Patients and tissue specimens
A total of 40 paired NSCLC and their paracancerous lung tissues were obtained from patients who were diagnosed with NSCLC based on histopathological evaluations and had undergone surgery at Tianjin Medical University General Hospital (TJMUGH) between January 2010 and December 2011. Among these, 16 patients were diagnosed with primary LUAC and 24 with primary LUSC. In each patient, lung cancer staging was performed according to the AJCC Cancer Staging Manual, 8th edition, and findings of physical examination; surgical resection; and computed tomography of the chest, abdomen, pelvis, and brain. All collected tissue samples were immediately snap-frozen in liquid nitrogen and stored at −80°C prior to RNA extraction. Basic demographic and clinical information such as sex, age, smoking history, TNM stage, lymph node metastasis, and prognostic data were also collected from medical records.
Download ofThe Cancer Genome Atlas (TCGA) data and preprocessing of RNA-seq data
All next-generation RNA sequencing (RNA-seq) data corresponding to NSCLC cases in the TCGA database were analyzed using TCGA-Assembler. Data of a total of 513 patients with LUAD (including 59 paracancerous lung tissues) and 502 patients with LUSC (including 49 paracancerous lung tissues) were analyzed. Transcripts per million (TPM) was calculated and normalized using the Tag count comparison package (version 1.6.5; http://www.bioconductor.org/packages/release/bioc/html/TCC.html) [23]. The following clinicopathological data evaluated during the follow-up of each patient were obtained from the TCGA NSCLC clinical database: survival status, survival time, age, sex, histology, smoking status, lymph node metastasis, clinical stage, and tumor recurrence. TNM staging was performed according to the AJCC Cancer Staging Manual, 8th edition. Next, the patients were divided into high EZH2 (greater than or equal to the median) and low EZH2 (less than the median) expression groups. Kaplan–Meier survival curves and the log-rank test were performed to analyze univariate survival distribution.
Cells culture and treatment
The human LUAD cell line A549 was purchased from the American Type Culture Collection (Manassas, VA, USA). Other human LUAD cell lines (H1299, H1975, HCC827, H1650, and H2030) and the normal human bronchial epithelial cell line BEAS-2B were obtained from the Institute of Biochemistry and Cell Biology of the Chinese Academy of Sciences (Shanghai, China). The H460 and A549 cell lines harbor a KRAS mutation; HCC827 and H1650 harbor an EGFR exon 19 deletion; H1299 harbors a p53 deletion or rearrangement mutation; H1975 harbors EGFR exon 21 mutation (L858R) and exon 20 (T790m) mutations; H1792 harbors a TP53 splice mutation and EGFR exon 21 (L858R) and exon 20 (T790m) mutations; and H2030 harbors EGFR exon 21 (L858R) and KRAS mutations. All cell lines were maintained in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS), 100 IU/ml penicillin, and 100 μg/ml streptomycin (Invitrogen, Carlsbad, CA, USA). All cells were cultured at 37°C in humidified atmosphere containing 5% CO2. Cells were transfected with small interfering RNAs (siRNAs) specific for EZH2; 21-bp (including a 2-deoxynucleotide overhang) siRNA duplexes were purchased from RiboBio (Guangzhou, China). An unrelated siRNA containing a 19-bp scrambled sequence was used as a negative control siRNA. Transfection was performed using 10 nM siRNA oligonucleotide duplexes and Lipofectamine 2000 (Invitrogen) according to the manufacturer’s recommendations. Gefitinib (ZD1839), GSK343, and DZNep HCl were obtained from Selleck Chemicals LCC (Houston, TX, USA). Cells were seeded in different culture plates. When the cell confluence level exceeded 60%, EZH2-specific siRNA or scrambled control siRNA was transfected using Lipofectamine MAX (Invitrogen) according to the manufacturer’s instructions.
Cell Counting Kit-8 (CCK-8) assay
CCK-8 assay (Beyotime) was performed according to the manufacturer’s instructions as a quantitative measure of cell proliferation and determinant of the half-maximal inhibitory concentration (IC50) of each drug in each cell line. Briefly, 5000 cells were seeded in each well of a 96-well flat-bottomed plate and incubated for 24 h. Next, different concentrations of gefitinib, GSK343, DZNep, GSK+gefitinib (G+g), and DZNep+gefitinib (D+g) were added to different wells. After 48 h of incubation at 37°C, 10 μl CCK-8 (5 mg/ml) was added to each well and the cells were incubated for another 1 h. Absorbance was measured at 450 nm using a microplate reader (SpectraMax M5, Molecular Devices, Sunnyvale, CA, USA). The experiments were repeated at least three times.
5-Ethynyl-2′-deoxyuridine (EdU) staining
EdU staining involves the incorporation of the nucleoside analog uridine into a newly synthesized DNA strand and detection of the S phase of the cell cycle. This assay can also indirectly reflect cell proliferation. The Cell-Light™ EdU stain kit was obtained from RiboBio, and staining was performed according the manufacturer’s instructions. Briefly, the cells were incubated with 50 μM EdU for 2 h, washed twice with phosphate-buffered saline (PBS), and fixed with 4% paraformaldehyde. Next, the proliferated cells were infiltrated with 0.5% Triton X-100, washed twice with PBS, and stained. Subsequently, the cells were analyzed using a fluorescence microscope equipped with a single interference filter set. The red (Apollo)-stained, EdU-labeled cells represent cells is in a state of active division. Blue color represent nuclear staining with Hoechst 33342.
Colony formation assay
Regarding colony formation assay, 500 cells were seeded into each well of a 6-well plate and incubated at 37°C for 24 h. Next, the cells were treated with gefitinib, GSK343, DZNep, G+g, or D+g in medium and incubated at 37°C for 48 h. The concentration of each drug was set as half the IC50 value, and normal medium was used as a negative control (NC). After 48 h of drug treatment, the plates were washed twice with PBS, filled with fresh drug-free medium, and incubated for another 21 days. Finally, the plates were washed twice with PBS and stained with 0.5% crystal violet at room temperature for 30 min. The number of colonies (defined as >50 cells) was calculated in each group.
Flow cytometry analysis of apoptosis and cell cycle
Cells (2 × 105 cells/well) were seeded into 6-well plates and cultured for 24 h. Different treatments (NC, GSK343, DZNep, gefitinib, G+g, and D+g) were added at various concentrations, after which the cells were cultured for another 48 h. Regarding apoptosis assay, cells were stained using the Annexin V-FITC Apoptosis Analysis Kit (BD Biosciences, San Jose, CA, USA) and analyzed using the FACSAria™ flow cytometer (BD Biosciences). Regarding cell cycle assay, cells were trypsinized and fixed in 70% ice-cold ethanol overnight. Subsequently, cells were treated with DNase-free ribonuclease (TaKaRa, Beijing, China), stained with propidium iodide (PI; BD Biosciences), and analyzed using the FACSAria™ flow cytometer (BD Biosciences) equipped with ModFit LT (Topsham, ME, USA).
Wound healing assay
H1299 cells were seeded in six-well plates. Once the cells reached 90%–100% confluency, a 200-µL sterile pipette tip was scraped across the confluent cell layer to create an artificial wound. The suspended cells were removed by washing with sterile PBS. Subsequently, medium containing different drugs (NC, GSK343, DZNep, gefitinib, G+g, and D+g) was added to the wells, and the plates were cultured for another 48 h. The initial gap length after scratching (0 h) and residual gap length (24–48 h) were observed under an inverted microscope. Wound closure was calculated using the following formula: (0-h wound width − 24-h wound width) / 0-h wound width. The results of three independent experiments were averaged.
Transwell invasion assay
Regarding cell invasion assay, the upper surfaces of the polycarbonic membranes of Transwell® filters (Corning, NY, USA) were coated with 100 µl of 300 µg/mL Matrigel. Next, 1 × 105 cells were seeded in each upper chamber along with serum-free medium mixed with different drug treatments, and 600 µL medium containing 10% FBS were added to each lower chamber. The cells were incubated at 37°C in a humidified incubator with 5% CO2 for 24 h. Cells that migrated to the under membranes of the filters were fixed in methanol and stained with 0.1% crystal violet. The Nikon TE2000 microscope (Tokyo, Japan) at 100× magnification was used to collect five randomly selected visual field images. The experiments were performed in triplicate.
Tumor xenografts and OS analysis
Female BALB/c athymic nude mice (4–5 weeks old) were purchased from the Experimental Laboratory Animal Center of Beijing University (Beijing, China). All mice were maintained under specific pathogen-free conditions and were examined prior to starting the study to ensure that they were healthy and could be adapted for tumor implantation. A total of 2 × 106 A549 cells were subcutaneously injected into the left flank of each mouse to establish a tumor xenograft model. When the tumors reached a volume of 200 mm3, the mice were randomly distributed into the following six groups (10 mice/group). A vehicle group was administered 0.5% methylcellulose 400 (Wako Pure Chemical Industries, Ltd., Osaka, Japan) and sodium lactate buffer via daily oral gavage. A GSK343 group was administered 4 mg/kg GSK343 in 100 μL of corn oil (Santa Cruz Biotechnology, Dallas, TX, USA) via intraperitoneal injection every alternate day. a DZNep group was administered 2 mg DZNep/kg in 100 μL of corn oil via intraperitoneal injection every alternate day. A gefitinib group was administered 100 mg/kg gefitinib/day dissolved in 0.5% methylcellulose 400 and sodium lactate buffer via daily oral gavage. A G+g group was administered 4 mg/kg GSK343 in 100 μL corn oil via intraperitoneal injection every alternate day and 100 mg/kg gefitinib/day in 0.5% methylcellulose 400 and sodium lactate buffer via daily oral gavage. A D+g group was administered 2 mg/kg DZNep in 100 μL corn oil via intraperitoneal injection every alternate day and 100 mg/kg gefitinib/day in 0.5% methylcellulose 400 and sodium lactate buffer via daily oral gavage. All four groups received treatment for 28 days. Tumor size was measured every alternate day. Tumor volume (V) was calculated by measuring tumor length (L) and width (W) with a caliper and using the following formula: V = (L × W2) × 0.5 [24].
Immunohistochemistry
Xenograft tumor tissues were fixed overnight in 4% formalin, dehydrated with ethanol, and embedded in paraffin. Next, 4-µm-thick tissue slices were first dewaxed with xylose and rehydrated using an alcohol solution gradient. After a 10-min blocking step with normal goat serum, the tissue slices were incubated with a primary antibody specific for EZH2 (Cell Signaling Technology, Beverly, MA, USA) for 1 h at room temperature, washed with PBS, and incubated with a horseradish peroxidase (HRP)-conjugated secondary antibody (ZSJQ Corp, Beijing, China) for 60 min. Finally, the sections were incubated with 3,3′-diaminobenzidine for 3 min at room temperature and counterstained with hematoxylin.
RNA extraction and quantitative PCR (qPCR) assays
Total RNA was extracted from patient tissue samples or cultured cells using the TRIzol reagent (Invitrogen) according to the manufacturer’s instructions. RNA was quantified using a spectrophotometer (Beckman, USA), and RNA quality was assessed using denaturing 1.2% agarose gel electrophoresis. Subsequently, 2 μg of extracted RNA was reverse transcribed in a final reaction volume of 20 μl using random primers under standard conditions specified for the PrimeScript RT reagent Kit (TaKaRa) and reverse transcriptase (Promega, Beijing, China). We used the Power SYBR Green PCR Master Mix (Applied Biosystems, Foster City, CA, USA) to determine EZH2 and EGFR expression levels according to the manufacturer’s instructions. Results were normalized to the expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). qPCR assays and data collection were performed using the ABI 7500 real-time PCR system (Applied Biosystems). All gene primers were obtained from SBS Genetech (Beijing, China). Performance analysis was conducted using the 2−ΔΔCT method.
Protein isolation and western blotting
Cells were washed twice with ice-cold PBS and lysed in RIPA buffer [(1× PBS, 1% NP40, 0.5% sodium deoxycholate, 0.1% sodium dodecyl sulfate (SDS)) supplemented with 2 mM PMSF (Thermo Fisher Scientific, Inc., Waltham, MA, USA). We used a BCA protein assay kit (Pierce Biotechnology, Rockford, IL, USA) to measure protein concentrations. Next, 30 μg of each protein extract was denatured and separated using 10% SDS-polyacrylamide gel electrophoresis and electrophoretically transferred onto polyvinylidene fluoride membranes (Millipore, Lake Placid, NY, USA). The membranes were blocked in 5% non-fat milk and incubated overnight at 4°C in dilutions of the following primary antibodies: rabbit anti-human EZH2 (dilution, 1:1000; Cell Signaling Technology), mouse anti-human β-actin (1:3000; Sigma-Aldrich, St. Louis, MO, USA), rabbit anti-human EGFR (1:1000; Cell Signaling Technology), rabbit anti-human phospho (p)-EGFR (1:1000; Cell Signaling Technology), rabbit anti-human AKT (1:1000; Cell Signaling Technology), rabbit anti-human p-AKT (1:1000; Cell Signaling Technology), rabbit anti-human P38-MAPK (1:1000; Cell Signaling Technology), rabbit anti-human p-P38-MAPK (1:1000; Cell Signaling Technology), rabbit anti-human BCL-2 (1:1000; Cell Signaling Technology), rabbit anti-human Bax (1:1000; Cell Signaling Technology), rabbit anti-human caspase-3 (1:1000; Cell Signaling Technology), rabbit anti-human P62 (1:1000; Cell Signaling Technology),rabbit anti-human mTOR (1:1000; Cell Signaling Technology), and rabbit anti-human p-mTOR (1:1000; Cell Signaling Technology). Subsequently, the membranes were exposed to an HRP-conjugated secondary antibody (1:1000 dilution; Thermo Fisher Scientific, Inc.) at room temperature for 1 h. Finally, the Pierce ECL Substrate (Thermo Fisher Scientific, Inc.) was used to visualize the bands.
Statistical analysis
Statistical analysis was performed using the SPSS software package, version 21.0 (IBM Corp., Armonk, NY, USA). Data are presented as mean ± standard deviation of independent experiments. Kaplan–Meier survival analysis was performed to estimate OS. Univariate cox regression analysis and subsequent multivariate analysis using the likelihood ratio were performed to identify significant variables. The statistical significance of differences between two groups was analyzed using Student’s t-test. All P-values obtained in this study were two tailed, and the statistical significance level was set at P-value of <0.05.