International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Kidney cancer remains one of the most prevalent and deadliest tumors in urological oncology, with ~155,702 mortalities and 434,419 new cases recorded worldwide in 2022 (1). Clear cell renal cell carcinoma (ccRCC) accounts for >70% of all kidney cancer cases and is the most common subtype (2). ccRCC typically exhibits aggressive growth, frequent metastasis and substantial immune infiltration, with ~30% of patients experiencing recurrence after total nephrectomy (3,4). Conventional radiation therapy and chemotherapy have demonstrated limited efficacy in the treatment of all renal cell carcinoma subtypes (5). Immunotherapy and targeted therapy have shown considerable potential in patients with ccRCC. However, owing to low response rates, immunotherapy may not be suitable for all individuals (6). In addition, patients receiving targeted therapy are prone to resistance, limiting the efficacy of existing treatments and complicating ccRCC control (7). The outcomes of patients with ccRCC are primarily affected by drug resistance and metastasis (8). Therefore, to effectively treat metastatic ccRCC, it is clinically important to identify the most preferable biomarkers and immune-related therapeutic targets.
Human guanylate-binding protein 5 (GBP5), which belongs to the guanylate-binding protein (GBP) family, is classified under the dynamin superfamily of large GTPases that are induced by interferons (9,10). It is regarded as a key regulator of immune responses in neoplastic diseases (9). Studies have shown that GBP5 exerts antiviral activity and influences innate immunity and inflammation (11,12). Due to the notable functions of other members of the GBP family in proliferation and invasion, GBP5 may also serve a major role in malignancy (13). Previous studies have supported the key role of GBP5 in cancer progression. In gastric adenocarcinomas, GBP5 expression is markedly upregulated, contributing to cancer development through the Janus kinase 1 (JAK1)-STAT1/GBP5/C-X-C motif chemokine ligand 8 (CXCL8) signaling circuit (14,15). In addition, GBP5 enhances the migration, invasion and proliferation of glioblastoma cells, promoting glioblastoma progression through the proto-oncogene tyrosine-protein kinase Src/ERK1/2/MMP3 pathway (16). Furthermore, in triple-negative breast cancer cells exhibiting high GBP5 expression, GBP5 knockdown notably reduces the number of migrating cells, the activity of the IFN-γ/STAT1 and TNF-α/NF-κB signaling axes and the expression of programmed death-ligand 1 (PD-L1) (17).
Previous reports have outlined the relevance of GBP5 in tumor immunity. Several studies have indicated that GBP5 expression is associated with the extent of immune cell infiltration in breast cancer (17), melanoma (18), colon cancer (19), ovarian cancer (20), hepatocellular cancer (21) and small-cell lung cancer (22), emphasizing the role of GBP5 in shaping the tumor immune microenvironment (TME). In oral squamous cell carcinoma, GBP5 serves as an immune-related biomarker that induces NF-κB activation and facilitates radioresistance, PD-L1 upregulation and tumor metastasis (23). In hepatocellular carcinoma and ovarian cancer, GBP5 not only activates the PI3K-AKT signaling pathway but also modulates tumor metabolism and immune evasion through the regulation of DNA methylation and micro-RNA networks (24,25). Furthermore, in cutaneous melanoma and ovarian cancer, GBP5 induces pyroptosis through the JAK2-STAT1-caspase-1 axis, therefore influencing cell death and immune responses. Overall, GBP5 modulates the immune microenvironment which further influences the prognosis of diverse tumor types (20,26). In ccRCC, immune-suppressive cells, such as regulatory T cells and myeloid-derived suppressor cells, induce immune dysfunction, leading to poor therapeutic outcomes (27). However, the precise function of GBP5 is still not fully understood in ccRCC.
The present study employed R software together with a variety of bioinformatics tools to integrate and analyze data, aiming to comprehensively investigate the role of GBP5 in ccRCC. Specifically, its differential expression patterns, associations with clinicopathological features, prognostic and diagnostic significance, protein-protein interaction networks, biological functions, and potential involvement in tumor immune cell infiltration were examined. Furthermore, in vitro cellular experiments were performed to validate and further explore the impact of GBP5 on the biological behavior of ccRCC. To the best of our knowledge, the results of the present study demonstrate the biological function and clinical significance of GBP5 and its influence on tumor immunity in ccRCC for the first time. Fig. 1 illustrates the study design and workflow, highlighting the step-by-step process from data acquisition to experimental validation. The present study provides novel insights into the function of GBP5 in ccRCC, potentially offering new biomarkers and therapeutic targets for the diagnosis and treatment of ccRCC.
GEPIA2 (http://gepia2.cancer-pku.cn/) analyzes gene expression differences based on tumor samples and normal samples from both The Cancer Genome Atlas (TCGA; http://portal.gdc.cancer.gov) and the Genotype-Tissue Expression (https://www.gtexportal.org) databases (28). In the present study, this tool facilitated the comparison of GBP5 expression between renal tumor tissue and adjacent normal tissue.
From the GEO database (http://www.ncbi.nlm.nih.gov/geo), two datasets were acquired. GSE53757 (29) included GBP1-6 mRNA data from 72 ccRCC and 72 normal tissues. GSE36895 (30) provided GBP1-6 mRNA data from 23 ccRCC samples and 23 normal tissues. Analysis of GBP1-6 mRNA levels was conducted using these two datasets.
As an online tool, the KM plotter (https://kmplot.com/analysis) provided survival analysis capabilities for 21 tumor types by associating gene expression profiles with clinical prognosis (31). Using this resource, the KIRC dataset was selected in the pan-cancer module, and the ‘auto select best cutoff’ option was applied for patient grouping to evaluate the overall survival of GBP1-7.
GeneMANIA (http://www.genemania.org) is a publicly accessible tool for analyzing genetic and protein interactions, co-expression, pathways and gene co-localization (32). The potential interactions of GBP5 with related genes were assessed using this platform.
TIMER (https://cistrome.shinyapps.io/timer/) is based on six algorithms used to evaluate immune infiltration levels (33). The impact of GBP5 expression on immune cell infiltration in kidney renal clear cell carcinoma (KIRC) was analyzed in the present study. TIMER2.0 (http://timer.comp-genomics.org/) was also employed to investigate GBP5 expression differences between tumor tissue and adjacent tissue, alongside its association with immune cell gene markers in KIRC (34).
Based on single-cell RNA sequencing, the TISCH2 database (http://tisch.comp-genomics.org/) is designed to analyze the TME at single-cell resolution (35). The distribution and expression patterns of GBP5 in the TME were analyzed using the GSE111360 (36) and GSE121636 (37) datasets.
TCGA-KIRC RNA-sequencing data were downloaded from TCGA database. Transcript per million format expression data and the corresponding clinical information were extracted. Boxplots illustrating GBP5 expression across different clinicopathological features, including tumor-lymph node-metastasis (TNM) stages, were generated using the ‘ggplot2 v3.3.5’ R package (RStudio, Inc.). Receiver operating characteristic (ROC) curves were plotted to assess the diagnostic value of GBP5 in ccRCC. Based on the criteria of log2 fold change ≥2 and the adjusted P-value of <0.05, GBP5-related differentially expressed genes (DEGs) were identified and visualized through a volcano plot plotted using the aforementioned ‘ggplot2’ R package. Gene Ontology (GO) biological process enrichment analysis was performed using the DAVID database (https://david.ncifcrf.gov/tools.jsp). Gene set enrichment analysis (GSEA) was conducted to explore the relevant molecular signaling pathways. Data from the Tracking Tumor Immunophenotype (TIP) database (http://biocc.hrbmu.edu.cn/TIP) were used for immune cell infiltration analysis (38). The relationship between GBP5 expression and immune cell infiltration and circulation was assessed using Spearman's rank correlation analysis.
A total of eleven pairs of clinical specimens were obtained from the University of Hong Kong-Shenzhen Hospital (Shenzhen, P.R. China) between August 2024 and November 2024. Patients were eligible if no radiotherapy or chemotherapy had been administered prior to biopsy, the pathological diagnosis confirmed ccRCC and no other malignant tumors were present. The Ethics Committee of the University of Hong Kong-Shenzhen Hospital approved the collection of renal carcinoma tumor tissues and adjacent normal tissues (approval no. 2024256), ensuring that all procedures complied with the ethical guidelines of the Declaration of Helsinki. All the donors signed an informed consent form.
Human ACHN, Caki-1, 786-O and human proximal tubular epithelial (HK-2) cell lines were procured from Procell Life Science & Technology Co., Ltd. RPMI 1640 complete medium (Procell Life Science & Technology Co., Ltd.) was used to culture the 786-O and ACHN cells. For Caki-1 cells, McCoy's 5A complete medium (Procell Life Science & Technology Co., Ltd.) was used. For HK-2 cells, Dulbecco's modified eagle medium (Gibco; Thermo Fisher Scientific, Inc.) was utilized. A 10% fetal bovine serum solution (FBS; Procell Life Science & Technology Co., Ltd.) and 1% antibiotics solution (100 µg/ml streptomycin and 100 U/ml penicillin) were added to prepare the complete medium. Cultures were placed at 37°C in a 5% CO2 incubator, Mycoplasma contamination was checked in all cell lines and short tandem repeats analysis was used for authentication.
Lentivirus overexpression, GBP5 overexpression (GBP5 OE; 47597-2; order of vector elements: Ubi-MCS-3FLAG-SV40-puromycin), vector controls (CON254), GBP5 short hairpin RNA (shRNA; order of vector elements: hU6-MCS-CBh-gcGFP-IRES-puromycin) and negative control shRNAs (shNC) were produced by Shanghai GeneChem Co., Ltd. The target sequences of the shRNAs were as follows: shNC (CON313), 5′-TTCTCCGAACGTCACGT-3′; the GBP5-specific shRNA shGBP5#1 (103584–1), 5′-CTGGAAATAGATGGGCAACTT-3′ and the GBP5-specific shRNA shGBP5#2 (103585–1), 5′-TGCCTCATCGAGAACTTTAAT-3′. Caki-1 and 786-O cells were transfected according to manufacturer's instructions. Cells infected with lentivirus were treated with puromycin (2 µg/ml) for 7 days, followed by evaluation of target gene knockdown and overexpression efficiency using western blot analysis. The stable cell lines obtained after successful selection and maintained within 50 passages were used for subsequent experiments.
Tissue and cellular protein lysates were prepared using radioimmunoprecipitation assay buffer (cat. no. P0013B; Beyotime Institute of Biotechnology) supplemented with protease and phosphatase inhibitors (cat. no. P1048, Beyotime Institute of Biotechnology). Protein concentrations were determined using the bicinchoninic acid method (cat. no. P0012; Beyotime Institute of Biotechnology). The proteins were resolved using 8–10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis, the proteins (10–50 µg) were then transferred on to nitrocellulose membranes (cat. no. 66485; Pall Life Sciences). The membranes were blocked with 5% skimmed milk for 1 h at 20–25°C, after which the membranes were incubated at 4°C for 8–12 h with primary antibodies against CD163 (1:1,000; cat. no. R381830; Chengdu Zen-BioScience Co., Ltd.), GAPDH (1:5,000; cat. no. D110016-0100; Sangon Biotech Co., Ltd.), β-actin (1:5,000; cat. no. D110022-0100; Sangon Biotech Co., Ltd.), Flag (1:5,000; cat. no. 20543-1-AP; Proteintech Group, Inc.) and GBP5 (1:2,000; cat. no. 13220-1-AP; Proteintech Group, Inc.). The membranes were incubated with secondary antibodies [horseradish peroxidase (HRP)-conjugated goat anti-rabbit IgG; 1:5,000; cat. no. 511203; Chengdu Zen-BioScience Co., Ltd.] and HRP-conjugated goat anti-mouse IgG (1:5,000; cat. no. 511103; Chengdu Zen-BioScience Co., Ltd.) at 20–25°C for 2 h. Protein signals were detected using the ChemiDoc™ MP enhanced chemiluminescence system (Bio-Rad Laboratories, Inc.) and quantified using the ImageJ software (version 1.40 g; National Institutes of Health).
786-O and Caki-1 cells were seeded in 12-well plates and cultured until they reached full confluence. A 1,000 µl pipette tip was used to create a linear scratch in the cell monolayer. The wells were rinsed twice with PBS to remove detached cells and the adherent cells were maintained in a medium containing 1% FBS. Images of the wound area were captured at 0, 10, 12, 24 and 36 h using time-lapse imaging under a light microscope (magnification, ×4).
Transwell assay was performed to assess the migratory potential of ccRCC cells. Cell migration was evaluated using uncoated Transwell chambers (cat. no. 14341, 8 µm pore size; LABSELECT®; Lanjieke Technology Co., Ltd.) and cell invasion was assessed using Matrigel-coated inserts (Corning, Inc.) pre-incubated at 37°C for 30 min. A total of 3×104 786-O and Caki-1 cells were seeded into the upper chambers of 24-well plates containing serum-free medium. Culture medium supplemented with 15% FBS was added to the lower chambers, making up 500 µl as a chemoattractant. After incubation at 37°C for 24 h for the invasion assay and 12 h for the migration assay, the cells that had moved to the lower surface of the membrane were fixed with 4% paraformaldehyde at 20–25°C for 30 min, followed by crystal violet staining at 20–25°C for 10 min. Following staining, the cells were imaged using a light microscope (magnification, ×10) and cell counts were performed in three or more randomly selected fields of view.
GraphPad Prism (version 7.0; GraphPad; Dotmatics) was used for statistical analyses. Each experiment was performed in triplicate. Continuous variables are presented as mean ± standard deviation. Both unpaired and paired two-tailed Student's t-tests were used for comparisons between groups. For comparisons involving multiple groups, Tukey's post hoc test was applied after one-way ANOVA. Spearman's or Pearson's correlation coefficients were determined, depending on suitability, to perform correlation analyses. P<0.05 was considered to indicate a statistically significant difference. Additionally, the following criteria indicated statistical significance: *P<0.05, **P<0.01, ***P<0.001 and ****P<0.0001.
Research suggests that the GBP gene family members regulate the biological behavior of various cancers, indicating their role in cancer initiation and progression (12). Therefore, the GEPIA2.0 tool was initially applied to assess the expression of the GBP family members in ccRCC. Results revealed that GBP1-7 exhibited a consistent expression pattern, characterized by upregulation in tumor tissues (Fig. 2A-G). Notably, the upregulation of GBP1, GBP2 (39), GBP4 and GBP5 was statistically significant. This observation was consistent with the results of the analysis of ccRCC datasets from the GEO database, further reinforcing their validity (Fig. 2H and I). Furthermore, the Kaplan-Meier survival analysis results indicated that elevated levels of GBP2 (39) and GBP5 were associated with a poor prognosis, whereas low expression of GBP3, GBP4 and GBP7 was associated with unfavorable outcomes (Fig. 2J-P). Collectively, these findings support the hypothesis that the GBP gene family is critically involved in ccRCC, with individual members exhibiting distinct clinical relevance. Research specifically targeting GBP5 in ccRCC has been limited. Therefore, the present study focused on elucidating the function of GBP5 in ccRCC.
To comprehensively assess the expression profile of GBP5 across multiple tumor types, the TIMER database was employed. Analysis showed a marked increase in GBP5 expression across a wide range of malignancies, including KIRC and kidney renal papillary cell carcinoma (Fig. 3A). To investigate the clinical relevance of GBP5 in ccRCC, an in-depth analysis was performed using transcriptomic and clinical data from TCGA. Elevated GBP5 expression was significantly associated with notable clinicopathological factors, including tumor size (T stage), lymph node involvement (N stage), distant metastasis (M stage) and both clinical and pathological stage classifications (Fig. 3B-E). Notably, GBP5 expression increased progressively with advancing tumor stage. In addition, univariate Cox regression analysis indicated that elevated GBP5 expression was associated with unfavorable clinical characteristics in patients with ccRCC (Fig. 3F). To evaluate the diagnostic potential of GBP5 in ccRCC, ROC curve analysis was performed, revealing a good diagnostic efficacy with an area under the curve (AUC) value of 0.934 (Fig. 3G), suggesting high sensitivity and specificity. These results imply that GBP5 is a promising diagnostic biomarker involved in the progression of ccRCC.
To further investigate the potential biological function of GBP5 in ccRCC, DEG analysis was performed and the results were visualized using a volcano plot (Fig. 4A). Notably upregulated and downregulated genes were identified using the criteria: Log2 fold change >2 and adjusted P-value <0.05. Enrichment analysis was performed on these DEGs, revealing significant associations with various GO terms. Specifically, GO biological process analysis revealed significant enrichment in ‘humoral immune response’, ‘positive regulation of cell activation’, ‘positive regulation of leukocyte activation’ and ‘immune response-regulation signaling pathways’ (Fig. 4B). The enriched GO cellular component terms were the ‘immunoglobulin complex’, ‘external side of plasma membrane’, ‘plasma membrane signaling receptor complex’ and ‘T cell receptor complex’-related components (Fig. 4C). Regarding GO molecular functions, enrichment was observed in functions such as ‘antigen binding’, ‘immunoglobulin receptor binding’, ‘passive transmembrane transporter activity’ and ‘channel activity’-associated functions (Fig. 4D).
Subsequently, GeneMANIA was used to build a protein-protein interaction network for GBP5 in order to identify interacting proteins potentially involved in tumorigenesis. Results indicated that GBP5 exhibited strong co-expression with proteins such as GBP1, GBP2, NLR family pyrin domain containing 3 (NLRP3) and GBP4 (Fig. 4E), implying a potential interaction between GBP5 and these genes, which could promote tumor progression.
To further elucidate the biological pathways regulated by GBP5 across the high and low expression groups, GSEA was performed. As shown in Fig. 4F, several biological pathways showed notable enrichment in the high GBP5 expression group, including IFN-γ response, IL-6/JAK/STAT3 signaling, inflammatory response, G2/M damage checkpoint and TNF-α signaling through NF-κB. By contrast, pathways such as oxidative phosphorylation, fatty acid metabolism, adipogenesis, estrogen response late and the p53 pathway were enriched in the low GBP5 expression group (Fig. 4G). According to these findings, GBP5 may influence ccRCC progression through mechanisms related to tumor immunity, cell cycle regulation and inflammation.
To investigate whether GBP5 affects immune cell infiltration in ccRCC, the TIMER online tool was used to explore the relationship between GBP5 expression and the abundance of various immune cell types. In ccRCC, a strong positive correlation was observed between GBP5 expression and the levels of immune cell infiltration, including B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells (Fig. 5A). These findings suggest that GBP5 serves a role in immune cell infiltration in ccRCC. TIMER2.0 was then utilized to conduct a deeper analysis of the association between GBP5 expression and marker genes of various immune cells (Table I). The data revealed strong positive correlations between GBP5 and marker genes of T cells, CD8+ T cells, M2 macrophages and exhausted T cells, further supporting the involvement of GBP5 in immune regulation in ccRCC.
Table I.Correlation analysis between guanylate-binding protein 5 and immune cell markers in the Tumor Immune Estimation Resource 2.0 database. |
To validate the relationship between GBP5 expression and immune cell infiltration in ccRCC, two single-cell RNA datasets of ccRCC were analyzed from the TISCH2 database. GBP5 expression was assessed in different TME-associated cell subsets. The results demonstrated that in both KIRC GSE111360 and GSE121636 datasets, GBP5 was highly expressed in CD4+ T cells, proliferating T cells, regulatory T cells, CD8+ T cells and natural killer (NK) cells (Fig. 5B and C).
Further analysis of the immunological characteristics of GBP5 in ccRCC was conducted. Results indicated a strong positive correlation between GBP5 and various immune cells, including macrophages, NK cells and CD8+ T cells (Fig. 5D). To validate the findings from public databases, western blotting of tumor and adjacent normal tissues from 11 patients with ccRCC was performed. In ccRCC tissues, the levels of GBP5 and CD163 were markedly higher compared with those in adjacent normal tissues. The relative expression levels of GBP5 and CD163 exhibited a positive correlation (Fig. 5E). These findings underscore the association between GBP5 and M2 macrophages and suggest a potential interaction between them.
Several in vitro experiments were performed to investigate the role of GBP5 in ccRCC cells. Western blotting was performed to assess GBP5 expression in Caki-1, 786-O, ACHN and HK-2 cells. Findings demonstrated that GBP5 protein levels were markedly higher in renal cancer cell lines compared with HK-2 cells (Fig. 6A). For subsequent experiments, 786-O and Caki-1 cell lines were selected.
The efficiency of lentivirus-mediated gene silencing was evaluated using western blotting. As shown in Fig. 6B, two lentiviral constructs carrying different shRNA sequences targeting GBP5 were tested in Caki-1 and 786-O cells. Both shRNA constructs effectively knocked down GBP5 expression, with shGBP5#1 showing improved knockdown efficiency. Thus, the shGBP5#1 lentivirus was selected to establish stable GBP5 knockdown cell lines. Similarly, GBP5 OE cell lines were generated using a lentiviral system. Following the exogenous introduction of GBP5 into 786-O and Caki-1 cells, western blotting analysis confirmed a significant upregulation of GBP5 expression (Fig. 6C).
To explore the biological function of GBP5 in cancer progression, a wound healing assay was performed. Notably, the suppression of GBP5 expression significantly decreased the migratory capacity of the cells (Fig. 7A). By contrast, GBP5 overexpression enhanced the wound-closure rate (Fig. 7B). These findings were consistently observed in Caki-1 and 786-O cell lines, indicating that the GBP5-mediated regulation of cell migration is not cell-type specific.
The Transwell migration assays demonstrated that GBP5 knockdown inhibited cell migration, whereas GBP5 overexpression significantly promoted it (Fig. 7C and D). In addition, the Matrigel-coated Transwell invasion assays demonstrated that GBP5 depletion markedly suppressed the invasive capabilities of Caki-1 and 786-O cells, whereas the overexpression of GBP5 exerted the opposite effect, significantly enhancing cell invasion (Fig. 7C and D).
ccRCC is the predominant subtype of RCC. It is associated with a poor prognosis and also lacks effective biomarkers. The primary cause of ccRCC-related mortality is the low rate of early detection and scarcity of effective treatments for patients with advanced or metastatic disease (40,41). Consequently, it is key to identify reliable biomarkers for early detection and prognosis, along with the development of innovative therapeutic approaches to enhance the clinical outcomes of ccRCC.
GBP5, which is induced by IFN-γ and classified within the large GTPase family, is one of seven GBPs with strong sequence homology that contribute to host defense mechanisms (42). The function of GBP5 in cancer, particularly ccRCC, remains largely unclear. In the present study, GBP5 was recognized as a potential immunotherapeutic target, exhibiting upregulation in several types of cancer. Previous studies have highlighted the involvement of GBP5 in the progression of oral squamous cell carcinoma (23), hepatocellular carcinoma (43), gastric cancer (15) and glioblastoma (16). In the present study, GBP5 was characterized as a novel biomarker for ccRCC involved in the tumorigenesis and progression of this malignancy. Findings show that GBP5 expression was notably increased in ccRCC tissues and was also associated with a poor prognosis. With this, the results of the bioinformatics analysis demonstrated that GBP5 exhibited stage- and grade-specific expression, making it a useful biomarker for diagnosing and predicting the prognosis of patients with ccRCC. However, GBP5 has been reported to serve an opposite role in ovarian cancer (44), likely due to the complexity of ovarian tumorigenesis and the organ-specific characteristics, such as lineage-specific transcription factors (45).
Cancer progression involves a complex array of immune evasion mechanisms that collectively shape the immunological characteristics of the TME. Tumor-associated immune cells have been explored as potential therapeutic biomarkers for cancer (46). In response to pathogen- or danger-associated molecular patterns, GBP5 serves a key role in the activation of the NLRP3 inflammasome (47). It primarily mediates inflammation and activates macrophages in the innate immune system (48). Previous studies have suggested that GBP5 may be involved in tumor immunity (17–22). The efficacy of both targeted therapies and immunotherapies is influenced by the TME immune landscape (49). Various immune cell types such as T cells, regulatory tumor-associated macrophages (TAMs), T regulatory cells (Tregs), cancer-associated fibroblasts and myeloid-derived suppressor cells, serve a role in the TME and contribute to immune cell infiltration, which is regulated by tumor and immune-modulating factors (50,51). Associations between GBP5 expression and several immune cell markers were observed, including CD8+ T cells, Tregs, T helper type 1 cells, exhausted T cells, M2 macrophages and monocytes. Patients with ccRCC showing elevated programmed cell death protein 1 expression alongside CD8+ T cell and Treg infiltration tended to have worse clinical outcomes (52–54). Tregs may contribute to tumor progression and immune escape by interacting with the TME and suppressing antigen-presenting cell maturation (55). Although CD8+ T cell infiltration is elevated in the ccRCC microenvironment, it does not appear to enhance prognosis, most likely because of functional T cell exhaustion (56). Moreover, analysis revealed a positive association between GBP5 expression and CD163, an established marker of M2-polarized TAMs. M2-like TAMs are commonly found in tumors and facilitate tumor progression, cell proliferation, angiogenesis, invasion and metastasis by secreting diverse cytokines and chemokines (57–60). These findings indicate that GBP5 is a possible driver of ccRCC progression through TAMs.
Cancer metastasis is the primary cause of mortality in patients with cancer (61). Regarding cancer biology, GBP5 has been implicated in various malignant behaviors, including tumor invasion, migration, epithelial-mesenchymal transition, cell proliferation and supporting properties of cancer stem cells (16,23). Similarly, GBP5 was found to be involved in the regulation of renal cancer cell invasion and migration, perhaps explaining why patients with high GBP5 expression in ccRCC exhibit a poor prognosis. With regards to the molecular mechanisms underlying the function of GBP5 in tumorigenesis, previous research has demonstrated that GBP5 might enhance tumor progression in breast and oral cancers by upregulating PD-L1 expression and activating NF-κB signaling (17,23). Additionally, GBP5 accelerates gastric cancer development by engaging in a JAK1-STAT1/GBP5/CXCL8-mediated positive feedback circuit (15). In the present study, through bioinformatic approaches, GBP5 was found to associate with key oncogenic signaling cascades such as the INF-γ response, IL-6/JAK/STAT3 axis and the TNF-α/NF-κB network. These pathways are recognized for their essential roles in both cancer development and immune regulation (62,63). However, the precise molecular mechanisms by which GBP5 modulates these pathways in RCC warrants further investigation.
The present study has some limitations. Firstly, the publicly available ccRCC datasets used in the analyses were limited in scope, which may have introduced potential errors or biases. Secondly, while the pro-migratory and pro-invasive roles of GBP5 in renal cancer cells have been confirmed in vitro, the underlying molecular mechanisms remain unexplored and no in vivo validation has been performed. Finally, although GBP5 appears to be related to tumor immune cell infiltration, the present study lacks supporting in vitro and in vivo experimental evidence. These limitations must be considered when interpreting the findings and they will be addressed in future studies to provide a deeper understanding of the role of GBP5 in immune cell infiltration and metastasis in ccRCC.
Overall, using bioinformatic analyses and in vitro experiments, the upregulation of GBP5 in ccRCC was confirmed. Moreover, immune cell infiltration in ccRCC is associated with the upregulation of GBP5. In addition, GBP5 facilitates renal cancer cell migration and invasion, suggesting its usefulness as a prognostic biomarker for poor clinical outcomes and a promising immunotherapeutic target in ccRCC.
We gratefully acknowledge the Clinical, Translational and Basic Research Laboratory of the University of Hong Kong-Shenzhen Hospital for providing infrastructure support and research resources essential to this study.
The present study was supported by the Medical Scientific Research Foundation of Guangdong (grant no. B2023384).
The data generated in the present study may be requested from the corresponding authors.
XC, SJY, XX and CL participated in the study design, conducted the experiments, collected and analyzed the data and drafted the initial manuscript. Bioinformatics analysis and some experimentation was performed by XC, SJY, SY, WZ and ZL. SJY and CL were also in charge of revising the manuscript, supervising the study and offering technical experimental support. XC, SJY, XX and CL confirm the authenticity of all the raw data. All authors were involved in preparing, proofreading and submitting the manuscript. All authors read, confirmed and approved the final version of the manuscript, and are accountable for the manuscript's content.
The collection of renal carcinoma tumor tissues and adjacent normal tissues was approved by the Ethics Committee of the University of Hong Kong-Shenzhen Hospital (approval no. 2024256) and was performed in accordance with the guidelines of the Declaration of Helsinki. All donors signed informed consent forms.
Not applicable.
The authors declare that they have no competing interests.
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI | |
|
Shuch B, Amin A, Armstrong AJ, Eble JN, Ficarra V, Lopez-Beltran A, Martignoni G, Rini BI and Kutikov A: Understanding pathologic variants of renal cell carcinoma: Distilling therapeutic opportunities from biologic complexity. Eur Urol. 67:85–97. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Braun DA, Hou Y, Bakouny Z, Ficial M, Sant' Angelo M, Forman J, Ross-Macdonald P, Berger AC, Jegede OA, Elagina L, et al: Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma. Nat Med. 26:909–918. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Choueiri TK and Motzer RJ: Systemic therapy for metastatic renal-cell carcinoma. N Engl J Med. 376:354–366. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Maines F, Caffo O, Veccia A, Trentin C, Tortora G, Galligioni E and Bria E: Sequencing new agents after docetaxel in patients with metastatic castration-resistant prostate cancer. Crit Rev Oncol Hematol. 96:498–506. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Choueiri TK, Fishman MN, Escudier B, McDermott DF, Drake CG, Kluger H, Stadler WM, Perez-Gracia JL, McNeel DG, Curti B, et al: Immunomodulatory activity of nivolumab in metastatic renal cell carcinoma. Clin Cancer Res. 22:5461–5471. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Abah MO, Ogenyi DO, Zhilenkova AV, Essogmo FE, Ngaha Tchawe YS, Uchendu IK, Pascal AM, Nikitina NM, Rusanov AS, Sanikovich VD, et al: Innovative therapies targeting drug-resistant biomarkers in metastatic clear cell renal cell carcinoma (ccRCC). Int J Mol Sci. 26:2652024. View Article : Google Scholar : PubMed/NCBI | |
|
Linehan WM and Ricketts CJ: Decade in review-kidney cancer: Discoveries, therapies and opportunities. Nat Rev Urol. 11:614–616. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Praefcke GJK and McMahon HT: The dynamin superfamily: Universal membrane tubulation and fission molecules? Nat Rev Mol Cell Biol. 5:133–147. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Vestal DJ: The guanylate-binding proteins (GBPs): Proinflammatory cytokine-induced members of the dynamin superfamily with unique GTPase activity. J Interferon Cytokine Res. 25:435–443. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Shenoy AR, Wellington DA, Kumar P, Kassa H, Booth CJ, Cresswell P and MacMicking JD: GBP5 promotes NLRP3 inflammasome assembly and immunity in mammals. Science. 336:481–485. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Tretina K, Park ES, Maminska A and MacMicking JD: Interferon-induced guanylate-binding proteins: Guardians of host defense in health and disease. J Exp Med. 216:482–500. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Fellenberg F, Hartmann TB, Dummer R, Usener D, Schadendorf D and Eichmüller S: GBP-5 splicing variants: New guanylate-binding proteins with tumor-associated expression and antigenicity. J Invest Dermatol. 122:1510–1517. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Patil PA, Blakely AM, Lombardo KA, Machan JT, Miner TJ, Wang LJ, Marwaha AS and Matoso A: Expression of PD-L1, indoleamine 2,3-dioxygenase and the immune microenvironment in gastric adenocarcinoma. Histopathology. 73:124–136. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Cao FY, Wang CH, Li X, Ma MZ, Tao GC, Yang C, Li K, He XB, Tong SL, Zhao QC, et al: Guanylate binding protein 5 accelerates gastric cancer progression via the JAK1-STAT1/GBP5/CXCL8 positive feedback loop. Am J Cancer Res. 13:1310–1328. 2023.PubMed/NCBI | |
|
Yu X, Jin J, Zheng Y, Zhu H, Xu H, Ma J, Lan Q, Zhuang Z, Chen CC and Li M: GBP5 drives malignancy of glioblastoma via the Src/ERK1/2/MMP3 pathway. Cell Death Dis. 12:2032021. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng SW, Chen PC, Lin MH, Ger TR, Chiu HW and Lin YF: GBP5 repression suppresses the metastatic potential and PD-L1 expression in triple-negative breast cancer. Biomedicines. 9:3712021. View Article : Google Scholar : PubMed/NCBI | |
|
Deng Z, Liu J, Yu YV and Jin YN: Machine learning-based identification of an immunotherapy-related signature to enhance outcomes and immunotherapy responses in melanoma. Front Immunol. 15:14511032024. View Article : Google Scholar : PubMed/NCBI | |
|
Elsayed I, Elsayed N, Feng Q, Sheahan K, Moran B and Wang X: Multi-OMICs data analysis identifies molecular features correlating with tumor immunity in colon cancer. Cancer Biomark. 33:261–271. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zou C, Shen J, Xu F, Ye Y, Wu Y and Xu S: Immunoreactive microenvironment modulator GBP5 suppresses ovarian cancer progression by inducing canonical pyroptosis. J Cancer. 15:3510–3530. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Xiang S, Li J, Shen J, Zhao Y, Wu X, Li M, Yang X, Kaboli PJ, Du F, Zheng Y, et al: Identification of prognostic genes in the tumor microenvironment of hepatocellular carcinoma. Front Immunol. 12:6538362021. View Article : Google Scholar : PubMed/NCBI | |
|
Tong Q, Li D, Yin Y, Cheng L and Ouyang S: GBP5 expression predicted prognosis of immune checkpoint inhibitors in small cell lung cancer and correlated with tumor immune microenvironment. J Inflamm Res. 16:4153–4164. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Chiu HW, Lin CH, Lee HH, Lu HW, Lin YHK, Lin YF and Lee HL: Guanylate binding protein 5 triggers NF-κB activation to foster radioresistance, metastatic progression and PD-L1 expression in oral squamous cell carcinoma. Clin Immunol. 259:1098922024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen HQ, Zhao J, Li Y, He LX, Huang YJ, Shu WQ, Cao J, Liu WB and Liu JY: Gene expression network regulated by DNA methylation and microRNA during microcystin-leucine arginine induced malignant transformation in human hepatocyte L02 cells. Toxicol Lett. 289:42–53. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Mitra A, Ghosh S, Porey S and Mal C: GBP5 and ACSS3: Two potential biomarkers of high-grade ovarian cancer identified through downstream analysis of microarray data. J Biomol Struct Dyn. 41:4601–4613. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Shi Z, Gu J, Yao Y and Wu Z: Identification of a predictive gene signature related to pyroptosis for the prognosis of cutaneous melanoma. Medicine (Baltimore). 101:e305642022. View Article : Google Scholar : PubMed/NCBI | |
|
Diaz-Montero CM, Rini BI and Finke JH: The immunology of renal cell carcinoma. Nat Rev Nephrol. 16:721–735. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Tang Z, Kang B, Li C, Chen T and Zhang Z: GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res 47 (W1). W556–W560. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
von Roemeling CA, Radisky DC, Marlow LA, Cooper SJ, Grebe SK, Anastasiadis PZ, Tun HW and Copland JA: Neuronal pentraxin 2 supports clear cell renal cell carcinoma by activating the AMPA-selective glutamate receptor-4. Cancer Res. 74:4796–4810. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Peña-Llopis S, Vega-Rubín-de-Celis S, Liao A, Leng N, Pavía-Jiménez A, Wang S, Yamasaki T, Zhrebker L, Sivanand S, Spence P, et al: BAP1 loss defines a new class of renal cell carcinoma. Nat Genet. 44:751–759. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Lánczky A and Győrffy B: Web-based survival analysis tool tailored for medical research (KMplot): Development and implementation. J Med Internet Res. 23:e276332021. View Article : Google Scholar : PubMed/NCBI | |
|
Warde-Farley D, Donaldson SL, Comes O, Zuberi K, Badrawi R, Chao P, Franz M, Grouios C, Kazi F, Lopes CT, et al: The GeneMANIA prediction server: Biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res 38 (Web Server Issue). W214–W220. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, Li B and Liu XS: TIMER: A web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 77:e108–e110. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, Li B and Liu XS: TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res 48 (W1). W509–W514. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
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(D1): D1425–D1431. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, Liu IH, Chiou SH, Salahudeen AA, Smith AR, et al: Organoid modeling of the tumor immune microenvironment. Cell. 175:1972–1988.e16. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Borcherding N, Vishwakarma A, Voigt AP, Bellizzi A, Kaplan J, Nepple K, Salem AK, Jenkins RW, Zakharia Y and Zhang W: Mapping the immune environment in clear cell renal carcinoma by single-cell genomics. Commun Biol. 4:1222021. View Article : Google Scholar : PubMed/NCBI | |
|
Xu L, Deng C, Pang B, Zhang X, Liu W, Liao G, Yuan H, Cheng P, Li F, Long Z, et al: TIP: A web server for resolving tumor immunophenotype profiling. Cancer Res. 78:6575–6580. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ye S, Li S, Qin L, Zheng W, Liu B, Li X, Ren Z, Zhao H, Hu X, Ye N and Li G: GBP2 promotes clear cell renal cell carcinoma progression through immune infiltration and regulation of PD-L1 expression via STAT1 signaling. Oncol Rep. 49:492023. View Article : Google Scholar : PubMed/NCBI | |
|
Cerbone L, Cattrini C, Vallome G, Latocca MM, Boccardo F and Zanardi E: Combination therapy in metastatic renal cell carcinoma: Back to the future? Semin Oncol. 47:361–366. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Braun DA, Bakouny Z, Hirsch L, Flippot R, Van Allen EM, Wu CJ and Choueiri TK: Beyond conventional immune-checkpoint inhibition-novel immunotherapies for renal cell carcinoma. Nat Rev Clin Oncol. 18:199–214. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Santos JC and Broz P: Sensing of invading pathogens by GBPs: At the crossroads between cell-autonomous and innate immunity. J Leukoc Biol. 104:729–735. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Fu J, Qin W, Tong Q, Li Z, Shao Y, Liu Z, Liu C, Wang Z and Xu X: A novel DNA methylation-driver gene signature for long-term survival prediction of hepatitis-positive hepatocellular carcinoma patients. Cancer Med. 11:4721–4735. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zahra A, Dong Q, Hall M, Jeyaneethi J, Silva E, Karteris E and Sisu C: Identification of potential bisphenol A (BPA) exposure biomarkers in ovarian cancer. J Clin Med. 10:19792021. View Article : Google Scholar : PubMed/NCBI | |
|
Arner EN and Rathmell WK: Mutation and tissue lineage lead to organ-specific cancer. Nature. 606:871–872. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Gajewski TF, Schreiber H and Fu YX: Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 14:1014–1022. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Liu P, Ye L, Ren Y, Zhao G, Zhang Y, Lu S, Li Q, Wu C, Bai L, Zhang Z, et al: Chemotherapy-induced phlebitis via the GBP5/NLRP3 inflammasome axis and the therapeutic effect of aescin. Br J Pharmacol. 180:1132–1147. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou L, Zhao H, Zhao H, Meng X, Zhao Z, Xie H, Li J, Tang Y and Zhang Y: GBP5 exacerbates rosacea-like skin inflammation by skewing macrophage polarization towards M1 phenotype through the NF-κB signalling pathway. J Eur Acad Dermatol Venereol. 37:796–809. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wilkie KP and Hahnfeldt P: Tumor-immune dynamics regulated in the microenvironment inform the transient nature of immune-induced tumor dormancy. Cancer Res. 73:3534–3544. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Heidegger I, Pircher A and Pichler R: Targeting the tumor microenvironment in renal cell cancer biology and therapy. Front Oncol. 9:4902019. View Article : Google Scholar : PubMed/NCBI | |
|
Najjar YG, Rayman P, Jia X, Pavicic PJ Jr, Rini BI, Tannenbaum C, Ko J, Haywood S, Cohen P, Hamilton T, et al: Myeloid-derived suppressor cell subset accumulation in renal cell carcinoma parenchyma is associated with intratumoral expression of IL1β, IL8, CXCL5, and Mip-1α. Clin Cancer Res. 23:2346–2355. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Dai S, Zeng H, Liu Z, Jin K, Jiang W, Wang Z, Lin Z, Xiong Y, Wang J, Chang Y, et al: Intratumoral CXCL13+CD8+T cell infiltration determines poor clinical outcomes and immunoevasive contexture in patients with clear cell renal cell carcinoma. J Immunother Cancer. 9:e0018232021. View Article : Google Scholar : PubMed/NCBI | |
|
Giraldo NA, Becht E, Pagès F, Skliris G, Verkarre V, Vano Y, Mejean A, Saint-Aubert N, Lacroix L, Natario I, et al: Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer. Clin Cancer Res. 21:3031–3040. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Giraldo NA, Becht E, Vano Y, Petitprez F, Lacroix L, Validire P, Sanchez-Salas R, Ingels A, Oudard S, Moatti A, et al: Tumor-infiltrating and peripheral blood t-cell immunophenotypes predict early relapse in localized clear cell renal cell carcinoma. Clin Cancer Res. 23:4416–4428. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Paluskievicz CM, Cao X, Abdi R, Zheng P, Liu Y and Bromberg JS: T Regulatory cells and priming the suppressive tumor microenvironment. Front Immunol. 10:24532019. View Article : Google Scholar : PubMed/NCBI | |
|
Braun DA, Street K, Burke KP, Cookmeyer DL, Denize T, Pedersen CB, Gohil SH, Schindler N, Pomerance L, Hirsch L, et al: Progressive immune dysfunction with advancing disease stage in renal cell carcinoma. Cancer Cell. 39:632–648.e8. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mantovani A, Marchesi F, Malesci A, Laghi L and Allavena P: Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 14:399–416. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Pittet MJ, Michielin O and Migliorini D: Clinical relevance of tumour-associated macrophages. Nat Rev Clin Oncol. 19:402–421. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Xiang X, Wang J, Lu D and Xu X: Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct Target Ther. 6:752021. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng W, Ye S, Liu B, Liu D, Yan R, Guo H, Yu H, Hu X, Zhao H, Zhou K and Li G: Crosstalk between GBP2 and M2 macrophage promotes the ccRCC progression. Cancer Sci. 115:3570–3586. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Meirson T, Gil-Henn H and Samson AO: Invasion and metastasis: The elusive hallmark of cancer. Oncogene. 39:2024–2026. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yu H, Pardoll D and Jove R: STATs in cancer inflammation and immunity: A leading role for STAT3. Nat Rev Cancer. 9:798–809. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Johnson DE, O'Keefe RA and Grandis JR: Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 15:234–248. 2018. View Article : Google Scholar : PubMed/NCBI |