2.1 Patient Selection and Specimen Collection
A cohort of thirty-one HCC patients, who underwent interventional surgical procedures at the Third Affiliated Hospital of Sun Yat-sen University between 2017 and 2019, contributed tissue samples for this study. All participating patients received postoperative pathological validation to confirm HCC diagnosis. The study’s design and execution received the endorsement of the Ethics Committee at the Third Affiliated Hospital of Sun Yat-sen University (II2023-284-01).
2.2 Data Acquisition and Analytical Procedures
TCGA Database Analysis: We procured RNA-sequencing data (in FPKM format) alongside clinical records for 50 normal samples and 374 HCC patients from the Liver Hepatocellular Carcinoma (LIHC) project housed within The Cancer Genome Atlas (TCGA) database. ROC Curve Generation: Utilizing the R package “ROCR”, we constructed Receiver Operating Characteristic (ROC) curves. Differential Expression Analysis: The “limma” package in R facilitated the comparative analysis of gene expression data between groups demarcated by median NOL9 expression levels. Criteria for differentially expressed genes (DEGs) were set at |logFoldChange| > 0.5 and an adjusted p-value (adj.P.Val) < 0.05.
Functional Enrichment Analysis: We employed the “clusterProfiler” package in R to conduct Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on DEGs significantly associated with NOL9 expression. The top 30 terms, filtered by an adj.P.Val < 0.05 and counts > 2, were cataloged based on gene count.
Correlation Analysis: The interplay between NOL9 DNA methylation and its transcript expression in the TCGA-LIHC dataset was explored. Kaplan-Meier (KM) survival analyses, executed using the Survival and Survminer software packages, gauged the potential influence of NOL9 DNA methylation/expression on HCC patient prognosis.
Gene Expression Profiling Interactive Analysis (GEIPA): The mRNA expression interrelation between NOL9 and ZNF384 was scrutinized via the GEIPA database (http://gepia.cancer-pku.cn/).
Transcription Factor Binding Site Prediction: The JASPAR software facilitated predictions regarding transcription factor binding sites.
2.3 ssGSEA Enrichment Scores of Ribosome Biogenesis Gene Sets
To perform ssGSEA analysis, we first downloaded the gene expression data from the GEO database, including GSE83148 with 127 hepatitis B (HB) cases, GSE17548 with 10 hepatitis B virus (HBV)-related HCC cases, and GSE14520 with 160 HBV-related HCC cases. Subsequently, we computed the ssGSEA enrichment scores using the “GSEABase” and “limma” packages in R software.
For each sample, we normalized the gene expression data before calculating the ssGSEA enrichment scores using predefined gene sets (GOPB_ribosome_biogenesis and GOBP_regulation_of_ribosome_biogenesis) obtained from the MSigDB database. The ssGSEA enrichment scores were calculated based on a comparison of the gene expression levels of the gene set members with those of the background gene set.
To compare the differences between HBV and HBV-related HCC, we employed the Wilcoxon rank-sum test to assess the statistical significance of the ssGSEA enrichment scores between the two groups. A similar analysis was conducted using the gene expression data from the TCGA-LIHC dataset, comparing cancer tissues and adjacent non-tumor tissues in HCC.
All statistical analyses were performed using R software, and a P-value of < 0.05 was considered statistically significant.
2.4 Cell Culture and Proliferation Assays
Human HCC cell lines, namely HepG2, Huh7, and HEK293T, were sourced from the Hepatology Laboratory of the Third Affiliated Hospital of Sun Yat-sen University. Cells were cultured in DMEM supplemented with 10% FBS and 1% Penicillin/Streptomycin.
CCK-8 Assay: Cells (1,000/well) were seeded into 96-well plates. Post cell attachment (designated as 0h), the medium was replaced with CCK-8 containing media. After a 1-hour incubation at 37°C, absorbance was measured at 450nm using a microplate reader.
EdU Assay: Cells (2 × 10^4/mL) were seeded into 48-well plates and subjected to the EdU Cell Proliferation Kit as per the manufacturer's instructions. Post Hochest staining for 3 minutes, cellular imaging was conducted using fluorescence microscopy.
Colony Formation Assay: Cells (1,000/well) were seeded into 6-well plates. Following a culture period of 14–20 days, cells underwent fixation and staining. Colonies comprising more than 50 cells were enumerated.
2.5 Immunohistochemistry Analysis
Paraffin-embedded tissue sections were deparaffinized in an oven. Endogenous peroxidase activity was quenched using 3% hydrogen peroxide. Antigen retrieval was achieved using EDTA repair solution under high-pressure and high-temperature conditions. Tissue sections were then demarcated using a histochemical pen, approximately 0.5cm from the tissue radius. The primary antibody targeting NOL9 (1:200, # abs139241, absin, China) was applied in accordance with the manufacturer's guidelines. Following primary antibody incubation and subsequent washing, sections were treated with appropriate secondary antibodies. Microscopic imaging was conducted post-staining. Scoring Methodology: Ten distinct fields of view were randomly chosen for evaluation. Immunohistochemical staining intensity was categorized into four tiers: no staining (scored as 1), weak staining (scored as 2), moderate staining (scored as 3), and strong staining (scored as 4). The proportion of positively stained cells was graded as follows: <25% (scored as 1), 25%-50% (scored as 2), 50%-75% (scored as 3), and > 75% (scored as 4). The final score for each sample was computed as the product of staining intensity and the positive rate.
2.6 Western Blot Analysis
HCC tissues and cells were lysed using RIPA lysis buffer. Proteins were separated via SDS-PAGE and subsequently transferred onto polyvinylidene fluoride (PVDF) membranes. Post-blocking, membranes were probed overnight at 4°C with primary antibodies: NOL9 (1:1000, # ab140597, Abcam, UK), GAPDH (1:3000, # ab8245, Abcam, UK), beta-tubulin (1:5000, # A12289-50, ABclonal, China), Rb (1:1000, # 9309S, CST, USA), P-Rb (1:1000, # 8516T, CST, USA), CDK6 (1:1000, # PA5-27978, ThermoFisher, USA), Cyclin D1 (1:1000, # 2978T, CST, USA), and beta-catenin (1:1000, # 9562S, CST, USA). Secondary antibodies, either goat anti-rabbit IgG-HRP (1:5000, # E030120-01, SaiguoTech, Guangzhou, China) or goat anti-mouse IgG-HRP (1:5000, # E030110-01, SaiguoTech, Guangzhou, China), were applied for 1 hour at room temperature. Protein bands were visualized using the Excellent Chemiluminescent Substrate detection kit and quantified via grayscale analysis with ImageJ (version 1.8.0).
2.7 Quantitative Real-time PCR (qRT-PCR)
Total RNA was isolated from 1 × 10^6 cells using the RNA Quick Purification kit. Subsequently, 1µg of RNA was reverse transcribed into cDNA employing the Fast Reverse Transcription Kit (5 × Mix with gDNA Remover). qRT-PCR was executed using the Takara TB Green Premix Ex Taq II, adhering to a 3-step cycling protocol and manufacturer's guidelines. Primer sequences are provided in Supplementary Table 2.
2.8 Cell Transfection
Cells were seeded in 6-well plates and, upon reaching 60% confluence, were transfected with either target gene-specific siRNA or control siRNA (Ribobio, Guangzhou, China) using Lipofectamine 3000 (Invitrogen, MA, USA). Post 24-hour incubation, qRT-PCR verified target gene knockdown. At the 48-hour mark, Western blotting assessed knockdown efficiency. Only cultures exhibiting an 80% or greater reduction in target gene mRNA and protein expression, relative to controls, proceeded to subsequent experiments.
2.9 Lentivirus Package and Stable Cell Lines
For the creation of stable NOL9-overexpressing or knockdown cell lines, 293T cells were employed to package recombinant lentiviruses harboring the NOL9 gene or specific shRNAs against NOL9 (shNOL9-#1, shNOL9-#2). A lentiviral vector with a non-specific sequence served as a control. Target cells were incubated in a serum-rich medium supplemented with viral particles and polybrene. Puromycin (InvivoGen, France) was used to select stably transfected cells.
2.10 Methylation Specific PCR (MSP)
Genomic DNA was isolated from 5 × 10^6 cells utilizing the Promega Wizard Genomic DNA Purification Kit, adhering to the manufacturer's guidelines. Subsequently, bisulfite conversion of the DNA was executed using the EpiTect Bisulfite Kits (Qiagen). Quantitative real-time PCR assessed the methylation status of the NOL9 promoter regions. Primer sequences are delineated in Supplementary Table 2.
2.11 Flow Cytometry for Cell Cycle and Apoptosis Analysis
For cell cycle assessment, post 24-hour serum starvation, cells were collected, fixed in 70% ethanol overnight at 4°C, and stained with propidium iodide (PI) prior to flow cytometry. Apoptosis was evaluated by staining cells with PI and Annexin V-APC as per the manufacturer's protocol (KeyGEN, China).
2.12 Luciferase Reporter Assays
Cells were plated in 12-well dishes and, post 24 hours, subjected to plasmid transfection as indicated. 36 hours post-transfection, cellular lysates were analyzed for luciferase activity using the Dual-Luciferase Reporter Assay System (Promega, # E1910) following the manufacturer’s instructions
2.13 Chromatin Immunoprecipitation (CHIP)
Cells underwent cross-linking with 1% formaldehyde for 10 minutes, terminated with glycine. Lysates were sonicated to yield chromatin fragments, which were subsequently immunoprecipitated using the ZNF384 antibody (# ab176689, Abcam, UK). IgG served as a negative control. ChIP primer sequences can be found in Supplementary Table 2.
2.14 TOP/FOP-Flash Wnt Reporting Analysis
HCC cells were cultured in 12-well cell culture dishes for TOP/FOP-flash Wnt reporter assays. The cell transfection density was about 50%. We used the TOP/FOP-flash plasmid (Beyotime, Shanghai, China) to estimate the transcriptional activity of beta-catenin/TCF in cells. First, OE-NOL9/si-NOL9, TOP/FOP-flash and renin luciferase reporter plasmid (pRL-TK) were co-transfected into HCC cells. Forty-eight hours after transfection, we treated the cells using a dual luciferase assay kit (Promega, # E1910). Next, we used a microplate reader to estimate luciferase activity.
2.15 In vivo Tumorigenesis Assay
The tumorigenesis experiment utilized BALB/c nude mice aged 4 to 6 weeks. Each mouse was injected with 5 × 10^6 cells subcutaneously on the right hind side. Successful injections were achieved in three mice per group. Tumor size was determined using the formula: volume (cm^3) = (length x width^2)/2. Following the experiment, the mice were euthanized in a humane manner, and the tumors were surgically removed and weighed. The Animal Experimentation Ethics Committee at The Third Affiliated Hospital of Sun Yat-sen University approved the use of mice, and the Laboratory Animal Center staff provided appropriate care for all animals.
2.16 RNA-sequencing Analysis
Total RNA was extracted from both control HCC cells and NOL9-knockdown HCC cell lines using Trizol, with three biological replicates performed. The qualified RNA was then sent to Guangzhou Kidio Biotechnology Co., Ltd. (Guangzhou, China) for RNA sequencing (RNA-seq) on the Illumina sequencing platform. Subsequent data analysis was completed on Kidio’s cloud platform, OmicShare.
2.17 Statistical Analysis
Data sourced from TCGA and GEO database were integrated and processed using R software (R version 4.1.0) and GraphPad Prism (version 9.0). In vitro experiments were conducted a minimum of three times, with results presented as mean ± standard deviation (SD). Differences between two groups were evaluated using Student’s t-test, while the association between clinical attributes and NOL9 expression was determined using the Chi-squared test. For comparisons involving three or more groups, one-way or two-way ANOVA was employed for parametric data, and Kruskal-Wallis tests for nonparametric data. Pearson’s correlation assessed gene correlations. Hazard ratios (HR) were estimated with a 95% confidence interval (CI). A two-tailed P-value ≤ 0.05 was deemed statistically significant.