Volume 2025, Issue 1 9943789
Research Article
Open Access

Integrative Mendelian Randomization and Transcriptome Analysis Unveil Dual Pathways in Prostate Cancer Etiology

Dongdong Wang

Dongdong Wang

Department of Pharmacy , Quzhou People’s Hospital , The Quzhou Affiliated Hospital of Wenzhou Medical University , Quzhou , China , qzhospital.com

Search for more papers by this author
Mingwei Zhan

Mingwei Zhan

Department of Urology , Jinling Hospital , School of Medicine , Nanjing University , Nanjing , China , nju.edu.cn

Search for more papers by this author
Pengfei Liu

Pengfei Liu

Department of Urology , Jinling Hospital , School of Medicine , Nanjing University , Nanjing , China , nju.edu.cn

Search for more papers by this author
Yi Yu

Corresponding Author

Yi Yu

Department of Reproductive Medicine , The First Affiliated Hospital of Ningbo University , Ningbo , China , nbu.edu.cn

Search for more papers by this author
Rijian Guan

Corresponding Author

Rijian Guan

Department of Urology , Quzhou People’s Hospital , The Quzhou Affiliated Hospital of Wenzhou Medical University , Quzhou , China , qzhospital.com

Search for more papers by this author
First published: 30 April 2025
Academic Editor: Dawei Cui

Abstract

Background: Prostate cancer remains a significant global health challenge, influenced by both genetic and environmental factors. Despite extensive research, the specific metabolic pathways contributing to its pathogenesis are not fully understood.

Methods: We obtained significant single nucleotide polymorphisms (SNPs) associated with 1400 metabolites and 731 immune cells from the GEWAS database and published literature. Using an integrative approach that combines single-cell and bulk transcriptome analysis with Mendelian randomization (MR) and machine learning, we identified significant SNPs related to metabolic and immune profiles, with a specific focus on glutathione metabolism and its downstream metabolite cysteinylglycine disulfide. Advanced statistical techniques, including inverse variance weighted MR, were employed to explore causal relationships between these metabolic markers and prostate cancer risk. Additionally, we investigated the role of ATP6V1G1 in prostate cancer cell lines through RNA interference and various functional assays.

Results: Our MR analysis indicated that elevated cysteinylglycine disulfide levels are protective against prostate cancer. Conversely, higher monocyte counts were associated with increased cancer risk, suggesting a dual pathway influence on disease etiology through immune modulation and metabolic dysregulation. Machine learning algorithms further validated these associations and identified potential biomarkers for prostate cancer susceptibility. In vitro experiments demonstrated that silencing ATP6V1G1 significantly reduced the proliferation and migration of prostate cancer cells, highlighting its oncogenic potential.

Conclusion: The study highlighted a significant association between prostate cancer occurrence, glutathione metabolism, and monocyte activity. The critical interplay between glutathione metabolism and immune cell dynamics in prostate cancer is underscored, proposing novel biomarkers for early diagnosis and potential therapeutic targets. Notably, ATP6V1G1 emerged as a key gene with significant diagnostic and prognostic value, offering new prospects for therapeutic intervention in prostate cancer.

1. Introduction

Prostate cancer is the leading cause of cancer-related deaths among men in Western countries, predominantly affecting men aged 45–60 years [1, 2]. Diagnosis typically involves methods such as prostate-specific antigen (PSA) testing and prostate biopsies, which can be invasive and often lead to overdiagnosis [3, 4]. The etiology of prostate cancer is complex, involving an interplay of genetic, environmental, and lifestyle factors that contribute to its heterogeneous nature and epidemiological disparities across different populations.

Genetic predisposition plays a significant role in prostate cancer, as evidenced by family inheritance and twin studies that highlight the genetic contribution to this disease. Androgen biosynthesis and metabolism, mediated through the androgen receptor pathway, are crucial in the development of prostate epithelium and cancer cells [5]. Understanding these genetic and molecular interactions is critical for developing targeted therapeutic and preventive strategies [6].

This study focused on the metabolic pathways involved in prostate cancer, with a particular emphasis on glutathione metabolism. Glutathione, the most abundant intracellular antioxidant, plays a pivotal role in cellular defense against oxidative stress. Glutathione is a tripeptide composed of L-glutamate, cysteine, and glycine [7]. It exists in both reduced (GSH) and oxidized (GSSG) states, maintaining a balance crucial for cell survival and function. In cancer cells, this balance is often disrupted, leading to an altered redox state that can promote tumor growth and resistance to chemotherapy. Abnormalities in glutathione levels and its related enzymes are linked to the etiology and progression of prostate cancer [8, 9].

Mendelian randomization (MR) is a powerful analytical tool that uses genetic variants as instrumental variables to infer causal relationships between modifiable exposures and clinical outcomes [10, 11]. This approach leverages the random assortment of genes at conception, which mitigates confounding factors typically encountered in observational studies [12]. MR provides a robust framework to explore how specific genetic and metabolic factors, such as those involved in glutathione metabolism, contribute to disease risk and progression.

By integrating MR with comprehensive metabolic profiling, this research aims to delineate the complex genetic and metabolic landscape of prostate cancer. This approach allows us to pinpoint metabolic alterations at the genomic level that could be leveraged for targeted intervention, ultimately paving the way for personalized medicine strategies in oncology.

2. Method

2.1. Data Acquisition and Processing for Mendelian Analysis

We obtained SNP data for 1400 metabolites and 731 immune cells from the GEWAS database, identifying statistically significant SNPs [13, 14]. The filtering criteria were set with p values of 1e − 5 for metabolites and 5e − 5 for immune cells, with additional filters applied to remove SNPs directly impacting prostate cancer while eliminating confounding factors. From the IEU database, we retrieved information on prostate cancer (ICD10: C61, malignant neoplasm of the prostate) with GWAS ID: ukb-b-1392 for subsequent univariate Mendelian analysis.

2.2. Single Factor and Two-Step Mendelian Analysis

We used the TwoSampleMR package to analyze SNPs, which had been filtered to remove confounding factors. Additionally, we performed the MR-PRESSO test for pleiotropy, obtaining exposure factors associated with prostate cancer incidence after multiple hypothesis testing. We downloaded raw data related to cysteinylglycine disulfide levels from the EMBL’s European Bioinformatics Institute and obtained original data on HLA DR++ monocytes from the IEU database online. After intersecting SNPs between these datasets, we conducted a Mendelian analysis to understand the relationship between these factors.

2.3. Single-Cell Data Integration and Cell Type Identification

Single-cell data were sourced from the Gene Expression Omnibus (GEO) database (GSE193337), including both normal and tumor samples. Quality control was rigorously enforced, setting thresholds for mitochondrial genes (≤ 10%), ribosomal genes (≤ 30%), hemoglobin genes (≤ 5%), and the total number of genes per cell (200–4000). Following quality control, data normalization was performed using the “NormalizeData” function. We used the “FindVariableFeatures” function to identify the top three thousand most variable genes, followed by PCA for dimensionality reduction. Sample integration and cell clustering were accomplished using the Harmony algorithm and the KNN approach, respectively.

2.4. Pathway and Transcription Factor Analysis

We extracted the cluster monocyte from the samples and used “FindMarkers” to identify differentially expressed genes between the meta high and meta low groups, setting the logFC threshold to 0. Genes obtained from this analysis were saved for subsequent machine learning tasks, with a log2FC threshold of 0.25 and filtering out ribosomal and red blood cell genes. Next, we performed Gene Set Enrichment Analysis (GSEA) on the two groups, identifying upregulated and downregulated pathways. Further analysis focused on transcription factor activity using Dorothea, identifying the top 20 transcription factors with the greatest changes in each group. Subsequently, we conducted cell communication analysis using the CellCall package, visualizing communication pathways relevant to our monocyte group of interest. A Sankey diagram was created to illustrate communication between the meta high group and plasma cells. Finally, we performed pseudotime analysis on monocytes. We reperformed dimensionality reduction and clustering on the selected cluster and integrated the data using Harmony. The “dims” parameter was set to 15, followed by UMAP visualization. We used feature plots to display glutathione metabolism pathway activity and simulated the monocyte cell development process using the vector method.

2.5. Machine Learning Techniques for Data Analysis

We utilized machine learning algorithms, including CatBoost, NGBoost, and XGBoost, to analyze the dataset further. Models were trained on the GSE46602 dataset and validated on the GSE70768 dataset. We analyzed immune infiltration results using the IOBR package. Then, we established the training model, evaluated the model, calculated the AUC value, and generated ROC curves. This multitier approach allowed us to rigorously test the predictive power of our model and to identify the most significant genes involved in prostate cancer.

2.6. Cell Lines and Culture

This study utilized human prostate cancer cell lines LNCaP, PC3, DU145, and VCaP as well as the normal cell line PrECLH. All cell lines were purchased from the cell bank of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum and incubated at 37°C in a 5% CO2 atmosphere.

2.7. Small Interfering RNA (siRNA) Knockdown

ATP6V1G1 expression was silenced using siRNA. Cells were treated with three different siRNAs (siControl, siATP6V1G1#1, siATP6V1G1#2). siRNA transfections were carried out using Lipofectamine 2000 according to the manufacturer’s guidelines.

2.8. Colony Formation Assay

The proliferative capacity of cells was assessed by a colony formation assay. Treated cells were seeded into 6-well plates at 1000 cells per well. After incubating for 14 days, cells were fixed with formaldehyde and stained with crystal violet. Visible colonies were counted using a microscope.

2.9. Wound-Healing Assay

Cell migration capability was assessed by a scratch assay. Cells were seeded in 6-well plates until confluence and then scratched with the tip of a 200-μL pipette. Floating cells were removed, fresh medium was added, and images were taken at 0 and 24 h to measure the degree of scratch closure using image analysis software.

3. Results

3.1. MR Identifies Protective and Risk Factors for Prostate Cancer

Our analysis via MR revealed that elevated monocyte counts were identified as a potential risk factor, positively correlated with increased prostate cancer risk (b = 0.010, p = 0.046) (Figures 1(a), 1(b), 1(c), 1(d), 1(e)). Conversely, a statistically significant inverse association between cysteinylglycine disulfide levels and the risk of prostate cancer was found (IVW method: b = −0.0024, SE = 0.000635, p = 2.14 × 104), indicating that higher levels of this metabolite may confer protection against the disease. The proportion of variance explained (PVE) was substantial at 46.24%, and the false discovery rate (FDR) was controlled at 4.98%, underlining the robustness of this finding. This finding aligns with the known roles of inflammation and immune surveillance in cancer progression (Figures 1(f), 1(g), 1(h), 1(i), 1(j)). Further mediation analysis indicated that monocytes may elevate prostate cancer risk by decreasing levels of cysteinylglycine disulfide (Figures 1(k), 1(l), 1(m), 1(n), 1(o)). These results collectively underscore the intricate interplay between metabolic dysregulation and immune responses in the etiology of prostate cancer, highlighting potential targets for therapeutic intervention and risk assessment.

Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.
Details are in the caption following the image
Univariate Mendelian and mediated Mendelian outcomes. (a) Mendelian outcomes for monocyte and prostate cancer; (b) monocyte forest plot; (c) monocyte funnel plot; (d) monocyte leave-one-out plot; (e) monocyte scatter plot; (f) Mendelian outcomes for cysteinylglycine disulfide levels and prostate cancer; (g) cysteinylglycine disulfide levels forest plot; (h) cysteinylglycine disulfide levels funnel plot; (i) cysteinylglycine disulfide levels leave-one-out plot; (j) cysteinylglycine disulfide levels scatter plot; (k) Mendelian outcomes for monocyte and cysteinylglycine disulfide levels; (l) forest plot of monocyte and cysteinylglycine disulfide levels; (m) funnel plot of monocyte and cysteinylglycine disulfide levels; (n) leave-one-out plot of monocyte and cysteinylglycine disulfide levels; (o) scatter plot of monocyte and cysteinylglycine disulfide levels.

3.2. Single-Cell Analysis Reveals Differential Pathway Activation

We conducted a comprehensive single-cell analysis using samples obtained from the GEO database, focusing on monocytes to further explore their role in prostate cancer via immune and metabolic pathways. Initial quality control, normalization, and batch correction processes were applied rigorously, utilizing the Harmony algorithm to ensure consistent data quality across samples (Figures 2(a), 2(b)). The dimensionality reduction techniques, t-SNE and UMAP, effectively segregated the cell populations, allowing for detailed visualization and characterization of cellular subtypes within the prostate cancer and control groups (Figures 2(c), 2(d)). The analysis highlighted a notable increase in the proportion of monocytes within the tumor microenvironment compared to normal tissue, suggesting their enhanced role in tumor pathology (Figure 2(e)). We specifically analyzed glutathione and its metabolite, cysteinylglycine, within these cells. Pathway analysis, informed by gene sets from MsigDB, showed distinct metabolic activity profiles; monocytes in the tumor environment exhibited heightened activity in pathways related to glutathione transport, disulfide bond formation, metabolism, conjugation, and recycling. These findings were visualized using bubble plots, which clearly demonstrated the enhanced metabolic pathway activity in monocytes from prostate cancer samples compared to those from normal controls (Figures 2(f), 2(g), 2(h), 2(i), 2(j)). Furthermore, the overall pathway activity across different cell types was quantitatively assessed, with monocytes showing the highest activity particularly in metabolism and disulfide bond formation pathways (Figure 2(k)). The results suggested that monocytes may contribute to the oxidative stress response within the tumor microenvironment through altered glutathione metabolism, potentially affecting the overall progression and therapeutic response of prostate cancer. This enhanced metabolic activity in monocytes could serve as a novel biomarker for early diagnosis and as a potential therapeutic target in prostate cancer management. Further analysis divided cells into two groups, meta high and meta low, focusing on the distribution of other metabolites and visualizing the differences. It was observed that CCl4 was more prevalent in the meta low group (Figures 2(l), 2(m)), suggesting that CCl4 may act as an inhibitor in the meta high group, potentially impeding the monocyte-mediated metabolism of glutathione.

Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.
Details are in the caption following the image
Single-cell analysis and analysis of cysteinylglycine metabolism pathway activity. (a), (b) Single-cell quality control results. (c), (d) t-SNE and UMAP single-cell dimensionality reduction clustering, displaying Seurat cluster. (e) Distribution of immune cells in the normal group and control group. (f) Bubble plot of glutathione transport. (g) Bubble plot of disulfide bond formation. (h) Bubble plot of glutathione metabolism. (i) Bubble plot of glutathione conjugation. (j) Bubble plot of glutathione recycle. (k) Bubble plot of the average activity of each pathway (l), (m) CCl4 expressed in both groups.

3.3. GSEA, Single-Cell Communication, and Transcription Factor Analysis

Continuing with the analysis, the GSEA identified key pathways differentially expressed between the “meta high” and “meta low” monocyte groups (Figure 3(a)). The “meta high” group exhibited upregulation in critical metabolic pathways, including those related to copper, zinc, and calcium ion metabolism, as well as elevated activity in glucose and amino sugar metabolism and lipoprotein clearance pathways. Conversely, the “meta low” group showed a downregulation of pathways involved in inositol lipid–mediated signaling and hydrogen peroxide metabolism, indicating a potential suppression of cellular defense mechanisms against oxidative stress. Transcription factor analysis using the Dorothea database revealed significant differences in the activation of transcription factors between the two groups. In the “meta low” group, PRDM14, a transcription factor known for its tumor-suppressing functions, was the most upregulated, suggesting a potential role in inhibiting tumor progression [15]. In contrast, the “meta high” group showed higher expression levels of MYC, a well-known oncogene, possibly reflecting a higher proliferative capacity and less differentiated state of the tumor cells [16] (Figure 3(b)). Single-cell communication analysis provided insights into the cellular interactions within the tumor microenvironment. Enhanced communication pathways were observed between monocytes in the “meta high” group and other cell types such as epithelial cells, vascular endothelial cells, and smooth muscle cells, suggesting a complex interplay that may promote tumor growth and metastasis (Figures 3(c), 3(d), 3(e), 3(f)). Plasma cells exhibit stronger cell communication with the meta high group, indicating that plasma cells may play a promoting role in the cysteinylglycine metabolism pathway of monocytes. Then, monocyte cells were reclustered (Figures 3(g), 3(h)). Pseudotime analysis sheds light on the developmental trajectory of monocytes within the tumor microenvironment. As monocytes matured, there was a marked increase in the activity of the cysteinylglycine metabolism pathway, indicating a progressive adaptation of these cells as the tumor evolves (Figures 3(i), 3(j), 3(k)). Through feature plot analysis, we observed that the activity of the cysteinylglycine metabolism pathway increases as cells mature. This reflects an enhancement in both monocyte maturity and cysteinylglycine metabolism pathway activity as tumors progress, aligning with our earlier analyses.

Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.
Details are in the caption following the image
GSEA, single-cell CellCall, and pseudotime analysis. (a) GSEA of the meta high and meta low groups; (b) heatmap of differentially expressed genes; (c) heatmap of CellCall; (d) dot plot of CellCall; (e) visualization of CellCall; (f) Sankey diagram between meta high group and plasma cells; (g), (h) reclustering of monocyte cells; (i), (j) activity of the glutathione metabolism pathway; (k) pseudotime analysis of monocytes.

3.4. Bulk Data Analysis and Immune Infiltration Analysis

In the examination of bulk RNA sequencing data, we applied rigorous preprocessing and batch correction techniques to ensure data integrity across the different datasets (Figures 4(a), 4(b)). Subsequent Lasso regression analysis allowed us to identify significant genes that were used as final variables in organizing the training and validation sets (Figures 4(c), 4(d)). The deployment of machine learning techniques, specifically CatBoost, NGBoost, and XGBoost, further validated our findings. Each method was meticulously tested against the training and validation datasets, with the results displaying high AUC values, underscoring their predictive reliability. CatBoost emerged as the most effective, demonstrating the highest AUC value of 0.94, followed by XGBoost and NGBoost with AUC values of 0.88 and 0.86, respectively (Figures 4(e), 4(f), 4(g), 4(h)). The consistency of gene expression profiles across different datasets confirms the robustness of our analytical approach and the clinical relevance of these immune-related gene signatures.

Details are in the caption following the image
Analysis of immune infiltration and its correlation. (a), (b) Batch correction of bulk data; (c), (d) LASOO-selected feature variables; (e) ROC curve of XGBoost; (f) ROC curve of NGBoost; (g) ROC curve of CatBoost; (h) comparison between three methods.
Details are in the caption following the image
Analysis of immune infiltration and its correlation. (a), (b) Batch correction of bulk data; (c), (d) LASOO-selected feature variables; (e) ROC curve of XGBoost; (f) ROC curve of NGBoost; (g) ROC curve of CatBoost; (h) comparison between three methods.
Details are in the caption following the image
Analysis of immune infiltration and its correlation. (a), (b) Batch correction of bulk data; (c), (d) LASOO-selected feature variables; (e) ROC curve of XGBoost; (f) ROC curve of NGBoost; (g) ROC curve of CatBoost; (h) comparison between three methods.
Details are in the caption following the image
Analysis of immune infiltration and its correlation. (a), (b) Batch correction of bulk data; (c), (d) LASOO-selected feature variables; (e) ROC curve of XGBoost; (f) ROC curve of NGBoost; (g) ROC curve of CatBoost; (h) comparison between three methods.
Details are in the caption following the image
Analysis of immune infiltration and its correlation. (a), (b) Batch correction of bulk data; (c), (d) LASOO-selected feature variables; (e) ROC curve of XGBoost; (f) ROC curve of NGBoost; (g) ROC curve of CatBoost; (h) comparison between three methods.
Details are in the caption following the image
Analysis of immune infiltration and its correlation. (a), (b) Batch correction of bulk data; (c), (d) LASOO-selected feature variables; (e) ROC curve of XGBoost; (f) ROC curve of NGBoost; (g) ROC curve of CatBoost; (h) comparison between three methods.
Details are in the caption following the image
Analysis of immune infiltration and its correlation. (a), (b) Batch correction of bulk data; (c), (d) LASOO-selected feature variables; (e) ROC curve of XGBoost; (f) ROC curve of NGBoost; (g) ROC curve of CatBoost; (h) comparison between three methods.
Details are in the caption following the image
Analysis of immune infiltration and its correlation. (a), (b) Batch correction of bulk data; (c), (d) LASOO-selected feature variables; (e) ROC curve of XGBoost; (f) ROC curve of NGBoost; (g) ROC curve of CatBoost; (h) comparison between three methods.

3.5. Screening of Key Genes and Clinical Prognostic Features

Our models highlighted the importance of specific genes, including ATP6V1G1 and CAPG, pinpointed in the NGBoost model as crucial for their predictive capabilities (Figure 5(a)). In the TCGA database, we further analyzed the diagnostic value of the genes, with TXN having the highest AUC (AUC = 0.928), followed by ATP6V1G1 (AUC = 0.925) (Figure 5(b)). The expression levels of different genes showed statistically significant differences between cancerous and adjacent noncancerous tissues (Figure 5(c)). Considering that ATP6V1G1 consistently demonstrated high diagnostic value across various models, we selected ATP6V1G1 as the key gene for further analysis. The Kaplan–Meier (KM) curve indicated that patients with high ATP6V1G1 expression have a poorer prognosis (Figure 5(d)). Additionally, ATP6V1G1 was significantly overexpressed in advanced T and N stages (Figures 5(e), 5(f)). In terms of progression-free interval (PFI), ATP6V1G1 expression was higher in patients who have died (Figure 5(g)). Figures 5(h), 5(i) demonstrate the differential expression of ATP6V1G1 in prostate cancer tissues and adjacent normal tissues via immunohistochemistry (IHC). The staining conferred a significant overexpression of ATP6V1G1 in prostate cancer tissues compared to normal tissues. Western blot analysis of prostate cancer cell lines (PC-3 and DU-145) and the normal prostate epithelial cell line (RWPE-1) further corroborates these findings, showing elevated ATP6V1G1 protein levels in the cancer cell lines. GAPDH was used as the loading control (Figure 5(j)). In summary, ATP6V1G1 was a key gene with significant diagnostic and prognostic value in cancer.

Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.
Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.
Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.
Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.
Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.
Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.
Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.
Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.
Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.
Details are in the caption following the image
Screening of key genes and clinical prognostic features. (a) Importance of genes identified by the NGBoost model, highlighting ATP6V1G1 and CAPG; (b) ROC curve for ATP6V1G1 in the TCGA database with high diagnostic accuracy; (c) expression levels of key genes in cancerous versus noncancerous tissues; (d) Kaplan–Meier survival curve showing higher ATP6V1G1 expression correlates with poorer prognosis; (e), (f) ATP6V1G1 expression in different T and N stages; (g) higher ATP6V1G1 expression in patients who have died, indicating prognostic value; (h) ATP6V1G1 expression in normal prostate tissue by IHC; (i) ATP6V1G1 expression in prostate cancer tissue by IHC; (j) western blot analysis of ATP6V1G1 in prostate cancer cell lines (PC-3, DU-145) and normal cell line (RWPE-1), with GAPDH as the loading control.

3.6. Immunological Characteristics of ATP6V1G1

To further analyze the immunological characteristics of ATP6V1G1, we divided the samples into high- and low-expression groups based on the median expression level. Using the ssGSEA algorithm, we found that most immune cells were significantly overexpressed in the low-expression group, and ATP6V1G1 showed a significant negative correlation with most immune cells (Figures 6(a), 6(b)). Additionally, most immune checkpoints were significantly overexpressed in the low-expression group, with ATP6V1G1 showing a significant negative correlation with these checkpoints (Figures 6(c), 6(d)). Tumor immune phenotype tracking, which involves monitoring and analyzing the immune characteristics of tumors over time, is crucial in tumor treatment and disease progression. We observed that the low-expression group exhibited higher expression levels across most steps compared to the high-expression group (Figure 6(e)). Furthermore, in the immune function scores, the low-expression group showed higher expression (Figure 6(f)). In the tumor microenvironment scores, the high-expression group exhibited significantly higher expression in tumor purity but lower expression in StromalScore, ImmuneScore, and ESTIMATEScore (Figure 6(g)). Given the critical role of immune checkpoints in determining immunotherapy response, we used the TCIA database and found that CTLA4 and PD1 immunotherapy were more effective in the low-expression group (Figures 6(h), 6(i), 6(j)). Immunotyping assessment showed that ATP6V1G1 had the highest expression in C4 (lymphocyte depleted) (Figure 6(k)). C4 indicates low levels of lymphocyte infiltration and high cell cycle activity, potentially reflecting an immunosuppressive microenvironment where tumor cells can grow more easily and evade immune surveillance. Therefore, ATP6V1G1 emerges as a promising target for immunotherapy, offering new prospects and pathways for advancing immunotherapeutic strategies.

Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.
Details are in the caption following the image
Immunological characteristics of ATP6V1G1. (a) Immune cell expression levels in high versus low ATP6V1G1 groups; (b) negative correlation between ATP6V1G1 and immune cells; (c) immune checkpoint expression in high versus low ATP6V1G1 groups; (d) negative correlation between ATP6V1G1 and immune checkpoints; (e) tumor immune phenotype tracking showing higher expression in the low ATP6V1G1 group; (f) higher immune function scores in the low ATP6V1G1 group; (g) tumor microenvironment scores indicating higher tumor purity but lower StromalScore, ImmuneScore, and ESTIMATEScore in the high ATP6V1G1 group; (h) greater effectiveness of CTLA4 and PD1 immunotherapy in the low ATP6V1G1 group; (i) highest ATP6V1G1 expression in C4 (lymphocyte depleted), indicating an immunosuppressive microenvironment.

3.7. Exploring the Role of ATP6V1G1 in Prostate Cancer Cell Lines

This study investigated the expression patterns and functions of ATP6V1G1 in prostate cancer cell lines through a series of experiments. Figures 7(a), 7(b) indicate that ATP6V1G1 was upregulated in prostate cancer tissues and cell lines, suggesting a potential role in promoting the progression of prostate cancer. ATP6V1G1 mRNA expression was silenced using two different siRNAs (siATP6V1G1#1 and siATP6V1G1#2) in DU145 and PC3 cells. The results, as shown in Figures 7(c), 7(d), demonstrated a significant reduction in ATP6V1G1 mRNA levels compared to the control group, confirming the effectiveness of the siRNA approach. The CCK8 and colony formation assay results, depicted in Figures 7(e), 7(f), 7(g), 7(h), revealed that silencing ATP6V1G1 significantly reduced the proliferative capacity of both DU145 and PC3 cells. The wound healing assay results evaluated the migration capacity of DU145 and PC3 cells following ATP6V1G1 knockdown. The migration distance was notably decreased in cells treated with siATP6V1G1#1 and siATP6V1G1#2 compared to the control, indicating that ATP6V1G1 supports the invasive properties of prostate cancer cells (Figures 7(i), 7(j)). These results collectively highlighted the oncogenic potential of ATP6V1G1 in prostate cancer, impacting both cell proliferation and migration.

Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.
Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.
Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.
Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.
Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.
Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.
Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.
Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.
Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.
Details are in the caption following the image
Role of ATP6V1G1 in prostate cancer cell lines. (a), (b) mRNA expression levels of ATP6V1G1 in prostate cancer tissues and cell lines compared to normal tissues and PrEC LH cell line; (c), (d) effects of ATP6V1G1 knockdown by siRNA on mRNA levels in DU145 and PC3 cells; (e), (f) CCK8 assay results in prostate cancer cell lines; (g), (h) colony formation assay results following ATP6V1G1 silencing in prostate cancer cell lines; (i), (j) wound healing assay results in prostate cancer cell lines.

4. Discussion

Prostate cancer is a widespread health issue affecting millions of men globally [17, 18]. It is the second most common cancer in men after lung cancer, comprising 7% of newly diagnosed cancers in men globally and rising to 15% in developed regions. Each year, over 1.2 million new cases of prostate cancer are diagnosed globally, with more than 350,000 deaths attributed to the disease annually. This makes prostate cancer one of the leading causes of cancer-related death among men [19, 20].

MR studies have been widely used to explore the causes of various diseases, but their combination with tissue cell single-cell analysis is still relatively uncommon [21, 22]. Our study integrated MR results from both single-cell and bulk data levels, ultimately validating the reliability of associated genes through machine learning, providing a more comprehensive understanding of the causes of prostate cancer.

Our findings reveal that elevated levels of cysteinylglycine disulfide, a storage form of cysteinylglycine and a metabolite of glutathione, potentially reduce the risk of prostate cancer, highlighting its protective role. This association underscores the importance of glutathione metabolism in prostate cancer, as fluctuations in the levels of cysteinylglycine disulfide correlate with changes in glutathione metabolism. This is supported by our MR analysis, which robustly associates higher levels of this metabolite with a decreased incidence of the disease. Conversely, the increase in monocyte counts correlates with heightened cancer risk, suggesting that immune system dysregulation contributes to tumor progression. These results emphasize the importance of targeted therapies that could modulate the glutathione metabolism pathway, potentially curbing prostate cancer development.

The integration of single-cell RNA sequencing and bulk transcriptome analysis has provided a comprehensive view of the cellular and molecular landscapes within prostate tumors [23, 24]. Particularly, the differential pathway activation in monocytes underlines the metabolic plasticity of these cells in the tumor microenvironment. Our machine learning models corroborate these findings, identifying gene signatures that predict disease presence and progression with high accuracy. Such predictive biomarkers are invaluable for developing personalized medicine approaches, enhancing both diagnostic precision and treatment efficacy.

Notably, our study highlights the role of ATP6V1G1, which emerged as a key gene with significant diagnostic and prognostic value. In vitro experiments demonstrated that silencing ATP6V1G1 significantly reduced the proliferation and migration of prostate cancer cells, suggesting its oncogenic potential. ATP6V1G1 is known to play a critical role in the acidification of intracellular organelles and the regulation of autophagy, processes that are often dysregulated in cancer. Studies have shown that overexpression of ATP6V1G1 is associated with poor prognosis in various cancers, including breast cancer and glioma [25, 26]. This gene offers new prospects for therapeutic intervention in prostate cancer, underscoring the importance of further research on its role in disease progression.

Despite these advances, several limitations must be acknowledged. The causal relationships derived from MR rely on the assumption that the genetic variants used as instrumental variables affect the disease outcome only through the exposure being studied. Unmeasured confounding or pleiotropy could skew these interpretations. Additionally, while our models demonstrate significant predictive power, the external validity of these findings needs to be tested in broader, more diverse populations to confirm their generalizability. Future research should focus on validating these metabolic and immune signatures in longitudinal studies, potentially integrating more comprehensive genetic, environmental, and lifestyle data. This would not only confirm the clinical relevance of our findings but also might unveil new therapeutic targets within the intricate network of pathways influencing prostate cancer.

5. Conclusion

This study revealed the dual roles of cysteinylglycine disulfide and monocytes in prostate cancer, showing that elevated cysteinylglycine disulfide levels protect against the disease, while increased monocyte counts heighten risk. Additionally, ATP6V1G1 was identified as a key gene with significant diagnostic and prognostic value, suggesting its potential as a therapeutic target. The integration of MR, transcriptome analysis, and machine learning provided a comprehensive understanding of the metabolic and immune pathways involved in prostate cancer. Future research should validate these findings in diverse populations to confirm their clinical relevance and potential for personalized treatment strategies.

Conflicts of Interest

The authors declare no conflicts of interest.

Author Contributions

Rijian Guan: conceptualization, writing – original draft, funding acquisition, and software. Dongdong Wang: investigation, data curation, writing – original draft, and formal analysis. Mingwei Zhan: investigation, data curation, writing – original draft, and resources. Pengfei Liu: writing – review and editing, investigation, validation, and visualization. Yi Yu: writing – review and editing, supervision, project administration, and funding acquisition. Dongdong Wang and Mingwei Zhan contributed equally to this work.

Funding

This work was supported by the Zhejiang Medical Association Clinical Research Fund project (2022ZYC-A119).

Acknowledgments

The authors have nothing to report.

    Data Availability Statement

    The datasets analyzed during the current study are publicly available from the following repositories: GEWAS: https://www.ebi.ac.uk/gwas/; IEU OpenGWAS Project: https://gwas.mrcieu.ac.uk/; MSigDB (Molecular Signatures Database): https://www.gsea-msigdb.org/gsea/msigdb/; TCGA (The Cancer Genome Atlas): https://portal.gdc.cancer.gov/; GEO (Gene Expression Omnibus): https://www.ncbi.nlm.nih.gov/geo/.

      The full text of this article hosted at iucr.org is unavailable due to technical difficulties.