Patients and Specimens. A total of 219 HCC tissues were obtained from surgical resections of patients in Eastern Hepatobiliary Surgery Hospital (EHBH, Shanghai, China) from 2010 to 2018. The procedure of human specimen collection was approved by the Ethics Committee of EHBH. A Tissue microarray containing 134 HCC tissues from patients with follow-up information (Cohort A) was used for survival analysis. A total of 97 patients at high risk of recurrence that received sorafenib therapy after hepatectomy in EHBH from 2015 to 2018 were included in Cohort B. Recurrence-free survival (RFS) and overall survival (OS) analysis were performed using the Kaplan-Meier method. RFS was defined as the interval between the date of surgery and recurrence. OS was defined as the interval between the date of surgery and death. If recurrence was not diagnosed, patients were censored on the date of the last follow-up.
Cell lines and cell culture. HepG2 (p53-wild), Hep3B (p53-null), Huh7 (p53-Y220C), HCT116 (p53-wild), HT29 (p53-R273H) and PLC/PRF/5 (p53-R249S) cells were purchased from Shanghai Cell resource center of Chinese Academy of Sciences. JHH1 (p53-wild) cells were kindly provided by Haojie Jin at the Shanghai Cancer Institute. HEK293T cells were prechased from the American Type Culture Collection (ATCC). HepG2, Hep3B, Huh7, PLC/PRF/5, JHH1 and HEK293T cells were cultured in DMEM (BasialMedia, with 4.5 g/L glucose, 4mM L-glutamine, and 0.11 g/L pyruvate). HCT116 and HT29 cells were cultured in RPMI-1640 (BasialMedia, with 2 g/L glucose and 300mg/L L-glutamine). Culture medium was supplemented with 10% Foetal Bovine Serum (Biological Industries, 04-001-1ACS), penicillin (10000U/mL)/streptomycin (10000ug/mL) and amphotericin B (25 ug/mL) (BasialMedia). Mycoplasma Elimination Reagent (InvivoGen, 25 mg/mL) was used for the last 3 days before freezing. Fresh cells were recovered every month to maintain cell viability and a low number of cell lines passage. Cells were maintained at 37℃ in an atmosphere of humidified air containing 5% CO2. Cells were routinely passaged every 2 days and not allowed to grow to confluence.
Real-time PCR analysis. Total RNA was isolated from cells using TRIzol Reagent (ThermoFisher, 15596-018) and RNeasy Mini Kit (Qiagen, 74104). For RT-PCR, mRNA was reverse transcribed into cDNA using the M-MLV system (Promega, M1701). Real-time PCR was performed on LightCycler 480 II system using a SYBR Green PCR Master Mix (Roche), with 18S as a reference control. Primers are listed in Supplementary Table 3.
Cell proliferation, colony formation and viability assay. Cell proliferation was measured using a CCK-8 Cell Counting Kit (Vazyme, A311) according to the manufacturer’s instructions. For colony formation assay, cells were plated in 6-well plates at a density of 3000–5000 cells/well and treated with drugs 24 h later. Colonies were stained with crystal violet after 12–14 days and then counted by using Image J for quantification. For cell viability assessment, cells were cultured in 96-well plates and tested using the CellTiter-Glo luminescent cell viability assay (Promega, G7571).
Flow cytometry. Cells (30000–50000 cells/well) were seeded in 6-well plates and treated with drugs for 48h. For tumor cell death detection, cells were resuspended in 500µl phosphate buffer saline (PBS) and incubated with PropidiumIodide (PI) (Sigma-Aldrich, P4170) for 30 minutes and then analyzed by the flow cytometry performed with a FACSVerse flow cytometer (BD Biosciences). For lipid peroxidation assay, cells were resuspended in 500µl PBS containing 20mM C11-BODIPY 581/591 (ABclonal, RM02821) and incubated for 1h at 37℃ in a cell culture incubator. The signals from both non-oxidized C11 (wave length > 580 nm) and oxidized C11 (wave length 505–550 nm) were monitored. The data were normalized to control samples as shown by relative lipid peroxidation.
Western blotting, co-immunoprecipitation and antibodies. Cells were lysed in RIPA Lysis Buffer (Beyotime, P0013B) on ice and centrifuged at 12,000 rpm 4°C for 15 minutes. Protein concentrations were measured using a BCA protein assay kit (Thermo Scientific, 23225). Blotting was performed following the standard procedures and detected using an Odyssey fluorescence scanner (Li-Cor, Lincoln, Neb). Antibodies used are URI (1:1000, Proteintech, 11277-1-AP,), SREBP1 (1:200, Santa Cruz, sc-13551), SCD1 (1:1000, ABclonal, A16429), FASN (1:1000, ABclonal, A19050), FADS2 (1:1000, ABclonal, A10270), GPX4 (1:1000, ABclonal, A11243), GCLM (1:1000, ABclonal, A11444), GCLC (1:1000, ABclonal, A4499), SLC7A11 (1:1000, Proteintech, 26864-1-AP), ACSL4 (1:1000, Proteintech, 22401-1-AP), MDM2 (1:1000, ABclonal, A13327), Myc-Tag (1:1000, ABclonal, AE010), Flag-Tag (1:1000, ABclonal, AE005), His-Tag (1:1000, ABclonal, AE003), HA-Tag (1:1000, ABclonal, AE008), ACC (1:1000, CST, 3676), TRIM28 (1:1000, Proteintech, 15202-1-AP,), P53 (1:1000, Proteintech, 10442-1-AP), P21 (1:1000, Proteintech, 10355-1-AP), USP5 (1:1000, ABclonal, A4202), USP7 (1:1000, ABclonal, A13564), USP14 (1:1000, ABclonal, A19589), Actin (1:5000, Proteintech, 66009-1-ig), GAPDH (1:5000, ABclonal, AC001).
For co-immunoprecipitation assay, cell lysates were prepared with IP lysis buffer (Beyotime, P0013) and incubated with antibodies overnight at 4°C, followed by the addition of Mag 25K/Protein A/G (Enriching, LM220622) for another 4h. Immune complexes were isolated by centrifugation and proteins were eluted into loading buffer, followed by western blotting.
Luciferase reporter assay. To assess SCD1 gene promotor activity, cells were co-transfected with SCD1 luciferase reporter plasmid, with Renilla control reporter as an internal control. After 48 h, cells were lysed in a Passive Lysis 5X Buffer (Promega, E1941), and the enzymatic activity of luciferase were measured using a Dual-Luciferase Assay kit (Vazyme, DL101-01), according to the manufacturer's protocol.
Chromatin immunoprecipitation (ChIP) assay. ChIP assay was performed using the ChIP Assay Kit (Beyotime, P2078). Briefly, 1×107 cells were cross-linked with formaldehyde/PBS for 10 min at 4°C, and then added glycine solution, reaction at room temperature for 5 min. Subsequently, samples were sonicated in lysis buffer to obtain 200-1,000 bp DNA fragments to be immunoprecipitated with 2 µg of mouse SREBP1 (Santa Cruz, sc13551), mouse P53 (Santa Cruz, sc126) or mouse IgG antibodies. The primer sequences specific to the promoter region of gene are listed in Supplementary Table 3.
Tumor xenograft experiment. All mice were treated according to protocols approved by the Navy Medical University Animal Care Facility and the National Institutes of Health guidelines. For cell line derived xenografts (CDX) models, approximately 1×107 HepG2-Ctrl and HepG2-shURI cells in 0.1 mL PBS were injected subcutaneously into the right flank of 6-week-old male nude mice. Tumor volume was measured using a caliper every 3 days and calculated using the modified ellipsoidal formula: tumor volume (mm3) = (length×width2) /2. After tumour establishment, mice were randomly assigned to once per day treatment. Mice were sacrificed by cervical dislocation when the volume reached 1-1.5 cm3 and tumor tissues were harvested for further study.
Immunohistochemistry and Scoring. 10% formalin-fixed paraffin-embedded tissues were sectioned (4µm) and stained with hematoxylin and eosin (H&E) for histological analysis or used for immunohistochemistry (IHC). For IHC, endogenous peroxidases were inactivated by 3% hydrogen peroxide and nonspecific signals were blocked by 1% BSA. Sections were incubated with primary antibody at 4°C overnight, HRP-conjugated secondary antibody at 37°C for 1h, and subsequently stained with DAB substrate. Counterstaining was performed with hematoxylin, and mounted with a mounting medium. Antibodies are listed in Supplementary Table 4.
H-score assay was performed, ranging from 0 to 300. URI and SCD1 staining was scored according to four categories: 0 for ‘no staining’, 1 + for ‘light staining’, 2 + for ‘intermediate staining’; and 3 + for ‘dark staining’. The percentage of cells at different staining intensities was determined by visual assessment, with the score calculated using the formula 1 × (% of 1 + cells) + 2 × (% of 2 + cells) + 3 × (% of 3 + cells).The outcome-based discriminatory threshold IHC H-score for this analysis was set at 200 and samples were then classified as either low (H-score < 200) or high (H-score ≥ 200) for URI and SCD1 protein expression.
Multiplex immunohistochemistry/immunofluorescence (mIHC/IF) staining. To investigate the expression of URI and SCD1 in patient tissues, multiplex immunohistochemistry/immunofluorescence (mIHC/IF) was conducted using a tissue microarray. Briefly, endogenous peroxidases were blocked by 3% hydrogen peroxide and nonspecific signals were blocked by 1% BSA. The slides were immersed in Tris-EDTA buffer to perform heat-induced antigen retrieval. The slides were then incubated with anti-URI (Proteintech, 11277-1-AP), anti-SCD1 (Abcam, ab236868) antibodies. Signal detection were performed using a TSA kit (AKOYA) and nuclei were counterstained with DAPI. Slides were imaged and scanned using a slice scanner (Pannoramic MIDI: 3Dhistech, Hungary) and images were analyzed via HALO 2.0 Area Quantification algorithm (Indica Labs; Corrales, NM), at Nanjing Freethinking Biotechnology Co., Ltd. (China).
RNA sequencing. Total RNA was isolated using a RNeasy mini kit (Qiagen, Germany) from three biological replicates. TruSeq Stranded Total RNA Sample Preparation kit (Illumina, USA) was used to prepare the strand-specific libraries following the manufacturer’s instructions. Briefly, using the oligo (dT) beads to enrich mRNA. Following purification, the mRNA is fragmented into small pieces using divalent cations under 86℃ for 6 min. The cleaved RNA fragments are copied into first strand cDNA using reverse transcriptase and random primers. This is followed by second strand cDNA synthesis using DNA Polymerase I and RNase H. These cDNA fragments then go through an end repair process, the addition of a single ‘A’ base, and then ligation of the adapters. Products are then purified and enriched with PCR to create the final cDNA library. Purified libraries were quantified by Qubit 2.0 Fluorometer (Life Technologies, USA) and validated by Agilent 2100 bioanalyzer (Agilent Technologies, USA) to confirm the insert size and calculate the mole concentration. Cluster was generated by cBot with the library diluted to 10 pM and then were sequenced on the Illumina NovaSeq 6000 (Illumina, USA). The library construction and sequencing was performed at Shanghai Biotechnology Corporation.
For each sample, 33-95M RNA-seq clean reads were obtained that mapped to homo sapiens using HISAT2. Sequencing read counts were calculated using Stringtie (v.1.3.0). Then expression levels from different samples were normalized by the Trimmed Mean of M values (TMM) method. The normalized expression levels of different samples were converted to FPKM (Fragments Per Kilobase of transcript per Million mapped fragments). The edgeR package of R was used to analyze the difference between intergroup gene expression. The P-value threshold was determined by controlling the FDR (False Discovery Rate) with the Benjamini algorithm. The corrected P-value is called the q-value. Differentially expressed genes (DEGs) were defined as transcripts with a fold change in expression level (according to the FPKM value) greater than 2.0 and a q-value less than 0.05. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and GO enrichment analysis was performed with the clusterProfiler package of R and the enrichment criteria including a q-value < 0.05. Heatmaps of specific genes were generated using the pheatmap package of R. Gene set enrichment analysis (GSEA) was performed using GSEA software.
CUT&Tag library construction. HepG2 cells were treated with 5µM Nutlin-3 for 4h. Dead cells were identified by trypan blue staining. Ensure that the cell viability is greater than 95% before detection. Hyperactive™ In-Situ ChIP Library Prep Kit (TD901-TD902, Vazyme Biotech) for Illumina was used to perform CUT&Tag assay47. Briefly, concanavalin A-coated magnetic beads (ConA beads) were added to resuspended cells and incubated at room temperature to bound cells. Non-ionic detergent Digitonin was used to permeate cell membrane. Then, mouse p53 antibody (sc126, Santa), secondary antibody and the Hyperactive pA-Tn5 Transposase were incubated with the cells that were bounded by ConA beads in order. Therefore, the Hyperactive pA-Tn5 Transposase can exactly cut off the DNA fragments that were bound with target protein. In addition, the cut DNA fragments can be ligated with P5 and P7 adaptors by Tn5 transposase and the libraries were amplified by PCR with the P5 and P7 primers. The purified PCR products were evaluated using the Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA). Finally, these libraries were sequenced on the Illumina NovaSeq6000 platform and 150 bp paired-end reads were generated for the following analysis by Shanghai OEbiotech company.
Lipidomics by liquid chromatography coupled to mass spectrometry (LC-MS). Lipid extraction and mass spectrometry-based lipid detection were performed by BioNovaGene (Suzhou, China). Briefly, take an appropriate amount of sample in a 2 mL EP tube, add 750 µL of Chloroform methanol mixed solution (2:1), vortex for 30 s. Add 2 steel beads, put them into the tissue grinder, and grind at 50Hz for 60s, repeated 2 times. Put on the ice for 40 min, add 190 µL H2O, vortex for 30 s, put on the ice for 10min. Centrifuged at 12000 rpm for 5min at room temperature and transfer 300 µL lower layer fluid into a new centrifuge tube. Then add 500 µL of Chloroform methanol mixed solution (2:1), vortex for 30 s. Centrifuged at 12000 rpm for 5min at room temperature and transfer 400µL lower layer fluid into the same centrifuge tube above. Samples were concentrated to dry in vacuum. Dissolve samples with 200µL isopropannol, and the supernatant was filtered through 0.22 µm membrane to obtain the prepared samples for LC-MS.
Chromatographic separation was accomplished with an ACQUITY UPLC® BEH C18 (100 × 2.1 mm, 1.7 µm, Waters) column maintained at 50°C. The temperature of the autosampler was 8°C. Gradient elution of analytes was carried out with acetonitrile:water = 60:40(0.1%formic acid + 10mM ammonium formate)(C) and isopropanol: acetonitrile = 90:10(0.1%formic acid + 10mM ammonium formate)(D) at a flow rate of 0.25 mL/min. Injection of 2 µL of each sample was done after equilibration. An increasing linear gradient of solvent C (v/v) was used as follows: 0–5 min, 70%-57% C; 5-5.1 min, 57%-50% C; 5.1–14 min, 50%-30% C; 14-14.1 min, 30% C; 14.1–21 min, 30%-1% C; 21-24min, 1% C; 24-24.1 min, 1%-70% C; 24.1–28 min, 70% C. The electrospray tandom mass spectrometry (ESI-MSn) experiments were used with the spray voltage of 3.5 and − 2.5 kV in positive and negative modes, respectively. Sheath gas and auxiliary gas were set at 30 and 10 arbitrary units, respectively. The capillary temperature was 325°C. The Orbitrap analyzer scanned over a mass range of m/z 150–2000 for full scan at a mass resolution of 35,000. Data dependent acquisition (DDA) MS/MS experiments were performed with HCD scan. The normalized collision energy was 30 eV. Dynamic exclusion was implemented to remove some unnecessary information in MS/MS spectra.
Targeted medium- and long-chain fatty acids quantitation. LC-MS analysis of medium- and long-chain fatty acids in HepG2-Ctrl and HepG-shURI cells was performed by Shanghai Sensichip Infotech Co. (Shanghai, China). Briefly, all the samples were mixed with 600 µL 50% acetonitrile, vortex for 1min, and centrifuged at 12000 rpm, 4℃ for 15 min. Then, 200ul of supernatant was added with 100 µL 200 mM 3-NPH and 100 µL 120 mM EDC (6% pyridine, 400 ng/mL acetic acid-D3) and was incubated at 40℃ for 1min. Samples were centrifuged at 12000 rpm, 4℃ for 15 min and their supernatants were transferred to tubes for LC-MS analysis. LC-MS data was acquired on AB SCIEX 5500 QQQ (Applied Biosystems, Foster City, CA, USA) mass spectrometer coupled with high-performance liquid chromatography (HPLC) system ACQUITY UPLC. (Waters, Milford, MA, USA). The column for chromatographic separation was a ACQUITY UPLC BEH Amide Column (2.1 x 100 mm, 1.7 µm). For determination of relative metabolite abundances, the total abundances were normalized to an internal standard (acetic acid-D3) and the weight for cell extracts.
Proteins LC-MS/MS Analysis. For in-gel tryptic digestion, gel pieces were destained in 50 mM NH4HCO3 in 50% acetonitrile. Gel pieces were dehydrated with 100 µL of 100% acetonitrile for 5 min, the liquid removed, and the gel pieces rehydrated in 10 mM dithiothreitol and incubated at 37°C for 60 min. Gel pieces were again dehydrated in 100% acetonitrile, liquid was removed and gel pieces were rehydrated with 55 mM iodoacetamide. Samples were incubated at room temperature, in the dark for 45 min. Gel pieces were washed with 50 mM NH4HCO3 and dehydrated with 100% acetonitrile. Gel pieces were rehydrated with 10 ng/µL trypsin resuspended in 50 mM NH4HCO3 on ice for 1 h. Excess liquid was removed and gel pieces were digested with trypsin at 37°C overnight. Peptides were extracted with 50% acetonitrile/5% formic acid, followed by 100% acetonitrile. Peptides were dried to completion and resuspended in 2% acetonitrile/0.1% formic acid.
LC-MS/MS Analysis
The tryptic peptides were dissolved in 0.1% formic acid (solvent A), directly loaded onto a home-made reversed-phase analytical column (15-cm length, 75 µm i.d.). The gradient was comprised of an increase from 6–23% solvent B (0.1% formic acid in 98% acetonitrile) over 16 min, 23–35% in 8 min and climbing to 80% in 3 min then holding at 80% for the last 3 min, all at a constant flow rate of 400 nl/min on an EASY-nLC 1000 UPLC system.
The peptides were subjected to NSI source followed by tandem mass spectrometry (MS/MS) in Q ExactiveTM Plus (Thermo) coupled online to the UPLC. The electrospray voltage applied was 2.0 kV. The m/z scan range was 350 to 1800 for full scan, and intact peptides were detected in the Orbitrap at a resolution of 70,000. Peptides were then selected for MS/MS using NCE setting as 28 and the fragments were detected in the Orbitrap at a resolution of 17,500. A data-dependent procedure that alternated between one MS scan followed by 20 MS/MS scans with 15.0s dynamic exclusion. Automatic gain control (AGC) was set at 5E4.
The resulting MS/MS data were processed using Proteome Discoverer 1.3. Trypsin/P (or other enzymes if any) was specified as cleavage enzyme allowing up to 2 missing cleavages. Mass error was set to 10 ppm for precursor ions and 0.02 Da for fragment ions. Carbamidomethyl on Cys were specified as fixed modification and oxidation on Met was specified as variable modification. Peptide confidence was set at high, and peptide ion score was set > 20.
DNA electrophoresis. 10× TAE buffer were diluted into 1× TAE for working concentration. 2% agarose gel was made using 1× TAE buffer with additional 10µl SolarRed (10000×, Solarbio, G5560). The separation of DNA fragments were achieved in 2% agarose gels in 1× TAE buffer using a DNA marker (Vazyme, DL5000) at 120V for 25 minutes.
Recombinant Myc-URI and His-TRIM28 production. The recombinant Myc-URI and His-TRIM28 were obtained from AtaGenix Laboratories Co. (Wuhan, China). Briefly, the URI and TRIM28 cDNA was obtained from the National Center of Biotechnology Information. The cDNA was cloned in a plasmidic expression vector (pET28b) harboring a histidine tag. After DNA sequence verification, the plasmid was transferred into Escherichia coli. The bacteria starter was obtained by incubation at 37°C for 4 h. Recombinant protein was obtained using a Ni-NTA Superflow column (Qiagen, Hilden, Germany).
Data collection. Transcriptome data were downloaded from the GEO database (https://www.ncbi.nlm.nih.gov/geo/), and the dataset of GSE96794, GSE143233 and GSE25097 was used in this study.
Statistical analyses. All the statistical analyses were performed using GraphPad Prism8 software (GraphPad Software, Inc.). Two tailed t-test and Mann-Whitney test were used to compare shURI vs. Ctrl groups. ANOVA models were used to compare continuous outcomes across multiple experimental groups. Kaplan- Meier analysis with log-rank tests was used to determine disease-free survival (DFS) and overall survival (OS). The data represent mean values of at least three independent experiments. Error bars represent mean ± SEM or median ± SEM, as appropriate. Statistical significance was set at p < 0.05.