Patient samples
A total of 55 gastric tumor tissues and matched adjacent normal tissues were evaluated in this study. Tissues were obtained from patients who underwent surgery at the Department of General Surgery, China-Japan Union Hospital of Jilin University, China between 2015 and 2018. After surgical dissection, patient samples were immediately frozen by liquid nitrogen and stored at -80°C. Patients did not receive chemo- or radio- therapy before surgery. This study was approved by the ethics committee from the Institutional Review Board of the China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, P. R. China. All participants gave written informed consent.
Cell culture and reagents
Five gastric cancer cell lines (AGS, BGC-823, MKN-45, MGC-803 and SGC-7901) and one normal gastric mucosa endothelial cell line GES-1 were purchased from the Cell Research Institute of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in RPMI 1640 medium (Thermo Fisher Scientific, Inc., Carlsbad, CA, USA) with 10% fetal bovine serum (FBS) (Thermo Fisher Scientific, Inc., Carlsbad, CA, USA) and 100 units/ml penicillin plus 100 μg/ ml streptomycin (Thermo Fisher Scientific, Inc., Carlsbad, CA, USA) at 37°C in a humidified cell culture incubator with 5% CO2. The 5-Fu resistant gastric cancer cell line AGS/5-Fu R was established according to previous description by continually exposing to increased concentrations of 5-Fu (30). The acquired 5-Fu resistant cells were re-selected by treating with 5-Fu at 300 uM every two months. The rabbit anti-GLUT1 (#73015), rabbit anti-Hexokinase 2 (#2867), rabbit anti-LDHA (#3582) and rabbit anti-β-actin (#4970) antibodies were purchased from Cell Signaling Technology (Danvers, MA, USA). Mouse anti-PTBP1 (#32-4800) monoclonal antibody was purchased from Thermo Fisher Scientific (Shanghai, China). 5-Fu, 2-DG and Oxamate were purchased from Sigma-Aldrich (Shanghai, China).
Construction and transfections of Plasmid DNA, siRNA and miRNA
The pCDNA3.1-PTBP1 overexpression plasmid was constructed according to previous description (33). Fragment encoding the full-length cDNA of PTBP1 was ligated into the pcNDA3.1 vector. The small interfere RNA (siRNA) targeting SNHG16 was designed and synthesized by Sangon Biotech (Shanghai, China). The PTBP1 shRNA was synthesized by ABLife Biotech (Wuhan, China). Silence sequence of PTBP1: GCGTGAAGATCCTGTTCAATA. Sense and antisense strands were annealed to be shRNA. Vector pGFP-B-RS was digested by HindⅢ and BamHⅠ at 37℃ for 2h~3h. A linearized vector DNA digested by HindⅢ and BamHⅠ and insert shRNA were ligase by T4 DNA Ligase (NEB). The interference sequence of shRNA was verified by Sanger sequencing. Control miRNA and miR-506-3p precursor were purchased from GenePharma Co. (Shanghai, China). Transfections were performed using the Lipofectamine 2000 (Invitrogen, California, USA) according to the manufacturer’s instructions. MiRNAs or siRNAs were transfected at 50 nM. Plasmid DNA was transfected at 2 µg. After 48 h, cells were collected for the following experiments.
RNA isolation and qRT-PCR
Total RNA was isolated from surgically resected gastric tumors or cultured gastric cancer cells using the TRIZOL reagent (Ambion, USA). The RNA was further purified with two phenol-chloroform treatments and then treated with RQ1 DNase (Promega, Madison, WI, USA) to remove DNA. The quality and quantity of RNA samples were assessed by measuring the absorbance at 260 nm/280 nm (A260/A280) using Smartspec Plus (BioRad, Hercules, CA, USA). First-strand cDNA was synthesized using 0.5 ug total RNA using a SuperScript First-Standard Synthesis System for RT-PCR (Invitrogen, Carlsbad, CA, USA) following the manufacturer’s protocol. qRT-PCR experiments were performed using the SYBR Green qPCR Master Mix (ThermoFisher Scientific, Shanghai, China). Samples were analyzed using an ABI Prism 7700 Sequence Detection System (Applied Biosystems, Foster City, NJ, USA). Primers for qRT-PCR were: miR-506-3p: Forward: 5’- GCCACCACCATCAGCCATAC-3’, Reverse: 5’-GCACATTACTCTACTCAGAAGGG-3’, SNHG16: Forward: 5’-GCAGAATGCCATGGTTTCCC-3’, Reverse: 5’- GGACAGCTGGCAAGAGACTT-3’, PTBP1: Forward: 5’-GCATCGACTTTTCCAAGCTC-3’, Reverse: 5’-GGAAACCAGCTCCTGCATAC-3’, GLUT1 Forward: 5’- TTGCAGGCTTCTCCAACTGGAC-3’, Reverse: 5’-CAGAACCAGGAGCACAGTGAAG-3’, HK2: Forward: 5’-TACACTCAATGACATCCGAACTG-3’, Reverse: 5’-CGTCCTTATCGTCTTCAATATCC-3’, LDHA: Forward: 5’-ATGAAGGACTTGGCGGATGA-3’, Reverse: 5’-ATCTCGCCCTTGAGTTTGTCTT-3’, β-actin: Forward: 5’-CTGAGAGGGAAATCGTGCGT-3’, Reverse: 5’-CCACAGGATTCCATACCCAAGA-3’. The expression of β-actin was used to normalize the relative expression levels. Human U6 was an internal control for miRNA detection. The thermal profile was set as follows: 95°C for 1 min and 40 cycles at 95° C for 15 s, 58°C for 20 s, and 72°C for 20 s. Experiments were performed in triplicate and repeated three times. The results were analyzed using the 2−ΔΔCt method.
RNA-Seq analysis
The quality and quantity of the purified RNA samples were redetermined by measuring the absorbance at 260 nm/280nm (A260/A280) using Smartspec Plus (BioRad, USA). The integrity of RNA was further verified by 1.5% agarose gel electrophoresis. For each sample, 1μg of the total RNA was used for RNA-seq library preparation by VAHTS Stranded mRNA-seq Library Prep Kit (Vazyme Biotech Co., Ltd, Nanjing, China). Polyadenylated mRNAs were purified and fragmented, and then converted into double strand cDNA. After the step of end repair and A tailing, the DNAs were ligated to VAHTS RNA Adapters (Vazyme Biotech Co., Ltd, Nanjing, China). Purified ligation products corresponding to 200-500 bps were digested with heat-labile UDG, and the single strand cDNA was amplified, purified, quantified and stored at -80℃ before sequencing. For high-throughput sequencing, the libraries were prepared following the manufacturer's instructions and applied to Illumina HiSeq X Ten system for 150 nt paired-end sequencing.
RNA-Seq raw data clean and alignment
Raw reads containing more than 2-N bases were first discarded. Then adaptors and low-quality bases were trimmed from raw sequencing reads using FASTX-Toolkit (Version 0.0.13). The short reads less than 16nt were also dropped. After that, clean reads were aligned to the GRch38 genome by tophat2 (34) allowing 4 mismatches. Uniquely mapped reads were used for gene reads number counting and FPKM calculation (fragments per kilobase of transcript per million fragments mapped).
Functional enrichment analysis
To sort out functional categories of DEGs, Gene Ontology (GO) terms were identified using KOBAS 2.0 server (35). Hypergeometric test and Benjamin-Hochberg FDR controlling procedure were used to define the enrichment of each term.
Differentially Expressed Genes (DEG) analysis
The R/Bioconductor package edgeR (36) was utilized to screen out the differentially expressed genes (DEGs). A false discovery rate <0.05 and fold change >2 or < 0.5 were set as the cut-off criteria for identifying DEGs.
Bioinformatics analysis
The interaction between lncRNA-SNHG16 and miR-506-3p was predicted by starBase of ENCORI (http://starbase.sysu.edu.cn/). The correlation between SNHG16 and PTBP1 expressions in gastric cancer patient was analyzed by starBase of ENCORI. The binding of miR-506-3p and PTBP1 was predicted from starBase of ENCORI. Survival rates according to SNHG16, miR-506-3p and PTBP1 expressions in gastric cancer patients were analyzed by www.kmplot.com. 3’UTRs of glycolysis enzymes were obtained from NCBI. Binding motifs of PTBP1 were analyzed from starBase of ENCORI. p<0.05 was considered statistical significance.
RNA immunoprecipitation (RIP)
RNA was isolated from GC cells using a Rneasy Mini kit (Qiagen, Hilden, Germany). GC cells were lysed in RIP lysis buffer from the Magna RIPTM RNA-binding protein immunoprecipitation kit (Millipore, Bedford, MA, USA). Anti-IgG (ab172730, Abcam) or anti-PTBP1 (#32-4800, Thermofisher) antibody with A/G immunomagnetic beads were premixed in immunoprecipitation buffer. Cell lysates were mixed with above reaction to immuno-precipitate PTBP1-RNA complexes at 4 oC for 16 hours. Proteinase K was then added into the reaction. RNA samples were purified. The enrichments of GLUT1, HK2 and LDHA mRNAs which bond with PTBP1 were analyzed by qPCR and RT-PCR. Experiments were repeated three times.
RNA pull-down assay
RNA pull-down assay was performed according to previous report. Briefly. 3’UTRs of glycolysis enzymes and the negative control (antisense 3’UTR and scramble) were in vitro transcribed and biotin labeled using a biotin RNA labeling mix (Roche, Shanghai, China). Mixture was treated with RNase-free DNase I and purified by a RNeasy mini kit (Qiagen, Hilden, Germany). Proteins from GC cell extracts were incubated with the above biotinylated RNAs at 50 pmol/mg followed by incubation with streptavidin agarose beads (Invitrogen, Carlsbad, CA, USA). After washing, protein samples were detected by Western blot. Experiments were repeated three times.
RNA stability assay
GC cells were first transfected with control siRNA or siPTBP1 for 48 hours. Actinomycin D (ActD) was added at a final concentration of 5 μg/ml and incubated for various periods. Total RNA was isolated and transcripts of glycolysis enzymes were examined by qRT-PCR. β-actin was an internal control.
Seahorse metabolic flux analysis
Extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) were detected using the Seahorse XFp Analyzer (Agilent, Santa Clara, CA, USA) and detection kits from Agilent Technologies Inc. (Santa Clara, CA, USA) according to the manufacturer’s instructions. Equal number of cells from each treatment were analyzed. Results were normalized by protein concentrations. Experiments were performed in triplicate and repeated three times.
Glucose uptake and lactate production
Gastric cancer cells were seeded on a 12-well plate (1x105 per well). The detections of glucose uptake and lactate production were performed using the Glucose Uptake Colorimetric Assay Kit (Applygen Technologies, Beijing, China) and the L-lactate assay kit (BioVision, Milpitas, CA, USA) according to the manufacturer’s instructions. Results was normalized by the cell number of each well. Relative glycolysis rate was calculated from the absorbance of drug-treated cells/untreated cells. Experiments were performed in triplicate and repeated three times.
Luciferase assay
Gastric cancer cells (5 × 104 cells/well) were seeded in 24-well plates and cultured for 24 hours. Cells were then co-transfected with control miRNA or miR-506-3p plus 50 ng pGL3-reporter luciferase reporter containing WT- or Mut- SNHG16 or PTBP1 3’UTR using Lipofectamine 2000 (Thermo Fisher Scientific Inc., Waltham, MA, USA). Forty-eight hours post-transfection, luciferase activity was measured using a Dual luciferase reporter assay system (Promega, Madison, WI, USA) according to the manufacturer’s instructions. Firefly luciferase activity was normalized to that of the Renilla luciferase. Experiments were performed in triplicate and repeated three times.
Cell viability assay
Cell viability was examined by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay (Sigma-Aldrich, Shanghai, China). Gastric cancer cells (5x103 cells/well) were seeded into 96-well plates at 80% confluence for 24 hours. Cells were washed with PBS. medium was refreshed and MTT solution was added at 37 °C for 2 hours. Then, 150 μl DMSO was added to dissolve the formazan crystals. The optical density (OD) of formazan concentrations was determined at 570 nm using a microplate reader (Bio-Rad Laboratories). The OD values were normalized by cell numbers. Relative viability was obtained from the absorbance at 540 nm of drug-treated cells/the absorbance at 540 nm of untreated cells. Experiments were performed in triplicate and repeated three times.
Clonogenic assay
Anchorage-dependent GC cells (1 x 103 cells/well) were plated onto 6-well plate for 24 hours. After 5-Fu treatment, medium was refreshed and cells were cultured for additional 10 days then stained by 1% crystal violet solution. The survival clones were examined under microscopy. Experiments were repeated three times.
Cell apoptosis assay
GC Cells (3 × 105 per well) were seeded onto 6-well plates for 24 hours. Cell apoptosis rate was assessed by FITC-Annexin V/PI Kit (#556547, BD Biosciences, San Jose, CA, USA) according to the manufacturer’s instructions. Briefly, after treatments, cells were washed twice with cold PBS. Cells were stained with FITC-Annexin V (5 ml) and PI (5 ml) for 30 min at room temperature in dark. Apoptosis was analyzed using a BD Accuri C6 Flow cytometer (BD Biosciences, San Jose, CA, USA). Experiments were repeated three times.
Western blot
Proteins were extracted from cells using the RIPA lysis buffer (Beyotime Ltd., Shanghai, China) suppled with 1 x protease inhibitor cocktail (Sigma-Aldrich, Shanghai, China). Lysates were incubated on ice for 15 minutes and centrifugated at 10,000 g for 15 min at 4 °C. Protein concentrations were measured by Bradford method. Equal amounts of proteins were applied to a 10% sodium dodecyl sulfate-polyacrylamide gel (SDS-PAGE) and transferred to a nitrocellulose membrane. Membranes were blocked with 5% nonfat milk at room temperature for 1 hour. After complete washing by PBST, membranes were incubated with primary antibodies (1:1000 dilution) with gentle shaking at 4 °C overnight. After washing, membranes were incubated with HRP-conjugated goat anti-mouse or goat anti-rabbit antibody (1:3000 dilution) at room temperature for 1 hour. The detection of antibody-bound protein signals was performed using the enhanced chemiluminescence kit (Bio-Rad Ltd., USA). Experiments were repeated three times.
Xenograft mice model
All of the xenograft experiments were complied with the guidelines of the Institutional Animal Care and Use Committee of the China-Japan Union Hospital of Jilin University. Totally thirty-two six-week-old nude mice were used in this study. Mice were kept on a regular 12/12 hr light-dark cycle cages. Nude mice were separated to two groups (16 each) then were injected subcutaneously with AGS cells (8x106) transfected with control shRNA or PTBP1 shRNA to establish xenograft models. Mice from each xenograft group were randomly divided into two groups and treated as follows: PBS control, 5-Fu alone [40 mg/kg intraperitoneal (i.p.), 2 times/wk], sh PTBP1 alone and 5-Fu plus sh PTBP1. Mice mortality was monitored daily. Tumors from dead mice were collected at the death time point. After 80 days the survival mice were euthanized by CO2 method and the xenograft tumor tissues were dissected. Tumors were stored at -80°C for downstream analysis. Experimental protocol was carried out in accordance with the European Communities Council Directive of 24 November 1986 (86/609/EEC) and approved by an institutional review committee from Institutional Animal Care and Use Committee of the China-Japan Union Hospital of Jilin University.
Statistical analysis
Statistical difference was analyzed using the GraphPad Prism 7.0 software. Results are presented as the mean ± SD. The unpaired Student's t-test was used for the data analysis between two groups and significance among three or more groups was analyzed by one-way ANOVA followed by Bonferroni corrections. All experiments were performed in triplicate and repeated three times. A statistical difference of p < 0.05 was considered significant.