Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Letters
Join Editorial Board Propose a Special Issue
Print ISSN: 1792-1074 Online ISSN: 1792-1082
Journal Cover
December-2025 Volume 30 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
December-2025 Volume 30 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML

  • Supplementary Files
    • Supplementary_Data1.pdf
    • Supplementary_Data2.pdf
Article Open Access

Prognostic value of M2 macrophage‑related genes and their importance in the immunotherapy response in hepatocellular carcinoma

  • Authors:
    • Aiping Wang
    • Xiaojing Li
    • Zi Wang
    • Lan Chen
  • View Affiliations / Copyright

    Affiliations: Department of Gastroenterology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, Hubei 430015, P.R. China, Department of Pathology, Wuhan Puren Hospital, Puren Hospital Affiliated of Wuhan University of Science and Technology, Wuhan, Hubei 430080, P.R. China, College of Medicine, Hubei Three Gorges Polytechnic, Yichang, Hubei 443000, P.R. China
    Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 540
    |
    Published online on: September 22, 2025
       https://doi.org/10.3892/ol.2025.15286
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Liver hepatocellular carcinoma (LIHC) is a major contributor to cancer‑associated mortality worldwide, with a poor prognosis due to late‑stage diagnosis and limited therapeutic options. M2 macrophages serve crucial roles in the tumor microenvironment (TME), contributing to tumor progression, immune evasion and resistance to therapy. However, the prognostic importance of M2 macrophage‑related genes in LIHC and their potential for predicting responses to immunotherapy remain underexplored. A bioinformatics analysis was performed using The Cancer Genome Atlas data to identify M2 macrophage‑related genes in LIHC. Differential expression and weighted gene co‑expression network analysis were used to identify key gene modules and a prognostic model was developed and validated using Kaplan‑Meier analysis and receiver operating characteristic curves. The immune therapy response was assessed using tracking of indels by decomposition and Submap analyses. Killer cell lectin like receptor B1 (KLRB1) was knocked down in HuH‑7 cells and overexpressed in HuH‑1 cells to evaluate its effect on tumor cells and macrophage regulation. The effect on human umbilical vein endothelial cell tube formation was also assessed. A total of two M2 macrophage‑related genes (KLRB1 and phosphatidylinositol‑5‑phosphate 4‑kinase type 2α) were identified as notable prognostic biomarkers for LIHC. The prognostic model demonstrated certain predictive power, with high‑risk patients exhibiting markedly worse overall survival. This model was validated in external datasets and associated with immune infiltration patterns. Furthermore, low‑risk patients were more likely to respond to immune checkpoint blockade therapy. The inhibition of KLRB1 enhanced LIHC cell activity and increased macrophage polarization from the M0 phenotype to the M2 phenotype by regulating LIHC cell secretions. In conclusion, M2 macrophage‑related genes are valuable prognostic biomarkers in LIHC. The prognostic model effectively stratifies patients by survival risk and predicts immunotherapy responses, thereby highlighting the potential for improved TME‑targeted therapies in LIHC. The mechanism of KLRB1 regulation in LIHC‑macrophage interactions and its impact on LIHC activities were also evaluated.

Introduction

Liver hepatocellular carcinoma (LIHC) is a major contributor to cancer-associated mortality worldwide, accounting for a notable proportion of global cancer mortalities with a 5-year-survival probability of ~18%; of these occurrences, ~90% are classified as HCC (1). Despite advancements in surgical techniques, liver transplantation and systemic therapies, the prognosis for patients with LIHC remains poor (2). This poor prognosis is primarily due to late-stage diagnoses, high recurrence rates and resistance to conventional treatments (3,4). Furthermore, the lack of reliable early detection biomarkers and effective therapeutic options exacerbates the challenges in managing LIHC (5). Consequently, there is an urgent need to identify novel molecular biomarkers that can serve as prognostic indicators and guide therapeutic decision-making, thus ultimately improving patient outcomes.

The tumor microenvironment (TME) is a crucial driver of cancer progression, with macrophages (particularly M2 macrophages) serving a central role in modulating immune responses (6). M2 macrophages promote tumor growth, metastasis and immune evasion by secreting cytokines such as IL-10 and TGF-β, which suppress the activation of cytotoxic T cells and natural killer (NK) cells (7,8). Research has demonstrated that M2 macrophages notably influence the progression of LIHC by promoting tumor growth and metastasis (9). M2 macrophages are generally considered to possess immunosuppressive properties, which enable them to create a tumor-supportive microenvironment. These cells achieve this effect by secreting a variety of cytokines and chemokines that facilitate tumor cell survival and dissemination (10,11). Previous studies have revealed that M2 macrophages contribute to the initiation and progression of liver cancer via interactions with hepatic stellate cells within the LIHC microenvironment (12,13). This crosstalk not only enhances the polarization of M2 macrophages but also promotes tumor immune evasion by inhibiting the activity of CD8+ T cells. Consequently, therapeutic strategies targeting M2 macrophages may offer novel approaches for the treatment of LIHC (11,14). Furthermore, M2 macrophages contribute to resistance to chemotherapy and immunotherapy, thus positioning these cells as critical targets for novel therapeutic strategies (15). The elucidation of the molecular mechanisms underlying M2 macrophage polarization in LIHC may provide valuable insights into improving prognostic assessment and enhancing treatment efficacy.

The present study aimed to explore the prognostic significance of M2 macrophage-associated genes in LIHC and create a predictive model for patient outcomes. Using data from The Cancer Genome Atlas (TCGA) database, key genes associated with M2 macrophage infiltration were identified using the weighted gene co-expression network analysis (WGCNA) algorithm. A risk model was constructed based on these genes and validated using survival analysis. Additionally, the potential of these genes as therapeutic targets was examined by assessing their relationships with immune checkpoint expression and responses to immunotherapy. The findings offer valuable insights into the role of M2 macrophages in LIHC and identify potential biomarkers for prognosis and immunotherapy strategies.

Materials and methods

Data collection and preprocessing

The primary dataset used for data analysis included the LIHC cohort from the TCGA database, which included gene expression profiles and clinical data for patients with LIHC. The raw RNA sequencing (RNA-seq) data were processed and normalized to the transcripts per million format to standardize the gene expression levels across all samples. In addition, samples with a reported survival time of 0 days were excluded from the analysis to ensure the integrity and reliability of the survival data. To validate the prognostic model, two independent external validation datasets were used. The first dataset (GSE76427) was retrieved from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/), and the second dataset (LIRI-JP) was obtained from the International Cancer Genome Consortium (ICGC) database (https://dcc.icgc.org/).

Evaluation of macrophage immune infiltration in LIHC

To assess macrophage immune infiltration in LIHC and normal tissue samples, six widely used immune infiltration estimation methods were applied: CIBERSORT (16), xCell (17), TIMER (18), QUANTISEQ (19), EPIC (20) and MCPCOUNTER (21). These algorithms, which leverage transcriptomic data, were used to quantify the relative levels of macrophage infiltration in both LIHC and normal samples. Additionally, the present study focused on the differential expression of M2 macrophages and performed comparative analyses of gene expression between patients with LIHC and normal controls.

Network construction, module preservation and correlation analysis

WGCNA was employed to identify gene modules associated with M2 macrophages (22). After excluding outlier samples, the top 25% most variable genes were selected for subsequent analysis. The optimal soft threshold for constructing the network was determined by evaluating the scale-free topology model fit. A power value that resulted in a fit close to 0.9 was chosen. The gene co-expression network was subsequently constructed and modules were identified based on hierarchical clustering. Preservation analysis was performed to assess the reproducibility of the identified modules. The modules were categorized as being strongly preserved (Z >10), weakly preserved (2<Z<10) or not preserved (Z<2). Modules with poor preservation were excluded from further analysis. Finally, the correlation between M2 macrophage expression levels and the identified gene modules was assessed using various immune infiltration methods. The modules with the greatest correlations with M2 macrophages were selected for further modelling.

Prognostic gene selection and M2 macrophages-related model development

Univariate Cox regression analysis was performed on 292 genes to identify those associated with prognosis in LIHC. Genes with P-values <0.05 were selected for further investigation. To refine this selection, Least Absolute Shrinkage and Selection Operator (LASSO) regression was performed with 10-fold cross-validation to identify the optimal regularization parameter (λ). A λ-value that optimized the model fit while minimizing overfitting was chosen. Multivariate Cox regression was subsequently performed to evaluate the independent prognostic value of the selected genes. A prognostic risk score model was developed based on the expression levels of these genes, which were weighted by their respective regression coefficients. The risk score formula was calculated by multiplying the expression level of each gene by its corresponding coefficient. To assess the model's predictive accuracy, Kaplan-Meier survival analysis and time-dependent receiver operating characteristic (ROC) curves were used to evaluate overall survival (OS) and model performance in both the training cohort (GSE76427) and independent validation cohorts (LIRI-JP).

Functional enrichment analysis

To examine the differential activation of biological pathways between the two risk groups, gene set variation analysis (GSVA) was performed on the hallmark gene sets (23). GSVA scores were computed for each gene set and differential expression analysis was performed to identify the pathways with significant differential activation between the two groups. In addition, Gene Ontology (GO) analysis was performed to identify biological processes and cellular components that were differentially enriched in the two risk groups. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis with the package ‘clusterprofiler’ of R studio (24) was also performed to examine differences in signaling pathway activation between the groups. For all the analyses, statistical significance was assessed using the Benjamini-Hochberg method to correct for multiple comparisons.

Immune infiltration, tumor mutational burden (TMB) and immune checkpoint analysis

To assess the relationships between immune characteristics and risk stratification, the ESTIMATE algorithm (ESTIMATE: Home) was first applied to quantify the scores for each patient with LIHC. Pearson correlation analyses were subsequently performed to examine the associations between these scores and the prognostic risk score. The TMB was also evaluated for each patient and a correlation analysis was performed with the risk score to determine whether the TMB was associated with the risk profile. To further explore immune responses, we employed the ‘limma’ package of R studio to conduct differential expression analysis was performed on common inhibitory and stimulatory immune checkpoint genes with the threshold of |log2 fold change|>1.5 and P<0.05. The expression of these genes was compared between the two risk groups to investigate their potential roles in immune evasion and therapeutic resistance. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm was employed to assess the immune escape potential in both risk groups (25). Finally, the Submap algorithm was applied to predict the likelihood of patient response to immune checkpoint blockade (ICB) therapy based on the gene expression signatures (26).

Single-cell (sc)RNA-seq based on the tumor immune single cell Hub 2 (TISCH2) database

The killer cell lectin like receptor B1 (KLRB1), with the smallest P-value in the multivariate Cox regression, was analyzed across multiple single-cell datasets to investigate its role in the TME. Using the TISCH2 database (27), the expression of KLRB1 was assessed in six independent scRNA-seq datasets (http://tisch.compbio.cn/home/). The expression levels of KLRB1 were compared across various immune cell types, including CD8+ T cells, CD8+ exhausted T cells (Tex) and NK cells. Ligand-receptor interactions were also examined between different cell populations using a heatmap to visualize the strength of these interactions. The interactions were further represented as a network diagram to illustrate the intensity or activity of ligand-receptor signaling, specifically between CD8+ T cells and other cell types in the TME.

Pan-cancer expression and prognostic analysis of KLRB1

To investigate the potential role of KLRB1 in cancer biology, a pan-cancer analysis was performed using data from TCGA. First, KLRB1 expression across various cancer types (including LIHC) was assessed to identify significant differences in expression. The prognostic value of KLRB1 was subsequently examined by analyzing its expression in relation to clinical outcomes, including the disease-free interval (DFI), disease-specific survival (DSS), OS and platinum-free interval (PFI) parameters across various cancer types. A forest plot analysis was performed to further investigate the relationship between KLRB1 expression and OS across various cancers. Finally, the samples were categorized into high- and low-KLRB1 groups and differential expression analysis was performed. Gene Set Enrichment Analysis (GSEA) was subsequently used to identify enriched biological pathways in the high- and low-expression groups, thus providing insights into the potential mechanisms through which KLRB1 influences tumor progression.

Cell culture

The HuH-1 (CL-0811), HuH-7 (CL-0120), and THP-1 (CL-0233) cell lines were purchased from Procell Life Science & Technology Co., Ltd. The HuH-1 and HuH-7 cells were cultured in Dulbecco's Modified Eagle's Medium supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (all from GIBCO; Thermo Fisher Scientific, Inc.). The THP-1 cells were cultured in RPMI 1640 medium supplemented with 10% FBS and 1% penicillin/streptomycin (all from GIBCO; Thermo Fisher Scientific, Inc.). Cultures were maintained in a humidified incubator at 37°C with a 5% CO2 atmosphere to ensure optimal growth conditions.

Construction of KLRB1 knockdown and overexpression cell lines

A lentivirus-based short hairpin RNA (shRNA) strategy was employed using three distinct shRNA sequences to downregulate KLRB1 expression in HuH-7 cells. These shRNA sequences were cloned into the lentiviral vector pLKO.1. Simultaneously, full-length KLRB1 complementary DNA (cDNA) was synthesized by PCR using total RNA extracted from HuH-1 cells and cloned into the lentiviral overexpression vector pLV-CMV-MCS-PGK-Puro to induce KLRB1 overexpression in HuH-1 cells. Both vectors were obtained from Addgene (Beijing Zhongyuan Co.). For lentivirus production, 10 µg of pLV-CMV-MCS-PGK-Puro was co-transfected with 7.5 µg of packaging plasmid (pMDLg/pRRE; Addgene; Beijing Zhongyuan Co.), 2.5 µg of envelope plasmid (VSV-G; Addgene; Beijing Zhongyuan Co.) and 15 µg of a helper plasmid (pRSV-Rev; Addgene; Beijing Zhongyuan Co.) using the calcium phosphate transfection method as part of a 3rd generation lentiviral system. Transfection was performed at 37°C for 48 h. Lentiviral particles were generated by co-transfecting 293T cells (Procell Life Science & Technology Co., Ltd.) with a packaging plasmid mix (REV: VSVG: PMDL=2:3:5) using TurboFect transfection reagent (cat. no. R0532; Fermentas; Thermo Fisher Scientific, Inc.), with virus supernatant harvested 36–48 h post-transfection and filtered for use in infection. HuH-7 and HuH-1 cells were seeded in 6-well plates until reaching 60–70% confluency before infection with lentiviral particles at a multiplicity of infection of 10, supplemented with poly-L-lysine (8 µg/ml) to enhance transduction. Following 12–16 h of co-culture with the virus, the medium was replaced with fresh culture medium. After 48 h, stable transductants were selected using 2 µg/ml puromycin (the original concentration was 1 mg/ml) to confirm successful transduction. All procedures were conducted at 37°C with 5% CO2. The sequences utilized for KLRB1 knockdown and overexpression are shown in Table SI.

RNA extraction and reverse transcription quantitative PCR (RT-qPCR)

According to the manufacturer's protocol, total RNA was extracted from tissue samples using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Subsequently, a cDNA synthesis kit (cat. no. R212-01; Vazyme Biotech Co., Ltd.) was used to convert the RNA into cDNA. The qPCR experiments were conducted using SYBR Green qRT-PCR Master Mix (Vazyme Biotech Co., Ltd.), with GAPDH serving as the internal control for normalization and the 2−ΔΔCq method (28) was used to analyze the difference. The thermocycling protocol consisted of an initial pre-denaturation step at 95°C for 5 min, followed by 40 cycles of denaturation at 95°C for 10 sec, annealing at 60°C for 20 sec and extension at 72°C for 20 sec. The primer sequences used are listed in Table SII.

Cell proliferation assay

Cell proliferation was assessed using a Cell Counting Kit-8 (CCK-8; Beijing Solarbio Science & Technology Co., Ltd.) according to the manufacturer's instructions. A total of 2,500 cells were seeded into 96-well plates and cultured for 3 days. A total of 10 µl of CCK-8 solution was added to each well for 1 h before measurement of the absorbance at 450 nm at room temperature.

Transwell migration assay

A Transwell chamber (pore size, 8 µm; Corning, Inc.) was used to assess the migratory ability of the cells. Target cells (typically 1×105 cells per well) were seeded into the upper chamber of the Transwell system, with serum-free medium used as the inducer. The lower chamber contained complete culture medium supplemented with 10% serum (GIBCO; Thermo Fisher Scientific, Inc.) as a chemoattractant to guide cell migration. The cells were incubated at 37°C in a 5% CO2 incubator for 24 to 48 h, thus allowing the cells to migrate through the membrane into the lower chamber. After incubation, non-migrating cells in the upper chamber were gently removed with a cotton swab. The cells in the lower chamber were subsequently washed with PBS, fixed with methanol at room temperature for 30 min and stained with 0.1% crystal violet solution at room temperature for 10 min. Finally, the stained cells in the lower chamber were observed and counted under an inverted microscope to assess the migratory ability of the cells through the software of Image J (version 1.8.0_345; National Institutes of Health).

Annexin V/propidium iodide (PI) staining assay

To assess cell apoptosis, the Annexin V-FITC/PI (BioLegend, Inc.) dual-staining method was used. First, sh-KLRB1-2 and OE-KLRB1 cells were seeded into culture dishes and cultured until they reached 70–80% confluence. After treatment, the cells were collected and washed twice with PBS to remove residual culture medium. The cells were then resuspended in 1× binding buffer and stained with annexin V-FITC and PI solution. The mixture was gently vortexed and incubated at room temperature in the dark for 15 min. Annexin V-FITC binds to phosphatidylserine exposed on the surface of the cell membrane, thereby marking early apoptotic cells, whereas PI stains the nuclei of late apoptotic or necrotic cells with compromised membrane integrity. After staining, apoptosis was analyzed by flow cytometry (FACSAria III; BD Biosciences) and evaluation using FlowJo (version 10.8.1). The dual fluorescence signals from annexin V-FITC and PI allowed for the distinction between healthy cells, early apoptotic cells, late apoptotic cells and necrotic cells, thereby providing a comprehensive assessment of the apoptotic status of the cells.

Cell tube formation assay

Human umbilical vein endothelial cells (HUVECs) (PUMC-HUVEC-T1; cat. no. CL-0675) were obtained from Procell Life Science & Technology Co., Ltd. The third to the fifth passage of HUVECs in the logarithmic growth phase were used for the subsequent experiments. The cell tube formation assay involved seeding HUVECs onto a solidified layer of Matrigel® and incubation for 4 to 6 h at 37°C to allow tube formation in a 96-well plate. After the cells were incubated with the coculture supernatant of HuH-7 or HuH-1 cells for 8 h at 37°C, tube-like structures formed. After the incubation period, the formation of tube-like structures was examined under a fluorescence microscope. The degree of tube formation was quantified by assessing parameters such as the number of branches and total branching length through Image J (version 1.8.0_345).

qPCR detection of macrophage cell polarization

To evaluate the polarization status of M0 cells, the expression levels of polarization marker genes were assessed using qPCR. The THP-1 cells were cultured in RPMI 1640 medium supplemented with 10% FBS and 1% penicillin/streptomycin (all from GIBCO; Thermo Fisher Scientific, Inc.). Cultures were maintained in a humidified incubator at 37°C with a 5% CO2 atmosphere to ensure optimal growth conditions. THP-1 cells were first treated with 100 nM phorbol 12-myristate 13-acetate for 48 h to induce differentiation into M0 cells. The culture supernatants from sh-KLRB1-2 and OE-KLRB1 cells were subsequently added to the M0 cells for 24 h at 37°C, after which the gene expression of the M1 and M2 markers was measured. According to manufacturer's protocol, total RNA was extracted from the cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and reverse transcription was performed to generate cDNA according to the manufacturer's instructions using a reverse transcription kit (cat. no. R212-01; Vazyme Biotech Co., Ltd.). SYBR Green fluorescence dye (Vazyme Biotech Co., Ltd.) was used for qPCR. Specifically, the first stage involved pre-denaturation at 95°C for 5 min, for one cycle. Then the second stage began, which involved denaturation at 95°C for 10 sec, annealing at 60°C for 20 sec and extension at 72°C for 20 sec, for 40 cycles. The relative expression levels of the M1 and M2 marker genes were calculated using the 2−ΔΔCq method (28), with GAPDH serving as the internal control. The expression levels of both M1 markers (CD86 and IL-12) and M2 markers (IL-10 and CD206) were measured. The sequences of the primers utilized are listed in Table SII.

Western blot analysis

The total protein of cells was obtained using radioimmunoprecipitation assay buffer for 30 sec (cat. no. P0013B; Beyotime Institute of Biotechnology). The total protein was separated using 10% SDS-PAGE. The separated proteins were subsequently transferred to a polyvinylidene fluoride membrane (GE Healthcare). Following a 2-h blocking step at room temperature with 5% non-fat dry milk (Yili), the membranes were incubated with the primary antibody overnight at 4°C. The primary antibodies were as follows: Bax (1:500 dilution; cat. no. A12009), Bcl-2 (1:1,000 dilution; cat. no. A19693), C-caspase-3 (1:500 dilution; cat. no. A11319) and β-actin (1:10,000 dilution; cat. no. AC038; all from ABclonal Biotech Co., Ltd.). Subsequently, the membranes were incubated with the secondary antibody (1:10,000 dilution; cat. no. AB_1185567; goat anti-rabbit IgG; Invitrogen; Thermo Fisher Scientific, Inc.) for 2 h at room temperature. The blots were visualized using an enhanced chemiluminescence system (Beyotime Institute of Biotechnology) and analyzed densitometrically using ImageJ software (version 1.8.0_345).

Statistical analysis

All bioinformatics analyses in the present study were performed using R Studio version 4.3.1 (https://cran.rstudio.com/bin/windows/base/old/4.3.1/), while data were processed with GraphPad Prism 9.0 (Dotmatics). Statistical comparisons between two groups were assessed using an unpaired Student's t-test, whereas differences among multiple groups were evaluated by one-way ANOVA and the Bonferroni's test was used as the post-hoc test. P<0.05 was considered to indicate a statistically significant difference. To ensure reproducibility, all experiments were repeated at least three times, with results expressed as the mean ± standard deviation.

Results

Differential expression of M2 macrophages in patients with LIHC and normal controls

Macrophage infiltration levels in LIHC and normal tissue samples were evaluated using six immune infiltration analysis methods. The heatmap shown in Fig. 1A illustrates the distribution of infiltrating macrophages in both LIHC and normal samples across the different methods. Differential expression analysis of M2 macrophages (Fig. 1B) revealed significant differences between patients with LIHC and normal controls. Notably, all analysis methods indicated relevant findings, except for the MCPCOUNTER method, which did not show a statistically significant difference. Specifically, quantTIseq exhibited a significant decrease, while the other methods demonstrated increases in expression. Consequently, the results are inconsistent across the different approaches. These results underscore the potential involvement of M2 macrophages in the immune microenvironment of LIHC.

Macrophage infiltration and M2
macrophage differential expression in LIHC. (A) Heatmap
illustrating macrophage infiltration levels in LIHC and normal
tissue samples, as quantified by six different immune infiltration
analysis methods. The heatmap provides a visual representation of
macrophage levels across all methods. The red lines indicate the
distribution of infiltrating macrophages in both LIHC and normal
samples across the different methods. (B) Differential expression
analysis of M2 macrophages between patients with LIHC and normal
controls. Significant differences in M2 macrophage expression were
observed in LIHC samples compared with normal controls across all
methods, with the exception of the MCPCOUNTER method. LIHC, liver
hepatocellular carcinoma.

Figure 1.

Macrophage infiltration and M2 macrophage differential expression in LIHC. (A) Heatmap illustrating macrophage infiltration levels in LIHC and normal tissue samples, as quantified by six different immune infiltration analysis methods. The heatmap provides a visual representation of macrophage levels across all methods. The red lines indicate the distribution of infiltrating macrophages in both LIHC and normal samples across the different methods. (B) Differential expression analysis of M2 macrophages between patients with LIHC and normal controls. Significant differences in M2 macrophage expression were observed in LIHC samples compared with normal controls across all methods, with the exception of the MCPCOUNTER method. LIHC, liver hepatocellular carcinoma.

Identification of M2 macrophage-related gene modules

The optimal soft threshold for constructing the gene co-expression network was determined to be 14, which yielded a scale-free topology model fit value approaching 0.9 (Fig. 2A). A total of 16 distinct gene modules were identified using WGCNA (Fig. 2B). Preservation analysis indicated that four modules (grey, gold, midnight blue and black) demonstrated Z scores <2, thereby indicating poor preservation; thus, these modules were excluded from further analysis (Fig. 2C). Correlation analysis between M2 macrophage expression levels and the gene modules revealed that the light cyan and yellow modules were the most strongly correlated with M2 macrophages (Fig. 2D). Genes from these two modules were selected for subsequent prognostic modelling.

Identification and preservation of
gene modules associated with M2 macrophages. (A) Determination of
the optimal soft threshold for network construction based on the
scale-free topology model fit. A power value of 14 was selected,
achieving a model fit close to 0.9. (B) Dendrogram showing the
identification of 16 gene modules using weighted gene co-expression
network analysis. (C) Preservation analysis of the identified gene
modules. Modules with Z scores <2 (grey, gold, midnight blue and
black) were excluded due to poor preservation. (D) Correlation
analysis between M2 macrophage expression levels and the gene
modules. ME, module eigengene.

Figure 2.

Identification and preservation of gene modules associated with M2 macrophages. (A) Determination of the optimal soft threshold for network construction based on the scale-free topology model fit. A power value of 14 was selected, achieving a model fit close to 0.9. (B) Dendrogram showing the identification of 16 gene modules using weighted gene co-expression network analysis. (C) Preservation analysis of the identified gene modules. Modules with Z scores <2 (grey, gold, midnight blue and black) were excluded due to poor preservation. (D) Correlation analysis between M2 macrophage expression levels and the gene modules. ME, module eigengene.

Development and validation of the M2 macrophage-related genes prognostic model

Univariate Cox regression analysis identified 14 genes with significant associations with patient prognosis (P<0.05) in the LIHC cohort (Fig. 3A). LASSO regression analysis (with a λ value of 0.0514) revealed three genes for inclusion in the final model, including KLRB1 (Fig. 3B). Multivariate Cox regression analysis validated the independent prognostic value of these three genes. The prognostic risk score model was constructed based on the following formula: RiskScore=phosphatidylinositol-5-phosphate 4-kinase type 2α (PIP4K2A) × 1.0165 + KLRB1 × 0.8897 (Fig. 3C). Kaplan-Meier survival analysis revealed significant differences in survival between the two risk groups (Fig. 3D). The model's performance was validated using time-dependent ROC analysis, which demonstrated a certain predictive ability across three separate cohorts, including TCGA LIHC, GSE76427 (GEO) and LIRI-JP (ICGC) (Fig. 3E).

Development and validation of the M2
macrophage-related prognostic model. (A) Univariate Cox regression
analysis identified 14 genes with significant prognostic value
(P<0.05) in patients with LIHC. (B) Least Absolute Shrinkage and
Selection Operator-Cox regression was applied to refine the gene
set, selecting three key genes based on a λ value of 0.0514
(10-fold cross-validation). (C) Multivariate Cox regression
confirmed the independent prognostic significance of the two
selected genes. A prognostic risk score model was constructed using
the following formula: RiskScore = PIP4K2A × 1.0165 + KLRB1 ×
0.8897. (D) Kaplan-Meier survival curves comparing two risk groups
in the TCGA-LIHC cohort. A significant difference in overall
survival was observed between the groups. (E) Time-dependent
receiver operating characteristic curve analysis of the prognostic
risk score model, showing certain predictive performance across
three independent cohorts: TCGA-LIHC, GSE76427 (Gene Expression
Omnibus) and LIRI-JP (International Cancer Genome Consortium).
LIHC, liver hepatocellular carcinoma; TCGA, The Cancer Genome
Atlas; PIP4K2A, phosphatidylinositol-5-phosphate 4-kinase type 2α;
SH3BP1, SH3 domain binding protein 1; KLRB1, killer cell lectin
like receptor B1; AUC, area under the curve; TMSB4XP8, TMSB4X
pseudogene 8; STK10, serine/threonine kinase 10; PTGER4,
prostaglandin E receptor 4; PLEKHA2, pleckstrin homology domain
containing A2; MX2, MX dynamin like GTPase 2; MCUB, mitochondrial
calcium uniporter dominant negative subunit β; IL2RG, IL2 receptor
subunit γ; GMIP, GEM interacting protein; EAF2, ELL associated
factor 2; ADAM19, ADAM metallopeptidase domain 19; HR, hazard
ratio.

Figure 3.

Development and validation of the M2 macrophage-related prognostic model. (A) Univariate Cox regression analysis identified 14 genes with significant prognostic value (P<0.05) in patients with LIHC. (B) Least Absolute Shrinkage and Selection Operator-Cox regression was applied to refine the gene set, selecting three key genes based on a λ value of 0.0514 (10-fold cross-validation). (C) Multivariate Cox regression confirmed the independent prognostic significance of the two selected genes. A prognostic risk score model was constructed using the following formula: RiskScore = PIP4K2A × 1.0165 + KLRB1 × 0.8897. (D) Kaplan-Meier survival curves comparing two risk groups in the TCGA-LIHC cohort. A significant difference in overall survival was observed between the groups. (E) Time-dependent receiver operating characteristic curve analysis of the prognostic risk score model, showing certain predictive performance across three independent cohorts: TCGA-LIHC, GSE76427 (Gene Expression Omnibus) and LIRI-JP (International Cancer Genome Consortium). LIHC, liver hepatocellular carcinoma; TCGA, The Cancer Genome Atlas; PIP4K2A, phosphatidylinositol-5-phosphate 4-kinase type 2α; SH3BP1, SH3 domain binding protein 1; KLRB1, killer cell lectin like receptor B1; AUC, area under the curve; TMSB4XP8, TMSB4X pseudogene 8; STK10, serine/threonine kinase 10; PTGER4, prostaglandin E receptor 4; PLEKHA2, pleckstrin homology domain containing A2; MX2, MX dynamin like GTPase 2; MCUB, mitochondrial calcium uniporter dominant negative subunit β; IL2RG, IL2 receptor subunit γ; GMIP, GEM interacting protein; EAF2, ELL associated factor 2; ADAM19, ADAM metallopeptidase domain 19; HR, hazard ratio.

Differential pathway and functional enrichment between the two risk groups

GSVA of the hallmark gene sets revealed distinct differences in pathway activation between the two risk groups. The high-risk group exhibited upregulation of pathways associated with coagulation, allograft rejection and myogenesis, thus suggesting an active inflammatory and tissue remodeling environment. By contrast, the low-risk group exhibited increased activity in pathways involved in cell cycle regulation, including ‘E2F_TARGETS’, ‘G2M_CHECKPOINT’ and ‘MITOTIC_SPINDLE’, thereby indicating increased cell proliferation and division (Fig. 4A). GO enrichment analysis further highlighted functional differences between the groups. The high-risk group was significantly enriched for pathways associated with cellular polarity and membrane organization, including the ‘apical plasma membrane’ and ‘apical part of the cell’ (Fig. 4B). Conversely, the low-risk group was enriched in immune-related pathways, particularly those associated with immunoglobulin antigen binding, thus reflecting an active immune response (Fig. 4C). KEGG pathway analysis also revealed several signaling pathways, such as ‘Primary immunodeficiency’, ‘Graft-versus-host disease’ and ‘Hippo signaling pathway’, that were differentially enriched between the two risk groups. The KEGG pathways with similar biological functions were further categorized to highlight shared themes across the risk groups (Fig. 4D).

Differential pathway and functional
enrichment analysis. (A) GSVA of the hallmark gene sets revealed
significant differences in pathway activation between the two risk
groups. (B) GO functional enrichment analysis identified
significant enrichment for pathways related to cellular polarity
and apical membrane organization in the high-risk group. (C) GO
analysis of the low-risk group showed enrichment for immune
response-related pathways, including immunoglobulin antigen
binding. (D) KEGG pathway enrichment analysis identified
differential activation of signaling pathways between the two risk
groups. Pathways were categorized based on shared biological
functions to reveal common themes across the groups. GO, Gene
Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; GSVA, Gene
Set Variation Analysis; BP, biological process; CC, cellular
component; MF, molecular function.

Figure 4.

Differential pathway and functional enrichment analysis. (A) GSVA of the hallmark gene sets revealed significant differences in pathway activation between the two risk groups. (B) GO functional enrichment analysis identified significant enrichment for pathways related to cellular polarity and apical membrane organization in the high-risk group. (C) GO analysis of the low-risk group showed enrichment for immune response-related pathways, including immunoglobulin antigen binding. (D) KEGG pathway enrichment analysis identified differential activation of signaling pathways between the two risk groups. Pathways were categorized based on shared biological functions to reveal common themes across the groups. GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; GSVA, Gene Set Variation Analysis; BP, biological process; CC, cellular component; MF, molecular function.

Immunotherapy prediction analysis based on the TIDE and Submap algorithms

A strong negative correlation was observed between the risk score and all three ESTIMATE scores (immune, stromal and overall ESTIMATE scores), thereby indicating that higher scores were associated with lower levels of immune and stromal cell infiltration (Fig. 5A). By contrast, no significant correlation was observed between the risk score and the TMB, thereby suggesting that the TMB is not a major factor influencing risk stratification in the present cohort (Fig. 5B). Subsequently, differential expression analysis of common inhibitory immune checkpoint genes was performed. The results revealed that most inhibitory checkpoint genes (including IL2, CD40 ligand and CD70) were significantly differentially expressed, with higher expression levels being observed in the high-risk group (Fig. 5C). Conversely, the expression of stimulatory immune checkpoint genes did not significantly differ between the two groups (Fig. 5D). TIDE analysis revealed that low-risk patients exhibited significantly lower TIDE and dysfunction scores, thus indicating a potentially stronger immune response and a lower risk of immune escape. The exclusion score of low-risk patients was significantly higher, which was inconsistent with the overall prediction result. This may be due to reasons such as sample differences. Due to sample differences and other reasons, dysfunction and exclusion alone may not be able to make good predictions. Therefore, the TIDE total score, which combined the scores of dysfunction and exclusion, was usually adopted as the final prediction result of the TIDE analysis. (Fig. 5E). Finally, Submap analysis revealed that patients in the low-risk group were more likely to respond favorably to ICB therapy, thereby further suggesting that the low-risk group demonstrates greater potential for immunotherapy responsiveness (Fig. 5F).

Immune infiltration, TMB, immune
checkpoint expression and immunotherapy response in two risk
groups. (A) Correlation between the prognostic risk score and the
immune, stromal and ESTIMATE scores, calculated using the ESTIMATE
algorithm. (B) Correlation analysis between the risk score and TMB
showed no significant association. (C) Differential expression
analysis of inhibitory immune checkpoint genes revealed significant
differences between two risk groups. (D) Differential expression of
stimulatory immune checkpoint genes showed no significant
differences between the two groups. (E) TIDE analysis demonstrated
that low-risk patients had significantly lower TIDE scores,
suggesting they have an improved immune response and a reduced
potential for immune escape. (F) SubMap analysis predicted that
low-risk patients are more likely to respond to immune checkpoint
blockade therapy, highlighting their greater potential for
benefiting from immunotherapy. PD1-R indicates a response to PD1
treatment, while PD1-noR indicates no response to PD1 treatment.
CTLA4-R indicates a response to CTLA4 treatment, while CTLA4-noR
indicates no response to CTLA4 treatment. *P<0.05; **P<0.01;
***P<0.001; ****P<0.0001; t-test based P-value. TMB, tumor
mutational burden; TIDE, Tumor Immune Dysfunction and Exclusion;
PD1, programmed cell death protein 1; CTLA4, cytotoxic T-lymphocyte
associated protein 4.

Figure 5.

Immune infiltration, TMB, immune checkpoint expression and immunotherapy response in two risk groups. (A) Correlation between the prognostic risk score and the immune, stromal and ESTIMATE scores, calculated using the ESTIMATE algorithm. (B) Correlation analysis between the risk score and TMB showed no significant association. (C) Differential expression analysis of inhibitory immune checkpoint genes revealed significant differences between two risk groups. (D) Differential expression of stimulatory immune checkpoint genes showed no significant differences between the two groups. (E) TIDE analysis demonstrated that low-risk patients had significantly lower TIDE scores, suggesting they have an improved immune response and a reduced potential for immune escape. (F) SubMap analysis predicted that low-risk patients are more likely to respond to immune checkpoint blockade therapy, highlighting their greater potential for benefiting from immunotherapy. PD1-R indicates a response to PD1 treatment, while PD1-noR indicates no response to PD1 treatment. CTLA4-R indicates a response to CTLA4 treatment, while CTLA4-noR indicates no response to CTLA4 treatment. *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001; t-test based P-value. TMB, tumor mutational burden; TIDE, Tumor Immune Dysfunction and Exclusion; PD1, programmed cell death protein 1; CTLA4, cytotoxic T-lymphocyte associated protein 4.

Expression and ligand-receptor interactions of KLRB1 in single-cell datasets

To understand the role of KLRB1 in immune cell populations, its expression was analyzed in six distinct single-cell datasets from the TISCH2 database. KLRB1 was observed to be predominantly expressed in CD8+ T cells, CD8+ Tex and NK cells, thus indicating its potential involvement in immune regulation and response within these cell types (Fig. 6A). The heatmap displays strong ligand-receptor interactions between various immune cell types, with notable interaction intensities being observed between CD8+ T cells and other immune populations (Fig. 6B). Finally, the network diagram depicts the dynamic interactions between CD8+ T cells and surrounding cell populations, thus highlighting the strength and activity of these interactions. The analysis demonstrated that CD8+ T cells exhibit significant signaling crosstalk with other immune cell populations (Fig. 6C).

KLRB1 expression and ligand-receptor
interactions in the TME. (A) Expression of KLRB1 across six
single-cell RNA-sequencing datasets from the Tumor Immune Single
Cell Hub 2 database. KLRB1 was predominantly expressed in
CD8+ T cells, CD8+ Tex and NK cells. (B)
Heatmap showing the strength of ligand-receptor interactions
between different immune cell populations. (C) Network diagram
illustrating the intensity of ligand-receptor interactions between
CD8+ T cells and other cell populations in the TME.
KLRB1, killer cell lectin like receptor B1; TME, tumor
microenvironment; NK, natural killer; Tex, exhausted T cells; LIHC,
liver hepatocellular carcinoma; DC, dendritic cell; Treg,
T-regulatory cell.

Figure 6.

KLRB1 expression and ligand-receptor interactions in the TME. (A) Expression of KLRB1 across six single-cell RNA-sequencing datasets from the Tumor Immune Single Cell Hub 2 database. KLRB1 was predominantly expressed in CD8+ T cells, CD8+ Tex and NK cells. (B) Heatmap showing the strength of ligand-receptor interactions between different immune cell populations. (C) Network diagram illustrating the intensity of ligand-receptor interactions between CD8+ T cells and other cell populations in the TME. KLRB1, killer cell lectin like receptor B1; TME, tumor microenvironment; NK, natural killer; Tex, exhausted T cells; LIHC, liver hepatocellular carcinoma; DC, dendritic cell; Treg, T-regulatory cell.

Expression and prognostic importance of KLRB1

KLRB1 expression was first assessed in multiple cancer types using TCGA pan-cancer data. KLRB1 was observed to be significantly differentially expressed in a majority of cancer types, including LIHC, thereby highlighting its relevance in tumor biology (Fig. 7A). Prognostic analysis revealed that KLRB1 expression was associated with favorable outcomes (including DFI, DSS, OS and PFI) in most cancer types (Fig. 7B). Furthermore, high expression of KLRB1 was found to be closely related to poor OS (Fig. 7C). The LIHC samples were then stratified into high- and low-KLRB1 expression groups. GSEA of these groups revealed significant enrichment of specific biological pathways such as wnt-β-catenin signaling, TGF-β signaling and PI3K-AKT-mTOR signaling in both the high- and low-expression groups, thus suggesting that KLRB1 may serve a key role in regulating various tumor-related processes (Fig. 7D).

Pan-cancer analysis of KLRB1
expression and prognostic impact. (A) Expression of KLRB1 across
multiple cancer types in The Cancer Genome Atlas. Significant
differences in KLRB1 expression were observed in several cancer
types, including LIHC. (B) Prognostic analysis of KLRB1 expression
in pan-cancer datasets, including DFI, DSS, OS and PFI. (C) Forest
plot analysis of OS based on KLRB1 expression in pan-cancer. (D)
Gene Set Enrichment Analysis revealed significant biological
pathways enriched in both high- and low-expression groups,
suggesting that KLRB1 may regulate important tumor-related
processes. *P<0.05; **P<0.01; ***P<0.001; t-test based
P-value. KLRB1, killer cell lectin like receptor B1; LIHC, liver
hepatocellular carcinoma; DFI, disease-free interval; DSS,
disease-specific survival; OS, overall survival; PFI, platinum-free
interval; FDR, false discovery rate; NES, normalized enrichment
score; ACC, adrenocortical carcinoma; BLCA, bladder cancer; BRCA,
breast invasive carcinoma; CESC, cervical squamous cell carcinoma;
CHOL, cholangiocarcinoma; COAD, colon adenocarcinoma; DLBC, diffuse
large B-cell lymphoma; ESCA, esophageal carcinoma; GBM,
glioblastoma multiforme; HNSC, head and neck squamous cell
carcinoma; KICH, kidney chromophobe; KIRC, kidney clear cell
carcinoma; KIRP, kidney renal papillary cell carcinoma; LGG, brain
lower grade glioma; LIHC, liver hepatocellular carcinoma; LUAD,
lung adenocarcinoma; LUSC, lung squamous cell carcinoma; MESO,
mesothelioma; OV, ovarian serous cystadenocarcinoma; PAAD,
pancreatic adenocarcinoma; PCPG, pheochromocytoma and
paraganglioma; PRAD, prostate adenocarcinoma; READ, rectum
adenocarcinoma; SARC, sarcoma; SKCM, skin cutaneous melanoma; STAD,
stomach adenocarcinoma; TGCT, testicular germ cell tumor; THCA,
thyroid carcinoma; THYM, thymoma; UCEC, uterine corpus endometrioid
carcinoma; UCS, uterine carcinosarcoma; UVM, uveal melanoma.

Figure 7.

Pan-cancer analysis of KLRB1 expression and prognostic impact. (A) Expression of KLRB1 across multiple cancer types in The Cancer Genome Atlas. Significant differences in KLRB1 expression were observed in several cancer types, including LIHC. (B) Prognostic analysis of KLRB1 expression in pan-cancer datasets, including DFI, DSS, OS and PFI. (C) Forest plot analysis of OS based on KLRB1 expression in pan-cancer. (D) Gene Set Enrichment Analysis revealed significant biological pathways enriched in both high- and low-expression groups, suggesting that KLRB1 may regulate important tumor-related processes. *P<0.05; **P<0.01; ***P<0.001; t-test based P-value. KLRB1, killer cell lectin like receptor B1; LIHC, liver hepatocellular carcinoma; DFI, disease-free interval; DSS, disease-specific survival; OS, overall survival; PFI, platinum-free interval; FDR, false discovery rate; NES, normalized enrichment score; ACC, adrenocortical carcinoma; BLCA, bladder cancer; BRCA, breast invasive carcinoma; CESC, cervical squamous cell carcinoma; CHOL, cholangiocarcinoma; COAD, colon adenocarcinoma; DLBC, diffuse large B-cell lymphoma; ESCA, esophageal carcinoma; GBM, glioblastoma multiforme; HNSC, head and neck squamous cell carcinoma; KICH, kidney chromophobe; KIRC, kidney clear cell carcinoma; KIRP, kidney renal papillary cell carcinoma; LGG, brain lower grade glioma; LIHC, liver hepatocellular carcinoma; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; MESO, mesothelioma; OV, ovarian serous cystadenocarcinoma; PAAD, pancreatic adenocarcinoma; PCPG, pheochromocytoma and paraganglioma; PRAD, prostate adenocarcinoma; READ, rectum adenocarcinoma; SARC, sarcoma; SKCM, skin cutaneous melanoma; STAD, stomach adenocarcinoma; TGCT, testicular germ cell tumor; THCA, thyroid carcinoma; THYM, thymoma; UCEC, uterine corpus endometrioid carcinoma; UCS, uterine carcinosarcoma; UVM, uveal melanoma.

Regulation of tumors by KLRB1 and its associated secretory factors on macrophage polarization

qPCR was used to assess the mRNA levels of KLRB1 in HuH-7 and HuH-1 cells. The results revealed that KLRB1 expression was higher in HuH-7 cells compared with HuH-1 cells (Fig. 8A). To knock down KLRB1 in HuH-7 cells (sh-KLRB1-2), shRNA was used and the knockdown cell line was selected using puromycin. For stable overexpression of KLRB1 in HuH-1 cells (OE-KLRB1), lentiviral vectors were used, followed by puromycin selection to generate stable overexpression cell lines (Fig. 8B). To evaluate the impact of KLRB1 on tumor cell proliferation, CCK-8 assays were performed. The results demonstrated that KLRB1 knockdown significantly increased cell proliferation compared with the control group, whereas overexpression significantly decreased cell proliferation (Fig. 8C). Transwell assays were performed to assess the effect of KLRB1 on cell migration. KLRB1 knockdown enhanced cell migration, whereas KLRB1 overexpression reduced migration (Fig. 8D and E). Additionally, the influence of KLRB1 on cell apoptosis was examined. The flow cytometry results indicated that KLRB1 knockdown inhibited apoptosis, whereas KLRB1 overexpression promoted apoptosis (Fig. 8F). And through WB experiments, it was found that knocking out KLRB1 could significantly inhibit the expression of Bax and c-caspase-3, and enhance the expression of Bcl-2 and caspase-3, while overexpression achieved the opposite result (Fig. S1). Furthermore, the tube formation assay demonstrated that, compared with the controls, the culture supernatant from KLRB1 knockdown cells increased the tube forming ability of HUVECs, whereas the supernatant from KLRB1-overexpressing cells reduced the tube forming ability of HUVECs (Fig. 8G).

KLRB1 regulates the activity of LIHC
cells and its influence on macrophages through LIHC cells. (A) qPCR
detection of KLRB1 expression in HuH-7 and HuH-1 cells. (B)
Validation of shRNA knockdown KLRB1 in the HuH-7 cell line and
lentivirus overexpression of KLRB1 in the HuH-1 cell line. (C) Cell
proliferation curve of sh-KLRB1-2 and OE-KLRB1. Cell migration
assay of (D) sh-KLRB1-2 and (E) OE-KLRB1 cells (scale bars, 100
µm). (F) Annexin V-FITC/PI assessed apoptosis in sh-KLRB1-2 and
OE-KLRB1 transfected cells. (G) Tube formation assay of HUVECs
treated with sh-KLRB1-2 and OE-KLRB1 cell conditioned media (scale
bar, 200 µm). (H) qPCR detection of polarization markers in M0 to
M1/M2 cells, treated with sh-KLRB1-2 or OE-KLRB1 cell conditioned
media. *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001;
t-test based P-value. LIHC, liver hepatocellular carcinoma; PI,
propidium iodide qPCR, quantitative PCR; KLRB1, killer cell lectin
like receptor B1; sh, short hairpin; OE, overexpression; NC,
negative control; OD, optical density.

Figure 8.

KLRB1 regulates the activity of LIHC cells and its influence on macrophages through LIHC cells. (A) qPCR detection of KLRB1 expression in HuH-7 and HuH-1 cells. (B) Validation of shRNA knockdown KLRB1 in the HuH-7 cell line and lentivirus overexpression of KLRB1 in the HuH-1 cell line. (C) Cell proliferation curve of sh-KLRB1-2 and OE-KLRB1. Cell migration assay of (D) sh-KLRB1-2 and (E) OE-KLRB1 cells (scale bars, 100 µm). (F) Annexin V-FITC/PI assessed apoptosis in sh-KLRB1-2 and OE-KLRB1 transfected cells. (G) Tube formation assay of HUVECs treated with sh-KLRB1-2 and OE-KLRB1 cell conditioned media (scale bar, 200 µm). (H) qPCR detection of polarization markers in M0 to M1/M2 cells, treated with sh-KLRB1-2 or OE-KLRB1 cell conditioned media. *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001; t-test based P-value. LIHC, liver hepatocellular carcinoma; PI, propidium iodide qPCR, quantitative PCR; KLRB1, killer cell lectin like receptor B1; sh, short hairpin; OE, overexpression; NC, negative control; OD, optical density.

To investigate how KLRB1 affects the secretion of tumor cell-derived factors involved in macrophage polarization, culture supernatants were collected from KLRB1-knockdown and KLRB1-overexpressing cells and incubated with THP-1 cell-derived M0 macrophages. The polarization status of M1 and M2 macrophages was then assessed using qPCR. The results demonstrated that KLRB1 knockdown culture supernatant increased the expression of M2 markers CD206 and IL-10, whereas KLRB1-overexpressing cell culture supernatant reduced the expression of CD206 and IL-10. By contrast, the M1 markers CD86 and IL-12 were downregulated following KLRB1 knockdown culture supernatant treatment and upregulated after overexpression supernatant treatment (Fig. 8H). These findings suggest that KLRB1 may inhibit the polarization of M0 macrophages towards the M2 phenotype. In summary, the upregulation of KLRB1 inhibits the activity of LIHC cells and facilitates the polarization of macrophages from the M0 phenotype to the M1 phenotype through changes in the cell secretome.

Discussion

LIHC is one of the most prevalent and fatal malignancies worldwide, with a poor prognosis being observed, primarily due to late-stage diagnoses, high recurrence rates and resistance to conventional treatments (29). The TME serves a crucial role in the progression of LIHC, with macrophages (particularly M2 macrophages) being key contributors to tumor growth, metastasis and immune evasion (30). M2 macrophages are characterized by their immunosuppressive properties, which promote tumor progression through the cells, thereby fostering immune escape (31). Additionally, M2 macrophages facilitate angiogenesis and ECM remodeling, thus further enhancing the pro-tumorigenic environment in LIHC (32). Previous advances in bioinformatics have enabled the integration of large-scale genomic data, thus allowing for the identification of key molecular signatures associated with immune infiltration and tumor progression (33,34). Using techniques such as WGCNA, researchers have identified M2 macrophage-related genes as potential biomarkers for prognosis in LIHC (35). Given the pivotal role of M2 macrophages in modulating the TME and influencing immune responses, a deeper understanding of their gene expression profiles could offer novel insights into prognostic prediction and therapeutic strategies for LIHC.

In the present study, M2 macrophage-related genes that serve significant roles in the prognosis of LIHC were identified. Using bioinformatics analyses, a prognostic risk model was constructed based on these genes, which demonstrated promising predictive power in both the training and external validation datasets. These findings reinforce the growing body of evidence suggesting that M2 macrophages are key players in the TME of LIHC and contribute to tumor progression (9,36). However, unlike some studies that have established associations between high M2 macrophage infiltration and poor clinical outcomes, the present results focused on the identification of gene expression patterns associated with M2 macrophages that may provide more reliable prognostic information for patients with LIHC.

Although increased infiltration of M2 macrophages has been associated with poor prognoses in various cancers, the present study did not identify direct correlations between M2 macrophage infiltration and various clinical outcomes (such as OS or progression-free survival) in patients with LIHC. This discrepancy may be due to the complex nature of the liver TME, where multiple factors, including hepatic fibrosis, viral infections and cirrhosis, contribute to disease progression beyond macrophage infiltration. Therefore, although M2 macrophage-related genes demonstrate potential as biomarkers for predicting disease progression, additional studies are necessary to elucidate the precise mechanisms through which M2 macrophages influence the clinical course of LIHC.

Notably, the present study suggests that M2 macrophages may influence the efficacy of immunotherapies in LIHC. Although immune checkpoint inhibitors have demonstrated promising results in various cancers, their effectiveness in LIHC is often hindered by the immunosuppressive TME, which is partly shaped by M2 macrophages (37). The analysis revealed that the preoperative risk model, which was constructed based on M2 macrophage-related genes, was associated with improved immune responses and improved outcomes in the low-risk group. Therefore, the targeting of M2 macrophages or their associated signaling pathways may provide a promising strategy to increase the effectiveness of immunotherapy in LIHC, particularly in low-risk patients who are identified using this model.

Although M2 macrophages serve a critical role in the TME and have been associated with poor prognoses in various cancers, the present study identified KLRB1 as an important factor in LIHC. KLRB1 is primarily expressed on NK cells and certain T-cell subsets and has been implicated in immune regulation and tumor immune surveillance (38). In LIHC, KLRB1 may influence the tumor immune landscape by modulating immune cell interactions, thereby potentially affecting tumor progression and patient outcomes. The findings suggest that KLRB1 expression may serve a protective role in LIHC, as higher levels of this gene were observed to be associated with improved survival outcomes. This result indicates the involvement of KLRB1 in promoting antitumor immune responses. However, the precise mechanisms through which KLRB1 modulates tumor progression and immune evasion in LIHC remain to be fully elucidated. Based on the present research findings, it may be speculated that KLRB1 may promote the progression of LIHC disease by regulating the polarity transformation of macrophages. Therefore, the targeting of KLRB1 or its associated signaling pathways may represent a promising therapeutic approach to improve patient prognosis and enhance the efficacy of immunotherapies in LIHC.

KLRB1 serves a crucial role in tumor regulation, with previous studies involving LIHC and lung adenocarcinoma (LUAD) highlighting its complex dual function. The expression levels and functions of KLRB1 vary markedly across different tumor types. For instance, in breast cancer and LUAD, low expression of KLRB1 is typically associated with poor prognosis, and its functions involve immune regulation, cell proliferation and migration (39). Similarly, in other cancer types, low expression of KLRB1 is generally associated with poor survival outcomes. In conclusion, the present study provides strong evidence that KLRB1 serves a multifaceted role in regulating both tumor cell behavior and macrophage polarization. The upregulation of KLRB1 appears to inhibit the proliferation and migration of LIHC cells, promote apoptosis and facilitate the polarization of macrophages towards the M1 phenotype. As KLRB1 is a membrane protein, the conditioned media may also contain exosomes or microvesicles secreted by the KLRB1 knockdown or OE cells. These vesicles can carry not only proteins and lipids but also mRNA and miRNA that can have an impact on recipient cells, including macrophages. For instance, specific miRNAs may influence macrophage gene expression related to polarization. Furthermore, changes in KLRB1 expression can alter the metabolic state of the transfected cells, which in turn could affect the composition of metabolites and extracellular vesicles released into the conditioned media. This is typically more effective in combating tumors. These findings highlight the potential of KLRB1 as a therapeutic target in LIHC and emphasize the importance of understanding the complex interactions between immune cells and cancer cells in the development of novel cancer treatments.

Despite the promising results associated with the M2 macrophage-related prognostic signature, several limitations must be acknowledged in the current study. Firstly, the research primarily relied on data retrieved from public databases, which often contain relatively small sample sizes and may not be promptly updated. In addition, the focal point of the investigation involved M2 macrophage-related genes; however, the specific molecular mechanisms by which these genes regulate the progression of LIHC necessitate further in-depth explorations. Consequently, the interpretation of the results presented requires additional validation and enhancement. Furthermore, the prognostic signature was predominantly verified using public datasets, which may harbor intrinsic biases and do not provide clinical validation within independent, real-world patient cohorts. Secondly, although analyses of immune-cell infiltration and predictions regarding immunotherapy offer valuable insights, the exact mechanisms linking M2 macrophage-related genes to immune responses in LIHC remain inadequately understood and warrant additional experimental investigations. Furthermore, the present study suggested that KLRB1 may serve a role in modulating immune responses and facilitating the progression of LIHC. A thorough review of the literature regarding KLRB1 indicates that this gene represents a novel therapeutic target; however, the molecular mechanisms through which KLRB1 regulates LIHC remain largely unexplored and should be further investigated. Additionally, the present research did not evaluate potential interactions between other key genes and molecular pathways that could influence the progression of LIHC, thereby highlighting the need for further explorations in this area. Finally, although the prognostic signature demonstrates promise in guiding immunotherapy strategies, its clinical applicability is constrained by the absence of prospective clinical trials to validate its predictive power. Future studies should focus on validating the prognostic signature within larger, more diverse patient populations and elucidating the mechanistic roles of M2 macrophages in the context of LIHC.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

The present work was supported by the Evaluation of the Efficacy of Liver-Coursing Spleen-Rectifying Decoction in Treating Non-Alcoholic Fatty Liver Disease and its Effects on Intestinal Flora and Serum LPS of Patients (grant no. S202406060003).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

LC was involved in the study conception and design and reviewed the manuscript. AW, XL and ZW performed the literature search, performed experiments, acquired and analyzed data, performed the statistical analysis and wrote and reviewed the manuscript. LC serves as a guarantor for the integrity of the entire study. LC, AW, XL and ZW confirm the authenticity of the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

LIHC

liver hepatocellular carcinoma

TME

tumor microenvironment

NK

natural killer

TCGA

The Cancer Genome Atlas

GEO

Gene Expression Omnibus

ICGC

International Cancer Genome Consortium

WGCNA

weighted gene co-expression network analysis

ROC

receiver operating characteristic

scRNA-seq

single-cell RNA sequencing

Tex

CD8+ exhausted T cells

OS

overall survival

CCK-8

Cell Counting Kit-8

PI

propidium iodide

RT-qPCR

reverse transcription quantitative PCR

LUAD

lung adenocarcinoma

References

1 

Siegel RL, Giaquinto AN and Jemal A: Cancer statistics, 2024. CA Cancer J Clin. 74:12–49. 2024.PubMed/NCBI

2 

Zheng Z, Mei J, Guan R, Zhang J, Xiong X, Gan J, Li S and Guo R: A novel liver-function-indicators-based prognosis signature for patients with hepatocellular carcinoma treated with anti-programmed cell death-1 therapy. Cancer Immunol Immunother. 73:1582024. View Article : Google Scholar : PubMed/NCBI

3 

Antonius Y, Kharisma VD, Widyananda MH, Ansori ANM, Trinugroho JP, Ullah Md E, Naw SW, Jakhmola V and Wahjudi M: Prediction of aflatoxin-B1 (AFB1) molecular mechanism network and interaction to oncoproteins growth factor in hepatocellular carcinoma. J Pure Appl Microbiol. 16:1844–1854. 2022. View Article : Google Scholar

4 

Jia G, He P, Dai T, Goh D, Wang J, Sun M, Wee F, Li F, Lim JCT, Hao S, et al: Spatial immune scoring system predicts hepatocellular carcinoma recurrence. Nature. 640:1031–1041. 2025. View Article : Google Scholar : PubMed/NCBI

5 

Yu B and Ma W: Biomarker discovery in hepatocellular carcinoma (HCC) for personalized treatment and enhanced prognosis. Cytokine Growth Factor Rev. 79:29–38. 2024. View Article : Google Scholar : PubMed/NCBI

6 

Li M, Yang Y, Xiong L, Jiang P, Wang J and Li C: Metabolism, metabolites, and macrophages in cancer. J Hematol Oncol. 16:802023. View Article : Google Scholar : PubMed/NCBI

7 

Brockmann L, Tran A, Huang Y, Edwards M, Ronda C, Wang HH and Ivanov II: Intestinal microbiota-specific Th17 cells possess regulatory properties and suppress effector T cells via c-MAF and IL-10. Immunity. 56:2719–2735.e7. 2023. View Article : Google Scholar : PubMed/NCBI

8 

Pascual-García M, Bonfill-Teixidor E, Planas-Rigol E, Rubio-Perez C, Iurlaro R, Arias A, Cuartas I, Sala-Hojman A, Escudero L, Martínez-Ricarte F, et al: LIF regulates CXCL9 in tumor-associated macrophages and prevents CD8+ T cell tumor-infiltration impairing anti-PD1 therapy. Nat Commun. 10:24162019. View Article : Google Scholar : PubMed/NCBI

9 

Han S, Bao X, Zou Y, Wang L, Li Y, Yang L, Liao A, Zhang X, Jiang X, Liang D, et al: d-lactate modulates M2 tumor-associated macrophages and remodels immunosuppressive tumor microenvironment for hepatocellular carcinoma. Sci Adv. 9:eadg26972023. View Article : Google Scholar : PubMed/NCBI

10 

Li D, Zhang T, Guo Y, Bi C, Liu M and Wang G: Biological impact and therapeutic implication of tumor-associated macrophages in hepatocellular carcinoma. Cell Death Dis. 15:4982024. View Article : Google Scholar : PubMed/NCBI

11 

Zhang Y, Han G, Gu J, Chen Z and Wu J: Role of tumor-associated macrophages in hepatocellular carcinoma: impact, mechanism, and therapy. Front Immunol. 15:14298122024. View Article : Google Scholar : PubMed/NCBI

12 

Sun Y, Zhao M, Cheng L, He X, Shen S, Lv J, Zhang J, Shao Q, Yin W, Zhao F, et al: Reduction of alternative polarization of macrophages by short-term activated hepatic stellate cell-derived small extracellular vesicles. J Exp Clin Cancer Res. 44:1172025. View Article : Google Scholar : PubMed/NCBI

13 

Du Y, Wu S, Xi S, Xu W, Sun L, Yan J, Gao H, Wang Y, Zheng J, Wang F, et al: ASH1L in hepatoma cells and hepatic stellate cells promotes fibrosis-associated hepatocellular carcinoma by modulating tumor-associated macrophages. Adv Sci (Weinh). 11:e24047562024. View Article : Google Scholar : PubMed/NCBI

14 

Jeong JM, Choi SE, Shim YR, Kim HH, Lee YS, Yang K, Kim K, Kim MJ, Chung KPS, Kim SH, et al: CX 3 CR1 + macrophages interact with HSCs to promote HCC through CD8 + T-cell suppression. Hepatology. 82:655–668. 2025. View Article : Google Scholar : PubMed/NCBI

15 

Cai J, Song L, Zhang F, Wu S, Zhu G, Zhang P, Chen S, Du J, Wang B, Cai Y, et al: Targeting SRSF10 might inhibit M2 macrophage polarization and potentiate anti-PD-1 therapy in hepatocellular carcinoma. Cancer Commun (Lond). 44:1231–1260. 2024. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Aran D, Hu Z and Butte AJ: xCell: Digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 18:2202017. View Article : Google Scholar : PubMed/NCBI

18 

Mellman I, Chen DS, Powles T and Turley SJ: The cancer-immunity cycle: Indication, genotype, and immunotype. Immunity. 56:2188–2205. 2023. View Article : Google Scholar : PubMed/NCBI

19 

Finotello F, Mayer C, Plattner C, Laschober G, Rieder D, Hackl H, Krogsdam A, Loncova Z, Posch W, Wilflingseder D, et al: Molecular and pharmacological modulators of the tumor immune contexture revealed by deconvolution of RNA-seq data. Genome Med. 11:342019. View Article : Google Scholar : PubMed/NCBI

20 

Racle J, de Jonge K, Baumgaertner P, Speiser DE and Gfeller D: Simultaneous enumeration of cancer and immune cell types from bulk tumor gene expression data. Elife. 6:e264762017. View Article : Google Scholar : PubMed/NCBI

21 

Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F, Selves J, Laurent-Puig P, Sautès-Fridman C, Fridman WH and de Reyniès A: Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 17:2182016. View Article : Google Scholar : PubMed/NCBI

22 

Langfelder P and Horvath S: WGCNA: An R package for weighted correlation network analysis. BMC Bioinformatics. 9:5592008. View Article : Google Scholar : PubMed/NCBI

23 

Hänzelmann S, Castelo R and Guinney J: GSVA: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 14:72013. View Article : Google Scholar : PubMed/NCBI

24 

Wang G, Liu T and He WT: Visualization analysis of research hotspots and trends on gastrointestinal tumor organoids. World J Gastrointest Oncol. 16:2826–2841. 2024. View Article : Google Scholar : PubMed/NCBI

25 

Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, Li Z, Traugh N, Bu X, Li B, et al: Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med. 24:1550–1558. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Kalhor K, Chen CJ, Lee HS, Cai M, Nafisi M, Que R, Palmer CR, Yuan Y, Zhang Y, Li X, et al: Mapping human tissues with highly multiplexed RNA in situ hybridization. Nat Commun. 15:25112024. View Article : Google Scholar : PubMed/NCBI

27 

Han Y, Wang Y, Dong X, Sun D, Liu Z, Yue J, Wang H, Li T and Wang C: TISCH2: expanded datasets and new tools for single-cell transcriptome analyses of the tumor microenvironment. Nucleic Acids Res. 51:D1425–D1431. 2023. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Yang X, Yang C, Zhang S, Geng H, Zhu AX, Bernards R, Qin W, Fan J, Wang C and Gao Q: Precision treatment in advanced hepatocellular carcinoma. Cancer Cell. 42:180–197. 2024. View Article : Google Scholar : PubMed/NCBI

30 

Ruf B, Bruhns M, Babaei S, Kedei N, Ma L, Revsine M, Benmebarek MR, Ma C, Heinrich B, Subramanyam V, et al: Tumor-associated macrophages trigger MAIT cell dysfunction at the HCC invasive margin. Cell. 186:3686–3705.e32. 2023. View Article : Google Scholar : PubMed/NCBI

31 

Jin R, Neufeld L and McGaha TL: Linking macrophage metabolism to function in the tumor microenvironment. Nat Cancer. 6:239–252. 2025. View Article : Google Scholar : PubMed/NCBI

32 

Zeng W, Li F, Jin S, Ho PC, Liu PS and Xie X: Functional polarization of tumor-associated macrophages dictated by metabolic reprogramming. J Exp Clin Cancer Res. 42:2452023. View Article : Google Scholar : PubMed/NCBI

33 

Zhang D, Zhao K, Han T, Zhang X, Xu X, Liu Z, Ren X, Zhang X, Lu Z and Qin C: Bisphenol A promote the cell proliferation and invasion ability of prostate cancer cells via regulating the androgen receptor. Ecotoxicol Environ Saf. 269:1158182024. View Article : Google Scholar : PubMed/NCBI

34 

Cai J, Chen T, Jiang Z, Yan J, Ye Z, Ruan Y, Tao L, Shen Z, Liang X, Wang Y, et al: Bulk and single-cell transcriptome profiling reveal extracellular matrix mechanical regulation of lipid metabolism reprograming through YAP/TEAD4/ACADL axis in hepatocellular carcinoma. Int J Biol Sci. 19:2114–2131. 2023. View Article : Google Scholar : PubMed/NCBI

35 

Qu X, Zhao X, Lin K, Wang N, Li X, Li S, Zhang L and Shi Y: M2-like tumor-associated macrophage-related biomarkers to construct a novel prognostic signature, reveal the immune landscape, and screen drugs in hepatocellular carcinoma. Front Immunol. 13:9940192022. View Article : Google Scholar : PubMed/NCBI

36 

Chen S, Zhang P, Zhu G, Wang B, Cai J, Song L, Wan J, Yang Y, Du J, Cai Y, et al: Targeting GSDME-mediated macrophage polarization for enhanced antitumor immunity in hepatocellular carcinoma. Cell Mol Immunol. 21:1505–1521. 2024. View Article : Google Scholar : PubMed/NCBI

37 

Vizcaino Castro A, Daemen T and Oyarce C: Strategies to reprogram anti-inflammatory macrophages towards pro-inflammatory macrophages to support cancer immunotherapies. Immunol Lett. 267:1068642024. View Article : Google Scholar : PubMed/NCBI

38 

Mace EM: Human natural killer cells: Form, function, and development. J Allergy Clin Immunol. 151:371–385. 2023. View Article : Google Scholar : PubMed/NCBI

39 

Huang G, Xiao S, Jiang Z, Zhou X, Chen L, Long L, Zhang S, Xu K, Chen J and Jiang B: Machine learning immune-related gene based on KLRB1 model for predicting the prognosis and immune cell infiltration of breast cancer. Front Endocrinol (Lausanne). 14:11857992023. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Wang A, Li X, Wang Z and Chen L: Prognostic value of M2 macrophage‑related genes and their importance in the immunotherapy response in hepatocellular carcinoma. Oncol Lett 30: 540, 2025.
APA
Wang, A., Li, X., Wang, Z., & Chen, L. (2025). Prognostic value of M2 macrophage‑related genes and their importance in the immunotherapy response in hepatocellular carcinoma. Oncology Letters, 30, 540. https://doi.org/10.3892/ol.2025.15286
MLA
Wang, A., Li, X., Wang, Z., Chen, L."Prognostic value of M2 macrophage‑related genes and their importance in the immunotherapy response in hepatocellular carcinoma". Oncology Letters 30.6 (2025): 540.
Chicago
Wang, A., Li, X., Wang, Z., Chen, L."Prognostic value of M2 macrophage‑related genes and their importance in the immunotherapy response in hepatocellular carcinoma". Oncology Letters 30, no. 6 (2025): 540. https://doi.org/10.3892/ol.2025.15286
Copy and paste a formatted citation
x
Spandidos Publications style
Wang A, Li X, Wang Z and Chen L: Prognostic value of M2 macrophage‑related genes and their importance in the immunotherapy response in hepatocellular carcinoma. Oncol Lett 30: 540, 2025.
APA
Wang, A., Li, X., Wang, Z., & Chen, L. (2025). Prognostic value of M2 macrophage‑related genes and their importance in the immunotherapy response in hepatocellular carcinoma. Oncology Letters, 30, 540. https://doi.org/10.3892/ol.2025.15286
MLA
Wang, A., Li, X., Wang, Z., Chen, L."Prognostic value of M2 macrophage‑related genes and their importance in the immunotherapy response in hepatocellular carcinoma". Oncology Letters 30.6 (2025): 540.
Chicago
Wang, A., Li, X., Wang, Z., Chen, L."Prognostic value of M2 macrophage‑related genes and their importance in the immunotherapy response in hepatocellular carcinoma". Oncology Letters 30, no. 6 (2025): 540. https://doi.org/10.3892/ol.2025.15286
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team