Abstract Background Given the limitations of conventional therapies in prostate cancer (PCa) management, identifying novel biomarkers capable of predicting tumor prognosis and immunotherapy response is critically important. This article revealed the prognosis, immunological characteristics, and potential mechanisms of HKR1 in PCa via bulk and single-cell RNA sequencing (scRNA-Seq). Methods Bulk and scRNA-Seq analyses of HKR1 in PCa were collected from online databases. Differential expression and Cox regression analyses were carried out to evaluate its expression and prognosis values in PCa, respectively. Correlation analyses were performed to evaluate associations between HKR1 expression and enriched pathways, immune cell infiltration, and other relevant biological processes. Results HKR1 showed higher expression in PCa than in normal tissues, as verified by qPCR in both PCa cell lines and tissue samples (p < 0.05). ScRNA-seq analysis demonstrated HKR1 expression in malignant cells, epithelial cells, and immune cell populations. Moreover, PCa sufferers with higher HKR1 expressions were linked with poorer prognoses, and Cox regression analysis suggested it was an independent indicator in PCa (p < 0.05). Further, we shed light on the fact that the toll-like receptor, the TGF-beta, and the p53 pathways were significantly related to HKR1 expression in PCa. HKR1 was also found to be markedly linked to immunity in PCa (p < 0.05). Notably, we characterized two novel lncRNA-RBP-HKR1 regulatory axes that potentially modulate HKR1 transcriptional dynamics in prostate carcinogenesis. Conclusions HKR1 played an undeniable role in the prognosis and immunological potential of PCa, providing evidence for the molecular mechanisms of HKR1 in PCa. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-025-14230-9. Keywords: HKR1, Prostate cancer, Immunotherapy, Prognosis, ScRNA-sequencing Introduction Prostate cancer (PCa) was the most common type of urinary malignant tumor, with approximately 268,490 new cases diagnosed in 2022 [[32]1]. Numerous epidemiologic studies had demonstrated several risk factors of PCa, involving age, family history, lifestyle, environmental or occupational exposures, and so on [[33]2]. The clinical adoption of prostate-specific antigen (PSA) testing in the early 1990s marked a transformative advancement in prostate cancer care, significantly enhancing early detection capabilities and therapeutic outcomes [[34]3]. Currently, various clinical interventions were developed for PCa therapy, including radiation therapy, androgen deprivation therapy, chemotherapy, and novel immunotherapies [[35]4]. Although early-stage PCa sufferers’ five-year overall survival (OS) rate was satisfactory, a majority of patients were still prone to relapse and develop castration-resistant prostate cancer after surgery or endocrine therapy [[36]5]. Meanwhile, false-negative results in PCa patients with normal serum PSA levels could affect timely treatment and lead to a poor prognosis [[37]6]. PCa exhibits distinct biological features—including pronounced tumor heterogeneity, a heavily immunosuppressive microenvironment, and minimal lymphocytic infiltration—that collectively contribute to the limited efficacy of conventional immunotherapies [[38]7]. Immune checkpoint inhibitor monotherapy demonstrated limited clinical efficacy, while combination therapy with the PD-1 inhibitor nivolumab and the CTLA-4-targeted agent ipilimumab exhibited a partial response in clinical trials [[39]8]. In addition, the predominant tumor-associated antigens in PCa include prostate-specific antigen (PSA), prostate-specific membrane antigen (PSMA), and six-transmembrane epithelial antigen of the prostate-1 (STEAP1) et al., which garnered significant interest in therapeutic development, with major approaches encompassing vaccine-based strategies and CAR-T cell therapies [[40]9]. However, most of these therapeutic approaches remained in Phase I/II clinical trials, indicating a substantial pathway to clinical implementation [[41]10]. Therefore, it was essential to investigate new prognostic biomarkers to predict the occurrence of PCa and explore their clinical applications in PCa. Members of the Krüppel-associated box domain zinc finger (KRAB-ZNF) family served as epigenetic suppressors, increasing DNA methylation, which was discovered to be involved in proliferation, development, and even the carcinogenesis of cancer [[42]11, [43]12]. The human kruppel-related gene 1 (HKR1), as one member of the KRAB-ZNF family, was responsible for recruiting histone deacetylase complexes to domains of DNA-binding sites [[44]13, [45]14]. As reported, HKR1 played crucial roles in regulating DNA methylation, and the longitudinal association between HKR1 methylation level and physical development was also revealed [[46]15, [47]16]. Studies demonstrated a significant association between HKR1 methylation levels and phosphorylated tau deposition load [[48]17]. Moreover, Oguri et al. proposed that HKR1 was up-regulated in lung carcinoma in comparison with normal tissues [[49]18]. One whole-exome sequencing on PCa patients with a family history of the disease was performed, and a truncation mutation of HKR1 was identified, which might contribute to the occurrence of PCa [[50]19]. In conclusion, these findings revealed the intrinsic connection between HKR1 and various diseases, including PCa. Nevertheless, the associations between HKR1 and PCa remained unknown. More investigations were necessary to be carried out on the potential functions of HKR1 in PCa based on public databases. Meanwhile, the “starBase” database was utilized to identify potential upstream mechanisms of HKR1 in PCa. This study identified a novel prognostic biomarker for Pca and characterized its potential clinical utility in therapeutic decision-making. Materials and methods Acquisition and analysis of PCa bulk RNA-seq data The TCGA-PRAD bulk RNA-seq and clinical data ([51]http://cancergenome.nih.gov/; accessed on January 2023) were employed for the current study, with 499 PCa samples and 52 normal samples. Those samples without critical clinical data were eliminated. These data were further normalized and processed in the R statistical environment, as previously described [[52]20, [53]21]. We analyzed HKR1 mRNA expression across multiple cancer types using the TIMER database ([54]https://cistrome.shinyapps.io/timer/), while protein expression in prostate cancer was assessed through the HPA ([55]https://www.proteinatlas.org/) [[56]22]. Differential expression analysis of HKR1 among PCa and normal sample tissues was conducted via the “limma” R program with a threshold of p-values below 0.05. Acquisition and analysis of PCa single-cell RNA sequencing (scRNA-seq) data The PCa scRNA-seq data for HKR1 was acquired from the [57]GSE141445 dataset containing 13 PCa tumor samples from the GEO online website ([58]https://www.ncbi.nlm.nih.gov/geo/; accessed on January 2023). The prior article was cited for information on data processing and cell cluster annotations [[59]23]. Briefly, the measurement data were merged with the “Seurat” R package and subjected to principal component analysis. Using Seurat’s FindNeighbors function (v4.0, default parameters), we first computed the k-nearest neighbor (kNN) relationships between cells, then transformed these into a shared nearest neighbor (SNN) graph for subsequent graph-based clustering. With local neighborhoods of cells, the “Seurat” R package’s “FindClusters” function was used to cluster cells identified with accepted gene markers. Prognostic abilities assessment Patients from the TCGA-PRAD dataset were grouped into a low- or high-HKR1 group with the median expression level of HKR1, and K-M survival analysis was carried out to discover whether the OS rate was different between these two subgroups. Moreover, to investigate the clinical relevance of HKR1 in PCa, we used clinical information from the TCGA-PRAD dataset, including age, lymph nodes, tumor status, PSA value, Gleason’s grade, stage, T, and N. Due to the small sample size and non-normal distribution of the data, pairwise comparisons between groups were performed using the Wilcoxon rank-sum test, while comparisons involving more than two groups were conducted using the Kruskal-Wallis test. Further, COX regression analyses were conducted to investigate relationships among OS and above clinical characteristics, as well as HKR1 expression, to identify independent prognostic markers for PCa. A nomogram was depicted with the clinical characteristics mentioned above using the “rms” R software package, with ROC curves, C-index, and calibration curves evaluating its predictive performance. Gene Set Enrichment Analysis (GSEA) and protein-protein interaction (PPI) network To investigate the biological role of HKR1 in PCa, GSEA version 4.0.0 ([60]http://www.gsea-msigdb.org/gsea/login.jsp) was employed to perform KEGG pathway enrichment analysis. This approach identifies key signaling pathways associated with specific biological processes [[61]24]. The KEGG pathway analysis using the c2.cp.kegg.v7.1.symbols.gmt dataset revealed enriched pathways associated with HKR1 expression profiles in the TCGA-PRAD dataset. The cut-off criteria consisted of NES (normalized enrichment score) > 1.5 as well as p values < 0.05. Additionally, the STRING database was utilized to draw a network of PPI networks for HKR1 ([62]https://cn.string-db.org/). To obtain information on protein interactions, we set the organism filter to “Homo sapiens” and specified the minimum required interaction score as the median confidence level (0.4). Immune assessment and drug sensitivity Following the methodology described in previous study [[63]25], we assessed correlations between HKR1 expression and immunogenomic features—including immune cell infiltration, tumor neoantigen burden (TNB), microsatellite instability (MSI), tumor mutational burden (TMB), and immune checkpoint molecule (ICM) expression—using the Sangerbox platform ([64]http://www.sangerbox.com/tool). Statistical significance was set at p < 0.001. The tumor microenvironment, containing stromal and immune scores, was estimated using TCGA-PRAD transcriptome data by means of the “ESTIMATE” algorithm, and the immune cell infiltration levels of HKR1 in PRAD were conducted by the “TIMER” website. We also utilized the “CellMiner” online website ([65]https://discover.nci.nih.gov/cellminer/) to find correlations among HKR1 and the IC50 values of common molecular drugs. StarBase v2.0 website identified HKR1-related mechanisms in PCa To investigate upstream mechanisms regulating HKR1, the LncRNA/RNA binding protein (RBP)/HKR1 network was identified through the “StarBase” website ([66]https://starbase.sysu.edu.cn/), as described in previously published articles [[67]26, [68]27]. First, we conducted a medium stringency (≥ 5) and pan-cancer cut-off criterion (≥ 22) to search for HKR1-targeted RBPs. Further, the RBP-relevant LncRNAs were identified with a threshold of pan-cancer ≥ 10, medium stringency (≥ 5), and hub LncRNAs in TCGA-PRAD (|log[2](fold change)|≥1, FDR < 0.05, OS p < 0.05). In addition, LncRNAs were negatively correlated with the expression of HKR1 (p < 0.001; correlation coefficient > 0.3). Finally, we visualized these interaction networks using Cytoscape software (version 3.8.2). Cell culture and clinical samples PRAD cell lines (LNCaP and DU145) and a human normal prostate cell line (WPMY-1) were purchased from the Cell Bank of the Shanghai Institute of Biological Science (SIBCB; China) and cultured according to the instructions described on the SIBCB website ([69]https://www.atcc.org). The PRAD and paraneoplastic tissues were obtained from PRAD patients who underwent surgery. Our current study has been approved by the institutional review board, and all patients involved signed an informed consent form for the research only. RNA extraction and real-time qPCR Total RNAs from PCa samples and cell lines were acquired by TRIzol (Takara, Kusatsu, Japan). The PrimeScript RT reagent kit (Novabio, China) was utilized to synthesize the complementary DNA. Gene expression profiling was conducted via SYBR Green-based quantitative RT-qPCR, with amplification data acquired and analyzed using StepOne Software v2.3 for relative quantification. The relative expression level was normalized through the ACTB expression. The primer sequences were listed as follows: ACTB primer (forward): 5’-CATGTACGTTGCTATCCAGGC-3’; ACTB primer (reverse): 5’-CTCCTTAATGTCACGCACGAT-3’ (reverse); HKR1 primer (forward): 5’-CTCTGTCCAGAATCGAAGCCA-3’; HKR1 primer (reverse): 5’-GAGGATTTCCTGCCCATAAACTT-3’. Statistical analysis The R programming language version 4.2.1 was used in this article. To examine the differences between two or more subgroups, the “Wilcoxon” test was used. Statistical analyses considered results with p-values < 0.05 to be significant. Results Bulk RNA-seq data of HKR1 expression and its prognosis in PCa The HKR1 expressions in pan-cancer were described in Fig. [70]1A. It revealed that HKR1 was down-regulated in malignancies including CHOL, COAD, READ, LIHC, and STAD, while up-regulated in tumors like PRAD, LUSC, UCEC, THCA, and LUAD (p < 0.05). The qRT-PCR analysis revealed significantly higher HKR1 mRNA expression in LNCaP cells compared to WPMY-1 cells (Fig. [71]1B). Also, HKR1 was up-regulated in PRAD tissues in contrast to normal tissues (Fig. [72]1C). Figure [73]1D unequivocally demonstrated that HKR1 was up-regulated in PRAD samples in comparison to normal prostate tissues in the TCGA-PRAD dataset (p = 9.982e-06). Kaplan-Meier analysis revealed HKR1 expression level as a significant prognostic factor, with low-expressing patients exhibiting prolonged overall survival compared to high-expressing cases (p = 0.014, Fig. [74]1E). The HPA database suggested that HKR1 protein expression was low in normal prostate tissues compared to PRAD samples (Fig. [75]1F-G). Fig. 1. [76]Fig. 1 [77]Open in a new tab Bulk RNA-seq data and survival prognosis of HKR1 expression in PRAD. (A) Pan-cancer analysis revealed HKR1 was up-regulated in PRADS with TCGA database; (B) qRT-PCR results indicated that HKR1 was up-regulated in PCa cell line LNCaP compared with normal WPMY-1 cells; (C) qRT-PCR results indicated that HKR1 mRNA expression was higher in PRAD tissues compared with adjacent tissues; (D) HKR1 was up-regulated in tumor tissues compared with normal tissues in TCGA-PRAD; (E) Survival analysis revealed high HKR1 expression is correlated with a poor prognosis in TCGA-PRAD; (F-G) The protein expression of HKR1 was highly-expressed in PCa tissues compared with tissues from HPA database. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 scRNA-seq data of of HKR1 expression in PCa To evaluate the HKR1 expression in PRAD at the single-cell level, the [78]GSE141445 dataset containing 13 PRAD samples was further enrolled. After data processing, the cell clusters were annotated by UMAP plot, as presented in Fig. [79]2A. The accepted markers utilized for each cell cluster were detailed in Figure [80]S1. Single-cell resolution analysis revealed the expression pattern of HKR1 in PRAD, visualized through UMAP dimensionality reduction (Fig. [81]2B) and violin plots (Fig. [82]2C). The results indicated that HKR1 could be expressed in various cell clusters, including malignant cells, epithelial cells, and immune cells. Fig. 2. [83]Fig. 2 [84]Open in a new tab [85]GSE141445 scRNA-seq data of HKR1 expression in PRAD. (A) PRAD samples were annotated with eight cell clusters by UMAP plot; (B) The expression of HKR1 in PRAD at single-cell levels by UMAP plot; (C) HKR1 had relatively higher expressions in epithelial cells, and malignant cells at single-cell levels Correlation analysis between HKR1 expression and the clinicopathological characteristics By using clinical information from the TCGA-PRAD dataset, patients were grouped with different clinicopathological characteristics, containing age, race, PSA value, T, M, N, and so on. Correlation analyses between HKR1 expression and clinicopathological information in PRAD were further conducted, and the Wilcoxon rank sum and Kruskal-Wallis tests were utilized for comparison. Notably, we found that the expression of HKR1 was significantly linked with age, PSA value, and T stage in Fig. [86]3 (all p < 0.05). Fig. 3. [87]Fig. 3 [88]Open in a new tab Relationship of HKR1 expression with clinicopathological characteristics in PRAD. Correlation analysis between HKR1 expression with (A) age; (B) race; (C) PSA-value; (D) T; (E) M; (F) N; (G) gleason-score; (H) biochemical-recurrence; (I) cancer-status Prognostic values of HKR1 in PRAD Through univariate Cox regression analysis, we identified that HKR1 expression and PSA value could serve as prognostic indicators in PRAD. Meanwhile, Cox regression analysis demonstrated that HKR1 might be an independently prognostic biomarker in PRAD (Table [89]1, p < 0.05). These findings indicated the prognostic value of HKR1 in PRAD. Prognostic modeling was performed through nomogram construction (R ‘rms’ package, version 6.2-0) incorporating: (i) established clinical predictors (age, Gleason score, T stage, N stage, PSA), (iii) recurrence history, and (iv) HKR1 expression levels et al., demonstrating integrated predictive performance for PRAD survival outcomes (Fig. [90]4A). The AUC of this nomogram’s 5-year ROC curve measured 0.974 (Fig. [91]4B). The performance of this nomogram was proven by 5-year calibration curves (Fig. [92]4C). Table 1. Univariate and multivariate Cox analysis based on HKR1 and clinicopathologic characteristics of OS for PRAD ID Univariate analysis Multivariate analysis HR HR.95 L HR.95 H P value HR HR.95 L HR.95 H P value Age 0.972997 0.855266 1.106936 0.677408 1.041754 0.872796 1.24342 0.650502 Staged T 2.940790 0.470691 18.373505 0.248550 3.515307 0.11587 106.649139 0.470266 Staged N 5.648441 0.791027 40.333501 0.084305 1.284547 0.004915 335.687756 0.929734 Cancer status 5.238172 0.868125 31.606547 0.070957 4.737859 0.22449 99.992521 0.317409 Lymphnodes 1.981361 0.925045 4.243891 0.078496 1.158299 0.093983 14.275514 0.908701 Gleason score 1.31901 0.514518 3.381395 0.564308 0.467868 0.068884 3.177833 0.437105 Psa value 1.213885 1.031111 1.429057 0.019916 1.374506 0.949736 1.989256 0.091693 HKR1 1.636443 1.223566 2.18864 0.000900 1.729961 1.227366 2.438365 0.001749 [93]Open in a new tab Fig. 4. [94]Fig. 4 [95]Open in a new tab Prognostic values of HKR1 in PCa in PRAD. (A) A nomogram was construct to evaluate its performance in prediction the prognosis for PRAD; (B) The 5-year ROC curve of nomogram; (C) The 5-year calibration curve was constructed to evaluate the performance of the model HKR1 significantly associated with pathways in PRAD GSEA was utilized by us to obtain HKR1 significantly associated with pathways in PRAD. It revealed HKR1 was significantly associated with toll-like receptor (NES=-1.818, p < 0.05), TGF-beta (NES=-1.786, p < 0.05), nod-like receptor (NES=-1.829, p < 0.05), p53 (NES=-1.741, p < 0.05), cytosolic DNA sensing (NES=-1.772, p < 0.05), chemokine (NES=-1.962, p < 0.05), and B cell receptor (NES=-2.015, p < 0.05) pathways, as presented in Fig. [96]5; Table [97]2. Fig. 5. [98]Fig. 5 [99]Open in a new tab HKR1-related signaling pathways through GSEA. GSEA was performed to identify HKR1-relevant signaling pathways in PRAD including (A) B cell receptor signaling pathway; (B) chemokine signaling pathway; (C) cytosolic DNA sensing pathway; (D) nod-like receptor signaling pathway; (E) p53 signaling pathway; (F) TGF-beta signaling pathway; (G) toll-like receptor signaling pathway; (H) all of these seven pathways Table 2. Gene set enrichment analysis results MSigDB collection Gene set name NES NOM P-val FDR q-val c2.cp.kegg.v7.1 symbols.gmt KEGG_B_CELL_RECEPTOR_SIGNALING_PATHWAY -2.015 <0.05 0.011 KEGG_CHEMOKINE_SIGNALING_PATHWAY -1.962 <0.05 0.015 KEGG_CYTOSOLIC_DNA_SENSING_PATHWAY -1.772 <0.05 0.046 KEGG_NOD_LIKE_RECEPTOR_SIGNALING_PATHWAY -1.829 <0.05 0.034 KEGG_P53_SIGNALING_PATHWAY -1.741 <0.05 0.054 KEGG_TGF_BETA_SIGNALING_PATHWAY -1.786 <0.05 0.042 KEGG_TOLL_LIKE_RECEPTOR_SIGNALING_PATHWAY -1.818 <0.05 0.036 [100]Open in a new tab Correlations between HKR1 expression and molecular characteristics A PPI network was depicted via the “STRING” website tool, and the ten most relevant proteins of HKR1 were identified (Fig. [101]6A). Using Sangerbox, we examined potential associations between HKR1 expression and MSI, TMB, and TNB. However, no significant correlations were observed in PRAD (Fig. [102]6B-D). Fig. 6. [103]Fig. 6 [104]Open in a new tab Correlations between HKR1 expression with molecular characteristics. (A) A PPI networks was established to identify ten most HKR1-relevant genes; Correlations between HKR1 expression with (B) MSI; (C) TNB; (D) TMB Correlations between HKR1 expression and tumor immunity The immune subtype composition between two subgroups with HKR1 expression was compared, and it showed that there was no difference between the two subgroups (Fig. [105]7A, p = 0.065). The correlation analyses among immune cell infiltration levels and HKR1 expression were illustrated in Fig. [106]7B. Our analysis revealed significant correlations between HKR1 expression levels and tumor-infiltrating immune cells, particularly CD8 + T cells and CD4 + T cells (p < 0.05). Moreover, HKR1 expression was found to have remarkable associations with ESTIMATE, Immune, and Stroma Scores (Fig. [107]7C, p < 0.05). The associations between HKR1 expression and immunological checkpoint molecules were depicted in Fig. [108]7D. We discovered HKR1 expression was correlated with CD86, CD70, CD44, CD40, CD274, CD200, and CD27 (p < 0.05). Furthermore, HKR1 expression showed significant associations with multiple activated immune cell populations, including B cells, CD8 + T cells, CD4 + T cells, dendritic cells, and central memory CD4 + T cells (Fig. [109]7E, p < 0.05). Fig. 7. [110]Fig. 7 [111]Open in a new tab Correlations between HKR1 expression with tumor immunity. (A) The composition of pan-cancer immune subtypes between two subgroups based on HKR1 expression. Correlations analysis between HKR1 expression and (B) immune cells infiltration; (C) tumor microenvironment; (D) immune checkpoint molecules; (E) immune cells. *P < 0.05; **P < 0.01; ***P < 0.001 HKR1-related drugs based on cellminer database We evaluated the correlations between HKR1 expression and drug sensitivities (IC50 values) through “CellMiner”, with cut-off values of correlation coefficient ≥ 0.3 and p < 0.01. As shown in Fig. [112]8, the HKR1 expression in the samples was positively correlated with the IC50 values of chelerythrine, cladribine, and fludarabine (p < 0.01). Patients with low HKR1 expression showed better response to molecular drugs’ chemotherapies. Fig. 8. [113]Fig. 8 [114]Open in a new tab Correlation analysis of HKR1 expression and drug sensitivity of chemotherapy. Associations between HKR1 expression with IC50 values of (A) chelerythrine; (B) cladribine; (C) fludarabine in the CellMiner database Identification of LncRNA/RBP/HKR1 mRNA networks In this research, the network of LncRNA/RBP/HKR1 was evaluated through the website StarBase v2.0. The specific flowchart is summarized in Fig. [115]9A. The LncRNA/HNRNPA2B1/HKR1 was discovered following a second search of a chosen RBP-relevant LncRNA with Venn diagrams (Fig. [116]9B). Finally, we identified two regulatory axes—LINC00342/HNRNPA2B1/HKR1 and AP00468.1/HNRNPA2B1/HKR1—as potential upstream mechanisms controlling HKR1 expression in PRAD (Fig. [117]9C). Fig. 9. [118]Fig. 9 [119]Open in a new tab Identification of LncRNA/RBP/HKR1 mRNA networks. (A) Flowchart and (B) Venn diagrams of identification for HKR1-related mechanism of LncRNA/RBP/HKR1 mRNA networks. (C) LncRNA/RBP/HKR1 mRNA networks were pictured by Cytoscape 3.6.1 Discussion Millions of men were diagnosed with PCa disease around the world [[120]6]. On the basis of the genetic characteristics of tumors, it was feasible to evaluate the occurrence and progression of PCa. As noted, HKR1 was a member of the KRAB-ZNF gene family, playing important roles in various cancers [[121]28]. Our preliminary data indicated upregulated HKR1 expression in PCa, yet the functional and clinical significance of this observation requires systematic investigation. The scRNA-seq technology could detect the transcriptional landscape of target tissues at the single-cell level, which enabled the fine division of cell subtypes and revealed cellular heterogeneity [[122]29]. Nowadays, its application was very extensive, enabling researchers to gradually deepen their understanding of the tumorigenesis, tumor microenvironment, and immune response in PCa [[123]23, [124]30]. Our analysis of existing single-cell RNA sequencing (scRNA-seq) data revealed HKR1 was mainly expressed in epithelial and malignant cell subtypes in PRAD based on the existing scRNA-seq data. Song et al. had reported the heterogeneities of tumor-related epithelial cell states in PRAD [[125]31]. Nevertheless, whether abnormal expression of HKR1 in epithelial cells promotes prostate cancer needed further exploration. To shed light on various signaling pathways related to the targeted genes, GSEA was further employed [[126]32]. Moreover, we identified several signaling pathways associated with HKR1, including toll-like receptor, TGF-β, p53, cytosolic DNA sensing, NOD-like receptor, chemokine, and B cell receptor signaling pathways. Toll-like receptor 4 was reported to play a vital role in tumor progression through regulating the RAS/MAPK signaling pathway [[127]33]. Deng et al. reported that the TGF-beta-related regulatory network played a vital role in PCa progression [[128]34]. Consistent with findings by Wan and the colleagues, elevated p53 protein expression in prostate cancer cells demonstrated significant associations with enhanced proliferative, migratory, and adhesive capacities [[129]35]. Chemokines and their receptors were proven to contribute survival, proliferation, and invasion capabilities to PCa cells in both the primary and metastatic sites of cancer [[130]36]. The regulation of chemokines and their receptors may potentially contribute to combating tumor development. These findings suggested that HKR1 might take part in the occurrence or progress of PCa through the above signaling pathways, and further experiments were needed to confirm it in vitro. PCa, often characterized as an immunologically “cold” tumor, generally shows limited response to immune-related therapies, although a subset of patients may still benefit from immunotherapy. Thus, the evaluation of the correlation between tumor cells and the tumor microenvironment was crucial for understanding the occurrence, progression, and therapeutic response of PCa [[131]37]. In addition, immune cell infiltration, which was a vital component of the tumor microenvironment, was closely correlated with the progression and immunological responses of the tumor [[132]38]. Our results indicated that HKR1 was positively linked with the number of CD8^+ T cells. Also, HKR1 expression showed a strong correlation with ESTIMATE, stromal, and immune Scores, indicating the relevance of the microenvironment for prognosis and precision immunotherapy of PCa. A high level of stromal, immune and estimate scores had been proven to be linked to a poor prognosis in PCa [[133]39]. Moreover, we found HKR1 expression was significantly associated with immune cells and ICM, including CD86, CD70, CD44, CD40, CD274, CD200, activated CD4 T cells, and etc. These findings demonstrate a strong correlation between HKR1 and tumor immunity in PRAD, highlighting its potential as a target for immunotherapy. To discover HKR1-related drugs in PCa, the “CellMiner” dataset was applied to facilitate the process of researching and selecting anticancer medications [[134]40, [135]41]. Our results demonstrated a significant positive correlation of HKR1 expression with the IC50 of chelerythrine, cladribine, and fludarabine, which had the potential to restrain the progression of PRAD. As reported, chelerythrine could suppress the proliferation of PCa cells via regulating NF-κB pathways [[136]42]. Fludarabine was found to be a potential target of N-MYC overexpressing neuroendocrine PCa [[137]43]. Overall, it meant PRAD patients with abnormal expression of HKR1 were more likely to benefit from these molecular drug therapies. Furthermore, the “StarBase” website was used to explore the upstream mechanisms of HKR1 in PCa, revealing its correlation with several RBPs and associated LncRNAs. Eventually, we identified the LINC00342/HNRNPA2B1/HKR1 and [138]AP004608.1/HNRNPA2B1/HKR1 axes as potential mechanisms of HKR1 in PCa. Heterogeneous nuclear ribonucleoproteins A2B1 (HNRNPA2B1) was a pre-mRNA-binding protein that acted as a crucial RNA regulator of RNA N6-methyladenosine (m^6A), which was involved in the progress and occurrence of various tumors [[139]44, [140]45]. Analysis of datasets from the TCGA database suggests that HNRNPA2B1 may function as a crucial m6A regulator in prostate cancer, and knockdown of HNRNPA2B1 can suppress the migration of PCa cells in vitro [[141]46]. As for the related lncRNA, colorectal tumor cells were induced to metastasize and proliferate by LINC00342 via sponging miR-19a-3p to regulate NPEPL1 expression [[142]47], and silence of LINC00342 could inhibit the invasion, proliferation, and migration of AGS cells in vitro [[143]48]. Besides, [144]AP004608.1 was found to be up-regulated in PCa tissues compared with benign prostate tissues [[145]49]. Overall, we had identified two LncRNA/RBP/HKRI network axes that might act on PRAD. Nevertheless, the two upstream mechanisms of HKR1 still needed further experiments to verify in PRAD. These limitations should also be pointed out in this research. Firstly, most parts of the data were obtained using bioinformatics analysis, and the potential function of HKR1 expression and related mechanisms in PCa needed to be investigated in vivo. Secondly, the value of HKR1 in immunotherapy intervention for PCa still needed further clinical trials to verify. Conclusions In conclusion, HKR1 was found to be up-regulated in PCa, and it could serve as a potential prognostic biomarker for PCa. Moreover, HKR1 was closely associated with several signaling pathways, such as the toll-like receptor, TGF-beta, and p53, in PCa. Moreover, HKR1 was found to be significantly correlated with immunity, which could act as a latent target for the clinical intervention of PCa. Two LncRNA/RBP/HKR1 axes (LINC00342/HNRNPA2B1/HKR1 and AP00468.1/HNRNPA2B1/HKR1) were identified for their potential upstream regulatory mechanisms of HKR1 in PCa. These findings provide a novel prognostic indicator for PCa and reveal its potential impact on immunotherapy in this disease. Electronic supplementary material Below is the link to the electronic supplementary material. [146]12885_2025_14230_MOESM1_ESM.tif^ (870.9KB, tif) Figure S1. Accepted cell markers utilized for each cell cluster by dotplot in the GSE141445 scRNA-seq data of PRAD Acknowledgements