Human HCC tumor specimens
Fresh resected tumors were collected with informed consent from patients who were enrolled at Eastern Hepatobiliary Surgery Hospital (Shanghai, China) without preoperative treatment. Human specimen collection was approved by the Ethics Committee of Eastern Hepatobiliary Surgery Hospital. Clinical information is available in Supplementary Table.1. Each tumor specimen was divided into three pieces for organoids culture, histology analysis and WGS analysis, separately.
Tumor dissociation
Fresh resected tissue was washed with PBS (Gibco), cut into pieces in collagenase D (Roche; final concentration was 4 mg/mL), and incubated at 37°C with rotation. Incubation time was ranged from 30 to 90 min. The solvent for collagenase D consisted of Advanced DMEM/F-12 (Gibco), GlutaMAX (Gibco), HEPES (Gibco), Penicillin-Streptomycin (Gibco), 100 µg/mL primocin (Invivogen), 0.1 mg/mL DNase I (Sigma-Aldrich) and 10µM Y-27632 (Sigma-Aldrich). After the tissue digestion, DMEM-HG (Gibco) medium containing 10% FBS (Gibco), primocin, Y-27632 and Penicillin-Streptomycin was added to inactivate collagenase D. Cell suspension was filtered using a 70 µm Nylon cell strainer and centrifugated for 5 min at 300–400×g. Cell pellet was washed for twice with PBS, and the number of living cells were detected using Trypan Blue.
HCC patients-derived organoids culture
Cell pellet from tumor tissue dissociation was resuspended with optimized hepatobiliary tumor organoids culture medium, consisting of Advanced DMEM/F-12, Penicillin-Streptomycin, GlutaMAX, HEPES, 100µg/mL Primocin, B-27 supplement (Gibco), 1.56 mM N-Acetylcysteine (Sigma-Aldrich), 50 ng/mL mouse recombinant EGF (Gibco), 100 ng/mL recombinant human FGF10 (Peprotech), 1ng/mL recombinant human FGF-basic (Peprotech), 25 ng/mL recombinant human HGF (Peprotech), 10 µM forskolin (Selleck), 500nM A8301 (Tocris), 10 µM Y-27632, 10mM Nicotinamide (Sigma-Aldrich), 3nM Dexamethasone (Sigma-Aldrich), 2% (vol/vol) Rspondin-conditioned medium, 5% (vol/vol) Noggin-conditioned medium and 30% (vol/vol) Wnt3a-conditioned medium. The same volume of cold Matrigel Basement Membrane Matrix (CORNING) was added, and each 50µL of organoids-matrigel was pipetted to form a single dome onto suspended cell plate. After solidification at 37°C for 45 min, culture medium was added, and the cell plate was transferred to humidified 37°C/5% CO2 incubator. Fresh medium was replaced each 3 days during organoids culture.
For cell passage, organoids were harvested into a 15mL tube, and centrifugated for 5 min at 1300–1500 rpm. Cell pellet was washed using cold PBS to remove the residual matrigel. Organoids were digested with TrypLE Express (Gibco) at 37°C, then the same volume of DMEM-HG medium (containing 10% FBS) was added to terminate the digestion. Organoids were centrifugated for 5 min at 1300–1500 rpm, and the cell pellet was suspended with culture medium. The same volume of matrigel was mixed and the cell suspension was seeded onto the suspended cell plate.
Cell viability assay
Cells were pre-seeded (1000 cells/well) and cultured in 384-well microplates (CORNING) for 24h. For organoids, 10 µL of matrigel was dispensed into 384-well microplates and allowed to polymerize before seeding. Then, chemicals were added to the culture medium at the indicated concentration. The same volume of solvent was used as the control. After 2–3 days of treatment, cell viability was assayed using CellTiter-Glo® 3D Reagent (Promega) in accordance with the manufacturer’s instructions.
Cell transfection
For lentiviral transfection in organoids, cells were digested with TrypLE Express and suspended in culture medium. The lentivirus and polybrene (Beyotime) were added into the cell suspension, and was centrifugated for 55 min at 500×g. Cell pellet was suspended with culture medium, mixed with the same volume of matrigel, and seeded onto the suspended cell plate. For lentiviral transfection in cell lines, 5×105 cells were pre-seeded onto 60mm dish for 24h, and incubated with DMEM-HG conditional medium containing lentivirus (1–2×107 TU) and polybrene. After incubation for 48h, cells were passage onto 100mm dish and selected with puromycin (Beyotime).
For siRNA transfection, 1×105 cells were pre-seeded onto 6-well plate with 2mL medium for 24h. Transfection mixture was prepared containing 100pmol siRNA, 5µL jetPRIME (Polyplus) and 200µL jetPRIME buffer (Polyplus), and incubated for 10 to 15 min at room temperature. The transfection mixture was added into the cell plate containing medium (2mL) directly, and was incubated at 37°C. Expression levels of the target genes were examined by qPCR and immunoblotting after 24-48h.
Quantitive real-time PCR (qPCR)
Total RNA was isolated by RNAsimple Kit (TIANGEN) as per manufactures protocol. Total RNA (1 µg) was reverse transcribed by ReverTra Ace qPCR RT Kit (TOYOBO) according to the manufacturer’s guidelines. qPCR was performed with gene specific primers at 95°C for 10 sec, 60°C for 60 sec (40 cycles) in 10 µL reaction mix containing 5 µL SYBR Green master mix (Qiagen), 1µL cDNA, 0.5 µL forward primer, 0.5 µL reverse primer and 3µL RNase-Free ddH2O using a Roche thermocycler. β-actin was used as an internal control. Quantification was performed with the ΔΔCt method. See Supplementary Materials for the primer sequences used for qPCR analysis.
Immunoblotting
Cell lysates were prepared by scraping (adherent cells) or centrifugation (suspended organoids) into lysis buffer (Beyotime, containing 1 mM PMSF). Cell lysates were incubated at 4°C for 1h with rotation and centrifuged at 12000 rpm, 4°C for 15 min. The supernatant was transferred into a new tube, and the protein concentration was examined using BCA kit (Beyotime). The antigens transferred to nitrocellulose filter membranes were probed with the primary antibodies against PKM2 (Cell Signaling Technology), DHFR (Cell Signaling Technology), LMNB1 (Proteintech), GAPDH (Proteintech) and β-actin (Sigma-Aldrich).
Hematoxylin-eosin (HE) and immunohistochemistry staining
For HE, tumor tissues or organoids were fixed with 4% paraformaldehyde, dehydrated and embedded into paraffin blocks. Sections (4–5 µm) were deparaffinized and stained with hematoxylin-eosin for histological analysis.
For Immunohistochemistry, sections were deparaffinized and incubated in 3% H2O2 for 20 min. Sections were dipped in boiling citrate buffer solution (0.01M, pH = 6.0) for 5 min and repeated 2 times. After cooling to room temperature, sections were blocked with 1%BSA/PBS for 1h and incubated with the primary antibody against PKM2 (Cell Signaling Technology) and DHFR (Cell Signaling Technology) at 4°C overnight. Sections were incubated with the secondary antibody for 1h and stained with DAB. Image collecting was performed using the Leica Biosystems. IHC scores of specimens were in accordance to the percentage of positive cells and staining intensity.
Tumor growth studies
For the tumor growth of organoids, 5 weeks of male NSG mice were purchased from the Model Organisms of Shanghai. We subcutaneously implanted 2×106 parental or Len-R organoids in the dorsal side of mice. Tumor volumes were monitored for a period of 42 days.
For the tumor growth of cell lines, 5 weeks of female nude mice were purchased from the Model Organisms of Shanghai. We subcutaneously implanted 1×106 WT or PKM2-shRNA expressing cells. Mice were subjected to lenvatinib (10 mg/kg, 3 times a week) or solvent after tumor volumes reached 100mm3. Tumor volumes and body weights were monitored 2–3 times a week.
LC-MS/MS based metabolomics
For the sample preparation, cells were harvested by centrifugation and washed by PBS. Cell pellet was quenched rapidly by liquid nitrogen. 2-chloro-1-phenylalanine (0.3mg/mL) dissolved in methanol as internal standard and methanol: acetonitrile: water (2/2/1, vol/vol/vol) were added to each sample. Samples were placed at -20°C for 2 min, grinded at 60 Hz for 2 min, and ultrasonicated at ambient temperature for 5 min. Samples were centrifuged at 12000 rpm, 4°C for 10 min. Supernatant was transferred into a new 1.5 mL tube, while the cell pellet was added the mixture of methanol: acetonitrile: water (2:2:1, vol/vol/vol). Samples were vortexed for 2 min, ultrasonicated at ambient temperature for 5 min, and centrifuged at 12000rpm, 4°C for 10 min. Supernatant was transferred into the above 1.5 mL tube and added the same volume of chloroform. Samples were placed at -20°C for 30 min and centrifuged at 12000 rpm, 4°C for 5 min. Supernatants were collected using crystal syringes, filtered through 0.22 µm microfilters and transferred to LC vials. The vials were stored at -80°C until LC-MS analysis.
For the data preprocessing and statistical analysis, the acquired LC-MS raw data were analyzed by the Compound Discoverer software. The positive and negative data were combined to import into R ropls package. Principle component analysis and orthogonal partial least-squares-discriminant analysis were carried out to visualize the metabolic alterations among experimental groups, after mean centering and Pareto variance scaling, respectively. In this study, the default 7 round cross validation was applied with 1/seventh of the samples being excluded from the mathematical model in each round, in order to guard against overfitting. The differential metabolites were selected on the basis of the combination of a statistically significant threshold of variable influence on projection values obtained from the orthogonal partial least-squares-discriminant analysis model and p-Values from a two-tailed Student's t-test on the normalized peak areas, where metabolites with variable influence on projection values larger than 1.0 and p-Values less than 0.05 were considered as differential metabolites.
Extraction of nuclear and cytoplasmic components
Organoids were harvested by centrifuging at 500×g for 5 min and washed by suspending the cell pellet with cold PBS. The nuclei and cytosolic fractions were collected using NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo scientific) according to the manufacturer’s instructions.
CO-IP
PKM2-overexpressed cells were lysed in lysis buffer (20mM Tris (pH = 7.5), 150mM NaCl, 1% Triton X-100, sodium pyrophosphate, β-glycerophosphate, EDTA, Na3VO4, leupeptin) and rotated at 4°C for 1h. Cell lysates were centrifuged at 12,000 rpm for 15 min at 4°C to remove the cell debris, and supernatants were incubated with PKM2 antibody (Cell Signaling Technology) at 4°C overnight with rotation. Equivalent mass of IgG was as a control. Cell lysates were incubated with Protein A/G Magnetic Beads (MedChemExpress) for 2 hours at 4°C with rotation. Beads were collected by performing magnetic separation, and the protein complex was eluted from the Protein A/G Magnetic Beads at 95°C for 5min. Immunoblotting was performed with primary antibody against PKM2 and SMAD4 (Cell Signaling Technology).
CUT&Tag
The assay was performed by using CUT&Tag Kit (Yeasen Biotechnology, Shanghai) in accordance with the manufacturer’s guidelines. Cells were incubated with the primary antibody against SMAD4. Equivalent mass of IgG was as a control. Goat Anti-Rabbit IgG H&L (abcam) was performed as the secondary antibody. PCR or qPCR analysis was performed with specific primers to SBE motif at DHFR promoter region. See Supplementary Materials for the primer sequences used for the amplification of SBE motif.
Luciferase reporter assay
5×104 cells were pre-seeded onto 24-well plates containing 500µL culture medium for 24h. 50µL jetPRIME buffer was prepared containing 0.5µg reporter plasmid (backbone was pGL3-Basic), 0.05µg internal control plasmid pSV-Renilla and 1.5µL jetPRIME. The mixture was incubated at room temperature for 10min and added into 24-well plates containing culture medium. Cell plate was incubated at 37°C for 48 h. FLuc and RLuc activities were determined using the Dual-Luciferase Assay System (Promega).
scRNA-Seq data analysis
Gene count matrices for each single-cell sample were generated by aligning reads to the hg38 genome using 10X Genomics CellRanger software v.4.0.0. The generated cell-by-gene unique molecular identifier count matrix was analyzed using the Seurat R package v.4.1.0. We kept cells with number of expressed genes between 200 and 7000. The cells were further filtered by removing cells with more than 15% mitochondrial genes. We retained genes with expression in at least 3 cells. The unique molecular identifier counts were then normalized for each cell by the total expression, multiplied by 10,000 and log-transformed. We used Seurat’s default method to identify highly variable genes and scale data for regressing out variation from unique molecular identifier and mitochondrial genes. The scaled data with variable genes were used to principal component analysis. The top 20 principal components were chosen for further analysis, including clustering to identify cell populations. UMAPs were calculated in the Seurat R package using the top 20 PCs and min_dist = 0.75. CCA was used to perform batch-effect correction for each untreated and lenvatinib treated paired samples. Intra-tumor heterogeneity was measured by calculating the average distance between the normalized expression profiles of each cell and all other cells in the same sample. The cell stemness of indicated samples and subgroups were characterized by calculating the average expression level of CSCs markers. We performed pseudo-time analysis in each integrated sample using Monocle 3 R package v.1.2.7.
Gene set enrichment analysis
SsGSEA was performed using GSVA in R v.1.42.0 using single-cell expression matrix with integrated normalized values. We applied hallmark gene sets and canonical pathways from KEGG gene sets of the C2 collection of Molecular Signatures Database (MSigDB) to each single cell to obtain enrichment score of each signature. Progeny python package was utilized to get activity score of canonical cancer relating pathways.
Differential expression analysis
DEseq2 was applied to identify the differentially expressed genes with a p.adjust value < 0.01 and a |log2FC| >1. For the single cell dataset, differential marker genes were identified using FindMarkers function with Wilcox test (only.pos = TRUE, min.pct = 0.25, log2FC.threshold = 0.25).
Whole genome sequencing analysis
The sequencing data was analyzed using a custom pipeline. In brief, data was preprocessed by Trim Galore v.0.6.6 which is a Perl wrapper around two tools: Cutadapt and FastQC. Sequencing reads were aligned to the human genome (hg38) using Burrows-Wheeler Aligner Mem v.0.7.17. Duplications were marked using Picard Tools v.2.22.2. Insertion–deletion realignment and base recalibration were achieved using GATK v.4.2.0. Somatic variants were detected using an ensemble approach with one variant caller: MuTect. Variant annotation was performed using SnpEff v.4.3 to obtain the final set of mutation calls. We used a two-step approach: (1) to reduce false-positive calls, the mutant variant site contains at least one allele that pass filters; (2) to get significant variant calls, the “HIGH” and “MODERATE” impact mutant variants were kept. Only variants with the following functional classification were considered in this study: nonsynonymous single nucleotide variants, stop gain mutations, and frameshift mutations.