Introduction
Prostate cancer (PCa) is a worldwide healthcare problem, ranking among the most frequently diagnosed cancers in the male population. It arises in the prostate gland, which is responsible for seminal fluid synthesis. The onset of prostate cancer is influenced by several factors including genetic susceptibility, hormonal impact, and exposure to the environment [1-4]. Importantly, androgens, with particular reference to testosterone, play central roles in the tumorigenesis process as well as in cellular proliferation. In addition, the androgen receptor (AR) is responsible for prostate cancer cell survival, with its activation being a powerful promoter for tumorigenesis [5-7]. Recent studies suggest that pubertal timing may influence prostate cancer risk [8]. A Mendelian randomization study found that delayed puberty may reduce exposure to high androgen levels during prostate development, potentially decreasing prostate cancer risk later in life [9]. This underscores the importance of hormonal regulation in prostate carcinogenesis, highlighting how early-life exposures impact long-term prostate health [10]. As the disease advances, prostate cancer can evolve into castration-resistant prostate cancer (CRPC), a more aggressive form that emerges when tumors no longer respond to androgen deprivation therapies (ADT) [11]. This transition is driven by genetic alterations in key tumor suppressor genes, such as SPOP and BRCA2, which contribute to progression from hormone-sensitive prostate cancer to a castration-resistant phenotype [6,12,13]. Previous research has also uncovered key gene changes linked to the progression of castration-resistant prostate cancer, including mitochondrial adaptations and androgen receptor-independent drivers such as FABP5 [14]. The emergence of CRPC presents a major clinical challenge, as it remains largely incurable and associated with poor patient outcomes. The molecular landscape of prostate cancer is further complicated by the presence of distinct aggressive subtypes, such as neuroendocrine prostate cancer (NEPC) and aggressive variant prostate cancer (AVPC) [14,15]. These subtypes exhibit unique biological behaviors and treatment responses, necessitating the development of targeted therapies and personalized medicine approaches. Identifying biomarkers associated with these aggressive variants is crucial for improving patient stratification and therapy selection [16,17]. This study sought to examine differential gene expression patterns between castration-resistant prostate cancer cells (PC3M) and normal prostate epithelial cells (PNT2), with the aim of elucidating the key molecular mechanisms and signalling pathways underlying the progression to castration-resistant prostate cancer.
Inflammation and the tumor microenvironment in prostate cancer progression
Chronic inflammation is implicated in the onset and progression of prostate cancer. The inflammatory activity in the prostatic tissue in its non-neoplastic state correlates with an increased risk for high-grade prostate cancer [18]. This suggests that proinflammatory mediators may contribute to tumor initiation, necessitating further investigation into the molecular links between inflammation and prostate carcinogenesis [19]. Additionally, the tumor microenvironment (TME), particularly the reactive stroma, plays a pivotal role in prostate cancer progression and metastasis [20]. The interaction between cancer cells and stromal components influences tumor behavior, promoting angiogenesis, immune evasion, and therapy resistance [21]. These findings suggest that the therapeutic approach through the tumor microenvironment may provide new therapeutic targets, especially for CRPC.
Genetic factors and disparities in prostate cancer risk
Genetic susceptibility is a proven risk factor for prostate cancer, and familial clustering of the disease is common. Germline alterations in genes like HOXB13 and BRCA2 have been implicated in hereditary prostate cancer syndromes, highlighting the relevance of genetic testing and counseling in susceptible individuals. Furthermore, racial and ethnic differences in prostate cancer incidence and mortality imply that genetic, environmental, and lifestyle factors all influence disease risk and progression [22]. Understanding these disparities is essential for developing equitable screening strategies and precision medicine interventions.
Study rationale and experimental approach
This study endeavored to elucidate molecular mechanisms involved in prostate cancer by comparing gene expression between CRPC cells (PC3M) and normal prostate epithelial cells (PNT2). PNT2, a prostate epithelial-derived cell line, is commonly used as a control model in research related to prostate cancer. PC3M, originating from a castration-resistant prostate cancer tumor, is a typical castration-resistant phenotype given its ability to grow despite androgen deprivation therapy. This hallmark reflects advanced-stage prostate cancer. Through these two cell lines, the current study provides an important comparison to determine molecular alterations unique to the development of (CRPC) progression. One significant question in CRPC research is whether AR remains the major driver as an oncogene or if other pathways become activated in therapy-resistant tumors. To explore the issue, RNA-sequencing analysis was carried out in three biological replicates from each type of cell, which led to the construction of large gene expression profiles. Altogether, 1,000 genes with differing expressions, referred to as differentially expressed genes (DEGs), were identified based on a cut-off criterion set by a ≥1-fold change (FC) in their expressions with a p<0.05, as analyzed by Student's t-test. The identified DEGs were depicted in a heatmap, which displayed the most significantly up-regulated and down-regulated genes.
Gene ontology, pathway enrichment, and network analysis
Following the announced (DEGs), gene ontology (GO) enrichment analysis grouped these affected genes into certain biological processes, cellular components, and molecular functions, providing insights into gene alterations in cancer progression. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis mapped out signaling pathways enriched in prostate cancer cells, providing an extensive outline of oncogenic signaling networks. To explore functional gene interaction, weighted gene co-expression network analysis (WGCNA) was used, allowing the discovery of modules consisting of co-expressed genes. Hierarchical clustering dendrogram grouped genes with similar expressions, with dynamic tree cutting used to outline co-expression modules, as represented by distinct color in the dendrogram. By uncovering hub genes in relevant modules, this study highlights the importance of key regulatory genes responsible for the development of castration-resistant prostate cancer (CRPC).
Significance and implications
The findings of this study have far-reaching clinical implications. By pinpointing key differentially expressed genes (DEGs), this research identifies potential biomarkers for early prostate cancer detection, which is critical for improving treatment outcomes. Additionally, the DEGs and enriched pathways may serve as novel drug targets, facilitating the development of precision medicine strategies. Targeted therapies addressing these molecular alterations could lead to more effective, personalized treatments with fewer side effects than conventional approaches.
Materials and methods
Cell lines and culture
The study utilized two prostate cell lines as follows: the PNT2 prostate epithelial cell line, established from a donor without a history of prostate disease, and the highly aggressive, androgen receptor-negative PC3M prostate cancer cell line. Both cell lines were purchased from the American Type Culture Collection (ATCC). These cells were cultured as monolayer cultures at 37°C in a 5% CO2 atmosphere, using RPMI-1640 medium supplemented with L-glutamine, penicillin, streptomycin, and 10% fetal bovine serum. All experiments were performed using three independent biological replicates for each cell line. To minimize technical variability, cells were cultured under identical conditions using standardized passage numbers, medium batches, and plating densities.
RNA extraction, RNA library preparation, and NovaSeq sequencing
Total RNA was extracted from the frozen cell pellets using a Qiagen RNeasy Mini kit (Hilden, Germany: QIAGEN N.V.), in accordance with the manufacturer's protocol. The Qiagen FastSelect rRNA HMR kit (Hilden, Germany: QIAGEN N.V.) was employed to prepare the rRNA depletion sequencing library. Additionally, a NEBNext Ultra II RNA Library Production kit (Ipswich, MA: New England Biolabs) for Illumina was used to construct the RNA sequencing library. The libraries were verified using a Tapestation 4200 kit (Santa Clara, CA: Agilent Technologies, Inc.), quantified with a Qubit 2.0 Fluorometer (Waltham, MA: Thermo Fisher Scientific Inc.), and validated by quantitative PCR. Each sample achieved an average sequencing depth of approximately 30 million reads. Quality control was performed before and after alignment using FastQC (Cambridge, UK: Babraham Bioinformatics, Babraham Institute) and MultiQC (Stockholm, Sweden: Philip Ewels), with all samples achieving >90% Q30 scores and no significant contamination or adapter presence. The multiplexed sequencing libraries were then sequenced on the Illumina NovaSeq 6000 System (San Diego, CA: Illumina, Inc.), utilizing a 2×150 Pair-End Configuration, version 1.5 (San Diego, CA: Illumina, Inc.). The raw sequence data generated by the Illumina NovaSeq platform was converted into FASTQ files using the Illumina bcl2fastq (San Diego, CA: Illumina, Inc.) application version 2.20, and subsequently de-multiplexed based on the index sequence identification. The RNA sequencing data analysis involved the following steps: Trimmomatic was employed to trim the sequence reads, remove adapter sequences, and exclude low-quality nucleotides. Trimmomatic (v0.39) was run using the following parameters: sliding window=4:20, minimum read length=36 bp, and removal of Illumina adapters.
Statistical analysis
The data in this study were analysed using GraphPad Prism 9 software (San Diego, CA: GraphPad Software). Two-tailed paired Student's t-tests and one-way ANOVAs were employed to statistically compare group means, where appropriate. For the RNA-sequencing data, differentially expressed genes were identified using the DESeq2 package (Heidelberg, Germany: European Molecular Biology Laboratory {EMBL}), which internally applies the Benjamini-Hochberg procedure to adjust for multiple testing and control the false discovery rate. Genes exhibiting a fold change of 1 or greater and an adjusted p-value less than 0.05 were considered statistically significant. These widely accepted thresholds were adopted in the transcriptomic analyses to strike a balance between sensitivity and specificity in detecting biologically meaningful changes in gene expression [23].
Results
Differential expression between PNT2 and PC3M
The scatter plot and differentially expressed genes (DEGs) between PNT2 and PC3M cells are presented in Figures 1a, 1b. Based on an analysis of the scatter plot and RNA-seq heatmaps, the top 1,000 most significant DEGs (500 upregulated and 500 downregulated) were identified (Figures 1a, 1b). These results reveal substantial transcriptional differences between normal prostate epithelial cells (PNT2) and castration-resistant prostate cancer cells (PC3M), suggesting key molecular alterations associated with prostate cancer progression.
Figure 1
Scatterplot and differentially expressed genes between PNT2 and PC3M cell.
(a) Scatterplot depicts the differentially expressed genes (DEGs) profile between the PNT2 prostate epithelial cell line and the PC3M castration-resistant prostate cancer cell line. Genes that are upregulated in the PC3M cells are shown in red, while downregulated genes are represented in blue. This analysis has identified 1,000 differentially expressed genes between the two cell types. (b) Heatmap depicts the expression patterns of the identified DEGs, with upregulated genes in PC3M cells shown in red and downregulated genes in blue compared to the PNT2 cell line.
Top 10 differentially expressed genes
A comparison of gene expression patterns between PNT2 and PC3M cells identified the 10 most significantly upregulated and downregulated genes, which are presented in Figure 2. The top 10 DEGs in PC3M cells include MT-CO1, MT-CYB, MT-ND4, MT-CO2, MT-CO3, MT-ATP6, SNRNP70, SF3A2, TPT1, and SF3B2, all of which were significantly upregulated in PC3M and downregulated in PNT2 (Figure 2 and Table 1). These findings highlight key mitochondrial and splicing-related genes that may contribute to castration-resistant prostate cancer (CRPC) progression and survival mechanisms.
Figure 2
Top 10 DEGs between the PC3 and PNT2 prostate cancer cell lines.
Heatmap depicts the expression profiles of the 10 most DEGs between the PC3M castration-resistant prostate cancer cell line and the PNT2 normal prostate epithelial cell line. Upregulated genes in PC3M are represented in red, while downregulated genes are shown in blue in comparison to PNT2.
Table 1
List of the 10 most significantly differentially expressed genes (DEGs) between PNT2 and PC3M.
Gene ID | Gene name | Log2 fold change | PNT2 | PC3M |
ENSG00000198804 | MT-CO1 | 402370.8776 | 77.73854636 | 402370.8776 |
ENSG00000198727 | MT-CYB | 118563.7544 | 9.893996809 | 118563.7544 |
ENSG00000198886 | MT-ND4 | 215952.0172 | 21.20142173 | 215952.0172 |
ENSG00000198712 | MT-CO2 | 164826.8557 | 5.653712463 | 164826.8557 |
ENSG00000198938 | MT-CO3 | 112731.657 | 15.54770927 | 112731.657 |
ENSG00000198899 | MT-ATP6 | 147171.6308 | 8.480568694 | 147171.6308 |
ENSG00000104852 | SNRNP70 | 18850.87334 | 0 | 18850.87334 |
ENSG00000104897 | SF3A2 | 10509.64961 | 4.240284347 | 10509.64961 |
ENSG00000133112 | TPT1 | 29296.58454 | 14.13428116 | 29296.58454 |
ENSG00000087365 | SF3B2 | 21312.50256 | 5.653712463 | 21312.50256 |
GO and pathway analysis
To further explore the functional roles of the significant DEGs, gene ontology (GO) enrichment analysis was conducted to categorize genes into biological processes, molecular functions, and cellular components (Figures 3-5 and Tables 2-4). Additionally, KEGG pathway analysis delineated the signaling pathways enriched in the DEGs (Figure 6, Table 5). To identify key regulatory genes and potential therapeutic targets, a hub gene network and gene-drug interaction analysis were performed (Figure 7a-7e, Table 6). Analysis in Figure 3 revealed that DEGs were significantly enriched in biological processes such as cell cycle regulation, protein localization and transport, intracellular signaling, macromolecule biosynthesis, metabolic reprogramming, cellular response to stress, and organelle organization. These pathways play essential roles in cancer initiation and progression, promoting uncontrolled proliferation, metabolic shifts, stress adaptation, and oncogenic signaling activation. In Figure 4, GO analysis identified key cellular components associated with the DEGs, including the nucleoplasm, nuclear envelope, nuclear lumen, mitochondria, intracellular protein-containing complexes, and organelle membranes. These components contribute to genomic instability, transcriptional dysregulation, metabolic reprogramming, and structural modifications that facilitate cancer progression and therapeutic resistance. Figure 5 highlights the enrichment of molecular functions, including kinase binding, RNA binding, enzyme binding, nucleotide binding, ATP binding, nucleic acid binding, and transferase activity. These functions support tumor development by enhancing signal transduction, regulating gene expression, driving metabolic changes, and facilitating enzymatic activities critical for sustained proliferation and survival in malignant cells. To explore the signaling pathways activated in the DEGs, KEGG pathway analysis was performed (Figure 6, Table 5). The results demonstrated significant enrichment in pathways related to ubiquitin-mediated proteolysis, cell cycle regulation, spliceosome activity, ribosome function, protein processing in the endoplasmic reticulum, autophagy, microRNAs in cancer, and metabolic pathways. In Figures 7a-7e, further analysis of the differentially expressed genes through hub gene network analysis identified critical regulators involved in cellular communication and signal transduction, metabolic reprogramming and mitochondrial dysfunction, chromatin remodeling and DNA repair, and stress response and protein homeostasis. Figure 7a presents the ranked distribution of the DEGs, while Figures 7b-7e highlight major hub gene interaction networks. Figure 7b reveals genes linked to cellular communication and signal transduction, such as CD9, UBE2G2, and TRA2B. Figure 7c identifies DEGs associated with metabolic reprogramming and mitochondrial dysfunction, including MT-CO1 and ATP5-MC2. Figure 7d highlights key genes involved in chromatin remodeling and DNA repair, such as CHD7, CHD8, and CDC25B. Figure 7e explores DEGs related to stress response and protein homeostasis, including GAPDH and HSP90AA1. The 10 most important hub genes are summarized in Table 6, highlighting their functional roles and biological significance in castration-resistant prostate cancer progression.
Figure 3
Biological processes analysis.
Gene ontology (GO) enrichment analysis revealed the biological processes associated with the DEGs between the PC3M castration-resistant prostate cancer cell line and the PNT2 normal prostate epithelial cell line. The x-axis depicts the fold enrichment, while the y-axis lists the enriched biological processes. The colour gradient represents the adjusted p-value (-log10), and the dot size indicates the number of genes involved in each process.
FDR: false discovery rate; DEG: differentially expressed gene
Figure 4
Cellular components analysis.
Gene ontology (GO) enrichment analysis revealed the cellular components associated with DEGs between PC3M and PNT2 cells. The x-axis depicts the fold enrichment, while the y-axis lists the enriched biological processes. The colour gradient represents the adjusted p-value (-log10), and the dot size indicates the number of genes involved in each process.
FDR: false discovery rate; DEG: differentially expressed gene
Figure 5
Molecular function analysis.
Gene ontology (GO) enrichment analysis revealed molecular functions of DEGs between PC3M and PNT2 cells. The x-axis depicts the fold enrichment, while the y-axis lists the enriched biological processes. The colour gradient represents the adjusted p-value (-log10), and the dot size indicates the number of genes involved in each process.
FDR: false discovery rate; DEG: differentially expressed gene
Figure 6
Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis.
Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs between PC3M and PNT2 cells. The x-axis represents fold enrichment, while the y-axis lists the significantly enriched pathways. The x-axis depicts the fold enrichment, while the y-axis lists the enriched biological processes. The colour gradient represents the adjusted p-value (-log10), and the dot size indicates the number of genes involved in each process.
FDR: false discovery rate; DEG: differentially expressed gene
Figure 7
Hub gene and network analysis in prostate cancer.
(a) The analysis of differentially expressed genes DEG using hub gene network analysis identified key regulatory genes involved in prostate cancer progression. The bar plot displays the ranked gene significance, with hub genes highlighted in distinct colours based on their connectivity and functional importance. (b-e) Major hub gene interaction networks derived from DEG analysis: (b) A network of genes that regulate cellular communication and signal transduction pathways. (c) A network of genes that regulate metabolic reprogramming and mitochondrial dysfunction pathways. (d) A network of genes that regulate chromatin remodeling and DNA repair pathways. (e) A network of genes that regulates stress response and protein homeostasis.
DEG: differentially expressed gene
Table 2
The top 20 GO enrichment analysis of biological process in castration-resistant prostate cancer.
FDR: false discovery rate; GO: gene ontology
Enrichment FDR | N genes | Pathway genes | Fold enrichment | Pathway |
2.94E-193 | 3347 | 4179 | 1.326855 | Organelle organization |
1.25E-129 | 2635 | 3340 | 1.306996 | Macromolecule localization |
4.65E-129 | 2285 | 2837 | 1.334342 | Protein localization |
2.14E-126 | 3444 | 4550 | 1.253984 | Macromolecule modification |
3.02E-126 | 2944 | 3806 | 1.281472 | Cellular localization |
1.82E-120 | 1571 | 1857 | 1.401537 | Intracellular transport |
7.19E-117 | 3258 | 4310 | 1.252316 | Cellular protein modification process |
7.19E-117 | 3258 | 4310 | 1.252316 | Protein modification process |
1.30E-109 | 1636 | 1974 | 1.373018 | Cellular macromolecule localization |
1.37E-108 | 1624 | 1960 | 1.372683 | Cellular protein localization |
4.52E-99 | 2775 | 3660 | 1.256094 | Cellular component biogenesis |
2.22E-92 | 1853 | 2332 | 1.316398 | Cellular response to stress |
1.55E-91 | 1611 | 1989 | 1.341841 | Establishment of protein localization |
7.74E-88 | 2312 | 3018 | 1.269138 | Establishment of localization in cell |
8.04E-87 | 1515 | 1867 | 1.344338 | Protein transport |
2.20E-86 | 2287 | 2987 | 1.268444 | Catabolic process |
2.37E-86 | 2000 | 2566 | 1.29126 | Cellular catabolic process |
8.78E-85 | 1602 | 1997 | 1.328999 | Cell cycle |
3.45E-81 | 3390 | 4675 | 1.201319 | Organic cyclic compound biosynthetic process |
4.82E-81 | 3274 | 4498 | 1.205867 | Heterocycle biosynthetic process |
Table 3
The top 20 GO enrichment analysis of cellular component in castration-resistant prostate cancer.
FDR: false discovery rate; GO: gene ontology
Enrichment FDR | N genes | Pathway genes | Fold enrichment | Pathway |
4.25E-254 | 3948 | 4885 | 1.338914 | Nuclear lumen |
3.83E-241 | 3671 | 4519 | 1.345805 | Nucleoplasm |
1.01E-96 | 1498 | 1816 | 1.366584 | Mitochondrion |
4.12E-85 | 1278 | 1540 | 1.374834 | Catalytic complex |
8.17E-83 | 3008 | 4085 | 1.219905 | Organelle membrane |
2.19E-77 | 1156 | 1391 | 1.3768 | Microtubule cytoskeleton |
3.48E-64 | 1102 | 1352 | 1.350346 | Organelle envelope |
3.48E-64 | 1102 | 1352 | 1.350346 | Envelope |
1.06E-60 | 745 | 868 | 1.421925 | Microtubule-organizing center |
2.50E-59 | 888 | 1067 | 1.37876 | Nucleolus |
1.63E-55 | 1871 | 2504 | 1.237883 | Cytoskeleton |
1.63E-55 | 1102 | 1379 | 1.323907 | Nuclear protein-containing complex |
1.80E-54 | 713 | 839 | 1.407887 | Transferase complex |
1.23E-53 | 577 | 658 | 1.452747 | Centrosome |
8.85E-47 | 723 | 872 | 1.373606 | Mitochondrial envelope |
1.42E-43 | 452 | 512 | 1.462543 | Mitochondrial matrix |
9.46E-42 | 700 | 854 | 1.357939 | Intracellular protein-containing complex |
1.52E-41 | 655 | 792 | 1.370113 | Ribonucleoprotein complex |
2.82E-41 | 676 | 822 | 1.362433 | Mitochondrial membrane |
2.35E-38 | 748 | 932 | 1.329615 | Nuclear body |
Table 4
The top 20 GO enrichment analysis of molecular functions in castration-resistant prostate cancer.
FDR: false discovery rate; GO: gene ontology
Enrichment FDR | N genes | Pathway genes | Fold enrichment | Pathway |
2.65E-112 | 3287 | 4371 | 1.245831 | Nucleic acid binding |
3.29E-101 | 1778 | 2197 | 1.340732 | Enzyme binding |
1.44E-95 | 1537 | 1873 | 1.359491 | RNA binding |
1.77E-72 | 1839 | 2382 | 1.279028 | Nucleoside phosphate binding |
1.88E-72 | 1838 | 2381 | 1.27887 | Nucleotide binding |
3.17E-64 | 1977 | 2620 | 1.250102 | Anion binding |
1.14E-61 | 2049 | 2739 | 1.239339 | Small-molecule binding |
8.22E-60 | 1627 | 2122 | 1.27023 | Purine nucleotide binding |
2.34E-59 | 1925 | 2565 | 1.243322 | Transferase activity |
1.09E-58 | 1625 | 2124 | 1.267474 | Ribonucleotide binding |
1.42E-58 | 1613 | 2107 | 1.268265 | Purine ribonucleotide binding |
1.13E-57 | 1560 | 2033 | 1.27124 | Purine ribonucleoside triphosphate binding |
1.48E-57 | 3276 | 4636 | 1.170687 | Metal ion binding |
7.00E-57 | 3331 | 4726 | 1.167673 | Cation binding |
1.17E-48 | 1337 | 1743 | 1.270791 | Adenyl nucleotide binding |
1.45E-47 | 1325 | 1730 | 1.268849 | Adenyl ribonucleotide binding |
2.59E-46 | 1275 | 1662 | 1.270923 | ATP binding |
4.71E-42 | 658 | 794 | 1.372921 | Kinase binding |
1.07E-41 | 1817 | 2492 | 1.207945 | Carbohydrate derivative binding |
1.09E-40 | 1943 | 2689 | 1.197077 | DNA binding |
Table 5
The top 20 GO enrichment analysis of KEGG pathway in castration-resistant prostate cancer.
FDR: false discovery rate; GO: gene ontology; KEGG: Kyoto Encyclopedia of Genes and Genomes
Enrichment FDR | N genes | Pathway genes | Fold enrichment | Pathway |
4.62E-39 | 1163 | 1527 | 1.261772 | Metabolic pathways |
3.28E-19 | 302 | 363 | 1.37829 | Amyotrophic lateral sclerosis |
3.61E-18 | 314 | 383 | 1.358223 | Alzheimer disease |
4.34E-18 | 393 | 495 | 1.315308 | Herpes simplex virus 1 infection |
9.72E-18 | 257 | 306 | 1.3914 | Huntington disease |
1.85E-17 | 377 | 475 | 1.314886 | Pathways of neurodegeneration |
2.70E-17 | 214 | 249 | 1.423819 | Salmonella infection |
4.50E-17 | 131 | 141 | 1.539191 | Ubiquitin-mediated proteolysis |
1.41E-16 | 200 | 232 | 1.428178 | Thermogenesis |
1.53E-16 | 225 | 266 | 1.401332 | Parkinson disease |
3.51E-16 | 214 | 252 | 1.406868 | Endocytosis |
5.28E-15 | 150 | 169 | 1.470431 | Protein processing in endoplasmic reticulum |
1.79E-14 | 116 | 126 | 1.525203 | Cell cycle |
2.20E-14 | 406 | 530 | 1.269084 | Pathways in cancer |
5.98E-14 | 120 | 132 | 1.506078 | Spliceosome |
8.07E-14 | 205 | 246 | 1.380572 | Shigellosis |
1.45E-13 | 121 | 134 | 1.495963 | Ribosome |
3.74E-13 | 141 | 161 | 1.450887 | MicroRNAs in cancer |
1.07E-12 | 186 | 223 | 1.38181 | Chemical carcinogenesis |
1.29E-12 | 125 | 141 | 1.468693 | Autophagy |
Table 6
Key hub genes identified in the network analysis of castration-resistant prostate cancer (CRPC).
Gene name | Function | Network category |
CD9 | Cell adhesion and communication | Cellular communication |
UBE2G2 | Ubiquitination and protein degradation | Cellular communication |
TRA2B | RNA splicing and regulation | Cellular communication |
MT-CO1 | Mitochondrial respiration | Metabolic reprogramming |
ATP5-MC2 | ATP synthesis | Metabolic reprogramming |
CHD7 | Chromatin remodeling | DNA repair and chromatin remodeling |
CHD8 | Epigenetic regulation | DNA repair and chromatin remodeling |
CDC25B | Cell cycle regulation | DNA repair and chromatin remodeling |
GAPDH | Glycolysis and energy metabolism | Stress response and protein homeostasis |
HSP90AA1 | Protein folding and stress response | Stress response and protein homeostasis |
These findings provide a comprehensive molecular landscape of castration-resistant prostate cancer, revealing key pathways and gene networks that contribute to tumor progression, therapeutic resistance, and potential precision medicine strategies. The identified critical regulators play pivotal roles in various hallmarks of cancer, including sustained proliferative signaling, metabolic reprogramming, genomic instability, and evasion of cell death. These insights offer valuable opportunities for the development of targeted therapeutic approaches and precision medicine strategies for the management of castration-resistant prostate cancer.
Discussion
This study provides a comprehensive molecular characterization of castration-resistant prostate cancer progression by examining DEGs between normal prostate epithelial cells and CRPC cells. The findings highlight significant molecular alterations associated with CRPC, particularly in metabolic reprogramming and cell cycle regulation. These results offer valuable insights into potential therapeutic targets and biomarkers that could aid in early detection, prognosis, and treatment of CRPC. To further investigate these molecular changes, differential expression analysis and gene network analysis were performed. Figure 2 presents a heatmap of the 10 most significantly differentially expressed genes, highlighting genes with the highest magnitude of transcriptional change between PNT2 and PC3M cells based on statistical significance. However, while differential gene expression analysis identifies genes with substantial expression differences, it does not necessarily highlight genes that are functionally central in cancer progression. Therefore, a gene network analysis was conducted to identify key regulatory genes based on their connectivity within biological pathways. The hub gene analysis revealed critical regulators involved in cellular communication, metabolic reprogramming, chromatin remodeling, and stress response mechanisms. These key molecular pathways and signaling networks play pivotal roles in driving the progression and therapeutic resistance of CRPC, making them promising targets for the development of novel treatment strategies. Although the current study utilizes immortalized cell lines, which may not fully capture the multifaceted nature of patient-derived tumors, future investigations will aim to validate the top candidate genes using CRISPR-Cas9 knockout techniques. This will be followed by transcriptomic analyses, in vivo experimentation, and clinical correlation studies to evaluate the relevance of these genes as potential therapeutic targets and biomarkers.
Dysregulation of key molecular pathways in CRPC
Functional enrichment analysis revealed that biological processes such as cell cycle regulation, metabolic adaptation, intracellular signaling, and macromolecule biosynthesis were significantly altered in CRPC cells. The GO enrichment analysis identified pathways related to cell cycle progression, intracellular transport, and protein localization, which are critical for maintaining the proliferative capacity of CRPC cells (Figure 3). The observed upregulation of cell cycle-related genes such as CDC25B and GTSE1 (Figure 7d) suggests that cell cycle dysregulation plays a key role in castration resistance, consistent with previous studies on CRPC progression [24-27].
Moreover, KEGG pathway analysis demonstrated significant enrichment in ubiquitin-mediated proteolysis, autophagy, and protein processing pathways, indicating that CRPC cells employ alternative mechanisms for protein degradation and cellular homeostasis in response to metabolic and therapeutic stress (Figure 6). These findings align with emerging evidence suggesting that alterations in protein degradation pathways contribute to therapy resistance in prostate cancer [28].
Cellular communication and signal transduction mechanisms in CRPC
Analysis of the gene interaction network depicted in Figure 7b revealed key genes with pivotal roles in cellular communication and signal transduction pathways, including CD9, UBE2G2, and TRA2B. These genes modulate critical cellular processes such as cell-cell adhesion, cell migration, and the ubiquitin-proteasome system, thereby enhancing the survival, proliferation, and metastatic potential of castration-resistant prostate cancer cells [29]. The protein CD9 is involved in controlling exosome communication and the epithelial-to-mesenchymal transition (EMT), processes that contribute to cancer progression. Similarly, the enzyme UBE2G2 is associated with protein degradation pathways, affecting tumor growth and therapy resistance in androgen-independent prostate cancer [30]. Additionally, the splicing regulator TRA2B has been linked to alternative splicing that drives cancer pathways and metastasis in various cancer types, including prostate cancer. Disruptions or dysregulations in these signaling pathways can have a significant influence on the progression, invasiveness, and aggressiveness of CRPC, making them potential therapeutic targets for intervention and management of this advanced prostate cancer subtype [31].
Metabolic reprogramming and mitochondrial involvement in CRPC
One of the most striking findings was the differential expression of mitochondrial genes, including MT-CO1, MT-CYB, MT-ND4, and MT-ATP6, which were significantly upregulated in PC3M cells. Mitochondrial dysfunction has been increasingly recognized as a hallmark of cancer, particularly in CRPC, where metabolic reprogramming enables tumor cells to thrive under nutrient-deprived and hypoxic conditions [32,33]. The upregulation of these mitochondrial genes suggests that CRPC cells rely more heavily on oxidative phosphorylation (OXPHOS), which has been proposed as a metabolic adaptation in androgen-independent prostate cancer [34,35]. Recent studies have demonstrated that detrimental alterations in mitochondrial complex I and IV genes, including MT-ND4 and MT-CO1, are linked to enhanced oxidative phosphorylation activity and increased aggressiveness in high-grade and treatment-resistant prostate cancers. Additionally, such dysregulation appears to promote elevated ATP production and reactive oxygen species generation, both of which contribute to resistance against apoptosis and hormonal therapy [36]. These findings align with the emerging concept that mitochondrial remodelling is central to the progression of castration-resistant prostate cancer, suggesting that targeting mitochondrial metabolism either through OXPHOS inhibitors or modulation of mitochondrial gene expression may represent a promising therapeutic approach for suppressing tumor growth and overcoming drug resistance [37,38].
Chromatin remodeling and DNA repair pathways in CRPC
Dysregulation of chromatin remodelling and DNA repair genes, including CHD7, CHD8, and CDC25B, significantly contributes to the progression of castration-resistant prostate cancer. Alterations in the expression or function of these genes can lead to aberrant gene expression patterns and compromised genomic integrity, ultimately driving the advancement of CRPC (Figure 7d) [39]. Recent studies have highlighted the involvement of CHD7 in the transcriptional regulation of cancer-associated genes, with evidence suggesting that its dysregulation may contribute to altered chromatin architecture and oncogenic signaling in advanced prostate malignancies [40]. CHD8 has also been implicated in CRPC, where its increased nuclear expression correlates with poor prognosis and enhanced metastatic potential, supporting its role in disease progression [41]. Furthermore, CDC25B, a cell cycle phosphatase frequently overexpressed in high-grade prostate tumors, has been shown to act as a co-activator of the androgen receptor (AR), thereby promoting AR-mediated transcription even under androgen-deprived conditions [42]. Targeting these critical chromatin remodelling and DNA repair pathways, particularly in tumors with impaired DNA repair mechanisms, aligns with the success and potential of therapies such as PARP inhibitors in the management of prostate cancer [43,44]. Exploiting the vulnerabilities of CRPC cells resulting from dysfunctional DNA repair could provide novel therapeutic avenues to improve patient outcomes.
Stress response and protein homeostasis in CRPC
Genes central to stress responses and protein homeostasis, such as GAPDH and HSP90AA1, were identified as key players (Figure 7e). These genes play a crucial role in protecting CRPC cells from stress-induced damage and apoptosis, thereby contributing to therapy resistance [45,46]. Recent research has shown that increased expression of GAPDH in cancer cells improves their survival under low oxygen conditions and contributes to drug resistance. This is achieved through GAPDH's interaction with the AKT signalling pathway, which promotes cell proliferation and tumor growth [47]. Furthermore, HSP90AA1, a molecular chaperone, is vital for preserving protein balance in CRPC cells. Its heightened expression is linked to unfavourable prognosis and has been implicated in stabilising client proteins that are crucial for cancer cell survival under stressful conditions [48]. Modulating these stress response and protein homeostasis mechanisms could potentially enhance the effectiveness of conventional therapies by disrupting the critical survival pathways in castration-resistant prostate cancer cells [49].
Conclusions
This study provides a comprehensive molecular analysis of castration-resistant prostate cancer (CRPC) by comparing gene expression profiles between normal prostate epithelial cells (PNT2) and CRPC cells (PC3M), revealing significant alterations in cell cycle regulation, metabolic pathways, DNA repair mechanisms, and stress response networks. These changes underscore the complexity of CRPC biology and contribute to the progression and therapeutic resistance of the disease. The identified molecular alterations offer insights into potential therapeutic targets and biomarkers for early detection, prognosis, and treatment strategies. Highlighting the need for targeted therapeutic approaches based on detailed molecular profiles, this research emphasizes that further investigation is crucial to translate these findings into clinical practice, aiming to improve outcomes for patients with CRPC.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2025, Naeem et al. This is an open access article distributed under the terms of the Creative Commons Attribution License CC-BY 4.0., which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Prostate cancer is a global health issue and one of the most common reasons for cancer-related mortality. This research aimed to investigate the molecular mechanisms underlying the progression to castration-resistant prostate cancer (CRPC). Differential gene expression was analyzed by contrasting the PNT2 prostate epithelial cell line and the PC3M CRPC cell line. RNA sequencing was performed on three biological replicates of each cell type, and 1,000 differentially expressed genes were identified with a fold change ≥1 and a p<0.05. A heatmap was generated to visualize the gene expression profiles, and the top 10 significantly altered genes were identified. Functional enrichment analysis, including Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis, was conducted to map the biological processes, cellular components, and molecular functions associated with the differentially expressed genes. Furthermore, weighted gene co-expression network analysis was utilized to identify co-expression modules and significant genes, thereby highlighting the top 10 most significant differentially expressed genes in significant pathways. The findings indicate substantial molecular alterations associated with the development of castration-resistant prostate cancer, with major pathways including metabolic deregulation pathways and cell cycle regulation. The identified differentially expressed genes (DEGs) and pathways provide significant insights into disease progression and potential therapeutic targets. These findings contribute to the understanding of prostate cancer at the molecular level and can be used to identify new diagnostic and therapeutic strategies. However, further validation is required to determine the clinical significance of these targets in the treatment of CRPC.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details
1 Basic Sciences, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Jeddah, SAU, Basic Sciences, King Abdullah International Medical Research Center, Jeddah, SAU