Patients and tissue specimens. A total of 105 cases of ESCC tissues and paired adjacent non-tumor tissues were obtained from patients admitted to the Fourth Hospital of Hebei Medical University between 2012 and 2016 with a confirmed diagnosis of ESCC who received surgical resection as their first treatment. Each surgically excised specimen was placed in liquid nitrogen immediately after acquisition and stored at -80°C for RNA extraction. Treatment records, clinical history, and follow-up data of patients were retrieved from the case management system in the medical record room. Grading and staging were undertaken according to the 8th edition AJCC/UICC staging of cancers of the esophagus and esophagogastric junction. A positive family history of upper gastrointestinal cancer (UGIC) was defined as a self-reported history of esophageal/cardia/gastric cancer in at least one of the first-degree relatives or two second-degree relatives. Written informed consent was obtained from all patients, and the study was approved by the Ethics Committee of the Fourth Hospital of Hebei Medical University.
Cell culture and X‑ray irradiation. Human esophageal cancer cell lines KYSE170, TE1, KYSE150, KYSE109 and human normal esophageal epithelial cell line HEEpiC were purchased from China Center for Type Culture Collection (CCTCC, Wuhan, China). The ESCC cell lines were cultured in RPMI 1640 (Gibco, Invitrogen, Life Technologies, Germany) medium supplemented with 10% fetal bovine serum (Invitrogen, Carlsbad, CA, USA) at 37°C with 5% CO2. The HEEpiC cells were cultured according to the manufacturer’s instructions.
Irradiation was performed at room temperature with a 6-MV Varian linear accelerator (Varian Medical Systems, Palo Alto, CA, USA) at a dose rate of 5 Gy/min and a source-skin distance of 100 cm. After irradiation, cells were recovered in respective growth medium for the indicated time until harvesting.
RNA isolation and quantitative real-time polymerase chain reaction (qRT-PCR) assay. Total RNA was isolated from tissue samples and cell lines using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) and the RNA was reverse transcribed into cDNA with Transcriptor First Strand cDNA Synthesis Kit (Roche, Basel, Switzerland) according to the manufacturer’s instruction. The qRT-PCR was performed in triplicate with StepOnePlus Real-Time PCR System (Applied Biosystems) using GoTap® qPCRMaster Mix (Promega, Madison, WI, USA). Expression of genes was normalized to the expression of GAPDH. Data were analyzed and showed with 2−ΔΔCt method. All primers used for qRT-PCRs were listed in Supplementary Table 4.
Cell transfection. FOXK1 siRNAs, CDK4 siRNAs, and CDC25A siRNAs were purchased from Ribbio (Guangzhou, China). The siRNA sequences were shown in Supplementary Table 5. For overexpression of FOXK1, CDK4, and CDC25A, the cDNAs encoding them were PCR-amplified and subcloned into pcDNA3.1 vector (Invitrogen, Carlsbad, CA, USA). For transient transfections, Lipofectamine 2000 reagent (Invitrogen, Carlsbad, CA, USA) was used according to the manufacturer’s instructions. The cells were collected after transfection with siRNA oligonucleotides (100 nM) or plasmids (2 mg) for 48 h. The infection efficiency was validated using qRT-PCR or western blotting assays. To generate FOXK1 stably expressing cells, the cells were isolated using 800 µg/mL of G418 after transient transfection.
Western blotting assay. Total protein extraction was performed in RIPA lysis buffer with PMSF (Solarbio, Beijing, China). Protein concentration was determined using BCA Protein Assay (Multi Sciences, Hangzhou, China). Equal amounts of protein samples were electrophoresed on 10% sodium dodecyl sulfate-polyacrylamide electrophoresis gel and then transferred onto polyvinylidene fluoride membranes (Millipore, Billerica, MA, USA), followed by blocking with 5% skim milk for an hour. Afterwards, the membranes were probed with specific primary antibodies overnight at 4°C and then with secondary antibody (KPL, Milford, MA, USA) for an hour at room temperature. The primary antibodies used were listed in Supplementary Table 6. Then the signals were visualized using enhanced chemiluminescence reagent (Multi Sciences, Hangzhou, China).
Cell viability assay. Cell viability was assessed using an MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2‐(4‐sulfophenyl)‐2H‐tetrazolium) assay. A CellTiter 96® Aqueous One Solution Cell Proliferation Assay kit (Promega, Madison, WI, USA) was used according to the manufacturer's instructions. The transfected cells were seeded at 1×103 cells/well and cultured in 96‐well plates. MTS reagent (20 µl) was added to 100 µl culture medium at different time points and incubated for 2 h. Then, the cell content was calculated by detecting the absorbance of each well at 490 nm via a Spark® multimode microplate reader (Tecan).
For cell viability after irradiation, cells were plated in 96-well plates to a density of 5×103 cells/ml and incubated for 24 h at 37°C. The viability of cells irradiated at 8 Gy was determined by incubation with MTS reagent, same as above.
Colony formation assay. The 5×103 transfected cells were inoculated in triplicate in 6-well plates and cultured for approximately 7 days until colonies were visible. The colonies (containing 50 cells or more) were counted manually after fixed with 4% paraformaldehyde and stained with 0.1% crystal violet.
For colony formation assay after irradiation, cells stably expressing FOXK1 and their respective control cells were plated at a low density onto 6-well plates (500 cells/well), followed by graded single doses (0–8 Gy). Two weeks later, the cells were fixed and stained with crystal violet (0.6%). Colonies of ≥ 50 cells were scored as survivors. The mean survival data for each individual cell line were fitted to the linear quadratic (LQ) model: SF = 1-(1-e− D/D0)N, where SF is the survival fraction, D is the irradiation dose, D0 is the mean lethal dose, and N is the extrapolation number.
Wound-healing assay. Cell migration capacities were evaluated by wound healing assays. The transfected cells were seeded into 6-well plates until grown to confluence. A scratch wound was generated using a sterile 200-µL pipette tip, and floating cells were removed by washing with 1 × PBS. Migration was photographed at the indicated time points after scratching using an inverted microscope and the percent wound closure was calculated for 3 randomly chosen fields.
Cell invasion assay. Cell migration assays were performed in Matrigel-coated chambers (Corning Costar, Corning, NY, USA) with 8-µm pore membranes. The 1×105 transfected cells suspended in 200 µL of serum-free RPMI 1640 medium were added to the upper chamber while the lower compartment of the chamber was filled with 600 µL of the medium containing 10% FBS. After 24 h of incubation at 37°C, the non-migrating cells in the upper chamber were removed by a cotton swab and lower surface of the chamber was fixed with 4% paraformaldehyde and stained with crystal violet. Invading cells were scored by counting at least 5 fields per membrane under a light microscope.
Flow cytometry analysis of cell cycle and apoptosis. FITC Annexin V and propidium iodide (PI) (BD bioscience, CA, USA) double staining was used to detect cell apoptosis, and PI (Multi Science, Hangzhou, China) mono staining was applied to monitor cell cycle. After the transfected cells (with or without 8 Gy single-dose of irradiation) were harvested, the cell concentration was adjusted to 1 × 106 cells per ml and then washed twice with PBS. Thereafter, apoptosis was detected by adding 5 µl of FITC Annexin V and 5 µl of PI to 100 µl cell solution for staining in the dark at room temperature for 15 min. To detect cell cycle, the cells were stained with 500 µl PI (1 µg/ml) for 30 min at room temperature in the dark. Flow cytometer was used to detect cell apoptosis and cell cycle.
Dual-luciferase reporter assays. The CDC25A promoter construct was obtained from PCR amplified fragment from genomic DNA and was cloned into the pGL3-Basic vector. The KYSE170 cells overexpressing FOXK1 or KYSE150 cells with knockdown of FOXK1 and their respective control transfectants were plated at a density of 1 × 105 − 5 × 105 cells/mL into 24-well plates and transfected with 0.2 µg pGL3-CDC25A promoter luciferase plasmid and 0.02 µg SV40 Renilla luciferase plasmid. After 48 h, firefly and Renilla luciferase activities were assayed sequentially in each well by Dual-Luciferase Reporter Assay System (Promega, Madison, WI, USA). Transcriptional activity was calculated as the ratio of firefly luciferase activity (reporter) to Renilla luciferase activity (control). All luciferase assays were carried out in triplicate. The CDK4 promoter construct was generated similarly and conducted the dual-luciferase reporter assays as described above. The primers of a series of CDC25A and CDK4 promoter fragments are listed in Supplementary Table 7.
Chromatin immunoprecipitation (ChIP) assay. ChIP assay was performed using EZ-Magna ChIP A/G (17-10086, Upstate, Millipore, MA, USA) kit to study the enrichment of FOXK1 in CDK4 and CDC25A promoter regions according to the manufacturer’s instructions. Antibody against FOXK1 (ab85999; Abcam) was used for immunoprecipitation. Quantitative analysis of ChIP-derived DNA was performed by real-time qPCR analysis (primers in Supplementary Table 7).
Statistical analysis. Statistical analyses were conducted with GraphPad Prism 8.0 and SPSS 25.0. Student’s t-test or Pearson's χ2 test was used to determine statistical significance. Error bars show mean ± standard deviation (SD) of independent experiments. All tests were performed two-tailed and P values < 0.05 were considered statistically significant. Kaplan-Meier analyses were used for overall survival curves and P values were calculated with the log-rank test. Uni-/multivariate analyses were computed by Cox proportional hazards model. Spearman r correlation analysis was applied to calculate bivariate correlations among the study variables.
Ethics Declarations
Ethics declarations
All methods in this research were performed in accordance with the principles of the Declaration of Helsinki. The study was approved by the Ethics Committee of the Fourth Hospital of Hebei Medical University.