Clinical patient samples
This study was conducted after approval by the Ethic Committee of Shandong University, and informed written consent was obtained from all patients. All of the gastric carcinoma tissues and paired normal tissues were obtained from five patients treated in the First Affiliated Hospital of the Fourth Military Medical University. The protocol for proteomics analysis of the samples were approved by the Institutional Research Ethics Review Boards. To avoid necrotic and adjacent non-tumor tissues inclusion, cancer tissues were taken from the core area of tumor. Adjacent tissues were taken from non-tumor regions at least 3 cm away from the core area of tumor. All tissues were verified by two pathologists and frozen immediately in liquid nitrogen and stored at -80°C.
Cell lines and cell culture
The human GC cell lines AGS and HGC-27 (HGC) used in this study were obtained from ATCC. HGC cells were cultured in RPMI-1640 medium containing with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin. AGS cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Gibco) with 10% FBS and 1% penicillin/streptomycin (Gibco). All cells were cultured in a sterile incubator humidified atmosphere containing 5% CO2 at 37°C.
LC-MS/MS sample preparation
Proteins were extracted from five pairs of human gastric tumor and adjacent non-tumor sample using T-PER buffer (Thermo Fisher Scientific) with protease inhibitor cocktail (Sigma-Aldrich) and phosphatase cocktail (Sigma-Aldrich). The protein concentrations of tissue lysates were quantified by Bicinchoninic Acid (BCA) Protein Assay kit (Pierce, thermo scientific, Germany). Triplicate lysates and bovine serum albumin (BSA) standards were added to a 96-well plate which contained 100 μL of working reagent followed by incubation for 30 min at 37°C. The absorbance was measured at 562 nm on a Microplate Reader (Thermo Fisher Scientific). The linear standard curve formula of protein quantification was established by drawing the relationship between the blank corrected BSA standard absorbance and the BSA standard concentration. The absorbance of the blank corrected sample was put into the linear standard curve formula to calculate the protein concentration of the sample. Equal amounts of the lysates were mixed with 8M urea (UA) in Microcon devices (YM-30, Millipore). Proteins were reduced using 10 mM dithiothreitol and then alkylated using 50 mM iodoacetamide (IAA) in the dark. In the digestion step, proteins were digested with trypsin (Promega) at a protein:trypsin ratio of 25:1 overnight at 37°C. Peptides were desalted by ZipTip C18 pipette tips (Millipore), washed with 0.1% trifluoroacetic acid (TFA), and eluted with 50% methanol followed by dry in a SpeedVac for LC-MS/MS analysis.
High pressure liquid chromatography
The mixed peptides were separated by high pressure liquid chromatography (HPLC) on an EASY-nLC 1000 system (Thermo Scientific, San Jose, CA). In brief, first set 2% CH3CN in water, NH3H2O to adjust pH 10.0 as Phase A, 98% CH3CN in water as Phase B. Peptides were dissolved in 80 µL phase A and fractionated with a long C18 column (300 ´ Ø 0.075 mm, 3 μm; Reprosil, Germany) with a mobile phase gradient ( 0-3 min, 100% phase A, 3-5 min, 100% -70% phase A, 5-45 min, 70% -30% phase A, 45-55 min, 30-5% phase A, 55-60 min, 5% phase A). Finally, 55 components were collected and dried in SpeedVac. Using 50% CH3CN and 1% TFA in water to dissolve 55 components and merge them into 10 components which finally dried under vacuum in SpeedVac for subsequent mass spectrometry analysis.
Mass spectrometry analysis
The MS and MS/MS spectra were acquired by an LTQ-Orbitrap Elite mass spectrometer (Thermo Scientific, San Jose, CA) in data-dependent mode. The spray voltage was 2.1 kV and the capillary temperature 275°C. MS spectra were acquired in profile mode in the m/z range of 350-1800 at a resolution of 60,000 at 400 m/z. MS/MS fragmentation of the 30 most intense peaks was performed for every full MS scan in the collision-induced dissociation mode. Triple technical replicates were analyzed for each sample.
Protein sequencing alignment and single amino acid polymorphisms analysis
MS/MS spectra were searched against the human protein database using SEQUEST in Proteome Discover. Trypsin (full cleavage) was specified as cleavage enzyme allowing up to two missing cleavages. MS/MS spectra were searched with a maximum allowed deviation of 10 ppm for the precursor mass and 0.6 Da for fragment masses. The oxidation of various amino acid residues including methionine was selected as dynamic phosphorylation (+79.9663 Da), and the false discovery rate (FDR) was 1%. For phosphorylation analysis, an open search algorithm provided by Byonic and MODa was used. For clustering, mass shifts (delta masses) were divided into subgroups with 1 Da intervals bounded by n-0.5 and n+0.5 Da (n = -200 to 1000). Mass shifts in each mass window were analyzed by multivariate clustering using Gaussian mixture components. Clusters within each window were determined by the Bayesian Information Criterion (BIC). Next, clusters in each window were fitted individually with Gaussian regression to calculate the peak value (expected mass shifts), the SD, and the Goodness-of-Fit (R2). Based on the given delta mass cluster which reflected the phosphorylation modifications, all the confident peptides were classified and normalized according to the spectral count.
Protein-Protein Interaction (PPI) Network Analysis
Using the STRING database (http://string-db.org/), PPI analysis for the phosphorylated differential proteins was conducted. The threshold for PPI analysis used the default values in the database.
Gene Ontology (GO) Functional analysis
The upregulated (above FC 2, p<0.05) and the downregulated (below FC 0.5, p<0.05) genes were sorted into different groups based on the fold change in gene expression. Each group was subjected to gene ontology (GO) functional annotation analysis under biological processes category, cell component and molecular function using DAVID Bioinformatics Resources (http://david.abcc.ncifcrf.gov/home.jsp) to determine significantly enriched genes. Pathways were identified from the Kyoto encyclopedia of genes and genomes (KEGG) database.
Immunoprecipitation
Cell lysate of HGC was carried out using RIPA Lysis Buffer (weak, Beyotime, China) and then incubated with 1 µg of the BCLAF1 antibody overnight at 4°C on a rotating wheel. 30 µL of protein A/G beads (MCE, China) were added to lysates and incubated on a rotator at 4°C for 2 hours. The beads were collected by a magnetic rack, and washed three times with ice-cold PBS-T (0.01% Tween) buffer. Beads were incubated with loading buffer and boiled for 5 minutes followed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and western blot analysis.
Protein in-gel digestion
The SDS/PAGE gels were cut into small pieces, washed twice with 50 mM NH4HCO3 buffer at 4°C for 4 h and then dried with 100% CH3CN for 10 min. Sequencing grade modified trypsin (Promega) at a concentration of 10 μg/mL in 50 mM NH4HCO3 buffer was added for overnight digestion at 37°C. The peptides were extracted from the gel with 60% CH3CN in 0.1% TFA for 30 min. The extracted solution was dried under vacuum in SpeedVac for subsequent mass spectrometry analysis. The obtained data were submitted to Mascot software (Matrix Science) to search for phosphorylated residues on the BCLAF1 protein.
Antibodies
The specific antibody recognizing pBCLAF1(Ser290) was raised in rabbits against peptide coupling with PSQNS(p)PIH-KLH (corresponding to the residues 286–293 of human BCLAF1). The antibody was prepared and purified by ABclonal Biotech (China). Antibodies used in this study as follow: BCLAF1 (A300-608A) was purchased from Thermo Fisher Scientific, Inc. (USA); γ-H2AX (#80312), Anti-rabbit IgG (H+L, Alexa Fluor 549 Conjugate, #8889), Anti-mouse IgG (H+L, Alexa Fluor 488 Conjugate, #4408), were purchased from Cell Signaling Technology, lnc. (USA); GAPDH (60004-1-Ig), Flag (F1804), GFP (F1804) were purchased from Proteintech, Ltd. (Shanghai, China).
Human gastric Cancer Tissue Microarray, Immunohistochemistry, and Scoring
Human GC tissue microarray (TMA) that consisted of the gastric tumor tissue specimens (n = 95) and adjacent non-normal tissue specimens (n = 85) were purchased from Outdo Biotech, Ltd. (Shanghai, China). The pBCLAF1(Ser290) antibody was used at a 1:100 dilution. The assignment of staining intensity score was based on the staining intensity (no intensity: 0, weak intensity: 1+, moderate intensity: 2+, and strong intensity: 3+) and positive-staining score was based on the percentage of positive-staining (0% positive: 0, 1–25% positive: 1, 26–50% positive: 2, 51–75% positive: 3, and 76–100% positive: 4) by three experienced pathologists. The final staining index was calculated using the formula: positive-staining score × staining intensity score. For data analysis, staining scores <4 were defined as low expression, and scores > = 4 indicated high expression.
Lentiviral plasmid construction and infection
BCLAF1 Lentiviral short hairpin RNA (shRNA) and a negative control vector (NC) were purchased from GeneChem Co., Ltd. (Shanghai, China) and transduced into the HGC and AGS cell lines following the manufacturer’s instructions. The Flag-tagged WT, S290D, and S290A overexpressed lentiviruses were amplified, purified from GeneChem Co., Ltd. (Shanghai, China). The cDNA was subcloned into the GV492 plasmid (Ubi-MCS-3FLAG-CBh-gcGFP-IRES-puromycin) (Genechem) and then co-transfected into HEK293T cells with the lentiviral genomic plasmids. Lentiviral particles were obtained by collecting supernatant using the kit for ultracentrifugation concentration and purification of lentiviral particles. Cells were cultured in 6-well plates until 60% confluent and infected with lentivirus particles at a MOI of 50 in the presence of 10 g/ml polybrene for 48 h. Stable cells were maintained in medium containing 0.5 µg/mL of puromycin.
MTT assay
Three replicates of equal amounts of cells (3×103/well) were seeded into 96-well plates and incubated for various durations. The cells were incubated with 20 µL 3-(4, 5-dimethylthiazol- 2-yl)-2, 5-diphenyltetrazolium bromide (MTT, 5 mg/ml in PBS) for 4 hours at 37°C. Then, 150 µL dimethyl sulfoxide (DMSO) was added to the wells, and the optical density (OD) was detected at 490 nm by a microplate reader.
Colony formation assay
Exponentially growing cells (5×102) were seeded into 6-well plates for 10-14 days to form colonies. For the determination of colony formation, the cells were fixed in 4% polyformaldehyde, stained with 1% crystal violet. Colonies of at least 50 cells were counted. The mean ± SD from three independent experiments was determined.
5-Ethynyl-2′-deoxyuridine proliferation assay
The 5-ethynyl-2ʹ-deoxyuridine (EdU) assay was performed with an EdU Kit (Byotime, China). Cells were seeded onto 24-well plates and cultured in 0.5 mL medium for 24 hours, and then added 0.5 mL of 10 µM EdU into each well for 2 hours. Then, cells were fixed with 4% polyformaldehyde at room temperature for 15 mins and subsequently incubated with Apollo staining solution for 30 mins. The cell nuclei were stained with Hoechst 33342 (1:1000 in PBS). The proportion of EdU-positive cells was determined with a Zeiss LSM780 confocal microscope system (Carl Zeiss, Germany). Image J_v1.8.0 (National Institutes of Health, USA) was used to count the number of all cells and proliferating cells.
Irradiation
Cells were ionize-irradiated (3.5 Gy/min) at room temperature using X-RAD225 OptiMAX Biological Irradiator (Precision X-Ray Inc., USA). Once irradiated, the cells were immediately transferred to the incubator at 37°C in 5% CO2 until the designated harvest time.
Immunofluorescent staining
Cells grown on coverslips were fixed in 4% paraformaldehyde for 15 min at room temperature and then permeabilized with 0.1% Triton X-100 in PBS for 15 min. The cells were blocked with 5% goat serum for 30 min before incubation with primary antibodies overnight at 4°C. After washing with PBS, cells were incubated with the secondary antibodies for 1 h at room temperature. The cell nuclei were dyed with Hoechst 33342 (1:1000 in PBS). After a final wash with PBS, the slides were covered with anti-fade mounting medium (Beyotime, China). Microscopic analyses were captured at magnification (200×, 630×) with a Carl Zeiss LSM 780.
Flow cytometry
For cell apoptosis analysis, 1×106 cells were washed twice with PBS and stained in 100 µL binding buffer with 5 µL Annexin V-APC and 10 µL 7-ADD for 20 mins in the dark at room temperature. Annexin V/PI staining assays were performed following the manufacturer’s protocol (BD Biosciences, San Jose, CA, USA), Then, an additional 400 µL of binding buffer was added to the cell suspension and the samples were determined by CytoFLEX S flow cytometry (Beckman, USA).
Western blotting
Cell lysates were extracted using RIPA lysis buffer (Beyotime Biotechnology, China) with protease inhibitor cocktail (APExBIO, China) and phosphatase cocktail (APExBIO, China). The protein concentration was measured by a BCA protein assay kit. The proteins were mixed with 5× loading buffer (EpiZyme, China) and boiled at 95°C for 5 min followed by separate in 10% SDS-PAGE (EpiZyme, China). The protein bands were electrophoretically transferred onto polyvinylidene difluoride membranes (PVDF, Roche, USA). The PVDF membranes were blocked with 5% BSA for 1 h at room temperature and incubated overnight with primary antibodies in 1% BSA. After incubation with secondary antibody at a dilution of 1:10000 for 1 h at room temperature, protein bands were visualized by Odyssey infrared imaging system (Li-CorBioscences, USA).
Statistical analysis
All quantitative data are represented as mean values of at least 3 independent experiments ± standard deviation (SD). Multiple hypothesis testing based on FDR using the Perseus has been used to analyze the foldchange between cancer with para-cancerous tissues. Differences between groups were analyzed by Student’s t-test for two groups and one-way ANOVA for more than two groups. Survival analysis was performed using Kaplan-Meier method and compared with the log-rank test. Pearson chi-squared test and Fisher’s exact test were used to analyze the relationship between pSer290-BCLAF1 expression and clinical features. Cox proportional hazard model was used to determine factors related to patient survival. P < 0.05 was considered as statistically significant value. All statistical analyses were performed with GraphPad Prism 8.0.2 software.