Human samples and ethical approval
Tissues from 30 GC patients were collected for immunohistochemistry in our study. Patients diagnosed with gastric cancer after surgical resection and with complete follow-up data were included. The ethics committee of the Second Affiliated Hospital of the South China University of Technology approved the study. Informed consent was obtained from patients at the time of sample collection.
Data acquisition and analysis
Six scRNA-Seq datasets with both gastric malignant and normal cells data were accessed through the GEO database (GSE134520, GSE158631, GSE163558, GSE167297, GSE183904 and GSE206785). Those data were integrated using the anchors method in the R with “Seurat” and “Harmony” packages. Data were then log-normalized by normalization and scaled for downstream analysis. Further cell subpopulation classification, differentially expressed gene (DEG) analysis of each cluster, and marker gene screening were performed. We used the R package “Limma” to analyze RNA-seq datasets. TCGA-STAD data were downloaded from the TCGA database (https://portal.gdc.com). Overall survival (OS) and free-progression survival (FP) of METTL14 in GC patients were analyzed by using the Kaplan-Meier Plotter online website (https://kmplot.com/). The expression profiles of METTL14 and ATF5 in GC patients were retrieved from the cBioPortal database (https://www.cbioportal.org/) and additionally from GSE66229 data. Subsequently, we employed the Spearman method to scrutinize the correlation between them. Based on the median METTL14 expression, we divided GC patients in the TCGA-STAD cohort into the high METTL14 group and low METTL14 group and further compared the differential expression of the stemness index between the two groups, which was derived from the OCLR algorithm, as constructed by Malta et al.
Cell Lines
Human embryonic kidney cell line 293T, gastric cancer carcinoma cell lines (AGS, BGC823, MKN45, HGC27), and human gastric mucosal cell line GES-1 were obtained from the Global Bioresource Center (ATCC, USA). The cells were grown in a DMEM medium containing 10% fetal bovine serum under 37°C with 5% CO2. All cells were subjected to STR identification and were excluded from mycoplasma contamination.
Plasmid construction and cell transfection
The lentiviral small hairpin RNA plasmid targeting METTL14 was ordered from Guangzhou IGE Biotechnology Ltd. The targeting sequence was GGATGAACTAGAAATGCAA or GCTGGACTTGGGATGATATAT. METTL14 and ATF5 overexpression lentivirus were ordered from Guangzhou IGE Biotechnology Co. The METTL14 plasmid was selected to carry the Flag tag. The lentiviral plasmids were transfected into 293T cells for 48 h using a PEI reagent to produce lentivirus. For the construction of stable cell lines, GC cell lines were infected with lentivirus for 48 h, followed by puromycin treatment (2 μg/ml). Cell transfection efficiency was detected by qRT-PCR and western blot.
Quantitative real-time RT-PCR (qRT-PCR)
Total RNA was extracted from the cell lines using TRIzol reagent according to the manufacturer’s description. After detecting the RNA quality and concentration, 1000 ng total RNA was used for reverse transcription by the HiScript II 1st Strand cDNA Synthesis Kit. The target genes were amplified using ChamQ Universal SYBR qPCR Master Mix on the LightCycler® 96 Instrument. The relative levels of target gene expression were normalized to GAPDH and calculated using the 2-ΔΔCt formula. The primers used are listed in Supplementary Table S1.
Western blot
GC cells were lysed by RIPA reagent and then quantified using the BSA method. Denatured proteins were migrated in 10% SDS-PAGE gels and transferred to nitrocellulose membranes. After closing with 5% BSA, the membranes were incubated with primary antibodiesovernight at 4°C, washed, and conjugated with HRP-conjugated secondary antibodies. The details pertaining to all employed antibodies and their respective working concentrations were provided in Supplementary Table S2. Finally, proteins were detected using an enhanced chemiluminescence kit.
Cell proliferation assays
CCK8 assay was used to detect cell proliferation viability. Cells were prepared at a density of 2000 cells/well. 10% CCK8 solution was added to the cells and was allowed to incubate at 37°C for 2 hours. Cell viability was detected at 0, 24, 48, 72, and 96 hours. The absorbance of the cells was measured at 450 nm by a Biotek Cytation5 instrument. All experiments were performed in triplicate.
For colony formation assay, cells were prepared in 6-well plates (AGS: 250cells/well, BGC823: 500cells/well) and were replaced with fresh DMEM medium every 3 days. When the cells appeared as obvious colonies, they were fixed with methanol at room temperature for 15 minutes, and stained with 0.1% crystal violet at room temperature for 20 minutes. The number of colonies containing more than 50 cells was counted.
For the EdU staining assay, 2000 cells per well were seeded in a 24-well plate and incubated overnight at 37°C. The EdU staining solution was diluted with DEME to a final working concentration of 10 uM and then added to the cells. After setting at 37°C for 2 h, cells were fixed with formaldehyde for 15 min and permeabilized with PBS containing 0.3% Triton X-100 for 10 min. Click reaction solution was configured according to the instructions and incubated for 30 min at room temperature, protected from light. Proliferating cells were labeled with EdU, while nuclei were stained with Hoechst 33342. PBS was used to remove excess reagents between each operation. Photographs were taken using a fluorescent microscope and analyzed using Image J software.
Wound-healing assays
Briefly, monolayers of cells were scratched with a 10ul tip when they reached 90% fusion. Unattached cells were washed away with PBS and added to a serum-free DMEM medium. After photoing under the microscope, the cells continued to be cultured for 24 hours at 37℃. The wound closure was captured after 24 hours and analyzed by Image J software.
Transwell migration and invasion assays
Transwell assays were performed using a 24-well Transwell chamber system. For migration assays, 200 ul of serum-free medium containing 5×104 cells was added to the upper chamber, and 600 ul of regular medium with 10% FBS was added to the lower chamber. After incubating at 37°C for 24 h, cells were fixed with 4% paraformaldehyde for 15 min, stained with 0.1% crystal violet at room temperature for 20 min, with the non-migrating cells removed with a cotton swab. For the invasion assay, 50 ul matrigel with a dilution of 1:8 was first pre-coated on the chambers’ upper surface and solidified at 37°C for 1 h. Then 200 ul of GC cells (1×105 AGS; 2×105 BGC823) were seeded into the upper chamber, followed by 600 μl of complete medium placed in the lower chamber. After incubation at 37°C for 24 h-48 h, the invading cells in the lower chamber were fixed and stained. Migrated or invaded cells were captured by microscopy, and the number of these cells was counted by Image J software.
In vivo assays for tumor growth and metastasis
5-week-old female Balb/c nude mice were purchased from Hunan SJA Laboratory Animal Co., Ltd. For tumor growth assay, 2×106 GC cells were injected subcutaneously into the nude mice with 5 mice in each group. Tumor volume was measured every 3 days using the following formula: tumor volume = (length × width2)/2. At the end of the experiment, mice were euthanized to take off the tumor, followed by being pictured and weighed. For the in vivo metastasis assay, 1×106 GC cells were injected into nude mice through the tail vein. 4 weeks later, the mice were sacrificed, and the metastatic lung tumors were analyzed. All animal experiments were performed under the approval of the Laboratory Animal Ethics Committee, School of Medicine, South China University of Technology.
Immunohistochemistry
GC tissue and xenograft tumors were paraffin-embedded and sectioned. After dewaxing, hydration, and antigen repair, goat serum was used to block the non-specific antigen for 30 min. Diluted primary antibodies were added to the tissue overnight at 4°C, and the secondary antibody was subsequently labeled. The primary antibodies used in this study and their working concentration were listed in Supplementary Table S2. Proteins were stained with DAB buffer, while nuclei were stained with hematoxylin. The integrated score was defined as the value of multiplying intensity and area. The intensity score was set as follows: brown or dark yellow: 3 scores; yellow: 2 scores; light yellow: 1 score; gray: 0. The staining area was scored as follows: exceeding 50%: 3 scores; 25%-50%: 2 scores; 10%-25%: 1 score; <10%: 0. “Scores <3” was classified as the METTL14 low group, while “scores ≥3” was classed as high expression of METTL14.
Sphere formation assays and ELDA assays
GC cells were seeded at 1000 cells/well in 24-well ultra-low attachment plates for sphere formation assays. Cells were resuspended in DMEM/F12 medium supplemented with 1X B27, 20ng/ml bFGF, 20ng/ml EGF and 4ug/ml insulin. At 14 days, spheres larger than ~20 μm in diameter were photographed, and their diameters were measured for analysis.
For extreme limiting dilution analysis, cells were seeded at decreasing densities (20, 10, 5 cells/well into 96-well ultra-low attachment plates) in DMEM/F12 medium supplemented with various growth factors, with 10 replicates per group. At 14 days, any well containing one or more spheres was counted and analyzed on the ELDA website (http://bioinf.wehi.edu.au/software/elda/) for further analysis.
Drug resistance assays
The effect of 5-FU and Oxaliplatin on GC cells was estimated by cell viability. Briefly, GC cells were seeded into 96-well plates and cultured at 37°C until fusion reached approximately 80%. Different concentrations of DDP were added and then incubated for 24 h. Cell viability was subsequently assessed using the CCK8 assay.
Dot blot
Total RNA was extracted by using TRIzol reagent according to the manufacturer’s description mRNA was isolated and purified using the Dynabeads mRNA Purification Kit. After denaturation at 95°C for 3 min, 2ul of mRNA was added to Nitrocellulose membranes and placed at 37°C for 30 minutes allowing mRNA to cross-link with the membranes. The membranes were incubated with m6A antibody at 4°C overnight. After incubation with secondary antibodies, the membrane was visualized by ECL chemiluminescence.
Methylated RIP-qPCR (MeRIP-qPCR)
Total RNA from GC cells was extracted with TRIzol reagent and fragmented to approximately 300 nt. 10% of the fragmented RNA samples were used as the Input group according to the manufacturer’s instructions. 4ug of m6A antibody or IgG antibody was incubated with fragmented RNA overnight at 4°C, and then protein A/G magnetic beads were added to continue incubation at 4°C for 6 hours. The m6A-enriched RNA was extracted as previously described, and the m6A-mRNA enrichment was analyzed by reverse transcription qPCR. Specific primers were designed for MeRIP-qPCR analysis according to the information from a motif-dependent m6A site predictor SRAMP (http://www.cuilab.cn/sramp).
Dual-luciferase reporter assay
We designed the wild-type (WT) ATF5 sequence and the mutant (Mut) ATF5 sequence with a putative m6A site mutation. These sequences were separately cloned into the pGL3-Basic vector to generate the luciferase reporter clone. The Renilla luciferase plasmid was used as an internal control. The luciferase reporter plasmid and the control plasmid were co-transfected into GC cells. Firefly and Renilla luciferase activities were assayed 24 h later using a dual luciferase reporter system. A dual luciferase reporter assay was conducted with three biological repetitions each time.
Actinomycin D treatment
The stability of ATF5 mRNA was analyzed with Actinomycin D, a transcriptional inhibitor under a finial working concentration of 5 ug/mL. Cells were lysed with TRIzol reagent, and RT-qPCR was applied to detect RNA levels.
Chromatin immunoprecipitation-qPCR (ChIP-qPCR)
ChIP assay was performed using BersinBio ChIP Kit (Catalog Bes5001) according to the manufacturer’s protocol. Briefly, approximately 2 × 107 cells were fixed in 1% formaldehyde for 10 min at room temperature. After 1 ml ChIP lysis buffer treatment, we collected the nuclear pellet to conducting ultrasonic pyrolysis. Immunoprecipitation was carried out overnight with purified anti- H3K18la antibody (PTM-1427RM), anti-ATF5 antibody (sc-377168 X), or IgG antibody as a negative control. Protein A/G beads were used to pull down the antigen-antibody compounds and then washed with washing buffers. The DNA-protein crosslinks were reversed with 5 M NaCl at 65 °C for 6 h, and DNA from each sample was purified. qPCR was performed using 2 μL DNA samples with the following primers:
METTL14: forward 5′- GATAGCCGCTTGCAGGAGAT -3′, reverse 5′-GCATTCTCGATCCCTCCCAC -3′.
WDR74: forward 5′- CGGCCTTATCCTTCCCATCTT -3′, reverse 5′- AGGGCTGTTTCCAAAATATAGCCA -3′.
Statistical analysis
Statistical analyses were performed with GraphPad Prism 8.3 and R 4.0 software. Experiments were repeated independently at least thrice. Groups were classified based on the median METTL14 expressed. Two-tailed Student’s t-test was used to compare data between two groups, and ANOVA analysis was employed to compare among three or more groups. Overall survival and free-progression survival were analyzed using the Kaplan-Meier method. In addition, correlation analysis of gene expression was performed by Spearman’s method. P<0.05 was considered statistically significant (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001).