Patients and tissue specimens
10 pairs of CHOL tissues (T) and para-carcinoma tissues (P) were obtained from (Shandong Provincial Hospital Affiliated to Shandong First Medical University). The para-carcinoma tissues were used for control. Tissue chips were purchased from OUTDO BIOTECH (shanghai, China). Clinicopathologic characteristics of patients could be seen in Supplementary Table S1. Samples stained with hematoxylin and eosin were verified by experienced pathologists.
Microarray analysis
Gene expression profile was detected in 10 pairs of CHOL tissues and para-carcinoma tissues, as well as in RBE cells infected with shCtrl or shPNO1 by microarray analysis. The para-carcinoma tissues and cells transfected with shCtrl were used as the control group. The RNeasy kit (Sigma) method was implied to extract the total RNA, and the concentration of which was determined by Nanodrop 2000 (Thermo Fisher Scientific). RIN value was evaluated with Agilent 2100 and Agilent RNA 6000 Nano Kit (Agilent). RNA sequencing was performed with Affymetrix human GeneChip PrimeView according to the manufacturer’s instruction, and the outcomes were scanned by Affymetrix Scanner 3000 (Affymetrix). Welch t-test with Benjamin-Hochberg FDR. was used to perform the raw data statistical significance assessment. In addition, significant difference analysis and functional analysis based on KEGG pathway enrichment analysis were executed.
Cell culture and transfection
Human cholangiocarcinoma cell lines (HuCCT1, RBE, HuH28 and TFK-1) and HEK293T were purchased from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). Human biliary epithelial cells (HIBEpiC) were obtained from Fenghbio (Hunan, China). Wild-type RBE cells (RBE/p53+/+) and RBE cells lacking the p53 gene (RBE/p53-/-) were produced from laboratory. HuCCT1, RBE, HuH28, TFK-1, RBE/p53+/+ and RBE/p53-/- cells were grown in 90% RPMI-1640 with 10% FBS (Corning); HEK293T cells were grown in 90% DMEM with 10% FBS (Corning). Cells were grown at 37 ˚C in a humidified atmosphere of 5% CO2. All the plasmids involved in this paper were obtained from Genechem (Shanghai, China). The information of sequences could be found in Supplementary Table S2. Cell lines in logarithmic phase were infected with lentivirus vectors with polybrene at a multiplicity of infection of 20 for 12~16 h, and then maintained in the new fresh complete medium. After 72 hours of infection, when the infection efficiency of lentivirus on cells was observed to be over 85% using fluorescence microscope (Olympus), cells were harvested for the follow-up experiments. Cells transfected with shCtrl or si-Ctrl were used as the control group.
Real-time quantitative PCR
Total RNA from the cell lines or tissue samples was isolated, respectively using TRIZOL (Sigma), reverse transcription of RNA (2.0 µg) to cDNA was performed using the PrimeScript RT reagent kit (Takara) following the manufacturer’s instructions. RT-PCR was carried out using the SYBR Premix Ex Tag (Takara) on VII7 real time PCR instrument. 2-ΔΔCt Method was used to analysis the relative quantitative of target genes with GAPDH as the internal reference. Primers for genes were synthesized by Sangon and shown in Supplementary Table S3. The para-carcinoma tissues and cells transfected with shCtrl or si-Ctrl were used as the control group.
IHC staining
Tissue sections were incubated with PNO1 antibody (1:100, Sigma) at 4°C overnight, then washed with phosphate-buffered saline (PBS) for 3 times, and incubated with horseradish peroxidase (HRP) conjugated goat anti-mouse IgG polyclonal antibody for 30 min at room temperature. DAB and hematoxylin were used to stain tissue slides. IHC scoring of patients were determined based on the combined evaluation of the staining intensity and staining extent scores. The para-carcinoma tissues were used as the control group.
Western Blotting
Cells and tissues were lysed in ice-cold RIPA buffer (Millipore) and protein concentration was detected by BCA Protein Assay Kit (HyClone-Pierce). The para-carcinoma tissues and cells transfected with shCtrl or si-Ctrl were used as the control group. The same amount of total protein from each group was separated by 12% SDS-PAGE and transferred onto a PVDF membranes (Bio-Rad Laboratory). The membranes were blocked with primary antibodies in TBST plus 5% non-fat milk. After being washed with TBST, membranes were incubated with the appropriate secondary antibody. The signals were visualized using ECL-PLUS Kit (Amersham). Band intensities were quantified using Image J software (NIH). The information of antibodies was shown in Supplementary Table S4.
Celigo cell counting assay
72 h after the infection, cells were seeded onto a 96-well plate with the cell density of 2,000 cells/well/100 µL in RPMI-1640 medium containing 10% FBS and then further cultured for 5 days. Cells transfected with shCtrl were used as the control group. Cell counting was accomplished every day by Celigo image cytometer (Nexcelom Bioscience) and the cell proliferation curve was drawn.
CCK8 assay
Lentivirus infected CHOL cells were measured by CCK8 assay. Cells in the logarithmic phase were seeded onto 96-well plates (2,000 cells/well). CCK8 solution (10 µL, MCE) was incubated with cells for 2.5 h. The OD450 was measured by microplate reader (Tecan) and the cell viability ratio was calculated. Cells transfected with shCtrl were used as the control group.
Cell apoptosis
Infected cells were inoculated in a 6-well plate and cells were harvested when the cell density reached over 50 %. After being washed with 4°C ice-cold PBS, cells were centrifuged at 1000 rpm and resuspended with 200 µL binding buffer. Evaluation of apoptosis was performed by Annexin V-APC staining flow cytometry method (eBioscience) following the manufacturer’s protocol. Cells transfected with shCtrl were used as the control group.
Cell-cycle analysis
Infected cells were inoculated in a 6-well plate and cells were harvested when the cell density reached 70 %. After being washed with 4°C ice-cold PBS, cells were centrifuged at 1000 rpm and fixed with 70% ethanol at 4°C overnight. Then the cells were washed again extensively, and then incubated with FxCycle PI/RNase Staining Solution (Thermo Fisher Scientific) for 20 minutes. DNA content was analyzed by FACS and the proportion of DNA in different phases was analyzed using FlowJo version 10.0. Cells transfected with shCtrl were used as the control group.
Colony formation assay
Lentivirus infected cells were inoculated in a 6-well plate with 800 cells/well/ 2 mL complete growth medium. Medium was exchanged every 2-3 days. Cell clones were photographed by an Olympus digital camera, then fixed and stained by 4% paraformaldehyde and Giemsa (Solarbio), respectively. The numbers of colonies were counted using photoshop 2020. Cells transfected with shCtrl were used as the control group.
Transwell migration assay
Transwell assay was performed using Corning chamber. Lentivirus infected cells were seeded in a 24-well plate with 5×104 cells/well the upper chambers filled with 100 µL serum-free medium. 600 µL DMEM medium plus 30% FBS were filled in the lower chamber and incubated at 37°C for 24 h (HuCCT1: 48 h). At the end of incubation, floating cells were removed and cells in lower chamber were fixed and stained with crystal violet. Images of cells were taken and analyzed using NIH image J software. Cells transfected with shCtrl were used as the control group.
Promoter activity assay
RBE cells were transduced with a plasmid expressing luciferase reporter under the control of a p53-driven promoter and lentivirus encoding anti-PNO1 or control shRNAs (the control group) for 4–6 hours. After 48 hours, cells were washed twice with PBS, and harvested with 1× PLB from the Dual-Luciferase Reporter Assay System (Promega). Then cells were lysed, centrifuged, and the supernatant was collected. Aliquots of supernatant (40 µL) were added to 96-well plates, followed by 20 µL luciferase assay reagent (Promega) at room temperature. Luciferase activity was measured immediately using a luminometer (Orion II Microplate Luminometer, Berthold Detection Systems). Data were normalized to the results obtained for the internal control Renilla luciferase.
To examine whether the 5 predicted transcription factors have indeed modulated PNO1 transcription, plasmids were constructed via encoding the full-length predicted factors. A separate plasmid was constructed encoding luciferase reporter downstream of a 2.0-kb fragment of the PNO1 promoter. Plasmid integrity was confirmed by DNA sequencing. Briefly, HEK293T or RBE cells were co-transfected with each of 5 different overexpression plasmids or MYC overexpression plasmid (500 ng), Renilla luciferase reporter (20 ng), and luciferase reporter (500 ng) using Lipofectamine 3000 Transfection reagent (Thermo Fisher Scientific). Promoter activities were measured after 48-hour transfection.
Northern blot analysis of 18S rRNA
Extracted total RNA (10 µg /sample) was fractionated on a 1.2% agarose–formaldehyde gel and transferred to Hybond NÞ membranes (Amersham). BIO-labeled probe was purchased from Thermo Fisher Scientific. The membrane was hybridized overnight in hybridization buffer (Thermo Fisher Scientific), washed twice (5 minutes each) under low-stringency conditions (2 × SSC, 0.1% SDS), blocked with blocking buffer at 50 ˚C for 30 minutes, then incubated at 50 ˚C for 20 minutes with stabilized streptavidin–horseradish peroxidase conjugate (1:300). Membranes were exposed to a phosphor storage screen and visualized using the Chemi Doc XRS+ System (Bio-Rad). Cells transfected with shCtrl were used as the control group.
Ribosome profile analysis
Transfected RBE cells were treated with 100 µg/mL cycloheximide for 15 minutes, washed with ice-cold PBS containing 100 µg/mL cycloheximide, and then resuspended in hypotonic buffer [10 mmol/L HEPES (pH 7.9), 1.5 mmol/L MgCl2, 10 mmol/L KCl, 0.5 mmol/L dithiothreitol (DTT), 100 µg/mL cycloheximide, 40 U/mL RNase inhibitor, 1 × protease inhibitor cocktail]. Cells were centrifuged at 14,000 rpm for 15 minutes at 4 ˚C. Total protein (1 mg) was loaded onto a linear sucrose gradient (5%–50%) and centrifuged for 3 hours at 38,000 rpm at 4 ˚C. Cells transfected with shCtrl were used as the control group.
Protein synthesis assay
Global protein synthesis was assayed using a commercial kit (Cayman Chemical). Transfected RBE cells were incubated for 30 minutes with o-propargyl-puromycin in complete medium in 96-well plates (100 µL/well). Cells transfected with si-Ctrl were used as the control group. Cells were fixed with cell-based assay fixative (100 µL/well), stained with 5 FAM-azide (Cayman Chemical), and photographed under a fluorescence microscope (Accu-Scope). Images were collected using MetaMorph image acquisition software (Molecular Devices). Fluorescence intensity in single cells was quantitated using Image Pro Plus software (Media Cybernetics).
Xenograft tumor studies
All animal procedures were carried out in accordance with the Helsinki Declaration and were approved by the Ethics Committee of Shandong Provincial Hospital Affiliated to Shandong First Medical University (NO.2019-156). For cell line tumor xenografts, 0.2 mL exponentially growing lentivirus infected HuCCT1 cell suspensions at a destiny of 2× 107cell/ml were injected subcutaneously into BALB/c nude mice (4-6 weeks old). For metastasis mouse models, 0.1 ml lentivirus infected HuCCT1 cell suspensions at a destiny of 1× 107 cell/ml were injected intravenously into BALB/c nude mice (4-6 weeks old). For the PDX model, tumor tissues were obtained directly from the operating room, dissected carefully, trimmed into small pieces (approximately 2~3-mm3) under sterile conditions and implanted subcutaneously into the flanks of female NOD/SCID mice. After 2 months of tumor growth, the established PDX tumors were transplanted subcutaneously into the flanks of female NOD/SCID mice. When the tumors reached about 100 mm3, the mice received an intraperitoneal dose of bortezomib (0.5 mg/kg, twice a week) for bortezomib treatment, and the normal group were treated with Saline for comparison at the same time. Tumor growth was assessed twice a week using a caliper and tumor volumes (V) were estimated as V=π/6×L×W×W (L and W was tumor length and width, respectively). Mice were sacrificed at humane endpoints and the tumor tissues were removed for immunostaining.
Ki-67 staining assay
Tissue slides were blocked with 3% PBS-H2O2 and were incubated with primary antibody Ki-67 at 4°C overnight. Then slides were incubated with HRP goat anti-rabbit IgG at room temperature for 2 h. Finally, all slides were stained by Hematoxylin (# BA4041, Baso) and Eosin (# BA4022, Baso). Tumors transfected with shCtrl were used as the control group.
Statistical Analysis
Student’s T-Test and one-way ANOVA were used to analyze the statistical significance. All statistical analysis was performed using SPSS 22.0 (IBM, SPSS) and GraphPad Prism 8.01 (GraphPad Software). Each bar represents the mean values ± SD of three independent experiments. P < 0.05 was considered statistically significant. Kaplan-Meier survival analysis was performed using the log-rank test. The primary endpoint of the animal study was defined as the tumor growth, tumor volumes and KI67 expression in tumor tissues.