Abstract Objective Vehicle emissions (VEs) are considered important causes of air pollution in cities. This study aims to analyze the role of VEs in the progression of prostate cancer (PCa). Methods We used the CTD database to obtain genes associated with VEs in prostate cancer to explore the associations between VEs and prostate cancer. LASSO regression analysis was subsequently used to construct a novel VEs-related progression model in the TCGA and GEO databases. Differences in the progression-free interval (PFI), clinical characteristics and immune characteristics were compared among patients with different VEs score. Finally, we confirmed that the changes of DNMT3B after VEs mediation and that DNMT3B promoted the progression of prostate cancer by preliminary experiments. Results The analysis of VEs-enriched diseases revealed the most significant enrichment in the prostate cancer pathway. The GO enrichment analysis observed that VEs affected multiple signaling pathways and biological processes in PCa. On the basis of VEs-associated genes, we relied on the VEs scoring model to accurately evaluate the PFI (p < 0.05). In patients with high VEs scores, tumor mutation burden (TMB) (R = 0.29, p < 0.001) and the cytolytic activity (CYT) (R = 0.161, p < 0.001) scores were increased, as was the expression of immunosuppressive ligands. Functional experiments showed that knockdown of DNMT3B inhibited the proliferation, colony formation and migration of prostate cancer cells in vitro. In addition, we found that DNMT3B expression was significantly increased in tumor cells after VEs treatment. Conclusions This study presents unique opinions into the impact of VEs on prostate cancer progression and highlights the need for more in-depth exploration of the mechanistic link between VEs and prostate cancer progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-025-03906-8. Keywords: Vehicle emissions, Prostate cancer, Progression-free interval Graphical Abstract [34]graphic file with name 12935_2025_3906_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12935-025-03906-8. Introduction Vehicle emissions are important contributors to global air pollution, which has a serious adverse impact on human health [[35]1]. Most VEs originates from the combustion of liquid energy in vehicles, mainly including gasoline and diesel. VEs are composed of a variety of nontoxic and toxic components and particulates. Long-term excessive exposure to these toxic components can induce the development of a variety of serious diseases, such as cancer [[36]2, [37]3]. The study confirmed that VEs, rock sources and coal combustion accounted for more than 85% of the cancer risk [[38]4]. The main volatile organic compounds (VOCs) in VEs are benzene, toluene, ethylbenzene, and xylene [[39]5]. PM[2.5] in VEs can enter the body through breathing, leading to related diseases [[40]6]. Polycyclic aromatic hydrocarbons (PAHs) are easily absorbed into the blood circulation, and prolonged high exposure can lead to tumors at multiple sites, including the skin, lung, and bladder [[41]7]. Moreover, the results suggest that exposure to high levels of diesel engine emissions can increase the risk of bladder cancer [[42]8]. Ambient air pollution is a clear carcinogen of lung cancer, and diesel engine exhaust or emissions are classified as Group 1 carcinogens. There is also evidence of a positive association between the risk of death from lung cancer and the number of years of work with regular exposure to newly emitted vehicle exhaust [[43]9]. In addition, the chemistry of gasoline and diesel emissions has changed as engines and post-combustion control technologies have changed. Prostate cancer is a multifaceted health problem [[44]10]. In addition, environmental factors are one of the causes of prostate cancer [[45]11]. Studies have shown that organic pesticides, heavy metals, VEs, and polycyclic aromatic hydrocarbons are closely associated with prostate cancer [[46]12–[47]14]. Large amounts of ultrafine particles in exhaust fumes increase the risk of prostate cancer [[48]11]. Parent et al. reported that a positive association between elevated ambient NO[2] exposure and incident prostate cancer in Canada [[49]15]. Cohen et al. noted an increased risk of prostate cancer in Israeli men exposed to traffic-related air pollution [[50]16]. Furthermore, occupational exposure to diesel fuel or fumes is associated with a statistically significant increase in prostate cancer incidence [[51]17]. Similarly, exposure to aircraft emissions also significantly increased the risk of prostate cancer in male patients [[52]18]. Therefore, the long-term exposure risk of VEs is closely related to the occurrence of prostate cancer. At present, the understanding of the effect of automobile exhaust on prostate cancer is still limited. In this study, we demonstrated that automobile exhaust promoted prostate cancer progression. Various databases were used to search for and analyze the effects of automobile exhaust on prostate cancer, and experiments were also conducted to verify the findings. In this study, we constructed a PFI prediction model based on VEs related genes. The purpose of this study is to explain the relationship between VEs, genes and PFI by bioinformatics methods. Patients with high expression of related genes are more likely to be affected by VEs in the future, and the disease progresses more rapidly. We expect that this study will make valuable contributions to the field of prostate cancer-related environmental pollution research. Methods and materials Cell culture The human prostate cancer cell lines PC-3, DU145, LNCaP and 22Rv1 were purchased from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). PC-3, LNCaP and 22Rv1 cells were cultured in RPMI-1640 (Gibco, USA), whereas DU145 cells were cultured in DMEM (Gibco, USA). The medium was supplemented with 10% fetal bovine serum (ExCell Bio, China) and 1% penicillin/streptomycin (Servicebio, China). The cells were cultured at 37 °C in a humidified 5% CO[2] environment. Cell-line validation was performed with the use of short tandem repeat (STR) analysis. Passage numbers in 5–15 cells were used in the article to reduce genetic drift. RNA isolation and qRT‒PCR Total RNA was isolated and extracted using RNA-Quick purification kit (YiShan Biotech, China) according to the manufacturer’s instructions. Reverse transcription was then performed using Hifair^® III 1st Strand cDNA Synthesis SuperMix for qPCR (Yeasen, China). The expression of mRNAs in prostate cell lines was quantified via a CFX Connect Real-Time PCR Detection System (Bio-Rad, USA). The primer sequences for DNMT3B (forward primer: AGGGAAGACTCGATCCTCGTC, reverse primer: GTGTGTAGCTTAGCAGACTGG) and ACTIN (forward primer: TGGCACCCAGCACAATGAA, reverse primer: CTAAGTCATAGTCCGCCTAGAAGCA) were purchased from GenePharma (Shanghai, China). Transient transfection DNMT3B siRNA oligonucleotides (si-1: 5’-CCACCUUCAAUAAGCUCGUTT-3’, si-2: 5’-CGCCUCAAGACAAAUUGCUTT-3’) and control siRNA (si-NC: 5’-UUCUCCGAACGUGUCACGUTT-3’) were obtained from GenePharma (Shanghai, China). The cells were seeded in 6-well plates at a density of 200 000 cells per well. Transfection experiments were performed when the cell density had grown to 60-70%. Subsequently, 100 nM siRNA and 5 µl/ml Lipofectamine RNAiMAX (Invitrogen, USA) were mixed and allowed to stand for 30 min at room temperature. The mixture was added gently and slowly to the cell culture medium and incubated for 48 h for RNA extraction and cell function experiments. Cell viability measurement Cell viability was assessed by a CCK-8 (K1018, APExBIO, USA) assay and a colony formation assay. The cells were seeded at 1000 cells per well in 96-well and 6-well plates after treatment under the corresponding conditions. The absorbance values were measured every 24 h via a CCK-8 assay at a wavelength of 450 nm for 6 days. The culture medium was aspirated from a 96-well plate, and cell viability was measured after two hours of incubation with 100 µl of culture and 10 µl of CCK-8 reagent. Cells were seeded in 6-well plates and fresh medium was changed every 4 days. Macroscopic clones were formed after 2 weeks, the medium was discarded, fixed with 4% paraformaldehyde (Servicebio, China), and stained with 0.1% crystal violet. Transwell assay The cells were diluted in serum-free medium and seeded in Transwell chambers (#353097, Falcon, USA) at 50,000 cells per well. Chambers were placed in a culture plate containing complete medium to allow for the possible migration of cells across the membrane to the bottom chamber. After fixation with paraformaldehyde, the membranes were stained with 1% crystal violet. VEs collection VEs were collected using Tedlar air bag (W-3989, DuPont). VEs comes from a certain model of car, which has a total mileage of about 50,000 km and uses 92# gasoline of Sinopec. The collected gas was mixed one-to-one with the medium for 24 h and then used for experiments. Data acquisition and processing The genes of VEs mediated PCa were obtained from the Comparative Toxicogenomics Database. The transcriptome data, clinical data, and mutation data for PCa were obtained from The Cancer Genome Atlas (TCGA) database. The validation datasets [53]GSE116918 and [54]GSE94767 were downloaded from the Gene Expression Omnibus (GEO) database. Gene set enrichment The “limma” package was utilized for computing the LogFoldChange and p value for each gene in different groups. All genes were imported for gene set enrichment analysis (GSEA) via the “Pi” package. Gene set variation analysis (GSVA) was performed on VEs score samples via the GSVA package. Construction of the VE score First, the genes associated with VEs were analyzed by univariate Cox regression. Least absolute shrinkage and selection operator (LASSO) Cox regression analysis can perform variable selection and regularization, and by imposing a penalty term (λ × absolute slope) on the size of the model coefficients, overfitting can be avoided, which can further improve the accuracy and interpretability of the prediction model. Therefore, it is a sensible choice to build a prediction model based on VEs-associated genes. The LASSO regression algorithm was performed using the glmnet R package. In addition, 1000 replicates were performed for each cycle to prevent overfitting. According to the results of the operation, we find that the best λ value is between 1 and 6, that is, lambda.1se = 1, lambda.min = 6. Subsequently, cox regression analysis showed that DNMT3B, KDM2A, PAX9 and TSHZ1 were the genes used to construct the model. The VEs scoring formula was established as follows: graphic file with name d33e351.gif Statistical analysis All experiments were carried out for three times. The data in this research were statistically analyzed and visualized via R language and GraphPad Prism. All comparisons between groups were performed via the Wilcoxon test. If not stated otherwise, p < 0.05 was considered statistically significant. ns, not significant, * p < 0.05, ** p < 0.01, and *** p < 0.001. Results Expression landscape of VEs-associated genes in PCa We obtained 27 genes associated with VEs from the CTD database. (Supplement material 1 ). The levels of differences between tumor and normal tissues were further explored. The expression of B3GAT1, CHEK2, DNMT3B, MAP2K1, and RNF43 was markedly upregulated in tumor, whereas the expression of BCL2, CDKN1B, ITGB8, MBNL1, MECON, PAX9, PKNOX2, POU2F2, PPFIBP2, RNASEL, TCF4, TCF7L2, and TTC28 was markedly downregulated in tumor tissues (Fig. [55]1A). Furthermore, we investigated copy number changes (CNVs) in these genes and reported that CNVs were prevalent in 29 VEs-associated genes. Among them, KDM2A, PAX9, and MECOM presented extensive CNV gains, whereas RNLS, CHD3, CDKN1B, TSHZ1, BCL2, CDH1, TCF4, TTC28, PKNOX2, POU2F2, and CHEK2 presented CNV loss (Fig. [56]1B). The gene networks describe a comprehensive picture of VEs-associated gene interactions and their impact on the progression-free interval (PFI) in PCa (Fig. [57]1C). We subsequently performed an enrichment analysis of the effects of VEs on prostate cancer via the CTD database. Among the VEs-enriched diseases, the main correlation was prostate-related diseases, especially prostate cancer (Fig. [58]1D). In Fig. [59]1D, it was mainly shown that VEs mainly affect biological processes in GO enrichment analysis. In addition, VEs affected pathways related to the PI3K/AKT signaling pathway, BH3 proteins/BCL-2 members, and EGFR tyrosine kinase inhibitor resistance, as shown by pathway enrichment analysis (Fig. [60]1D). Fig. 1. [61]Fig. 1 [62]Open in a new tab The VEs-associated gene landscape in prostate cancer. (A) Comparison of VEs-associated genes between the tumor and normal groups. (B) Frequencies of CNV gain and loss among VEs-associated genes. (C) Expression and interaction of VEs-associated genes in prostate cancer patients. The size of each cell represents the impact of each gene on the patient’s PFI. (D) Disease, GO, and pathway enrichment analyses of genes associated with VEs Screening of genes for constructing the VEs score model Univariate Cox regression analysis was used to further investigate the associations between VEs-associated genes and the PFI. (Supplemental material 2 and Fig. [63]2A). Subsequently, LASSO analysis was used to construct the PFI model (Fig. [64]2B). Finally, DNMT3B, KDM2A, PAX9, and TSHZ1 were selected for PFI model construction (Supplemental material 3 and Fig. [65]2C). The sensitivity of the VEs score was assessed by comparing the concordance indices (C-index) values. Obviously, the C-index of the VEs scoring models were greater than those of the individual genes. (Fig. [66]2D). Kaplan-Meier analysis manifested that patients with high DNMT3B expression had relatively poor PFIs, whereas the opposite was observed for PAX9. However, the expression of KDM2A and TSHZ1 was not significantly associated with PFI ( Supplement material 4A ). GSEA indicated that high expression of DNMT3B was connected to E2F transcription factors, the G2/M checkpoint and mitotic spindle assembly (Fig. [67]2E). Moreover, high expression of PAX9 was connected to early response factors, the interferon gamma response and TNFα signaling (Fig. [68]2E). We subsequently performed protein‒protein interaction analysis for DNMT3B and PAX9 via GeneMANIA, aiming to explore the potential interactions between proteins related to these three proteins (Supplemental material and Fig. [69]2F). The crystal structure of the DNMT3B-DNMT3L complex was obtained from the Protein Data Bank. Fig. 2. [70]Fig. 2 [71]Open in a new tab Screening VEs score-related model genes. (A) Univariate Cox regression analysis of VEs-associated genes. (B) LASSO regression analysis of prognosis-related genes. (C) Multivariate Cox regression analysis of VEs-associated genes. (D) C-index curve of the PFI. (E) Kaplan‒Meier analysis of the PFI of prostate cancer patients with low and high expression of DNMT3B and PAX9 was performed with data from the TCGA database. (F) GSEA of DNMT3B and PAX9 in different expression groups. (G) Determination of genes that interact with DNMT3B and PAX9 in tumors via physical interactions. Crystal structure of the DNMT3B-DNMT3L complex The VEs score model as a predictor of the PFI Patients with high VEs scores had significantly worse PFIs in the TCGA database (Fig. [72]3A). In the two validation cohorts, we reached the same conclusion (Fig. [73]3B). The evaluation of clinical information for TCGA prostate cancer patients revealed that the VEs score was significantly different from the Gleason score, T stage, and N stage (Fig. [74]3C and D). We observed an increase in the VEs score with increasing Gleason score (Fig. [75]3C). GSVA revealed that in addition to E2F-related pathways and the G2/M checkpoint pathway, other signaling pathways were observably activated in the low VEs score group (Fig. [76]3E). However, according to the results of our analysis, patients with high VEs scores had poor PFIs. Moreover, the cell cycle signaling pathway was activated in the high VEs score group (Supplement material 4B). Androgen receptor (AR) pathway is the core factor in the pathogenesis of PCa. Nevertheless, androgen response was worse in the high VEs score group (Fig. [77]3E and Supplement material 4B). Subsequently, we explored the associations of AR signaling pathway-related genes with VEs scores, which generally showed a negative trend with AR (Fig. [78]3F). These results indicate that the high VEs score group has greater malignant potential. Furthermore, there was no difference in AR mutation among the different VEs score groups (Fig. [79]3I). We subsequently observed a highly positive correlation between TMB and VEs scores (Fig. [80]3G). The top 10 mutation genes in the low VEs score group included SPOP, TTN, FOXA1, KMT2D, MUC16, TP53, SYNE1, ATM, RP1, and RYR2. The top 10 mutation genes in the high VEs score group included TP53, TTN, SPOP, KMT2D, MUC16, FOXA1, KMT2C, LRP1B, PTEN, and SPTA1 (Fig. [81]3H). Fig. 3. [82]Fig. 3 [83]Open in a new tab VEs score model and associations with survival and clinical features. (A-B) Kaplan‒Meier analysis of the VEs score low and high groups in the TCGA, [84]GSE116918 and [85]GSE94767 cohorts. (C) VEs scores of different Gleason score groups in TCGA. (D) Differences in clinical characteristics between the low- and high-VEs score groups. (E) GSVA revealed pathways enriched with different VEs scores. (F) Pearson’s correlation analysis between the VEs score and the expression of AR signaling pathway-related genes. (G) Relationship between VEs score and TMB. (H) Top 10 mutated genes in the two VEs score groups. (I) Mutation comparison of the AR gene in different VEs score groups Predictive potential of the VEs score in the response to cancer immunotherapy To reveal the predictive effect of the VEs score on cancer immunotherapy efficacy, we downloaded and evaluated the activity score of the cancer immune cycle from the Tracking Tumor Immunophenotype database. The VEs score was positively correlated with the release of cancer cell antigens, priming and activation, T-cell recruitment, eosinophil recruitment, basophil cell recruitment, B-cell recruitment, Th2 cell recruitment, and Treg cell recruitment. However, Th17 cell recruitment, MDSC recruitment and recognition of cancer cells by T cells were inversely associated with the VEs score (Fig. [86]4A and Supplementary material 6 ). In addition to immune cells, immune-related regulatory factors also play a key role in immunotherapy [[87]19]. Except for CTLA4, there was no difference in the expression of other genes between the two groups. (Fig. [88]4B). Meanwhile, we investigated the association between the VEs score and major histocompatibility complex (MHC) and found that TNFSF14, TNFSF18, TNFSF25, and CD28 were increased in the high VEs score group. (Fig. [89]4C). In terms of the expression of CTLA4, the high VEs score group was more sensitive to immunotherapy (Fig. [90]4D). Moreover, the cytolytic activity (CYT) presented a positive correlation with VEs score (Fig. [91]4E). GSEA demonstrated that immune-associated pathways were upregulated in the high VEs score groups (Fig. [92]4F). Therefore, we believe that patients with high VEs scores have a more active immune status and are more suitable for combination immunotherapy. Fig. 4. [93]Fig. 4 [94]Open in a new tab Estimation of anticancer immune activity via the VEs score. (A) Heatmap of the seven-step cancer immunity cycle and VEs score. (B-C) The expression of immune checkpoint-related genes and the MHC gene set in the two VEs score groups. (D) IPS between the two VEs score groups. (E) Relationships between VEs scores and CYT scores. (F) GSEA for the high VEs score group Effect of the VEs-associated gene DNMTB3 on prostate cancer progression To explore the function of DNMT3B in PCa, two small interfering RNAs (siRNAs) targeting DNMT3B were transfected into PCa cells. The mRNA level of DNMT3B was markedly knocked down (Fig. [95]5A). Subsequently, CCK8 and colony formation assays were used to verify the interference of DNMT3B knockdown on proliferation. The proliferation ability of tumor cells was significantly inhibited when DNMT3B was knocked down (Fig. [96]5B and C). A cell migration assay revealed that decreased DNMT3B expression resulted in decreased migration (Fig. [97]5D). Therefore, the above results suggest that DNMT3B has a critical role in prostate cancer progression. Moreover, we observed that the expression level of DNMT3B was increased after VEs treatment ( Supplement material 7). Fig. 5. [98]Fig. 5 [99]Open in a new tab DNMT3B promotes the proliferation of prostate cancer cells in vitro. (A) qRT‒PCR analysis of DNMT3B expression levels in DNMT3B-knockdown and control cells. (B) Cell viability was assessed in DNMT3B-knockdown prostate cancer cells. (C) Colony formation assays were performed in DNMT3B‐knockdown prostate cancer cells. (D) Representative images of prostate cancer cells migration assays showing cell migration after downregulation of DNMT3B Discussion VEs are important contributors to air pollution and adversely affect air quality and human health [[100]1]. Outdoor air pollution is classified as a Group 1 carcinogen [[101]20]. In particular, traffic-related air pollutants contain a variety of carcinogenic compounds [[102]21]. In addition to increasing cancer risk, epidemiological studies have shown that these pollutants may trigger several other diseases, such as cardiovascular disease, stroke, respiratory infections, and adverse neurological effects [[103]22]. Some studies have revealed an association between prostate cancer and VEs or occupations exposed to VEs [[104]17]. The morbidity of PCa is high in Western populations. Lifestyle and environmental factors contribute to the pathogenesis of PCa [[105]23]. The role of environmental factors in tumorigenesis is an important area of research. Our study provides a new perspective on the association between VEs and prostate cancer. Using univariate Cox analysis and LASSO analysis, we created a score associated with VEs that divided the PCa cohort into two groups. The patients with high VEs scores had worse PFI. This finding was subsequently confirmed in an external validation dataset. Moreover, we observed that the androgen response pathway was inhibited in the high VEs group, which had a worse PFI. This may be caused by relevant mechanisms that are not AR-dependent. The research has shown that SIX2 promotes the progression of prostate cancer cells through Wnt/β-catenin signaling pathway [[106]24]. The non-genomic functions of AR have an important impact on the progression of prostate cancer [[107]25]. Similarly, epithelial-to-mesenchymal transition [[108]26] neuroendocrine transformation [[109]27] and glucocorticoid receptors [[110]28] are also critical for prostate cancer progression. Our results suggest that the VEs score has some value in evaluating the progression of PCa. These findings not only further support that VEs can significantly influence cancer prognosis but also highlight the potential benefit of our predictive scoring model in prostate cancer management. At present, immune checkpoint blockade has achieved clinical success in the treatment of a variety of malignant tumors, but it is still at a relatively basic stage in the treatment of prostate cancer. Characteristics such as heterogeneity, cold TME and low number of neoantigens lead to the inefficiency of prostate cancer immunotherapy [[111]29]. Notably, the VEs score was significantly associated with immune-related characteristics. Diesel exhaust particles can induce oxidative stress and pro-inflammatory response to mediate macrophage activation and dysfunction in the lung [[112]30]. Patients with higher VEs scores had more active immune statuses. Among them, T cells, Eosinophil cells, Basophils cells, B cells, Th2 cells and Treg cells were more active in the high VEs score group. T cells suppress anti-tumor immunity, which is a great challenge in cancer immunotherapy [[113]31, [114]32]. Shimon et al. revealed that Treg cells inhibit the CD8^+ T cells, drive the induction of thymocyte selection-related high mobility group protein (TOX), and mediate genuine CD8^+ T-cell exhaustion [[115]33]. Tumor-infiltrating B cells and plasma cells are emerging anti-tumor targets as participants in the anti-tumor response [[116]34]. Similarly, eosinophils infiltrate multiple solid tumor types and interact directly with tumor cells through non-mutually exclusive mechanisms [[117]35]. These hyperactive immune cells could serve as effective therapeutic targets in the future. In the clinic, the development of CD8^+ T cell-related targets, such as PD-1/PD-L1 and CTLA4, is more well developed [[118]36, [119]37]. However, the expression of CTLA4 was increased in the VEs high score group, which made it highly possible to benefit from ICI treatment. Meanwhile, we found that a high VEs score was connected with Treg cell recruitment. Therefore, the development of drugs targeting Treg cells for these patients can be more effective. Current extensive research on cancer biomarkers has accelerated the diagnosis and treatment of cancer. We used four potential biomarkers (DNMT3B, KDM2A, PAX9, and TSHZ1) to construct a novel VEs score-related prognostic model. DNMT3B is a member of the DNA methyltransferase family whose function is de novo methylation, and it mediates the occurrence of a variety of tumors [[120]38–[121]41]. Further experiments confirmed that DNMT3B could promote PCa progression. In addition, we proved that VEs could promote DNMT3B mRNA expression in vitro. Multiple components in VEs have been verified to promote DNMT3B expression. PM2.5 exposure enhanced DNMT3B expression through ROS/AKT [[122]42]. This implies that oxidative stress is involved in the regulation of DNMT3B. FAS-AS1 and DNMT3B formed a mutual inhibitory loop, and DNMT3B increased in benzene exposure to inhibit FAS-AS1 expression [[123]43]. Moreover, continuous PAHs exposure, especially Benzo[a]pyrene, significantly increased the expression of DNMT3A and DNMT3B [[124]44]. The limitation of this research is that although our results revealed a clear link between VEs exposure and gene expression patterns in prostate cancer. Other unmeasured environmental exposures may simultaneously correlate with vehicular exhaust exposure and prostate cancer progression. Furthermore, models of VEs exposure, such as in vitro treatment, may not fully replicate real-world environmental exposures. We preliminarily verified that DNMT3B promoted prostate cancer progression. However, DNMT3B may not be the sole epigenetic regulator influenced by vehicular exhaust. PAHs in VEs are known to promote tumorigenesis via activation of the aryl hydrocarbon receptor (AhR) pathway, an independent mechanism that could act as an uncontrolled confounder. Studies supported that the abnormal changes in single nucleotide polymorphisms of DNMT3B lead to DNA hypomethylation or hypermethylation [[125]45]. Hypermethylation of DNMT3B and a novel epigenomic subtype associated with somatic mutations were found in 22% of prostate cancer patients [[126]46]. In addition, coexisting pollutants such as polyphenols [[127]47] and organic pollutant [[128]48] can interfere with DNMT3B expression. Therefore, further studies on the regulatory mechanism of VEs and DNMT3B are needed in the future. Electronic supplementary material Below is the link to the electronic supplementary material. [129]Supplementary Material 1^ (8.7KB, xlsx) [130]Supplementary Material 2^ (9.9KB, xlsx) [131]Supplementary Material 3^ (8.7KB, xlsx) [132]Supplementary Material 4^ (462.6KB, docx) [133]Supplementary Material 5^ (12.3KB, xlsx) [134]Supplementary Material 6^ (792.4KB, docx) [135]Supplementary Material 7^ (3.3MB, docx) Author contributions Bingzheng An: Writing– original draft, Formal analysis. Shuo Chen: Supervision, Project administration. Chen Zhang: Validation, Software. Junyan Wang: Visualization, Formal analysis. Zhaoxin Guo: Supervision, Project administration. Ze Gao: Writing– review & editing, Resources, Project administration. Funding This work was supported by grants from Shandong Provincial Natural Science Foundation (No. ZR2023QH192). Data availability No datasets were generated or analysed during the current study. Declarations Ethics, consent to participate, and consent to publish Not applicable. Competing interests The authors declare no competing interests. Footnotes Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Bingzheng An and Shuo Chen contributed equally to this work. Contributor Information Zhaoxin Guo, Email: sdlzx2k@126.com. Ze Gao, gaoze1992@email.sdu.edu.cn. References