International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Globally, primary liver cancer ranks as the sixth most prevalent cancer and was the third leading cause of cancer-related deaths in 2022. Estimates indicate ~860,000 new diagnoses and ~750,000 fatalities worldwide per year (1). In total, 75–85% of all primary liver cancer cases are classified as hepatocellular carcinoma (HCC) (2). The principal treatment modalities currently available for HCC include surgical intervention, liver transplantation, chemotherapy and radiation therapy (3). However, ~70% of patients with HCC will experience a recurrence within 5 years despite notable therapeutic advancements in recent decades, which impacts their long-term survival outcomes and quality of life (4,5). For clinicians, recurrence-free survival (RFS) time serves as a critical parameter when formulating individualized therapeutic strategies for patients with HCC (6). Consequently, developing a robust predictive model for the probability of HCC recurrence represents a crucial clinical need.
Advances in large-scale sequencing technology have enabled the development of multiple prognostic models utilizing bulk RNA sequencing (RNA-seq) to predict HCC RFS. Gu et al (7) designed a six-long non-coding RNA signature for HCC RFS estimation and Wang et al (8) constructed a seven-gene model. However, despite the large amount of clinical information in the bulk dataset, limitations of this sequencing technology result in expression data that is the average of the expression in the bulk tissue rather than the expression at the cellular level. This introduces biological noise that may degrade model performance. Recently, single-cell (sc)RNA-seq has been increasingly applied in the study of HCC. Compared with bulk approaches, scRNA-seq distinguishes individual cell populations and captures their transcriptional activity at a cellular resolution, providing notably more information to researchers (9–11).
Researchers increasingly favor combined analytical frameworks incorporating both single-cell and bulk transcriptomic data, as they can take advantage of the respective strengths of both sequencing technologies. Zhou et al (12) identified prognostic ferroptosis biomarkers that could guide individualized therapeutic strategies for HCC by integrative analysis of transcriptomic data at different resolutions. Yu et al (13) characterized carcinoma-associated fibroblasts in HCC and established a clinically relevant risk assessment model. Furthermore, Wang et al (14) reported cellular heterogeneity and immune infiltration in HCC and uncovered how they contribute to the immunosuppressive microenvironment.
The present study combined single-cell and bulk transcriptomic datasets and implemented integrative analysis to establish an innovative predictive model for HCC recurrence outcomes.
The present study incorporated one scRNA-seq dataset and two bulk transcriptomic profiles from human HCC specimens. From the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/), the GSE242889 dataset was retrieved, which included single-cell transcriptomes from five patients with HCC with corresponding peritumoral controls (15). Among the five patients, three were men and two were women, with a median age of 55 years (range, 42–60 years). Bulk transcriptomic profiles with corresponding clinical data were acquired from two independent sources: The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) dataset (https://portal.gdc.cancer.gov/) and the GEO dataset, GSE76427 (16). For analytical purposes, the TCGA-LIHC cohort served as the training cohort, whereas the GSE76427 cohort was used for subsequent validation. Subsequently, data cleanup was performed. In the TCGA-HCC dataset, only the samples in which the histological type was HCC and the new event type was locoregional recurrence, extrahepatic recurrence or intrahepatic recurrence were included. In addition, for these two bulk RNA-seq datasets, cases missing either RFS data (follow-up time/status) or RNA-seq profiles data were excluded, even when clinical annotations were available. Finally, a total of 342 patients with HCC and 50 normal controls from the TCGA-LIHC cohort were included. The cohort comprised 230 men and 112 women, with a median age of 61 years (range, 16–90 years). Additionally, 108 patients with HCC from the GSE76427 cohort were analyzed, consisting of 86 men and 22 women, with a median age of 64 years (range, 14–93 years).
The single-cell transcriptome dataset, GSE242889, was merged using the Harmony algorithm (17). The samples were then analyzed as described in our previous study (18), with modifications. First, quality control measures excluded cells demonstrating either insufficient sequencing depth (<200 detected genes) or potential multiplets (>4,000 genes), along with those showing elevated mitochondrial transcript proportions (≥50%). Second, cellular clustering was performed using the FindClusters algorithm in Seurat (resolution parameter, 0.8) (19), which incorporates the first 17 principal components (PCs) as inputs.
After the cells in the GSE242889 dataset were classified and annotated, hepatocyte data were extracted for subsequent analysis. The FindMarkers algorithm in Seurat was employed to identify DEGs in hepatocytes derived from different origin samples (19). The statistical thresholds for DEGs were as follows: |log2fold-change (FC)|>0.5 and adjusted P<0.05. In the TCGA-LIHC dataset, differential expression analysis comparing tumor and normal samples was performed using the ‘limma’ R package (version 3.44.3) (20), and genes with |log2 FC|>1 and adjusted P<0.05 were regarded as DEGs. Subsequently, the associations between the gene expression profiles and HCC recurrence outcomes were assessed using the ‘survfit’ function in the ‘survival’ package (https://github.com/therneau/survival; version 3.2.7). The log-rank test result was interpreted with P<0.05 indicating statistical significance. The overlapping genes of these results were defined as the DEGs associated with RFS.
Survival-related DEGs were utilized as candidate biomarkers to construct a prognostic signature. Least absolute shrinkage and selection operator (LASSO) Cox penalized regression analysis was performed using the R package ‘glmnet’ (version 4.1) (21), and genes demonstrating non-zero coefficients were incorporated into the final risk score calculation. The risk score for each patient with HCC was calculated as follows: Risk score=Σ (βi × Expi), where βi represents the non-zero coefficients from LASSO Cox regression and Expi denotes the expression levels of selected genes. Subsequently, patients were dichotomized into high- and low-risk groups based on median risk score stratification, followed by Kaplan-Meier survival analysis.
To identify the underlying mechanisms that affect HCC, the DEGs between the high- and low-risk subgroups were obtained. Functional enrichment analysis was then performed using the ‘clusterProfiler’ package (version 4.7.1) to identify significantly enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) terms among these DEGs (22). Gene Set Enrichment Analysis (GSEA) was performed for the genes in the prognostic signature to further elucidate their biological functions. Additionally, the relative abundances of 22 immune cell subtypes within HCC samples were quantified using the CIBERSORT algorithm (23), and the immune-related score was calculated using the ‘estimate’ R package (version 1.0.13) (24).
To isolate independent recurrence predictors, univariate Cox regression was first applied, followed by multivariate analysis for 155 patients with HCC with complete clinical variables. These variables included age, sex, body mass index, tumor (T) stage, lymph node (N) stage, metastasis (M) stage, grade, pathological stage, α-fetoprotein level, Child-Pugh score and the risk score, which were classified using the appropriate guidelines (25–27).
Following prognostic signature development, external validation was performed using the independent GSE76427 cohort. The prognostic ability of the signature was first assessed using time-dependent receiver operating characteristic (ROC) curve evaluation at 6-, 12- and 24-month intervals. Patients were subsequently dichotomized into high- and low-risk subgroups using the median risk score as the cutoff threshold, followed by Kaplan-Meier survival curve analysis.
The present study was approved by the Ethics Committee of the Second Affiliated Hospital of Nanchang University (Nanchang, China; approval no. 2024–078). Tissue samples were collected at the Second Affiliated Hospital of Nanchang University (Nanchang, China) from February 2024 to June 2024. Patients that were diagnosed with HCC through pathological examination were included in the present study, while any patients with coexisting tumors were excluded from the study. A total of five HCC tissue samples and five adjacent tissue samples were obtained following documented informed consent from all study participants. Among the patients, there were four men and one woman, with a median age of 51 years (range, 38–65 years).
RNA was extracted and reverse transcribed from the HCC and normal tissues. For qPCR, the primers used are presented in Table I. As the immunoglobulin λ constant 2 (IGLC2) cDNA sequence was not available on the National Center for Biotechnology Information website, it was retrieved and aligned from Ensembl (http://ensemblgenomes.org/). The RT process proceeded with an initial primer annealing step at 25°C for 5 min, followed by the main cDNA synthesis reaction at 42°C for 30 min, which was completed by a brief 5-sec heat inactivation step at 85°C. The qPCR thermocycling conditions were as follows: Initial denaturation at 95°C for 30 sec. This was followed by 40 cycles of denaturation at 95°C for 15 sec and annealing/extension at 60°C for 30 sec. Finally, a melting curve analysis was conducted by heating the samples from 65 to 95°C. To ensure reliability, each experimental procedure was performed in triplicate, and the study was independently performed three times. Gene expression levels were quantified using RT-qPCR with a PrimeScript™ RT Master Mix (cat. no. RR036A; Takara Bio, Inc.) on an ABI 7500 Real-time PCR system detection instrument (Applied Biosystems; Thermo Fisher Scientific, Inc.), with relative expression determined using the 2−ΔΔCq method (28).
R software (https://www.r-project.org/; version 4.2.3) was utilized for statistical analysis. The data are presented as mean ± SD from experiments conducted more than three times. Differences between two groups were analyzed using a two-sided unpaired Student's t-test. The Kaplan-Meier method was employed to plot survival curves, and differences were assessed using the log-rank test. The Cox proportional hazards model was used to determine independent factors. P<0.05 was considered to indicate a statistically significant difference.
From the original 60,284 cells in the GSE242889 dataset, the quality control pipeline retained 57,219 cells that passed all quality thresholds for downstream processing (Fig. 1A-D). The single-cell data were subsequently segregated into 30 distinct clusters, from which 24,630 characteristic marker genes were detected (Fig. 1E; Table SI). The 10 most significant markers per cluster were visualized using a heatmap (Fig. 1F). The results revealed that clusters 0, 9, 10, 11, 17, 18, 22, 26 and 28, containing 20,317 cells, were hepatocytes; clusters 3, 6, 12, 19, 20 and 25, containing 10,299 cells, were monocytes; clusters 1, 7 and 21, containing 6,824 cells, were T cells; clusters 2 and 24, containing 8,693 cells, were macrophages; clusters 5, 16, 23 and 29, containing 4,667 cells, were B cells; clusters 8, 13 and 27, containing 3,749 cells, were endothelial cells; cluster 14, containing 1,299 cells, was tissue stem cells; cluster 15, containing 1,091 cells, was neutrophils; and cluster 24, containing 280 cells, was common myeloid progenitors (Fig. 1G).
Among the 20,317 hepatocytes in the GSE242889 dataset, 17,560 cells were from HCC samples, and 2,757 cells were from adjacent nontumor samples. A total of 2,320 DEGs were identified in these cellular populations, comprising 752 significantly upregulated genes and 1,568 significantly downregulated genes (Fig. 2A). In the TCGA-LIHC dataset, 5,586 DEGs were detected, with 2,637 demonstrating increased expression and 2,949 showing decreased expression levels (Fig. 2B). In addition, the analysis revealed that 4,971 genes were significantly associated with HCC recurrence outcomes. By intersecting these results, 53 RFS-related DEGs were identified that were incorporated into the prognostic signature (Fig. 2C).
Through regularization with LASSO Cox regression, a risk score prognostic signature based on six genes [cyclin-dependent kinase inhibitor 2A (CDKN2A), complement factor H-related 3 (CFHR3), cytochrome P450 family 2 subfamily C member 9 (CYP2C9), high mobility group box 2 (HMGB2), IGLC2 and Jupiter microtubule-associated homolog 1 (JPT1)] was constructed (Fig. 2D and E). The risk score was calculated as follows: Risk score=(0.009 × expression of CDKN2A)-(0.019 × expression of CFHR3)-(0.010 × expression of CYP2C9) + (0.019 × expression of HMGB2)-(0.012 × expression of IGLC2) + (0.048 × expression of JPT1). Time-dependent ROC analysis demonstrated robust predictive accuracy for RFS, with 6-, 12- and 24-month area under the curve (AUC) values of 0.747, 0.742 and 0.721, respectively (Fig. 3A). Furthermore, patients in the high-risk subgroup exhibited significantly worse survival outcomes than those in the low-risk subgroup (Fig. 3B).
Differential expression analysis identified 525 genes distinguishing high- and low-risk patients. KEGG pathway enrichment revealed predominant involvement in the ‘cell cycle’, ‘metabolism of xenobiotics by cytochrome P450’, ‘retinol metabolism’, ‘drug metabolism-cytochrome P450’, ‘bile secretion’ and ‘chemical carcinogenesis-DNA adducts’ (Fig. 4A). Furthermore, GO analysis was used to characterize the functional attributes of the identified DEGs systematically (Fig. 4C). Biological processes were predominantly associated with ‘nuclear division’, ‘chromosome segregation’ and ‘carboxylic acid biosynthetic process’, cellular components were enriched in ‘spindle’, ‘chromosomal region’ and ‘chromosome, centromeric region’, and molecular functions were significantly represented by ‘ATPase activity’, ‘iron ion binding’ and ‘monooxygenase activity’. The results of the GSEA analysis of the six genes are presented in Fig. S1. In addition, CIBERSORT analysis revealed that the high-risk cohort had significantly fewer naive and memory B cells, memory resting CD4+ T cells and resting mast cells than the low-risk cohort, indicating immunosuppression (Fig. 4B). Moreover, the stromal score, immune score and estimate score of the high-risk group were significantly lower than those of the low-risk group (Fig. 4D).
Univariate Cox regression revealed that T stage, pathological stage and the risk score were significant predictors of HCC recurrence (Fig. 5A). However, subsequent adjustment demonstrated that only the prognostic signature retained independent predictive value, suggesting that HCC management could be augmented by incorporating the molecular signature assessment for more precise recurrence prediction (Fig. 5B).
As an externally validated dataset, the GSE76427 dataset was used to independently assess robustness and clinical applicability of the prognostic signature. Time-dependent ROC analysis demonstrated strong discriminative ability at 6 months (AUC, 0.840), with a maintained predictive value at 12 months (AUC, 0.596) and 24 months (AUC, 0.557) (Fig. 3C). Consistent with these findings, Kaplan-Meier analysis revealed significantly worse outcomes in the high-risk patients compared with that in the low-risk patients (Fig. 3D), indicating the robust predictive capacity of the integrated single-cell and bulk transcriptome-derived signature.
The RT-qPCR results revealed that CDKN2A, HMGB2 and JPT1 were significantly upregulated in HCC tumor tissues compared with in adjacent normal tissues (Fig. 6A-C), whereas CFHR3, CYP2C9 and IGLC2 were significantly downregulated in tumor tissues, compared with that in adjacent normal tissues (Fig. 6D-F). Notably, these experimental findings corroborated those of the prognostic signature, reinforcing its validity and implying potential functional roles for the signature genes.
The recurrence of HCC severely impairs patient outcomes, representing a critical clinical challenge that demands resolution. Although several prognostic models for HCC RFS have been reported (8,29), there is a scarcity of models that combine single-cell and bulk RNA sequencing for this purpose. The present study first analyzed the scRNA-seq dataset GSE242889 and extracted hepatocyte data to identify DEGs. The DEGs associated with RFS from the bulk dataset TCGA-LIHC were simultaneously acquired. LASSO Cox penalized regression analysis was subsequently performed for these overlapping genes to establish the prognostic model. External validation was performed with the GSE76427 dataset, with additional experimental confirmation performed using RT-qPCR. The overall study design is presented in Fig. 7.
In the training dataset, a six-gene (CDKN2A, CFHR3, CYP2C9, HMGB2, IGLC2 and JPT1) prognostic signature was constructed to calculate risk scores. To further elucidate the biological functions of the six genes, GSEA was performed and the supporting literature was searched. A previous study suggested that, as an essential regulator of immune cell functionality, CDKN2A could influence the outcomes of patients with HCC through the modulation of tumor-associated macrophages (30). CFHR3 is a member of the complement factor H-related protein family (31). Wan et al (32) reported that knockdown of CFHR3 contributed to the invasion, proliferation and migration of HCC by promoting STAT3 protein phosphorylation. CYP2C9 mediates the breakdown of diverse carcinogens and pharmaceutical molecules and has also been reported as a biomarker for HCC diagnosis (33). HMGB2 is a highly conserved nuclear protein that is a member of the high mobility group protein family (34). Lu et al (35) reported that HMGB2 facilitates HCC progression by activating the zinc finger E-box-binding homeobox 1/vimentin axis. The results of the GSEA of the aforementioned four genes are presented in Fig. S1A-D. JPT1 is also known as HN1 and serves a critical role in HCC (36,37). Notably, in the present study, GSEA indicated that it affects multiple signaling pathways, such as fatty acid metabolism and bile acid metabolism. Research on IGLC2 in HCC has not been reported, to the best of our knowledge; however, the GSEA in the present study suggested that epithelial mesenchymal transition, inflammatory response and the interferon γ response could be modulated by IGLC2.
The results of the KEGG analysis revealed predominant enrichment of DEGs associated with the ‘cell cycle’, ‘metabolism of xenobiotics by cytochrome P450’, ‘drug metabolism-cytochrome P450’ and ‘chemical carcinogenesis-DNA adducts’. Several studies have suggested that the cell cycle is a critical regulator of tumor growth and that the microenvironment modulates HCC (38,39). Notably, in the present study, CIBERSORT analysis revealed that the high-risk cohort had fewer naive and memory B cells, memory resting CD4+ T cells and resting mast cells than the low-risk cohort, which are features of immunosuppression. Previous research has reported that CD4+ T cells serve a pivotal role in orchestrating the anti-HCC immune microenvironment, and their deficiency disrupts intercellular immunological crosstalk and facilitates malignant progression (40). Zhang et al (41) investigated the infiltration of B cells and their clinical significance in HCC and reported that high infiltration of naive and memory B cells was markedly associated with prolonged survival time. In addition, the stromal score, immune score and estimate score of the high-risk group in the presents study were lower than those of the low-risk group. These findings indicate that there is a significant difference in the immune microenvironment between the two groups, and that this difference may be an important factor influencing recurrence. Therefore, the aforementioned six genes could affect HCC RFS via these molecular pathways.
In the validation dataset, GSE76427, the AUCs for 6-, 12- and 24-month RFS were 0.840, 0.596 and 0.557, respectively. Kaplan-Meier analysis also revealed significantly worse outcomes in high-risk patients than in low-risk patients. In addition, Cox regression analysis revealed that only the prognostic signature retained independent predictive value. Notably, the expression levels of six genes were validated using RT-qPCR. The results indicate that the prognostic signature has good credibility and offers value as an independent predictor of the prognosis of patients with HCC. Unlike previously reported models for predicting RFS in patients with HCC, the prognostic signature in the present study was constructed by integrating scRNA-seq and bulk RNA-seq datasets. Using the prognostic signature, the individualized survival probability could be calculated for each patient according to the expression levels of the six genes. For patients at high risk of recurrence, the model could prompt physicians to pay greater attention to these patients and offer support for physicians in making clinical decisions.
In summary, the present study constructed a prognostic model for predicting RFS in patients with HCC via integrated analysis of scRNA-seq and bulk RNA-seq data. Unlike most studies, the present study combined the model with the clinical information of patients to evaluate whether the model is an independent prognostic factor affecting survival. Furthermore, experiments were used to assess the expression of the genes in the model using clinical samples. This may partially compensate for the shortcomings of other studies and greatly enhances the clinical applicability of the model in the present study. Moreover, the model could serve as a valuable reference tool for clinicians. However, there are several limitations of the present study. First, in the validation dataset, the time-dependent AUCs at 12 and 24 months (0.596 and 0.557, respectively) were relatively low, possibly because as the follow-up period increases, the proportion of patient deaths caused by non-tumor-related factors increases. Additionally, as the present study was retrospective, further prospective, large-scale trials are needed to validate the model. Finally, the present study did not perform an in-depth experimental exploration of how the six model genes affect HCC RFS, which needs to be further explored.
Not applicable.
The present study was funded by the National Natural Science Foundation of China (grant no. 91210050).
The data generated in the present study may be requested from the corresponding author.
LZ, WY and WL designed the study. WL, QH and ZL performed the bioinformatic and statistical analyses. HL and XG collected the stored clinical samples and acquired the data for analysis. WY, JM and QL performed the experiments. WY and WL wrote the original manuscript. LZ reviewed and revised the manuscript. LZ, WY and WL confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.
The present study was approved by the Ethics Committee of the Second Affiliated Hospital of Nanchang University (Nanchang, China; approval no. 2024-078). Documented written informed consent was obtained from all study participants.
Not applicable.
The authors declare that they have no competing interests.
|
DEGs |
differentially expressed genes |
|
FC |
fold-change |
|
GEO |
Gene Expression Omnibus |
|
GO |
Gene Ontology |
|
GSEA |
Gene Set Enrichment Analysis |
|
HCC |
hepatocellular carcinoma |
|
KEGG |
Kyoto Encyclopedia of Genes and Genomes |
|
LASSO |
least absolute shrinkage and selection operator |
|
RT-qPCR |
reverse transcription-quantitative PCR |
|
ROC |
receiver operating characteristic |
|
RFS |
recurrence-free survival |
|
RNA-seq |
RNA sequencing |
|
scRNA-seq |
single-cell RNA-seq |
|
TCGA |
The Cancer Genome Atlas |
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI | |
|
Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA and Bray F: Global burden of 5 major types of gastrointestinal cancer. Gastroenterology. 159:335–349.e15. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Daher S, Massarwa M, Benson AA and Khoury T: Current and future treatment of hepatocellular carcinoma: An updated comprehensive review. J Clin Transl Hepatol. 6:69–78. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Villanueva A: Hepatocellular carcinoma. N Engl J Med. 380:1450–1462. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Martínez-Chantar ML, Avila MA and Lu SC: Hepatocellular carcinoma: Updates in pathogenesis, detection and treatment. Cancers (Basel). 12:27292020. View Article : Google Scholar : PubMed/NCBI | |
|
Singal AG, Kudo M and Bruix J: Breakthroughs in hepatocellular carcinoma therapies. Clin Gastroenterol Hepatol. 21:2135–2149. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Gu JX, Zhang X, Miao RC, Xiang XH, Fu YN, Zhang JY, Liu C and Qu K: Six-long non-coding RNA signature predicts recurrence-free survival in hepatocellular carcinoma. World J Gastroenterol. 25:220–232. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang W, Wang L, Xie X, Yan Y, Li Y and Lu Q: A gene-based risk score model for predicting recurrence-free survival in patients with hepatocellular carcinoma. BMC Cancer. 21:62021. View Article : Google Scholar : PubMed/NCBI | |
|
Guo DZ, Zhang X, Zhang SQ, Zhang SY, Zhang XY, Yan JY, Dong SY, Zhu K, Yang XR, Fan J, et al: Single-cell tumor heterogeneity landscape of hepatocellular carcinoma: Unraveling the pro-metastatic subtype and its interaction loop with fibroblasts. Mol Cancer. 23:1572024. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Li R, Miao X, Zhou X, Wu B, Cao J, Wang C, Li S and Cai J: Integrated single cell analysis reveals an atlas of tumor associated macrophages in hepatocellular carcinoma. Inflammation. 47:2077–2093. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Wu L, Zhong Y, Zhou K, Hou Y, Wang Z, Zhang Z, Xie J, Wang C, Chen D, et al: Single-cell landscape of the ecosystem in early-relapse hepatocellular carcinoma. Cell. 184:404–421.e16. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Q, Tao C, Ge Y, Yuan J, Pan F, Lin X and Wang R: A novel single-cell model reveals ferroptosis-associated biomarkers for individualized therapy and prognostic prediction in hepatocellular carcinoma. BMC Biol. 22:1332024. View Article : Google Scholar : PubMed/NCBI | |
|
Yu L, Shen N, Shi Y, Shi X, Fu X, Li S, Zhu B, Yu W and Zhang Y: Characterization of cancer-related fibroblasts (CAF) in hepatocellular carcinoma and construction of CAF-based risk signature based on single-cell RNA-seq and bulk RNA-seq data. Front Immunol. 13:10097892022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang T, Dang N, Tang G, Li Z, Li X, Shi B, Xu Z, Li L, Yang X, Xu C and Ye K: Integrating bulk and single-cell RNA sequencing reveals cellular heterogeneity and immune infiltration in hepatocellular carcinoma. Mol Oncol. 16:2195–2213. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Li K, Zhang R, Wen F, Zhao Y, Meng F, Li Q, Hao A, Yang B, Lu Z, Cui Y and Zhou M: Single-cell dissection of the multicellular ecosystem and molecular features underlying microvascular invasion in HCC. Hepatology. 79:1293–1309. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Grinchuk OV, Yenamandra SP, Iyer R, Singh M, Lee HK, Lim KH, Chow PK and Kuznetsov VA: Tumor-adjacent tissue co-expression profile analysis reveals pro-oncogenic ribosomal gene signature for prognosis of resectable hepatocellular carcinoma. Mol Oncol. 12:89–113. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, Wei K, Baglaenko Y, Brenner M, Loh PR and Raychaudhuri S: Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods. 16:1289–1296. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Lai W, Xie R, Chen C, Lou W, Yang H, Deng L, Lu Q and Tang X: Integrated analysis of scRNA-seq and bulk RNA-seq identifies FBXO2 as a candidate biomarker associated with chemoresistance in HGSOC. Heliyon. 10:e284902024. View Article : Google Scholar : PubMed/NCBI | |
|
Butler A, Hoffman P, Smibert P, Papalexi E and Satija R: Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 36:411–420. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI | |
|
Friedman J, Hastie T and Tibshirani R: Regularization paths for generalized linear models via coordinate descent. J Stat Softw. 33:1–22. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M and Alizadeh AA: Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 12:453–457. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Yoshihara K, Shahmoradgoli M, Martínez E, Vegesna R, Kim H, Torres-Garcia W, Treviño V, Shen H, Laird PW, Levine DA, et al: Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 4:26122013. View Article : Google Scholar : PubMed/NCBI | |
|
Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, Meyer L, Gress DM, Byrd DR and Winchester DP: The eighth edition AJCC cancer staging manual: Continuing to build a bridge from a population-based to a more ‘personalized’ approach to cancer staging. CA Cancer J Clin. 67:93–99. 2017.PubMed/NCBI | |
|
Daniele B, Annunziata M, Barletta E, Tinessa V and Di Maio M: Cancer of the liver italian program (CLIP) score for staging hepatocellular carcinoma. Hepatol Res. 37 (Suppl 2):S206–S209. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Moris D, Martinino A, Schiltz S, Allen PJ, Barbas A, Sudan D, King L, Berg C, Kim C, Bashir M, et al: Advances in the treatment of hepatocellular carcinoma: An overview of the current and evolving therapeutic landscape for clinicians. CA Cancer J Clin. May 20–2025.(Epub ahead of print). PubMed/NCBI | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Kong J, Wang T, Shen S, Zhang Z, Yang X and Wang W: A genomic-clinical nomogram predicting recurrence-free survival for patients diagnosed with hepatocellular carcinoma. PeerJ. 7:e79422019. View Article : Google Scholar : PubMed/NCBI | |
|
Luo JP, Wang J and Huang JH: CDKN2A is a prognostic biomarker and correlated with immune infiltrates in hepatocellular carcinoma. Biosci Rep. 41:BSR202111032021. View Article : Google Scholar : PubMed/NCBI | |
|
Skerka C, Chen Q, Fremeaux-Bacchi V and Roumenina LT: Complement factor H related proteins (CFHRs). Mol Immunol. 56:170–180. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Wan Z, Li X, Luo X, Wang B, Zhou X and Chen A: The miR-590-3p/CFHR3/STAT3 signaling pathway promotes cell proliferation and metastasis in hepatocellular carcinoma. Aging (Albany NY). 14:5783–5799. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yu D, Green B, Marrone A, Guo Y, Kadlubar S, Lin D, Fuscoe J, Pogribny I and Ning B: Suppression of CYP2C9 by microRNA hsa-miR-128-3p in human liver cells and association with hepatocellular carcinoma. Sci Rep. 5:85342015. View Article : Google Scholar : PubMed/NCBI | |
|
Starkova T, Polyanichko A, Tomilin AN and Chikhirzhina E: Structure and functions of HMGB2 protein. Int J Mol Sci. 24:83342023. View Article : Google Scholar : PubMed/NCBI | |
|
Lu K, Zhao T, Yang L, Liu Y, Ruan X, Cui L and Zhang Y: HMGB2 upregulation promotes the progression of hepatocellular carcinoma cells through the activation of ZEB1/vimentin axis. J Gastrointest Oncol. 14:2178–2191. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Li M, Pang X, Xu H and Xiao L: CircSCMH1 accelerates sorafenib resistance in hepatocellular carcinoma by regulating HN1 expression via miR-485-5p. Mol Biotechnol. 67:304–316. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang R, Fu Y, Yao M, Cui X, Zhao Y, Lu X, Li Y, Lin Y and He S: The HN1/HMGB1 axis promotes the proliferation and metastasis of hepatocellular carcinoma and attenuates the chemosensitivity to oxaliplatin. FEBS J. 289:6400–6419. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Qu M, Zhang G, Qu H, Vu A, Wu R, Tsukamoto H, Jia Z, Huang W, Lenz HJ, Rich JN and Kay SA: Circadian regulator BMAL1::CLOCK promotes cell proliferation in hepatocellular carcinoma by controlling apoptosis and cell cycle. Proc Natl Acad Sci USA. 120:e22148291202023. View Article : Google Scholar : PubMed/NCBI | |
|
Wu D, Zhang C, Liao G, Leng K, Dong B, Yu Y, Tai H, Huang L, Luo F, Zhang B, et al: Targeting uridine-cytidine kinase 2 induced cell cycle arrest through dual mechanism and could improve the immune response of hepatocellular carcinoma. Cell Mol Biol Lett. 27:1052022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Meng L, Xu N, Chen H, Xiao Z, Lu D, Fan X, Xia L, Chen J, Zheng S, et al: Hepatocellular carcinoma-specific epigenetic checkpoints bidirectionally regulate the antitumor immunity of CD4 + T cells. Cell Mol Immunol. 21:1296–1308. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Ma L, Goswami S, Ma J, Zheng B, Duan M, Liu L, Zhang L, Shi J, Dong L, et al: Landscape of infiltrating B cells and their clinical significance in human hepatocellular carcinoma. Oncoimmunology. 8:e15713882019. View Article : Google Scholar : PubMed/NCBI |