Abstract
Hepatocellular carcinoma has a high incidence and poor prognosis. In this study, we investigated the value of T-cell-related genes in prognosis by single-cell sequencing data in hepatocellular carcinoma. Twelve cases of hepatocellular carcinoma single-cell sequencing were included in the study. The high dimensional weighted gene co-expression network analysis (hdWGCNA) was used to identify gene modules associated with CD4+ T cells, CD8+ T cells and exhausted T cells. Altered signaling pathway activity in exhausted T cells was uncovered by the AUCell algorithm. xCELL, TIMER, QUANTISEQ, CIBERSORT and CIBERSORT-abs were performed to explore immune cell infiltration. Immune checkpoint inhibitor genes and TIDE methods were used to predict immunotherapy response. Finally, immunohistochemistry and real-time PCR were used to verify gene expression. The hdWGCNA algorithm identified 40 genes strongly associated with CD4+ T cells, CD8+ T cells and exhausted T cells. Seven genes were finally selected for transcriptome data modeling. The results of the three independent datasets suggested that the model had strong prognostic value. Model genes were critical factors influencing CD4+ T cell and CD8+ T cell infiltration in patients. The efficacy of PD-1 immunotherapy was higher in patients belonging to the low-risk group. Alterations in signaling pathways’ activity within exhausted T cells were crucial factors contributing to the decline in immune function. Differential expression of seven genes in CD8+ T cells, CD4+ T cells and exhausted T cells were key targets for improving immunotherapy response in HCC.
Supplementary Information
The online version contains supplementary material available at 10.1038/s41598-025-88377-7.
Keywords: Hepatocellular carcinoma, Single cell, hdWGCNA, Immunotherapy
Subject terms: Cancer, Biomarkers, Gastroenterology, Oncology
Introduction
Hepatocellular carcinoma (HCC) is highly malignant and is the third leading cause of cancer-related deaths1. The incidence of virus-associated HCC remains high in Asia. However, the incidence of hepatocellular associated with alcoholic liver disease in Europe and the United States is increasing annually2,3. The extremely low rate of early diagnosis and poor prognosis of HCC make it a huge challenge that endangers human health4,5.
Chronic liver injury leads to the infiltration of a large number of immune cells, which ultimately leads to the occurrence of HCC6,7. The tumour microenvironment of HCC patients contains a high proportion of immune cells. The genome of cells in the tumor microenvironment and cell-cell interactions influence the drug resistance and progression of malignant cells8. T cells function as immunosurveillance, immunosuppression, and other complex functions in the tumor microenvironment9,10. CD4+ T cells enhance immune surveillance and can be involved in the killing of malignant cells11. CD8+ T cells are the primary cells that kill tumor cells12. However, CD4+ T and CD8+ T cells would lead to downregulation of immune function as they are regulated by the immunosuppressive components of the tumor microenvironment13. Under the continuous stimulation of tumor immune microenvironment, CD4+ T and CD8+ T cells gradually lost their immune surveillance function and became exhausted T cells. The increase of exhausted T cells would lead to immune evasion, which seriously affects the therapeutic effect and prognosis of the tumor patient14.
Single-cell RNA sequencing(scRNA-seq) technology has become the state-of-the-art approach for unravelling the heterogeneity and complexity of RNA transcripts within individual cells, as well as revealing the composition of different cell types and functions within highly organized tissues/organs/organisms15. Single-cell technology has made a huge leap forward in our understanding of the composition and interactions of the tumor immune microenvironment16. In HCC, the scRNA-seq technology motivates the exploration of genes characterizing T-cell subpopulations and hairy heterogeneous interactions17,18. High dimensional weighted gene co-expression network analysis (hdWGCNA), a comprehensive framework for analyzing co-expression networks in high-dimensional transcriptomics data such as single-cell and spatial RNA sequencing (RNA-seq), provides functions for network inference, gene module identification, gene enrichment analysis, statistical tests, and data visualization19. In ulcerative colitis, nonalcoholic fatty liver disease, breast cancer and meningiomas20–23, hdWGCNA has been shown to have excellent application value. However, hdWGCNA has not yet been used to explore HCC.
In this study, we explored the value of characterized genes among CD4+ T, CD8+ T and exhausted T cells in the immunotherapy and prognosis of patients with HCC by single-cell sequencing technology and high dimensional weighted gene co-expression network analysis (hdWGCNA).
Method
Data download
Single-cell sequencing data from the tumour cores of 12 patients with HCC were downloaded from GSE18990324 (https://www.ncbi.nlm.nih.gov/geo/). HCC transcriptome data include The Cancer Genome Atlas (TCGA) (https://portal.gdc.cancer.gov/), GSE1452025 (https://www.ncbi.nlm.nih.gov/geo/) and International Cancer Genome Consortium (ICGC) (https://dcc.icgc.org/).
Data processing
Read 10x Genomics single-cell sequencing data and created Seurat objects with the Seurat function. Application of the harmony package eliminated batch effects and integrated data from 12 single-cell sequencing samples. Cells were annotated by reference to previous literature reports26–29 and the “SingleR” method. Cells were annotated based on average expression of known cell lineage-specific marker genes of T cells (CD2, CD3E, CD3D, CD3G), B cells (CD79A, SLAMF7), tumor-associated fibroblasts (CAFs) (COL1A2, COL3A1, COL6A1), tumor associated macrophages (TAMs) (CD14, CD163, CD68, CSF1R), tumor associated endothelial cells (TAEs) (ENG, VWF, PECAM1) as well as epithelial cells (KRT18, KRT19, EPCAM). Subset function was used to extract T cell subpopulations and reclustered and re-annotated26–29. (CD4+ T cells: CD3D, CD4), (CD8+ T cells: CD3D, CD8A, GZMK), (Exhausted T cells: PDCD1, HAVCR2, LAG3), (Memory T cells: IL17RA), (Proliferative T cells: MKI67), (Regulatory cells: FOXP3).
HdWGCNA
The R package “hdWGCNA” was taken to construct a scale-free network at the single-cell level by high-dimensional weighted gene co-expression network analysis (hdWGCNA). The optimal soft threshold for the scale-free topological model ft was set to > 0.80 when the threshold was 8. The kME values were calculated over the entire single-cell dataset through the ModuleConnectivity function, with higher kME values indicating a higher probability of the gene being hub genes. The topN hub nodes of each module were extracted by GetHubGenes function. Select 20 genes with high kME in each module. The correlation between modules and cell types was shown through bubble plots.
Building and validating machine learning model
Multiple machine learning algorithms were used to construct stable and accurate prognostic models, with the TCGA data being the modeling group. GSE1452 and ICGC data were used as an external validation set to verify the stability of the model. A consensus model was constructed based on algorithms such as Random Survival Forest, Random Survival Forest, CoxBoost. A total of 101 algorithm combinations were performed to match the predictive model based on the leave-one-out cross validation (LOOCV) framework. The area under the receiver operator curves (ROC) were used to assess the judgmental ability of the model. The Kaplan-Meier curves were used to predict prognosis. Consistency index (C-index) was used to assess the accuracy of each model. Module genes of interest were included in machine learning models. Machine learning selected genes were modeled for prognosis by Least absolute shrinkage and selection operator (LASSO) and multifactor Cox regression analysis. Risk score = βgene A × expr gene A + βgene B × expr gene B+… + βgene N × expr gene N, expr is the mRNA expression of the pivotal gene and β is the corresponding regression coefficient in multivariate genetic Cox regression analysis.
Immune infiltration analysis
T cells are the main cell type that exercise the immune function of the body. In this study, we identified the main set of genes affecting T cell function based on HdWGCNA analysis. On this foundation, it was further analyzed whether the model genes reduced the abundance of immune cell infiltration. Variation in immune cell activity was evaluated by single-sample GSEA (ssGSEA). Xcell, timer, quantitative sequence, CIBERSORT, and CIBERSORT-abs methods were used to assess risk modeling and immune cell correlations.
Prediction of immunotherapy response
TIDE predicts patient immunotherapy response through immune dysfunction and rejection. Low TIDE scores indicate a lower likelihood of immune escape and immunotherapy efficacy in patients (http://tide.dfci.harvard.edu). Immune checkpoint inhibitor genes improve patient prognosis by enhancing immune function. Further analysis of the differences in expression of 47 immune checkpoint inhibitor genes between high and low risk groups.
Cellular communication analysis
Subpopulations of T cells interact and influence each other in a number of ways. Ligands and receptors are important mediators of intercellular information transfer. The expression level of genes is an important basis for predicting the pathways of interactions between cells. CellChatDB data collects the receptor and ligand information of cellular communication, which is widely used in the analysis of cellular communication. Extract expression matrix and classification information from harmony and apply “createCellChat” package to create cell chat object. identify overexpressed genes to screen ligand-receptor pairs and project the results to PPI network. Minimum cells = 3 to filter the communication relationship between low quality cells.
Signaling pathway analysis
AUCell calculated the fraction of each cell enriched in the Kyoto Encyclopedia of Genes and Genomes (KEGG) gene set based on the area under the curve. KEGG pathway gene set download from (Gene Set Enrichment Analysis) GSEA (https://www.gsea-msigdb.org/gsea/downloads.jsp). AUCell applies the area under the curve to calculate the enrichment of the input gene set in the cell. We used the AUCell algorithm to assess pathway activity in individual cells. Gene expression in CD4+ T cells, CD8+ T cells and exhausted T cells were extracted from Harmony objects. Comparison of changes in signaling pathway activity in CD4+ T cells, CD8+ T cells and exhausted T cells based on AUCell algorithm.
Real-time PCR and immunohistochemistry
Tissues from patients with HCC at the Renmin Hospital of Wuhan University were collected for validation of model gene expression. GAPDH expression was used as an endogenous gene. The cDNA was synthesized from total RNA using NovoScript® plus an all-in-one first strand cDNA synthesis kit (Novo protein). Human Protein Atlas(HPA) is a comprehensive resource of human protein information designed to provide a detailed description of human gene and protein expression patterns(https://www.proteinatlas.org/). The database integrates proteomic data from multiple tissue and cell types, including immunohistochemistry and high-throughput antibody preparation techniques. We used HPA database to analyze the differences in the expression of model genes in liver cancer tissues and adjacent tissues.
Statistical analysis
The Wilcoxon rank–sum test was used as a backup to analyze differences between two groups. Correlations between variables were explored using pearson or spearman coefficients. Continuous variables fitted to a normal distribution were compared using t-tests. COX regression analysis and LASSO regression analysis were used to screen for prognostic genes. The Kaplan-Meier method and log-rank test were used for prognostic analysis. All data analyses were conducted using R (4.1.2, https://www.r-project.org/) software. P value of < 0.05 was considered to indicate a statistically significant difference. (*P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001).
Results
Annotated T-cell subpopulations
Single-cell sequencing data from the core region of 12 cases of HCC were integrated using the “harmony” package. After rigorous quality control, a total of 41,184 cells and 22,552 different features were obtained. A total of 9 subpopulations of HCC cells in the core region were identified by “SingleR” and reference to previous literature. The T cell subpopulation was extracted by applying the subset function to obtain 34,070 T cells and 22,552 different features. After downscaling, clustering and re-annotation of the T cells, a total of 7 T cell subpopulations were obtained (Fig. 1A,B).
Fig. 1.
Identify new analysis modules based on the HdWGCNA algorithm. (A) MAP plot of 12 cases of single-cell sequencing data after dimensionality reduction, clustering and annotation. (B) Reduced-dimensional clustering and annotation for T cell subpopulations. (C) Heatmap of correlation between new modules. (D) Correlation analysis of new modules and T cell subsets.
hdWGCNA revealed that cluster 5 and 9 were the target modules
A high-dimensional weighted gene co-expression network analysis was applied to re-cluster the modules of T cell features. The threshold of the scale-free topological model ft exceeded 0.80 when the soft threshold was 8. HdWGCNA algorithm identified a total of 9 different T-cell related gene modules. The kME values for each module and the 10 genes with the largest kME values in each module were shown. The correlation heatmap demonstrated the kME values for each module and the correlation for each T cell subpopulation. Our study focused on modules 5 and 9, as they were found to have a negative correlation with exhausted T cells and a positive correlation with CD4+ T cells and CD8+ T cells (Fig. 1C,D).
Construction of prognostic signature
The first 20 genes in Modules 5 and 9 were selected for prognostic modeling. 40 genes went through the LOOCV framework of 101 predictive models and the C-index in the modeling and validation groups was calculated to select the optimal features. The 9 best gene combinations were selected by the machine learning algorithm. 9 model genes were analyzed by LASSO and multifactorial Cox regression analysis to establish a prognosis model. Finally, 7 genes were included in the risk model. The survival curves of TCGA, ICGC and GSE14520 datasets showed that the prognosis of the low-risk group was better than that of the high-risk group (Fig. 2D,F,H). Furthermore, the area under the ROC curve confirmed that the model had excellent prognostic ability (Fig. 2). The immunohistochemistry and real-time PCR showed that ZFP36 was low expressed in tumor tissues. However, CACYBP, CKS2, UBB, HSPA8, HSP90AA1 and CXCR4 were highly expressed in tumor tissues (Fig. 3).
Fig. 2.
Building and validating models. (A) Expression of model genes in single cells. (B) TCGA survival curves. 1-, 3- and 5-year time-dependent ROC curves from the TCGA database. (C) GSE14520 survival curves. 1-, 3- and 5-year time-dependent ROC curves from the GSE14520 database. (D) ICGC survival curves. 1-, 3- and 5-year time-dependent ROC curves from the ICGC database.
Fig. 3.
Gene expression analysis. (A–G) CACYBP, CKS2, UBB, HSPA8, HSP90AA1, ZFP36 and CXCR4 relative protein expression. (H) Real-time PCR analysis. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.
Effect of risk‑score on immune cell infiltration
Multiple algorithms were used to assess differences in immune infiltration in high and low risk groups. Macrophages, CD4+ T cells, and CD8+ T cells differed in infiltration between the high and low risk groups. Correlation analysis recommended that higher patient risk scores were associated with lower CD8+ T cell and CD4+ T cell infiltration (Fig. 4).
Fig. 4.
Immune cell infiltration analysis. (A) Spearman correlation analysis shows that risk scores are strongly related to tumor-infiltrating immune cells. (B–I) Correlation of risk models and immune cells based on XCELL, TIMER, QUANTISEQ, CIBERSORT and CIBERSORT-abs.
Immunotherapy analysis
Immunotherapy is increasingly valuable in patients with HCC. TIDE scores of patients showed higher scores in the high-risk group than in the low-risk group suggested that patients in the low-risk group had a higher benefit from immunotherapy. PD-1 was more suitable for patients in the low-risk group. Multiple immune checkpoint genes were differentially expressed in the two risk groups. Meanwhile, the results of correlation analysis confirmed that the model and immune checkpoint genes were highly correlated. The expression of model genes in exhausted T cells correlated extremely highly with PDCD1 (Fig. 5).
Fig. 5.
Immunotherapy analysis. (A,B) Correlation analysis and differential expression analysis between immune checkpoint genes and risk groups. (C) Prediction of immunotherapy for high-risk and low-risk groups. (D) Single-cell sequencing data reveals the correlation analysis results of model genes and PDCD1 expression in T cells. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.
Analysis of variation in the activity of exhausted T-cell signaling pathways
AUCell algorithm was based on gene expression to assess alterations in vital signaling pathways in exhausted T cells. Compared with CD4+ T cells and CD8+ T cells, T cell receptor signaling pathway, TGF beta signaling pathway, chemokine signaling pathway, WNT signaling pathway, ubiquitination signaling pathway, VEGF signaling pathway, MAPK signaling pathway and other pathways that were closely related to tumor development and progression were obviously altered in exhausted T cells (Fig. 6).
Fig. 6.
Single-cell sequencing analyzes the metabolic activity in different cell types. (A–C) The difference in activity of MAPK signaling pathway in different types of T cells. (D–F) The difference in activity of P53 signaling pathway in different types of T cells. (G) Differential analysis of CD4+ T cell and exhausted T cell signaling pathways. (H) Differential analysis of CD8+ T cell and exhausted T cell signaling pathways. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.
Analysis of cell-cell interactions
Weights and probabilities of intercellular signaling pathways in the tumor microenvironment of HCC were evaluated based on the Cellchat algorithm. Exhausted T cells hold a greater power in the communication between T cell subpopulations(Fig. 7A,B). In particular, the interactions between exhausted T cells and CD4+ T cells, CD8+ T cells and regulatory T cells were pronounced. MIF signaling pathway and IL16 signaling pathway were the main signaling pathways involved in the interactions between T cell subpopulations (Fig. 7).
Fig. 7.
Cellular communication analysis. (A) The number of interaction in cell-cell communication network. (B) The weight of interaction in cell-cell communication network. (C,D) Heatmap display of intercellular communication weights. (E,F) Cell-cell communication interaction in MIF and IL16 signaling pathway.
Discussion
HCC had a low rate of early diagnosis4. Patients with a few liver resections were faced with a high recurrence rate and a high rate of drug resistance after surgery30,31. It was crucial to explore innovative therapeutic modalities for HCC. The tumor microenvironment of HCC contained tumor cells and non-tumor cells. Inflammatory factors, cytokines, and chemokines secreted by cells in the tumor microenvironment function as essential components in the regulation of distant metastasis, immune escape, and drug resistance32,33. T cells serve as the main force in the regulation of immune function. Antigens activate precursor T cells to differentiate into effector T cells to perform immune functions34,35. However, rapid proliferation of tumor cells and long-term antigen exposure lead to a decrease in effector T cell activity and a loss of immune activity to exhausted T cells36,37. Exploring potential specific targets to activate exhausted T cells may be a new approach for tumor immunotherapy. Single-cell sequencing technology has stimulated the research of cellular subpopulations in the tumor microenvironment.
In this study, we firstly combined scRNA-seq technology with the hdWGCNA to explore the characteristics of T cell subsets in the HCC immune microenvironment. We found that the parts that were inversely related to exhausted T-cells and CD4+ T cells, CD8+ T-cells were considered as potential targets for reversing exhausted T cells. Through multiple machine learning algorithms, 7 genes (CACYBP, CKS2, UBB, HSPA8, HSP90AA1, ZFP36 and CXCR4) closely related to CD4+ T cells, CD8+ T cells and exhausted T cells were screened out. Further, the patients were grouped into high and low risk groups by model genes. The results of the TIDE score revealed that patients in the low-risk group were less likely to escape while receiving immunotherapy. This was associated with accelerated immune cell apoptosis, cyclin phosphorylation and ubiquitination in patients in the high-risk group. Model genes and immune checkpoint inhibitor genes had a high correlation. Model genes may be key targets for inhibiting T cell exhaustion.
CACYBP is a cell cycle-regulating calcineurin. It mainly acts with calcium-binding proteins to regulate the cytoskeleton, ubiquitination, and the cell cycle38,39. Lian et al. revealed that overexpression of CACYBP accelerated the phosphorylation process of P27Kip1, and knockdown of CACYBP resulted in decreased cell cycle protein expression and accelerated apoptosis40. Our study further revealed that CACYBP may be overexpressed in functional T cells accelerating the phosphorylation of immune function-related proteins leading to T cell exhaustion. CKS2 was highly expressed in HCC. High expression CKS2 promoted cancer cell proliferation41. HSPA8 was highly immunogenic and involved in autophagy and immune cell infiltration in high cell carcinoma42,43. The ubiquitin proteasome system was an essential pathway for the regulation of the cell cycle, apoptosis and immunomodulation. HSP90AA1 was a critical link in the regulation of ubiquitination in HCC44. ZFP36 was the key factor regulating cellular autophagy45. UBB were able to influence tumor heterogeneity and immune cell infiltration in HCC46. Patients with overexpressed CXCR4 had poor sorafenib efficacy. In contrast, CD8+ T cell infiltration was increased in the tumor microenvironment of HCC after blockade of the SDF-1α/ CXCR4 pathway47,48. CXCR4 may be a crucial gene contributing to T-cell exhaustion. The gene expression and metabolism of exhausted T cells were severely altered. TIGIT, LAG3, HAVCR2 and PDCD1 were the marker genes of exhausted T cells. In the exploration of variation in gene expression in exhausted T cells, it was found that the expression of model genes and marker genes in exhausted T cells were highly related. These results indicated that model gene expression variations contribute significantly to the functional evolution of exhausted T cells. Model genes may be potential targets for regulating the functional recovery of exhausted T cells.
Significant changes in tumor-related signaling pathways occurred in exhausted T cells. This study showed that WNT signaling pathway activity was higher in exhausted T-cells than in CD8+ T-cells. The active WNT signaling pathway may be a factor contributing to T cell depletion. This study also found that the VEGF signaling pathway or TGF beta signaling pathway was lower in both CD4+ T cells and CD8+ T cells than in exhausted T cells. Alterations in the activity of the VEGF signaling pathway or TGF beta signaling pathway in exhausted T cells leaded to change in immune function in the tumor microenvironment. The activity of ubiquitination signaling pathway or MAPK signaling pathway was elevated in exhausted T cells compared to CD4+ T cells and CD8+ T cells. The ubiquitination signaling pathway or MAPK signaling pathway may promote immune escape by increasing exhausted T-cells. This study also found that with the increase of exhausted T-cells, the MIF signaling pathway and IL16 signaling pathway involved in the interactions between T cell subpopulations showed active states. MIF contributed to anti-inflammatory, immune evasive, and immune tolerant phenotypes in both innate and adaptive immune cell types49. IL16 can significantly promote the proliferation of liver cancer cells50. Model genes may affect the dynamic changes of CD4+ T cells, CD8+ T cells and exhausted T cells, and change the composition of HCC immune microenvironment through these pathways. Reducing T cell exhaustion and reverting T cell immune function may become the critical for immunotherapy of HCC. We constructed prognostic modeling of T cell exhaustion-related gene modules in HCC patients by multiple machine learning algorithms and provided new targets for patient treatment and prognosis.
The findings of this study were innovative. However, there remained limitations. Firstly, this study only focused on T cells, other immune cells also played an irreplaceable role in tumor cellular immune elimination. Secondly, the findings lack validation at the animal level. Finally, the mechanism of T cell exhaustion needs to be further explored.
In conclusion, the hdWGCNA analysis of single-cell sequencing data from HCC identified gene modules related to CD8+ T cells, CD4+ T cells and exhausted T cells. These modular genes can predict patient prognosis, guiding immunotherapy and being a potential target for suppressing T-cell exhaustion. However, the results of the study required further experimental validation.
The primers used for RT‒PCR analysis:
CACYBP-F: CTTCTTCTCGCGGAGGCT.
CACYBP-R: CCTCTTCTAGATCTTTCTGTAGCGAT.
CKS2-F: TCTTCGCGCTCTCGTTTCAT.
CKS2-R: TGGGTAACATAACATGCCGGT.
UBB-F: GGCTATGAGGAATTTGGGGCT.
UBB-R: AGATCTGCATTTTGACCCCTCA.
HSPA8-F: CCTACACCCCAGCAACCAT.
HSPA8-R: GTGCTGGAAAACACCCACAC.
HSP90AA1-F: CCCAGAGTGCTGAATACCCG.
HSP90AA1-R: TAACAGGTGCCCTGCTTCTC.
ZFP36-F: ACTGCCATCTACGAGAGCCT.
ZFP36-R: ACTCAGTCCCTCCATGGTC.
CXCR4-F: TACCATGGAGGGGATCAGTGAAAA.
CXCR4-R: AACAAACGGCACCTCCTCC.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Abbreviations
- TCGA
The cancer genome atlas program
- GEO
Gene expression omnibus
- HCC
Hepatocellular carcinoma
- UMAP
Uniform manifold approximation and projection
- RT-qPCR
Real-time quantitative PCR
- ROC
Receiver operating characteristic
- ssGSEA
Single sample GSEA
- HdWGCNA
high-dimensional weighted gene co-expression network analysis
- LASSO
Least absolute shrinkage and selection operator
- KEGG
Kyoto encyclopedia of genes and genomes
- GSEA
Gene set enrichment analysis
Author contributions
Weixing Wang and Jia Yu contributed to the study’s inception and design. Rongqiang Liu, Jing Ye and Jianguo Wang equally contributed to the literature search, analysis and writing of the manuscript. Other authors contributed to the study design and study supervision. All authors approved the final version of the manuscript.
Data availability
All data was in the manuscript and can be obtained from the corresponding author.
Declarations
Competing interests
The authors declare no competing interests.
Footnotes
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
These authors contributed equally: Rongqiang Liu, Jing Ye and Jianguo Wang.
Contributor Information
Jia Yu, Email: yogaqq116@whu.edu.cn.
Weixing Wang, Email: wangwx@whu.edu.cn.
References
- 1.Nagaraju, G. P., Dariya, B., Kasa, P., Peela, S. & El-Rayes, B. F. Epigenetics in hepatocellular carcinoma. Semin Cancer Biol.86, 622–632. 10.1016/j.semcancer.2021.07.017 (2022). [DOI] [PubMed] [Google Scholar]
- 2.Ganesan, P. & Kulik, L. M. Hepatocellular Carcinoma: New Developments. Clin. Liver Dis.27, 85–102. 10.1016/j.cld.2022.08.004 (2023). [DOI] [PubMed] [Google Scholar]
- 3.Dimitroulis, D. et al. From diagnosis to treatment of hepatocellular carcinoma: an epidemic problem for both developed and developing world. World J. Gastroenterol.23, 5282–5294. 10.3748/wjg.v23.i29.5282 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kulik, L. & El-Serag, H. B. Epidemiology and management of Hepatocellular Carcinoma. Gastroenterology156, 477–491. .e471 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Anwanwan, D., Singh, S. K., Singh, S., Saikam, V. & Singh, R. Challenges in liver cancer and possible treatment approaches. Biochim. et Biophys. acta Reviews cancer. 1873, 188314. 10.1016/j.bbcan.2019.188314 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Makri, E., Goulas, A., Polyzos, S. A. & Epidemiology Pathogenesis, diagnosis and emerging treatment of nonalcoholic fatty liver disease. Arch. Med. Res.52, 25–37. 10.1016/j.arcmed.2020.11.010 (2021). [DOI] [PubMed] [Google Scholar]
- 7.Yuen, V. W. & Wong, C. C. Hypoxia-inducible factors and innate immunity in liver cancer. J. Clin. Investig.130, 5052–5062. 10.1172/jci137553 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sas, Z., Cendrowicz, E., Weinhäuser, I. & Rygiel, T. P. Tumor Microenvironment of Hepatocellular Carcinoma: challenges and opportunities for New Treatment options. Int. J. Mol. Sci.2310.3390/ijms23073778 (2022). [DOI] [PMC free article] [PubMed]
- 9.Bian, J. et al. T lymphocytes in hepatocellular carcinoma immune microenvironment: insights into human immunology and immunotherapy. Am. J. cancer Res.10, 4585–4606 (2020). [PMC free article] [PubMed] [Google Scholar]
- 10.Gao, S. et al. The pathogenesis of liver cancer and the therapeutic potential of bioactive substances. Front. Pharmacol.13, 1029601. 10.3389/fphar.2022.1029601 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Li, X. Y., Shen, Y., Zhang, L., Guo, X. & Wu, J. Understanding initiation and progression of hepatocellular carcinoma through single cell sequencing. Biochim. et Biophys. acta Reviews cancer. 1877, 188720. 10.1016/j.bbcan.2022.188720 (2022). [DOI] [PubMed] [Google Scholar]
- 12.Xu, C. et al. CD8(+) T cells mediate the antitumor activity of frankincense and myrrh in hepatocellular carcinoma. J. Translational Med.1610.1186/s12967-018-1508-5 (2018). [DOI] [PMC free article] [PubMed]
- 13.Yang, Y. et al. Analysis of single-cell RNAseq identifies transitional states of T cells associated with hepatocellular carcinoma. Clin. Translational Med.10, e133. 10.1002/ctm2.133 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Brunell, A. E., Lahesmaa, R., Autio, A. & Thotakura, A. K. Exhausted T cells hijacking the cancer-immunity cycle: assets and liabilities. Front. Immunol.14, 1151632. 10.3389/fimmu.2023.1151632 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jovic, D. et al. Single-cell RNA sequencing technologies and applications: a brief overview. Clin. Transl Med.12, e694. 10.1002/ctm2.694 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lei, Y. et al. Applications of single-cell sequencing in cancer research: progress and perspectives. J. Hematol. Oncol.14, 91. 10.1186/s13045-021-01105-2 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ma, L., Khatib, S., Craig, A. J. & Wang, X. W. Toward a liver cell atlas: understanding Liver Biology in Health and Disease at single-cell resolution. Semin. Liver Dis.41, 321–330. 10.1055/s-0041-1729970 (2021). [DOI] [PubMed] [Google Scholar]
- 18.Aliya, S., Lee, H., Alhammadi, M., Umapathi, R. & Huh, Y. S. An overview on single-cell technology for Hepatocellular Carcinoma diagnosis. Int. J. Mol. Sci.2310.3390/ijms23031402 (2022). [DOI] [PMC free article] [PubMed]
- 19.Morabito, S., Reese, F., Rahimzadeh, N., Miyoshi, E. & Swarup, V. hdWGCNA identifies co-expression networks in high-dimensional transcriptomics data. Cell. Rep. Methods. 3, 100498. 10.1016/j.crmeth.2023.100498 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Mo, S. et al. Single-cell dissection, hdWGCNA and deep learning reveal the role of oxidatively stressed plasma cells in ulcerative colitis. Acta Biochim. Biophys. Sin (Shanghai). 55, 1730–1739. 10.3724/abbs.2023237 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Li, R., Zhao, M., Miao, C., Shi, X. & Lu, J. Identification and validation of key biomarkers associated with macrophages in nonalcoholic fatty liver disease based on hdWGCNA and machine learning. Aging (Albany NY). 15, 15451–15472. 10.18632/aging.205374 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chen, Y. et al. Single-cell sequencing and bulk RNA data reveal the tumor microenvironment infiltration characteristics of disulfidptosis related genes in breast cancer. J. Cancer Res. Clin. Oncol.149, 12145–12164. 10.1007/s00432-023-05109-y (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zhang, X. Single-cell RNA sequencing identifies macrophage signatures correlated with clinical features and tumour microenvironment in meningiomas. IET Syst. Biol.17, 259–270. 10.1049/syb2.12074 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Roessler, S. et al. A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients. Cancer Res.70, 10202–10212. 10.1158/0008-5472.Can-10-2607 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ma, L. et al. Multiregional single-cell dissection of tumor and immune cells reveals stable lock-and-key features in liver cancer. Nat. Commun.13, 7533. 10.1038/s41467-022-35291-5 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Zhu, A. X. et al. Molecular correlates of clinical response and resistance to atezolizumab in combination with bevacizumab in advanced hepatocellular carcinoma. Nat. Med.28, 1599–1611. 10.1038/s41591-022-01868-2 (2022). [DOI] [PubMed] [Google Scholar]
- 27.Zheng, C. et al. Landscape of infiltrating T cells in Liver Cancer revealed by single-cell sequencing. Cell169, 1342–1356e1316. 10.1016/j.cell.2017.05.035 (2017). [DOI] [PubMed] [Google Scholar]
- 28.Xue, R. et al. Liver tumour immune microenvironment subtypes and neutrophil heterogeneity. Nature612, 141–147. 10.1038/s41586-022-05400-x (2022). [DOI] [PubMed] [Google Scholar]
- 29.Liu, Y. et al. Identification of a tumour immune barrier in the HCC microenvironment that determines the efficacy of immunotherapy. J. Hepatol.78, 770–782. 10.1016/j.jhep.2023.01.011 (2023). [DOI] [PubMed] [Google Scholar]
- 30.Yang, J. D. et al. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat. Rev. Gastroenterol. Hepatol.16, 589–604. 10.1038/s41575-019-0186-y (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Brown, Z. J. et al. Management of Hepatocellular Carcinoma: a review. JAMA Surg.158, 410–420. 10.1001/jamasurg.2022.7989 (2023). [DOI] [PubMed] [Google Scholar]
- 32.Lu, C. et al. Current perspectives on the immunosuppressive tumor microenvironment in hepatocellular carcinoma: challenges and opportunities. Mol. Cancer. 18, 130. 10.1186/s12943-019-1047-6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Feng, M., Pan, Y., Kong, R. & Shu, S. Therapy of primary Liver Cancer. Innov. (Cambridge (Mass. 1, 100032. 10.1016/j.xinn.2020.100032 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lu, F. et al. Cells derived from T cell-derived Induced Pluripotent Stem cells for the Treatment of Hepatocellular Carcinoma: potential and challenges. Front. Immunol.12, 690565. 10.3389/fimmu.2021.690565 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hao, L., Li, S. & Hu, X. New insights into T-cell exhaustion in liver cancer: from mechanism to therapy. J. Cancer Res. Clin. Oncol.149, 12543–12560. 10.1007/s00432-023-05083-5 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Saeidi, A. et al. T-Cell exhaustion in chronic infections: reversing the state of exhaustion and reinvigorating Optimal Protective Immune responses. Front. Immunol.910.3389/fimmu.2018.02569 (2018). [DOI] [PMC free article] [PubMed]
- 37.Fisicaro, P. et al. Targeting mitochondrial dysfunction can restore antiviral activity of exhausted HBV-specific CD8 T cells in chronic hepatitis B. Nat. Med.23, 327–336. 10.1038/nm.4275 (2017). [DOI] [PubMed] [Google Scholar]
- 38.Zhai, H. et al. Expression of calcyclin-binding protein/Siah-1 interacting protein in normal and malignant human tissues: an immunohistochemical survey. J. Histochem. Cytochemistry: Official J. Histochem. Soc.56, 765–772. 10.1369/jhc.2008.950519 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bhattacharya, S. et al. The modular structure of SIP facilitates its role in stabilizing multiprotein assemblies. Biochemistry44, 9462–9471. 10.1021/bi0502689 (2005). [DOI] [PubMed] [Google Scholar]
- 40.Lian, Y. F. et al. CACYBP enhances cytoplasmic Retention of P27(Kip1) to promote Hepatocellular Carcinoma Progression in the absence of RNF41 mediated degradation. Theranostics9, 8392–8408. 10.7150/thno.36838 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Li, Z. et al. EGFL7 promotes hepatocellular carcinoma cell proliferation and inhibits cell apoptosis through increasing CKS2 expression by activating Wnt/β-catenin signaling. J. Cell. Biochem.119, 10327–10337. 10.1002/jcb.27375 (2018). [DOI] [PubMed] [Google Scholar]
- 42.Xiang, X., You, X. M. & Li, L. Q. Expression of HSP90AA1/HSPA8 in hepatocellular carcinoma patients with depression. OncoTargets Therapy. 11, 3013–3023. 10.2147/ott.S159432 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wang, Y. et al. HBx-Induced HSPA8 stimulates HBV replication and suppresses ferroptosis to support Liver Cancer Progression. Cancer Res.83, 1048–1061. 10.1158/0008-5472.Can-22-3169 (2023). [DOI] [PubMed] [Google Scholar]
- 44.Shi, W. et al. FBXL6 governs c-MYC to promote hepatocellular carcinoma through ubiquitination and stabilization of HSP90AA1. Cell. Communication Signaling: CCS. 1810.1186/s12964-020-00604-y (2020). [DOI] [PMC free article] [PubMed]
- 45.Zhang, Z. et al. RNA-binding protein ZFP36/TTP protects against ferroptosis by regulating autophagy signaling pathway in hepatic stellate cells. Autophagy16, 1482–1505. 10.1080/15548627.2019.1687985 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Guo, C. et al. Deciphering the immune heterogeneity dominated by natural killer cells with prognostic and therapeutic implications in hepatocellular carcinoma. Comput. Biol. Med.158, 106872. 10.1016/j.compbiomed.2023.106872 (2023). [DOI] [PubMed] [Google Scholar]
- 47.Wang, Y. et al. CXCR4-guided liposomes regulating hypoxic and immunosuppressive microenvironment for sorafenib-resistant tumor treatment. Bioactive Mater.17, 147–161. 10.1016/j.bioactmat.2022.01.003 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ullah, T. R. The role of CXCR4 in multiple myeloma: Cells’ journey from bone marrow to beyond. J. bone Oncol.1710.1016/j.jbo.2019.100253 (2019). [DOI] [PMC free article] [PubMed]
- 49.Noe, J. T. & Mitchell, R. A. MIF-Dependent Control of Tumor Immunity. Front. Immunol.11, 609948. 10.3389/fimmu.2020.609948 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Takeba, Y. et al. Identification of interleukin-16 production on tumor aggravation in hepatocellular carcinoma by a proteomics approach. Tumour Biol.43, 309–325. 10.3233/TUB-211507 (2021). [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data was in the manuscript and can be obtained from the corresponding author.







