Clinical samples
Paraffin samples of 67 CRC tissues with complete clinical data were obtained from Nanfang Hospital, Southern Medical University. A total of 47 CRC tissues and adjacent normal mucosa tissues were also collected from CRC patients undergoing standard resection, without chemotherapy or radiotherapy immunotherapy, at Nanfang Hospital. All specimens were histopathologically confirmed by at least 2 pathologists and obtained with informed consent. This study was approved by Southern Medical University Institutional Board (Guangzhou, China).
Cell culture and treatment
The human CRC cell lines SW620, LS174T, SW480, HCT15, LOVO, SW837, HT29, HCT116, Colo205, RKO, and CaCO2 were obtained from the American Type Culture Collection (ATCC). SW620 and HT29 cells were cultured in DMEM medium (Gibco) with 10% FBS (Gibco) at 37°C with 5% CO2. SW480, Colo205, Ls174t, HCT116, LoVo, HCT15, RKO, and CaCO2 cells were cultured in RPMI 1640 medium (Gibco) with 10% FBS (Gibco) at 37°C with 5% CO2. All these cell lines were routinely authenticated for purity and being infection-free.
Quantitative real-time PCR (qRT-PCR)
Total RNA was isolated from CRC tissues and cell lines using Trizol reagent (Invitrogen). 1μg total RNA was reverse transcribed into cDNA by PrimeScript RT reagent Kit (Takara). The acquired cDNAs were used as templates for quantitative real-time PCR analysis using ChamQTM Universal SYBR Q-PCR Master Mix (Vazyme). The relative RNA expression levels were calculated using the 2-ΔΔCt method, with the levels normalized to GAPDH mRNA. Sequences of the real-time PCR primers were: IGF2BP3 (Forward, 5′-TATATCGGAAACCTCAGCGAGA-3′; Reverse, 5′- GGACCGAGTGCTCAACTTCT-3′); METTL14(Forward, 5′- AGTGCCGACAGCATTGGTG-3′; Reverse, 5′- GGAGCAGAGGTATCATAGGAAGC-3′); GADPH(Forward, 5′-ACAGTCAGCCGCATCTTCTT-3′; Reverse, 5′-GACAAGCTTCCCGTTCTCAG-3′);
Western blotting
Treated CRC cells were extracted by RIPA lysis buffer and quantified by BCA Protein Assay Reagent Kit (Thermo Fisher Scientific). 20-50 μg of protein was separated by SDS-polyacrylamide gels and then transferred to PVDF membranes (Millipore). The membranes were blocked with 5% nonfat milk for 1-2 hours and incubated with primary antibody at 4℃ overnight. Anti-α-tubulin monoclonal antibody (T6199; Sigma) was used as a loading control. The primary antibodies were IGF2BP3 (Abcam, ab208869, 1:400), EGFR (Abcam, ab52894, 1:1000), ERK (Cell Signaling Technology, #4695S, 1:200), p-ERK (Cell Signaling Technology, #4370S, 1:200), JUK (Cell Signaling Technology, #9251S, 1:200), p-JUK (Cell Signaling Technology, #9255S, 1:200), METTL14 (Solarbio, K107984P, 1:4000). Secondary antibodies (1:8000) were labeled with HRP. The ECL detection system (KeyGEN) was used for visualization.
Histology and immunohistochemistry
Tissue samples were fixed with 4 % paraformaldehyde. After incubation, the samples were washed and dehydrated in graded ethanol. After appropriate permeation in xylene, the fixed tissues were embedded in paraffin and followed by cutting 2.5 μm paraffin sections. Then they were de-paraffinized xylene twice and rehydrated in descending concentration of ethanol. Standard hematoxylin-eosin (HE) staining of paraffin-embedded tissue was used for histological examination.
The sections along the HE stained slides were further processed for immunohistochemistry. The protein expression levels of IGF2BP3, EGFR, and METTL14 were determined by IHC analysis. Briefly, the slides were incubated with primary antibodies: IGF2BP3 (Altas Antibodies, HPA002037, 1:200), EGFR (Abcam, ab52894, 1:1000), METTL14 (Solarbio, K107984P, 1:50), at 4 ℃ overnight. The subsequent steps were performed using the SP-9000 SPlink Detection Kit (ZSGB-BIO) according to the manufacturer’s instructions. The final stainings were scored according to immunoreactivescore (IRS) analysis: staining intensity (SI), 0 (no staining), 1 (weak), 2 (moderate), or 3 (strong); percentage of positive cells (PP), 0 (negative staining), 1 (1%-10% positive staining), 2 (10%-25% positive staining), 3 (25%-50% positive staining), and 4 (50%-75% positive staining). SI and PP were multiplied to give a final score. The tissues with a final score ≤3 were classified as ‘Low Expression’ and the tissues with a final score >3 were classified as ‘High Expression’.
Establishment of Stable IGF2BP3 and EGFR Knockdown and Overexpression cells
The IGF2BP3 and EGFR construct was generated by subcloning PCR amplified full-length human IGF2BP3 and EGFR cDNA into pEZ-Lv105. Stable knockdown of target genes was achieved by lenti-viral based short-hairpin RNA delivery. Target specific shRNAs were cloned into lenti-viral vector pLKO.1. Viral particles were packaged in 293FT and used to infect SW480 and HCT116 cell lines. Infected cells were selected by puromycin and expanded to form a stable sub-line. Knockdown efficiency was confirmed at both mRNA and protein levels. A non-target control shRNA purchased from Sigma-Aldrich was used as a negative control. The target sequence of IGF2BP3 was shRNA1 (Forward, 5′- CCGGCGGTGAATGAACTTCAGAATTCTCGAGAATTCTGAAGTTCATTCACCGTTTTTG - 3′; Reverse, 5′- AATTCAAAAACGGTGAATGAACTTCAGAATTCTCGAGAATTCTGAAGTTCATTCACCG-3′); shRNA2 (Forward, 5′- CCGGGCAGGAATTGACGCTGTATAACTCGAGTTATACAGCGTCAATTCCTGCTTTTTG
-3′; Reverse, 5′- AATTCAAAAAGCAGGAATTGACGCTGTATAACTCGAGTTATACAGCGTCAATTCCTGCTTTTTG -3′);
Establishment of IGF2BP3 Knockout Cells
IGF2BP3 knockout in SW480 was generated using CRISPR/Cas9 technology by lentiviral transduction and puromycin selection. Cas9 and single guide RNAs (sgRNAs) lentivirus were bought from Genechem. SW480 cells were first engineered to express Cas9. sgRNAs targeting IGF2BP3 were cloned into the lentiCRISPR vector. SW480 cells were transduced with lentiCRISPR-IGF2BP3 and selected with puromycin. The knockout cell clones were isolated by limited serial dilution. A monoclonal cell line was generated, and the IGF2BP3 knockout effect was confirmed using Western blot and sequencing analysis. The sgRNA sequence targeting IGFBP3 was listed as follows: sg1(Forward, 5′-CTAAACTTCCATGTTGGCTATTATTG-3′; Reverse, 5′-CAATATTTGGTTTCTTATCCCAAAG-3′); sg2(Forward, 5′- GAAATGGCCGCCCAGCAAAACC-3′; Reverse, 5′- CACATTCCCAAGTACTGTACAAG-3′). A non-target control shRNA was used as a negative control.
In vitro cell proliferation, migration, and invasion assays
A colony-forming assay was used to determine the proliferation of cells. Cells were seeded in 6-well culture plates at a concentration of 200 cells per well. The cells were cultured for 14 days and then washed two times with PBS, fixed in 4% (vol/vol) paraformaldehyde, and stained with 0.1% (wt/vol) crystal violet solution. The colonies were counted under the microscope (1 colony for more than 50 cells). For the MTT assay, 1000 cells were seeded and transfected in a 96-well plate. After 24h, 5 mg/ml MTT solution was added to each well and the cells were incubated with MTT for 4 h in standard conditions. Then, the supernatant was removed. In all wells, 10 μl DMSO for MTT crystals dilution were added. The absorbance was measured at 570 nm using an automated microplate reader. For soft agar assay, each well of a six-well culture dish was coated with 2 ml bottom agar mixture (DMEM or 1640, 1.32% (w/v) agar). After the bottom layer had solidified, 2 ml top agar-medium mixture (DMEM or 1640, 0.66% (w/v) agar) containing 1×105 cells were added, and the dishes were incubated at 37°C for 2 weeks. The plates were stained with Crystal Violet, then the number of colonies was counted by a dissecting microscope. Experiments were repeated three times.
Mouse experiments
In vivo experiments were all performed on 4-6-week-old female BABL/cAnN-nude mice bought from Southern Medical University Animal Center. For the subcutaneous implantation model, 2×106 cells were first suspended in 200 ul serum-free medium and then were injected subcutaneously in the right flank. After one week, the tumor volumes were measured every day. Mice were sacrificed at week 4 to harvest the tumor bulks. For in vivo cetuximab treatment, one week after subcutaneous implantation, mice were randomized into control (0.9% NaCl), low dose cetuximab (1 mg/kg), and high dose cetuximab (2 mg/kg) groups, intraperitoneal (i.p.) injection, every 3 days. Mice were sacrificed at week 14 to harvest the tumor bulks. Tumor volume was calculated by the formula V = ab2/2, where a and b are the tumor’s length and width, respectively.
Luciferase reporter assay
The fragments of EGFR-3’UTR containing the wild-type m6A motifs as well as mutant m6A motifs (m6A was replaced by T) were synthesized at HanYi Biology Technologies. The wild-type and mutant EGFR-3’UTR fragments were inserted into the upstream of pmirGLO-basic firefly luciferase vector. CRC cells were seeded in 24-well plates and transfected with 5ug wild-type or mutant EGFR-3’UTR. The relative luciferase activity was detected using the Dual-Luciferase Reporter Assay System (Promega) 48 hours later. Firefly luciferase activity and Renilla luciferase activity were measured using Synergy NE02 (BioTek). The results were shown in the form of relative firefly luciferase activity normalized to Renilla luciferase activity. All the experiments were repeated three times, and three replicates were conducted for each group.
mRNA stability analysis
Cells with or without IG2BP3 overexpressed and knockouted were directly treated with Actinomycin D for 0h, 3h, and 6h at a final concentration of 5 μg/mLand harvested at the indicated time points. Total RNA was extracted, and real-time PCR was conducted to quantify the relative level of EGFR mRNA. The degradation rate and half-life of EGFR mRNA were estimated according to the published paper. Briefly, the degradation rate of mRNA (Kdecay) was calculated by the following equation:
ln(C/C0) = -Kdacayt
t is the transcription inhibition time, and C is the mRNA level at the time t. C0 is the level of mRNA at 0 hour in the equation, which means the mRNA level before decay starts. Thus, the mRNA half-time (t1/2) can be calculated by the equation
ln(1/2) = -Kdecayt1/2
m6A dot blot
Polyadenylated mRNA was purified by GenEluteTM mRNA Miniprep Kit (Sigma) from previously isolated total RNA. The poly(A)+ RNA samples were loaded to Hybond-N+ membrane (GE Healthcare) and UV crosses with the nylon membrane. The membrane was then blocked with 5% nonfat milk for 1 hour and incubated with m6A antibody (Abcam, 190886) at 4℃, overnight. After incubating with horseradish peroxidase-conjugated anti-mouse IgG, the membrane was visualized with the ECL detection system. The relative signal density of each dot was quantified by Image J software. The results of m6A level were shown in the form of relative m6A dot blot density normalized to methylene blue staining density.
MeRIP and MeRIP-qPCR
Total RNA was extracted by Trizol reagent (Takara), and mRNA was purified using GenEluteTM mRNA Miniprep Kit (Sigma). RNA fragmentation reagents (NEB) were used to randomly fragment RNA. The specific anti-m6A antibody (NEB) was applied for m6A immunoprecipitation. Anti-m6A antibody was pre-bound to Protein G magnetic beads in reaction buffer for 30 minutes. The fragmented mRNA was incubated with m6A-antibody-bound protein G magnetic beads at 4℃ for 1 hour and washed with low salt reaction buffer and high salt reaction buffer. M6A-antibody-bound RNA was extracted from the Dynabeads using Buffer RLT (Qiagen) and further incubated with Dynabeads MyOne Silane (Life Technologies). The RNA and Dynabeads mixture were precipitated with 100% ethanol washed with 70% ethanol and then re-suspend with nuclease-free water. The supernatant was carefully collected after the beads were pulled to the side of the tube by a magnetic field. Real-time PCR was carried following m6A-IP to quantify the changes to m6A methylation of EGFR. Sequences of the real-time PCR primers of EGFR were: forward, 5′- GATGGGCAGGTCAGGAGA-3′; reverse, 5′- CCAGGCTATCAATCAGGAAG-3′).
DAA (3-Deazaadenosine) inhibit experiment
The DAA inhibit experiments were performed as described previously [15]. Cells were seeded in 96-well culture plates. 4ug/ml DAA (APExBIO, B6121) were added. MTT assay were then performed to calculate the inhibition rate.
Statistical analysis
Statistical analyses were carried out using SPSS 20.0. Data are presented as means±SD from three independent experiments. For continuous variables, data were analyzed by two-tailed unpaired Student’s t test between two groups and by one-way ANOVA for multiple comparisons. For categorical variables, Chi-square test was used. The overall survival rate curves based on Kaplan-Meier method were plotted using the log-rank test. The correlation between IGF2BP3 expression and EGFR and METTL14 expression was determined by Spearman correlation analysis. P values less than 0.05 were considered statistically significant.