whereby the S phase was calculated using an RFit model.

Measurement of apoptosis [flow-cytometric analysis with the Annexin V/propidium iodide (PI) assay]

Cells were plated and grown overnight until they reached 80% confluence and then treated with 15d-PGJ2. After 48 h, detached cells in the medium were collected, and the remaining adherent cells were harvested by trypsinization. The cells (1×105) were washed with PBS and resuspended in 250 μl of binding buffer (Annexin V-FITC kit; Becton-Dickinson) containing 10 μl of 20 μg/ml PI and 5 μl of Annexin V-FITC, which binds to phosphatidylserine translocated to the exterior of the cell membrane early in the apoptotic pathway as well as during necrosis. After incubation for 10 min at room temperature in a light-protected area, the samples were analyzed on a FACSCalibur flow cytometer (Becton-Dickinson). FITC and PI emissions were detected in the FL-1 and FL-2 channels, respectively. For each sample, data from 30,000 cells were recorded in list mode on logarithmic scales. Subsequent analysis was performed with CellQuest software (Becton-Dickinson).

Mitochondrial transmembrane potential (MTP)

Cells were prepared for FACS analysis as described above and stained using a Mitocapture Apoptosis Detection kit obtained from BioVision (Palo Alto, CA, USA) with a fluorescent lipophilic cationic reagent that assesses mitochondrial membrane permeability, according to the manufacturer’s recommendations.

Microarray analysis

Total RNA was extracted from the 15d-PGJ2-treated and untreated HHUA cells using an RNeasy mini kit (Qiagen, Valencia, CA, USA) in accordance with the manufacturer’s instructions. Prior to hybridization, the quantity and quality of the total RNA were evaluated using a spectrophotometer and a 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA), respectively. Cy3-labeled cRNA targets were generated using a Low RNA Input Fluorescent Linear Amplification kit (Agilent Technologies). A human 44 K oligoarray was used for hybridization, in accordance with the manufacturer’s recommendations (Agilent Technologies). A laser confocal scanner (Agilent Technologies) was used to measure signal intensities in the expression microarray analysis. Feature Extraction software (Version 9.1; Agilent Technologies) with the manufacturer’s recommended settings was applied for the microarray image analysis. Analysis of the microarray images was performed with GeneSpring 7.3.1 software (Agilent Technologies). For comparison among multiple arrays, probe set data were median-normalized/chip. The data were then centered across the genes in 6 normal controls, followed by filtering based on a signal intensity of ≥100, and contained no flagged values. Among these differentially expressed genes, those designated as ‘upregulated’ were overexpressed >2-fold in comparison with the controls (P<0.05), whereas those designated as ‘downregulated’ were underexpressed <0.75-fold compared with the controls (P<0.05). Annotations including chromosomal loci were provided by Agilent Technologies.

For Gene Ontology (GO) analysis, differentially expressed genes were defined as those with a >2-fold increase or decrease in expression relative to the controls. GO term enrichment in the upregulated or downregulated gene sets was assessed using the GOstat web tool (13).

Western blot analysis

Cells were washed twice in PBS, suspended in lysis buffer [50 mM Tris (pH 8.0), 150 mM NaCl, 0.1% SDS, 0.5% sodium deoxycholate, 1% NP-40, phenylmethylsulfonyl fluoride at 100 μg/ml, aprotinin at 2 μg/ml, pepstatin at 1 μg/ml and leupeptin at 10 μg/ml], and placed on ice for 30 min. After centrifugation at 15,000 × g for 15 min at 4°C, the suspension was collected. Protein concentrations were quantified using the Bio-Rad protein Assay Dye Reagent Concentrate (Bio-Rad Laboratories, Hercules, CA, USA) according to the manufacturer’s recommendations. Whole-cell lysates (40 μg) were resolved by SDS-polyacrylamide gel electrophoresis on a 4–15% gel, transferred onto a polyvinylidene difluoride membrane (Immobilon; Amersham, Arlington Heights, IL, USA), and probed sequentially with antibodies against anterior gradient homolog 3 (AGR3; 1:1,000; GeneTex, Irvine, CA, USA), aldo-keto reductase family 1 member C1 (AKR1C1; 1:1,000; GeneTex), aldo-keto reductase family 1 member C3 (AKR1C3; 1:1,000; ProteinTech, Chicago, IL, USA), α-1-microglobulin/bikunin precursor (AMBP; 1:1,000; Abnova, Taipei, Taiwan), complement component 3a receptor 1 (C3AR1; 1:1,000; Abnova), chondroadherin (CHAD; 1:1,000; Avia Systems Biology, San Diego, CA, USA), Fer3-like (Drosophila) (FERDL3; 1:1,000; Avia Systems Biology), ferritin, light polypeptide (FTL; 1:1,000; GeneTex), galactose-3-O-sulfotransferase 3 (GAL3ST3; 1:1,000; Avia Systems Biology), glutamate-cysteine ligase, modifier subunit (GCLM; 1:1,000; Abnova), heme oxygenase (decycling) 1 (HMOX1; 1:1,000; Abnova), intercellular adhesion molecule 4 (ICAM4; 1:1,000; Abnova), potassium voltage-gated channel, shaker-related subfamily, β member 1 (KCNAB1; 1:1,000; Osenses, Keswick, Australia), mitochondrial ribosomal protein L37 (MRPL37; 1:1,000; ProteinTech), nitric oxide synthase 2A (NOS2A; 1:1,000; Applied Biological Materials, Kampenhout, Belgium), phosphorylated eukaryotic translation initiation factor 4E (p-eIF4E; 1:1,000; Bioworld Technology, Minneapolis, MN, USA), pirin (PIR; 1:1,000; Avia Systems Biology), tripartite motif-containing 16 (TRIM16; 1:1,000; Avia Systems Biology), thioredoxin reductase 1 (TXNRD1; 1:1,000; ProteinTech), UDP glucuronosyltransferase 1 family, polypeptide A6 (UGT1A6; 1:1,000; LifeSpan Biosciences, Seattle, WA, USA) and GAPDH monoclonal antibody (mAb) (1:10,000; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). The blots were developed using an enhanced chemiluminescent (ECL) kit (Amersham). Band intensity was measured using the public domain Image program ImageJ version 1.44, and fold increase in expression as compared with control, untreated cells was calculated.

Statistical analysis

Data are presented as the means ± SD of representative experiments and were analyzed by the Bonferroni-Dunn test using StatView 4.5 software (Abacus Concepts, Berkeley, CA, USA). A P-value <0.05 was considered to indicate a statistically significant difference.

Results

Effects of 15d-PGJ2 on the proliferation and viability of endometrial cancer cell lines in vitro

The antitumor effects of 15d-PGJ2 on 3 endometrial cancer cell lines in vitro were examined using a WST-1 assay of the 2-day exposure to 15d-PGJ2. Significant inhibitory effects of 15d-PGJ2 on the cell growth were observed in all 3 endometrial cancer cell lines (Ishikawa, HHUA and HEC-59) (Fig. 1).

Cell cycle analysis of endometrial cancer cells following exposure to 15d-PGJ2

We then investigated whether 15d-PGJ2 would lead to the induction of apoptosis and/or cell cycle arrest in the endometrial cancer cells (Table I). 15d-PGJ2 led to an increase in the sub G0/G1 apoptotic cell population and the cell population in the G2/M phase of the cell cycle compared to treatment with the vehicle alone, with a concomitant decrease in the proportion of cells in the S phase.

Table I

Cell cycle changes in endometrial cancer cell lines.

Table I

Cell cycle changes in endometrial cancer cell lines.

Cell lineVehicle15d-PGJ2 (10 μM)
Ishikawa
 Sub G0/G1 (%)3.1±0.16.2±0.3a
 G0/G1 (%)53.4±12.435.0±17.7a
 S (%)36.1±5.638.4±7.5
 G2/M (%)10.5±7.726.6±11.8a
HEC-59
 Sub G0/G1 (%)2.7±0.10.5±0.1a
 G0/G1 (%)51.9±0.854.7±0.9
 S (%)35.8±0.631.6±1.2
 G2/M (%)12.3±0.313.6±0.7a
HHUA
 Sub G0/G1 (%)3.9±0.85.2±2.0a
 G0/G1 (%)53.5±3.644.0±8.1
 S (%)35.3±1.744.0±4.0
 G2/M (%)11.3±2.012.1±4.0

{ label (or @symbol) needed for fn[@id='tfn1-ijmm-31-04-0778'] } Cells were plated in 15d-PGJ2 wells and grown for 2 days, and cell cycle distribution was measured. Means ± SD are shown.

a P<0.05 compared to the vehicle.

Apoptotic changes in endometrial cancer cells treated with 15d-PGJ2

To assess the ability of the endometrial cancer cells to undergo apoptosis in response to 15d-PGJ2 exposure and to distinguish between the different types of cell death, we double-stained the 15d-PGJ2-treated cells with Annexin V and PI and analyzed the results using flow cytometry. Annexin V binding combined with PI labeling was performed for the distinction of early apoptotic (Annexin V+/PI) and necrotic (Annexin V+/PI+) cells. At increasing doses of 15d-PGJ2, a simultaneous increase in both the Annexin V+/PI fraction (early apoptotic) and Annexin V+/PI+ (regarded as necrotic) subpopulations was detected (Table II).

Table II

Cell death measured by Annexin V and mitochondrial transmembrane potential assay in endometrial cancer cell lines.

Table II

Cell death measured by Annexin V and mitochondrial transmembrane potential assay in endometrial cancer cell lines.

Assay/cell lineVehicle15d-PGJ2 (10 μM)
Annexin V assay
 Ishikawa
  Viable (LL) (%)92.5±0.148.4±1.6a
  Apoptosis (LR) (%)4.9±0.135.8±0.8a
  Necrosis (UR) (%)2.5±0.215.3±0.8a
 HEC-59
  Viable (LL) (%)86.7±0.356.3±1.0a
  Apoptosis (LR) (%)4.7±0.36.5±0.3
  Necrosis (UR) (%)5.4±0.116.7±0.4a
 HHUA
  Viable (LL) (%)79.2±8.561.9±5.9a
  Apoptosis (LR) (%)6.7±1.112.9±3.3a
  Necrosis (UR) (%)4.6±1.117.1±4.3a
MTP assay
 Ishikawa
  Viable (%)7644
  Apoptosis (%)2559
 HEC-59
  Viable (%)7758
  Apoptosis (%)2344
 HHUA
  Viable (%)6814
  Apoptosis (%)3487

{ label (or @symbol) needed for fn[@id='tfn3-ijmm-31-04-0778'] } Induction of apoptosis by15d-PGJ2 in endometrial cancer cells detected by Annexin V-FITC and PI staining (Annexin V assay). Cells were cultured with 10 μM of 15d-PGJ2 for 48 h, stained with Annexin V-FITC and PI, and analyzed by flow cytometry. Each experiment was repeated 3 times. Data represent the means ± SD. LL, lower left quadrant (percentage of viable cells); LR, lower right quadrant (early apoptotic cells); UR, upper right quadrant (secondary necrotic cells).

a P<0.05 compared to the vehicle. Effect of 15d-PGJ2 treatment on the mitochondrial transmembrane potential (MTP assay). Cells were cultured with 10 μM of 15d-PGJ2 for 48 h, and the MitoCapture intensity (representing MTP) was determined by flow cytometry.

Loss of MTP in response to treatment with 15d-PGJ2

It has been shown that the loss of MTP occurs prior to nuclear condensation and caspase activation and is linked to cytochrome c release in many, but not all, apoptotic cells (14,15). It was found that the treatment of endometrial cancer cells with 15d-PGJ2 resulted in the loss of MTP (Table II).

Differential gene expression in 15d-PGJ2-treated cells

In order to identify potential and novel target genes responsive to the anticancer effects in 15d-PGJ2-treated endometrial cancer cells, we examined the global changes in gene expression in the HHUA cells following treatment with 10 μM of 15d-PGJ2 for 48 h (Tables III and IV). Of the 44,000 genes, GO analysis was carried out on the genes upregulated and downregulated by the treatment (Tables V and VI).

Table III

Upregulated genes following treatment with 15d-PGJ2 in HHUA cells.

Table III

Upregulated genes following treatment with 15d-PGJ2 in HHUA cells.

Fold changesGene symbolDescriptionGenBankUniGeneMap
17.50674AKR1C1Aldo-keto reductase family 1, member C1 (dihydrodiol dehydrogenase 1; 20-α (3-α)-hydroxysteroid dehydrogenase)NM_001353Hs.46026010p15-p14
15.647071AKR1C3Aldo-keto reductase family 1, member C3 (3-α hydroxysteroid dehydrogenase, type II)NM_003739Hs.7818310p15-p14
6.0476165AMBP α-1-microglobulin/bikunin precursorNM_001633Hs.4369119q32-q33
5.751939HMOX1Heme oxygenase (decycling) 1NM_002133Hs.51758122q12
4.723847A_32_P15767117p11.2
4.683617TRIM16Tripartite motif-containing 16NM_006470Hs.12353417p11.2
4.5855446PIRPirin (iron-binding nuclear protein)NM_003662Hs.495728Xp22.2
4.380505UGT1A6UDP glucuronosyltransferase 1 family, polypeptide A6NM_001072Hs.6544992q37
4.2928677TXNRD1Thioredoxin reductase 1NM_003330Hs.65492212q23-q24.1
3.9529867GCLMGlutamate-cysteine ligase, modifier subunitNM_002061Hs.3155621p22.1
3.817845 ENST0000031348119p13.3
3.6058688FTLFerritin, light polypeptideNM_000146Hs.43367019q13.3-q13.4
3.4976888CR598364Full-length cDNA clone CS0CAP007YJ17 of Thymus of Homo sapiens (human)CR598364Hs.596052
3.4703205G6PDGlucose-6-phosphate dehydrogenaseNM_000402Hs.461047Xq28
3.410493SRXN1Sulfiredoxin 1 homolog (S. cerevisiae)NM_080725Hs.51683020p13
3.343625SPP1Secreted phosphoprotein 1 (osteopontin, bone sialoprotein I, early T-lymphocyte activation 1)NM_000582Hs.3134q21-q25
3.3256302A_24_P28168311q23.3
3.108579TXNRD1Thioredoxin reductase 1BG001037Hs.65492212q23-q24.1
3.0549212PFKFB3 6-Phosphofructo-2-kinase/fructose-2,6-biphosphatase 3NM_004566Hs.19547110p14-p15
3.0234468FLJ35767FLJ35767 proteinNM_207459Hs.23189717q25.3
2.9421628EPHX1Epoxide hydrolase 1, microsomal (xenobiotic)NM_000120Hs.896491q42.1
2.833596GCNT3Glucosaminyl (N-acetyl) transferase 3, mucin typeNM_004751Hs.19471015q21.3
2.699825OSGIN1Oxidative stress induced growth inhibitor 1NM_013370Hs.12805516q23.3
2.6840672GSRGlutathione reductaseBC035691Hs.2715108p21.1
2.6485877IKBKGInhibitor of κ light polypeptide gene enhancer in B-cells, kinase γNM_003639Hs.43505Xq28
2.6185443 ENST00000313774Homo sapiens glucosaminyl (N-acetyl) transferase 3, mucin type, mRNA (cDNA clone MGC:9086 IMAGE:3851937), complete cds. [BC017032]Hs.19471015q22.2
2.5760007DDCDopa decarboxylase (aromatic L-amino acid decarboxylase)NM_000790Hs.3596987p11
2.5224981LMNB1Lamin B1NM_005573Hs.894975q23.3-q31.1
2.5161438A_32_P79741q21.3
2.505143NADSYN1NAD synthetase 1AL512694Hs.55698611q13.4
2.4961286HSPA1AHeat shock 70 kDa protein 1ANM_005345Hs.5200286p21.3
2.47789GCLCGlutamate-cysteine ligase, catalytic subunitNM_001498Hs.6544656p12
2.361285CN27279717000600009278 GRN_PREHEP Homo sapiens cDNA 5′, mRNA sequence.CN2727979p23
2.3579373HSPA8Heat shock 70 kDa protein 8BU731317Hs.18041411q24.1
2.3155344C16orf28Chromosome 16 open reading frame 28NM_023076Hs.64353616p13.3
2.2990816ABCB6ATP-binding cassette, sub-family B (MDR/TAP), member 6NM_005689Hs.1079112q36
2.2683835ALDH3A2Aldehyde dehydrogenase 3 family, member A2NM_000382Hs.49988617p11.2
2.2379045 ENST00000238571Homo sapiens (clone zap3) mRNA, 3′ end of cds. [L40403]Hs.53111114q24.3
2.2241304GLAGalactosidase, αNM_000169Hs.69089Xq22
2.2137868PRDX1Peroxiredoxin 1NM_002574Hs.1809091p34.1
2.2122195ANGPTL4Homo sapiens angiopoietin-like 4 (ANGPTL4), transcript variant 2, mRNA [NM_016109]NM_016109Hs.961319p13.2
2.165274GCLCHlutamate-cysteine ligase, catalytic subunitM90656Hs.6544656p12
2.1589625THC2309960Q7ZX66 (Q7ZX66) RNPC7 protein (Fragment), partial (9%) [THC2309960]Hs.527551Xq23
2.150731THC2269657Q6QI74 (Q6QI74) LRRG00134, partial (10%) [THC2269657]chr10
2.1422243PLXND1Plexin D1NM_015103Hs.3016853q21.3
2.141769ABCB6ATP-binding cassette, sub-family B (MDR/TAP), member 6NM_005689Hs.1079112q36
2.1359584GSRGlutathione reductaseNM_000637Hs.2715108p21.1
2.119439A_24_P178167Xp11.23
2.1153674AIFM2Apoptosis-inducing factor, mitochondrion-associated, 2NM_032797Hs.65537710q22.1
2.0789819KCNMB4Potassium large conductance calcium-activated channel, subfamily M, β member 4NM_014505Hs.52552912q

Table IV

Downregulated genes following treatment with 15d-PGJ2 in HHUA cells.

Table IV

Downregulated genes following treatment with 15d-PGJ2 in HHUA cells.

Fold changeGene symbolDescriptionGenBankUniGeneMap
0.01FERD3LFer3-like (Drosophila)NM_152898Hs.5921687p21.1
0.025915636A_24_P9228937q11.21
0.027622959AGR3Anterior gradient homolog 3 (Xenopus laevis)NM_176813Hs.1006867p21.1
0.038513284MRPL37Mitochondrial ribosomal protein L37NM_016491Hs.5849081p32.1
0.05087885NOS2ANitric oxide synthase 2A (inducible, hepatocytes)NM_000625Hs.43438617q11.2-q12
0.095559224C18orf23Chromosome 18 open reading frame 23AK091537Hs.50111418q21.1
0.10365046 ENST00000329610Homo sapiens prepro-NPW mRNA for prepro-Neuropeptide W polypeptide, partial cds. [AB084276]Hs.23353316p13.3
0.12478433CASC1Cancer susceptibility candidate 1NM_018272Hs.40777112p12.1
0.14965945THC2368014AY320849 immunoglobulin κ chain variable region (Homo sapiens), complete [THC2368014]2p11.2
0.15056583WBSCR19Williams Beuren syndrome chromosome region 19NM_175064Hs.6454837p13
0.15237725KIAA1183KIAA1183 proteinAB033009Hs.719319q13.32
0.16197103A_24_P93235519p13.11
0.1684146THC2280638RL2A_HUMAN (P46776) 60S ribosomal protein L27a, partial (24%) [THC2280638]4q13.3
0.16913189AK022268CDNA FLJ12206 fis, clone MAMMA1000941AK022268Hs.6583693
0.17584784GAL3ST3 Galactose-3-O-sulfotransferase 3NM_033036Hs.20834311q13.1
0.18523274TMEM169Transmembrane protein 169NM_138390Hs.3349162q35
0.18725868THC2441492ALU7_HUMAN (P39194) Alu subfamily SQ sequence contamination warning entry, partial (12%) [THC2441492]19q13.12
0.19028467 ENST00000304181 GB|AJ009794.1|CAA08833.1 proline rich domain [NP101191]10q24.31
0.20250778PDPRPyruvate dehydrogenase phosphatase regulatory subunitNM_017990Hs.65524516q22.1
0.21064165THC228380910q24.1
0.21432775ZNF791Zinc finger protein 791NM_153358Hs.52254519p13.2-p13.13
0.21798867C3AR1Complement component 3a receptor 1NM_004054Hs.59114812p13.31
0.2198143CCDC110Coiled-coil domain containing 110NM_152775Hs.411014q35.1
0.2209758KCNAB1Potassium voltage-gated channel, shaker-related subfamily, β member 1BC043166Hs.6545193q26.1
0.22387888CHADChondroadherinNM_001267Hs.9722017q21.33
0.22510499ICAM4Intercellular adhesion molecule 4 (Landsteiner-Wiener blood group)NM_001544Hs.63160919p13.2-cen
0.22763024eIF4EEukaryotic translation initiation factor 4EBM981574Hs.2497184q21-q25
0.2376016XPNPEP1X-prolyl aminopeptidase (aminopeptidase P) 1, solubleNM_020383Hs.39062310q25.3
0.23924729CPXM1Carboxypeptidase X (M14 family), member 1NM_019609Hs.65934620p13-p12.3
0.24067116NDRG2NDRG family member 2NM_201535Hs.52520514q11.2
0.2438523A_24_P9168538q24.21
0.24945486FLJ21272Hypothetical protein FLJ21272AK024925Hs.6128911q21.2
0.25685737CSNK1G1Casein kinase 1, γ 1; Homo sapiens casein kinase 1, γ 1 (CSNK1G1), mRNANM_001011664Hs.25433515q22.1-q22.31
0.2718289CATSPER1Cation channel, sperm associated 1NM_053054Hs.18910511q12.1
0.27470103APOA4Apolipoprotein A-IVNM_000482Hs.59194011q23
0.2767344MMP1Matrix metallopeptidase 1 (interstitial collagenase)NM_002421Hs.8316911q22.3
0.29412216C15orf37Chromosome 15 open reading frame 37NM_175898Hs.51201515q25.1
0.3019541COPZ2Coatomer protein complex, subunit ζ 2NM_016429Hs.40843417q21.32
0.3044736RREB1Ras responsive element binding protein 1NM_002955Hs.2982486p25
0.31109598GMFGGlia maturation factor, γNM_004877Hs.521019q13.2
0.3145231MGC16121Hypothetical protein MGC16121BC007360Hs.416379Xq26.3
0.31809595MCCD1Mitochondrial coiled-coil domain 1NM_001011700Hs.5589226p21.33
0.3269747WBSCR27Williams Beuren syndrome chromosome region 27NM_152559Hs.6470427q11.23
0.33880442DHRS2 Dehydrogenase/reductase (SDR family) member 2NM_182908Hs.27249914q11.2
0.342074MDFIMyoD family inhibitorNM_005586Hs.5201196p21
0.3541897DHRS2 Dehydrogenase/reductase (SDR family) member 2NM_182908Hs.27249914q11.2
0.361466IFP38Homo sapiens IFP38 (IFP38), mRNA [NM_031943]NM_031943Hs.513128chr13
0.36588448 ENST00000329078Homo sapiens, Similar to spinster-like protein, clone IMAGE:4814561, mRNA, partial cds. [BC041772]Hs.55601517p13.2
0.36866197THC2433384ALU7_HUMAN (P39194) Alu subfamily SQ sequence contamination warning entry, partial (15%) [THC2433384]17p13.1
0.37413767BG182941Transcribed locusBG182941Hs.6352807

Table V

Permutation analysis of the correlation between GO terms and upregulated genes following treatment with 15d-PGJ2.

Table V

Permutation analysis of the correlation between GO terms and upregulated genes following treatment with 15d-PGJ2.

GO AccessionGO TermCorrected P-valueCount in selection
GO:0055114Oxidation reduction5.08E-0510
GO:0016491Oxidoreductase activity5.08E-0510
GO:0005829Cytosol3.99E-0413
GO:0051186Co-factor metabolic process0.0019679455
GO:0016209Antioxidant activity0.0019679454

GenesGO TermID Treatment/control

GO:0055114Oxidation reduction
 AKR1C1A_23_P25797117.29906688
 AKR1C3A_23_P13854115.00240358
 HMOX1A_23_P1208835.318975127
 TXNRD1A_23_P2045814.005567769
GO:0016491Oxidoreductase activity
 AKR1C1A_23_P25797117.29906688
 AKR1C3A_23_P13854115.00240358
 HMOX1A_23_P1208835.318975127
 TXNRD1A_23_P2045814.005567769
GO:0005829Cytosol
 AKR1C1A_23_P25797117.29906688
 HMOX1A_23_P1208835.318975127
 TXNRD1A_23_P2045814.005567769
 GCLMA_23_P1039963.688437908
GO:0051186Co-factor metabolic process
 AMBPA_23_P2565045.657345747
 HMOX1A_23_P1208835.318975127
 GCLMA_23_P1039963.688437908
 GCLMA_32_P1779533.255236172
GO:0016209Antioxidant activity
 TXNRD1A_23_P2045814.005567769
 SRXN1A_23_P3201133.1590489
 GSRA_32_P316182.458848635
 PRDX1A_23_P119952.077957762

Table VI

Permutation analysis of the correlation between GO terms and downregulated genes following treatment with 15d-PGJ2.

Table VI

Permutation analysis of the correlation between GO terms and downregulated genes following treatment with 15d-PGJ2.

GO AccessionGO TermCorrected P-valueCount in selection
GO:0002675Positive regulation of acute inflammatory response0.0649058452
GO:0010817Regulation of hormone levels0.0761631953
GO:0032101Regulation of response to external stimulus0.0761631953
GO:0002673Regulation of acute inflammatory response0.0761631952
GO:0002790Peptide secretion0.077977732

GenesGO TermID Treatment/control

GO:0002675Positive regulation of acute inflammatory response
 IL6A_23_P710370.404520132
 C3A_23_P1014070.450077889
GO:0010817Regulation of hormone levels
 DHRS2A_23_P3215010.311235885
 IL6A_23_P710370.404520132
 EDN1A_23_P2148210.461688691
GO:0032101Regulation of response to external stimulus
 IL6A_23_P710370.404520132
 C3A_23_P1014070.450077889
 EDN1A_23_P2148210.461688691
GO:0002673Regulation of acute inflammatory response
 IL6A_23_P710370.404520132
 C3A_23_P1014070.450077889
GO:0002790Peptide secretion
 IL6A_23_P710370.404520132
 EDN1A_23_P2148210.461688691
Effects of 15d-PGJ2 on the expression of novel proteins

To elucidate the common mechanism of action of 15d-PGJ2 in endometrial cancer, we examined the effects of 15d-PGJ2 on the expression of 20 proteins that were selected from the cDNA microarray data in 3 endometrial cancer cell lines using western blot analysis (Fig. 2 and Table VII). 15d-PGJ2 markedly upregulated the levels of AKR1C3 and downregulated the levels of AGR3 and NOS2A proteins in all 3 endometrial cancer cell lines.

Table VII

Results of western blot analysis in the 3 cell lines.

Table VII

Results of western blot analysis in the 3 cell lines.

NameHHUAIshikawaHEC-59
AGR3
AKR1C1-
AKR1C3
AMBPNE
C3AR1--
CHADNE
FERDL3--
FTLNE-
GAL3ST3
GCLM--
HMOX1--
ICAM4-NE
KCNAB1-
MRPL37NENE
NOS2A
p-elF4E-NE
PIR
TRIM16--
TXNRD1-
UGT1A6

[i] ↑, Upregulation; ↓, downregulation; -, no change; NE, no expression.

Discussion

In the present study, we demonstrated that 15d-PGJ2 inhibits cell viability in endometrial cancer cells. The prominent arrest of these cells in the G2/M phase of the cell cycle and the induction of apoptosis likely account for this inhibitory effect, suggesting that 15d-PGJ2 has anticancer activity.

In order to investigate the molecular mechanisms involved in the effects of 15d-PGJ2 on the cell cycle arrest and the induction of apoptosis, we investigated the global gene expression profile changes in HHUA endometrial cancer cells following treatment with 15d-PGJ2. Surprisingly, the expression of PPARγ or angiotensin II type 1 receptor (AT1R) was not altered, although 15d-PGJ2 has been characterized as a potent PPARγ ligand. To identify novel target genes of 15d-PGJ2, we focused on some GO terms of the numerous genes upregulated and downregulated by 15d-PGJ2 treatment in the HHUA cells. GO analysis revealed that oxidation reduction (GO:0055114) and oxidoreductase activity (GO:0016491) were enriched in genes that were overexpressed in the 15d-PGJ2-treated HHUA cells compared to the untreated HHUA cells. Both GO terms include AKR1C3.

AKR1C3 is a multifunctional enzyme involved in androgen, estrogen, progesterone and prostaglandin metabolism. AKR1C3-mediated steroid metabolism may play a critical role in the maintenance of viable normal and abnormal endometrial epithelium (16). AKR1C3 has been reported to play important roles in the physiology of endometrial cells and that suppressed AKR1C3 expression represents a feature that allows the differentiation of hyperplastic and neoplastic endometrial epithelium from normal endometrial epithelium (16). In the present study, we demonstrated that 15d-PGJ2 markedly upregulated the levels of the AKR1C3 protein in all 3 endometrial cancer cell lines. Based on these observations, it can be hypothesized that the 15d-PGJ2-induced anticancer activity may be mediated, at least in part, by the upregulation of AKR1C3 in human endometrial cancer cells.

We confirmed the downregulation of AGR3 using western blot analysis in all 3 cell lines examined. AGR genes, a protein disulfide isomerase (PDI) family, harbour core thioredoxin folds (CxxS motifs) that have the potential to regulate protein folding and maturation. AGR3 is overexpressed by a hormone (estrogen-receptor α)-independent mechanism, identifying a novel protein-folding associated pathway that can mediate resistance to DNA-damaging agents in human cancers (17). These findings indicate that the downregulation of AGR3 by 15d-PGJ2 may cause DNA-damage, leading to the apoptosis of endometrial cancer cells.

Nitric oxide, a reactive free radical, acts as a biological mediator in several processes, including neurotransmission and antimicrobial and antitumor activities. The NOS2A gene encodes a nitric oxide synthase which is expressed in the liver and is inducible by a combination of lipopolysaccharide and certain cytokines. A recent study revealed that NOS2 upregulation contributes primarily to the proliferation and tumor maintenance in highly tumorigenic human glioma stem cells (18). Therefore, our finding that 15d-PGJ2 downregulated NOS2A expression suggests that the eicosanoid may inhibit the proliferation and maintenance of endometrial cancer cells via NOS2A downregulation.

In conclusion, the data from the present study demonstrate that 15d-PGJ2 exhibits anti-proliferative activity, potently induces cell cycle arrest, and stimulates apoptosis in human endometrial cancer cells. These events were accompanied by the upregulation of AKR1C3 and the downregulation of AGR3 and NOS2A. It is suggested that 15d-PGJ2 may be a novel therapeutic option for the treatment of endometrial cancer.

References

1 

Reinhardt MJ: Gynecologic tumors. Recent Results Cancer Res. 170:141–150. 2008. View Article : Google Scholar

2 

Obel JC, Friberg G and Fleming GF: Chemotherapy in endometrial cancer. Clin Adv Hematol Oncol. 4:459–468. 2006.

3 

Hill EK and Dizon DS: Medical therapy of endometrial cancer: current status and promising novel treatments. Drugs. 72:705–713. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Ota K, Ito K, Suzuki T, et al: Peroxisome proliferator-activated receptor gamma and growth inhibition by its ligands in uterine endometrial carcinoma. Clin Cancer Res. 12:4200–4208. 2006. View Article : Google Scholar

5 

Xin B, Yokoyama Y, Shigeto T, Futagami M and Mizunuma H: Inhibitory effect of meloxicam, a selective cyclooxygenase-2 inhibitor, and ciglitazone, a peroxisome proliferator-activated receptor gamma ligand, on the growth of human ovarian cancers. Cancer. 110:791–800. 2007. View Article : Google Scholar

6 

Yang YC, Tsao YP, Ho TC and Choung IP: Peroxisome proliferator-activated receptor-gamma agonists cause growth arrest and apoptosis in human ovarian carcinoma cell lines. Int J Gynecol Cancer. 17:418–425. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Nosjean O and Boutin JA: Natural ligands of PPARgamma: are prostaglandin J(2) derivatives really playing the part? Cell Signal. 14:573–583. 2002.PubMed/NCBI

8 

Forman BM, Tontonoz P, Chen J, Brun RP, Spiegeiman BM and Evans RM: 15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPARγ. Cell. 83:803–812. 1995.PubMed/NCBI

9 

Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC and Lehmann JM: A prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor g and promotes adipocyte differentiation. Cell. 83:813–819. 1995.PubMed/NCBI

10 

Wang JJ and Mak OT: Induction of apoptosis in non-small cell lung carcinoma A549 cells by PGD2 metabolite, 15d-PGJ2. Cell Biol Int. 35:1089–1096. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Shin SW, Seo CY, Han H, et al: 15d-PGJ2 induces apoptosis by reactive oxygen species-mediated inactivation of Akt in leukemia and colorectal cancer cells and shows in vivo antitumor activity. Clin Cancer Res. 15:5414–5425. 2009.PubMed/NCBI

12 

Mansure JJ, Nassim R and Kassouf W: Peroxisome proliferator-activated receptor gamma in bladder cancer: a promising therapeutic target. Cancer Biol Ther. 8:6–15. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Beissbarth T and Speed TP: GOstat: find statistically overrepresented Gene Ontologies within a group of genes. Bioinformatics. 20:1464–1465. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Rimon G, Bazenet CE, Philpott KL and Rubin LL: Increased surface phosphatidylserine is an early marker of neuronal apoptosis. J Neurosci Res. 48:563–570. 1997. View Article : Google Scholar : PubMed/NCBI

15 

Chen Y, Kramer DL, Diegelman P, Vujcic S and Porter CW: Apoptotic signaling in polyamine analogue-treated SK-MEL-28 human melanoma cells. Cancer Res. 61:6437–6444. 2001.PubMed/NCBI

16 

Zakharov V, Lin HK, Azzarello J, et al: Suppressed expression of type 2 3alpha/type 5 17beta-hydroxysteroid dehydrogenase (AKR1C3) in endometrial hyperplasia and carcinoma. Int J Clin Exp Pathol. 3:608–617. 2010.PubMed/NCBI

17 

Gray TA, MacLaine NJ, Michie CO, et al: Anterior Gradient-3: a novel biomarker for ovarian cancer that mediates cisplatin resistance in xenograft models. J Immunol Methods. 378:20–32. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Eyler CE, Wu Q, Yan K, et al: Glioma stem cell proliferation and tumor growth are promoted by nitric oxide synthase-2. Cell. 146:53–66. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April 2013
Volume 31 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li H and Li H: 15-Deoxy-Δ12,14-prostaglandin J2 induces growth inhibition, cell cycle arrest and apoptosis in human endometrial cancer cell lines. Int J Mol Med 31: 778-788, 2013
APA
Li, H., & Li, H. (2013). 15-Deoxy-Δ12,14-prostaglandin J2 induces growth inhibition, cell cycle arrest and apoptosis in human endometrial cancer cell lines. International Journal of Molecular Medicine, 31, 778-788. https://doi.org/10.3892/ijmm.2013.1268
MLA
Li, H., Narahara, H."15-Deoxy-Δ12,14-prostaglandin J2 induces growth inhibition, cell cycle arrest and apoptosis in human endometrial cancer cell lines". International Journal of Molecular Medicine 31.4 (2013): 778-788.
Chicago
Li, H., Narahara, H."15-Deoxy-Δ12,14-prostaglandin J2 induces growth inhibition, cell cycle arrest and apoptosis in human endometrial cancer cell lines". International Journal of Molecular Medicine 31, no. 4 (2013): 778-788. https://doi.org/10.3892/ijmm.2013.1268