Abstract
Objectives
As a highly prevalent disease in older men, the incidence of benign prostatic hyperplasia (BPH) is increasing as aggravating aging of the global population. This study aimed to explore the pathogenesis of BPH and its potential biomarkers.
Methods
Expression of miR-96-5p/miR-181a-5p and glycolipid metabolism indicators (GLMI) were detected by collecting the peripheral blood samples from 100 healthy volunteers and 110 patients with BPH. The diagnostic value of clinical factors for BPH was evaluated through the receiver operating characteristic (ROC) curve. The inhibitors of miR-96-5p and miR-181a-5p were introduced to detect their roles in BPH pathogenesis and regulation of FOXP2 expression. Interaction between miR-96-5p/miR-181a-5p and FOXP2 mRNA was verified through dual luciferase assay. Silencing FOXP2 to explore its role in the progression of BPH.
Results
GLMI, miR-96-5p, and miR-181a-5p levels are high in BPH patients and increased with the severity of disease, and their blood levels are positively correlated with International Prostate Symptom Score (IPSS). MiR-96-5p combined with miR-181a-5p is a good prediction model for the occurrence and severe progression of BPH, with area under curve (AUC) of 0.901 and 0.927, respectively; and they combined with GLMI showed a high authenticity, with AUC of 0.927 and 0.962, respectively. MiR-96-5p/miR-181a-5p inhibited FOXP2 expression by adsorption of its mRNA, and their inhibition reduced the viability and promoted apoptosis of WPMY-1 and BPH-1 cells, which was reversed by the silence of FOXP2.
Conclusions
GLMI, miR-96-5p, and miR-181a-5p presented a close clinical correlation with BPH, which are valuable biomarkers of BPH, and these two miRNAs may contribute to BPH progression by regulating FOXP2 negatively.
Introduction
Hyperplasia of prostate tissue will lead to benign prostatic hyperplasia (BPH), a common urinary condition for which age is a major risk factor [1]. In 2021, BPH was one of the vital causes of age-standardized prevalence rate and age-standardized incidence rate globally, with an incidence of 5.53 %, or 5,531.88 BPH cases per 100,000 people [2]. BPH primarily causes lower urinary tract symptoms (LUTS), including those associated with an overactive bladder such as frequent urination, urgency, and nocturia, as well as dysuria-related symptoms like delayed and intermittent urination [3].
There are many studies on the etiology and pathogenesis of BPH. Recently, a strong correlation between metabolic syndrome (MS) and BPH has been widely recognized. The key characteristics of MS include abnormal blood glucose and lipid metabolism, central obesity, and hypertension [4]. A study by Fu et al. found that each component of MS was a significant risk factor for BPH [5]. Additionally, MS was closely associated with clinical predictors of BPH progression, such as enlarged prostate volume (PV), accelerated prostate growth, and increased International Prostate Symptom Score (IPSS) [6]. Therefore, this study evaluated the diagnostic value of blood glycolipid metabolism indicators (GLMI), including the levels of hemoglobin A1c (HbA1c), fasting blood glucose (FBG), total cholesterol (TC), and triglycerides (TG), in patients with BPH.
MicroRNAs (miRNAs) are non-coding/single-stranded RNA molecules with short length. They regulate gene expression post-transcriptionally and are implicated in various human diseases. However, research on miRNAs in BPH is limited. A miRNA microarray analysis of BPH patients and healthy men revealed that miR-96-5p, miR-181a-5p, miR-1271-5p, miR-106a-5p, miR-143-3p, miR-4428, and miR-21-3p were up-regulated, while miR-25, miR-486-3p, miR-16-5p, miR-30a-3p, let-7c, miR-19b-5p, miR-191, and miR-940 were lacking, in the BPH prostate tissues [7]. Based on these findings, we chose miR-96/181a-5p as the focus of this research to investigate their diagnostic potential in BPH.
In this study, we analyzed the predictive value of miR-96-5p/miR-181a-5p combined with GLMI in the occurrence and development of BPH by constructing the receiver operating characteristic (ROC) model. Besides, Forkhead box p2 (FOXP2) was identified as a target of miR-96-5p and miR-181a-5p. Although it has been reported in prostate-related malignancies, its role in BPH has not been studied. Here, we explored the role of miR-96-5p/miR-181a-5p-FOXP2 axis in the function of BPH-1 cells.
Materials and methods
Participants and clinical sample processing
The healthy volunteers with normal physical examinations (n=100) and patients with BPH (n=110) came from Zhangjiakou First Hospital. Their basic information was collected in Supplementary Table 1. Their blood samples and basic information were kept intact. Informed consent was obtained from all participants. The Ethics Committee of Zhangjiakou First Hospital approved this study. (Approval Number: 2023040, Date: 2023-06-28).
The blood samples were collected from the anterior elbow veins of the volunteers when they came to our hospital for examination, using EDTA anticoagulant tubes (purple, VACUETTE, Austria), potassium oxalate/sodium fluoride anticoagulant tubes (gray, VACUETTE), and serum tubes (red, VACUETTE). The whole blood samples in the purple tubes were used for HbA1c detection using high performance liquid chromatography in the Mindray glycated hemoglobin analyzer (China), with imprecision (coefficient of variation, CV) of 3.0 %, bias of 3.6 %. The whole blood samples in the grey tubes were centrifuged (1,000 g, 10 min) and the supernatant (plasma) was taken for FBG detection using the hexokinase method in the Beckman Coulter UniCel DxC 800 Synchron fully automatic biochemical analyzer (USA), with CV of 3.0 %, bias of 2.0 %. The whole blood samples in the red tubes were centrifuged (1,000 g, 10 min) and the supernatant (serum) was taken for TC and TG detection using the cholesterol oxidase and glycerol kinase methods in the Beckman Coulter UniCel DxC 800 Synchron fully automatic biochemical analyzer, with CV of 3.0 and 5.0 %, bias of 4.0 and 5.0 %, respectively. For preservation of samples, the separated serum samples were aliquoted into transparent sterile Eppendorf (EP) tubes (Corning, USA) and then stored at −80 °C for sample retention. The blood levels of HbA1c, FBG, TC, and TG were detected when the volunteers were admitted to the hospital. These indicators were collected from clinical laboratory records and defined as GLMI.
Evaluation of diagnostic value for BPH
The predictive value of clinical factors in BPH was evaluated through the ROC curve constructed in SPSS software. The multivariate binary logistic regression analysis was performed to evaluate the risk factors for BPH.
Real-time quantitative PCR (RT-qPCR)
When conducting the research, the serum samples were first placed at −20 °C for 24 h, and then completely thawed at 4 °C for RT-qPCR detection. Trizol reagent (Invitrogen, USA) was used for the RNA extraction of serum samples and cells. Total RNA was reverse transcribed into cDNA using the BeyoRT III cDNA First Strand Synthesis kit (Beyotime, China). RT-qPCR was performed using SYBR® Green real-time PCR Premix (Toyobo Biologics, Japan) in a transparent 96-well PCR microplate (Corning, USA). U6 Small Nuclear 1 (U6) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were used as internal reference genes. The relative expression of genes was measured by the 2−ΔΔCt. The primer sequences are shown below: miR-96-5p (F/R: 5′-CAGTCGTTTTTACACGATCAC-3′/3′-GGTCCAGTTTTTTTTTTTTTTTAAACC-5′); miR-181a-5p (F/R: 5′-CTCGCTTCGGCAGCACA-3′/5′-AACGCTTCACGAATTTGCGT-3′); FOXP2 (F/R: 5′-TTGGATGACCGAAGCACTG-3′/5′-AGGTTTGGGAGATGGTTTGG-3′); U6 (F/R: 5′- CTCGCTTCGGCAGCACATATACT-3′/5′-ACGCTTCACGAATTTGCGTGTC-3′); GAPDH (F/R: 5′-GTAACCCGTTGAACCCCATT-3′/5′-CCATCCAATCGGTAGTAGCG-3′).
Cell culture
Human normal prostatic matrix immortalized cells (WPMY-1) and human prostatic hyperplasia cells (BPH-1) were purchased from SUNNCELL (China) and were cultured using the special medium with catalog numbers SNLM-019 and SNLM-625, respectively, from the same company. The culture environment was 37 °C, containing 5 % CO2.
Cell transfection
The inhibitors of miR-96/181a-5p were from MedChemExpress (USA). The small interfering RNA for FOXP2 was purchased from RiboBio (China). Opti-MEM (Gibco, USA), RNA/plasmid, and Lipofectamine reagent (Invitrogen) were mixed for 15 min and then added to the cells into 96-well plates. After 8 h, the transfection mixture was removed and a fresh complete medium was added. The subsequent experiments were conducted 24–48 h later.
Evaluation of cell viability
The cell counting kit-8 (CCK-8) kit (Solarbio, China) was used for cell viability assessment. The experiment was carried out in a 96-well plate. After cell treatment, the old medium was replaced with 100 μL fresh medium containing 10 % CCK-8 to incubate for 2 h at 37 °C. Finally, the optical density (OD) at 450 nm was measured with an enzyme-labeled instrument to calculate cell viability according to this formula: Cell viability (%) = [OD (experimental group) - OD (blank)]/[OD (control group) – OD (blank)] × 100.
Evaluation of apoptosis
An Annexin V- fluorescein isothiocyanate (FITC) apoptosis detection kit (Beyotime, China) was used for apoptosis detection. The cell pellet was collected in an eppendorf (EP) tube. Annexin V-FITC binding solution (195 μL), Annexin V-FITC (5 μL), and PI (10 μL) were then added successively. After incubation for 30 min away from light, the apoptosis rate was measured using flow cytometry.
Databases
The targets of miR-96-5p and miR-181a-5p were predicted using the TargetScan8.0 database (https://www.targetscan.org/). The GeneCards database (https://www.genecards.org/) helped identify the genes associated with BPH. The GSE119195 dataset, from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/), provided a gene expression profile by analyzing healthy prostate samples from brain-dead young men and BPH tissue samples from surgery.
Western blotting
The whole cell lysate was obtained by cracking the cells in radio immunoprecipitation assay (RIPA) lysate for 30 min and centrifuging to obtain the protein supernatant. The proteins (50 μg/lane) were isolated using 10 % polyacrylamide gel and then transferred to the polyvinylidene fluoride (PVDF) membrane which was then enclosed with 5 % skim milk for 2 h at room temperature (RT). After incubation with primary antibodies of FOXP2 (1:1000, Abcam, UK) and GAPDH (1:2000, Affinity Biosciences, China) overnight at 4 °C, the PVDF membrane was incubated in the secondary antibody (1:10000, Beyotime, China) at RT for 2 h. After washing with TBST buffer, the protein signal was determined using an enhanced chemiluminescence (ECL) chemiluminescence kit (LABIO, China).
Dual luciferase assay
The luciferase vector of wild-type (wt)/mutant (mut) FOXP2 was co-transfected with the inhibitors of miR-96-5p/miR-181a-5p into cells. After 24 h, the cellular luciferase activity was measured using a Double-Luciferase Reporter Assay Kit (GENE CREATE, China).
Statistical analysis
The one-way ANOVA method was used to analyze more than two groups, while the student’s t-test compared two groups, utilizing GraphPad Prism 5 software. A p value of <0.05 was considered statistically significant.
Results
The diagnostic value of GLMI in BPH
Because of the high correlation between abnormal glucose/lipid metabolism and BPH, we constructed a ROC curve to predict the occurrence and development of BPH based on blood GLMI, which includes FBG, HbA1c, TC, and TG. Comparing blood GLMI levels between healthy volunteers and BPH patients, we found that their combination can predict BPH occurrence, with an AUC of 0.869 (95 % confidence interval (CI): 0.820–0.917) (Figure 1A). Additionally, evaluation of IPSS against blood GLMI in BPH patients shows that GLMI is a good classifier of BPH severity, with an AUC of 0.922 (95 % CI: 0.869–0.976) (Figure 1B). These findings suggest that GLMI has potential for diagnosing BPH.

The role of GLMI in diagnosing BPH. The ROC curve of GLMI in predicting the occurrence (A) and development (B) of BPH. AUC, area under the curve.
The diagnostic value of miR-96-5p/miR-181a-5p in BPH
MiR-96-5p and miR-181a-5p were found to be overexpressed in the blood of BPH patients (Figure 2A and B), and their blood level showed predictive accuracy for diagnosing BPH, with AUC of 0.845 (95 % CI: 0.791–0.899) and 0.855 (95 % CI: 0.803–0.907), respectively (Figure 2C and D). Combining these two microRNAs resulted in higher diagnostic accuracy with an AUC of 0.901 (95 % CI: 0.857–0.944) (Figure 2E). Elevated miR-96-5p/miR-181a-5p levels were observed in BPH patients with high IPSS scores (Figure 2F and G). The ROC curve analysis of IPSS and the levels of these microRNAs indicates that high miR-96-5p or miR-181a-5p level could predict the malignant progression of BPH (AUC=0.853 [95 % CI: 0.779–0.927]/0.847 [95 % CI: 0.775–0.920]) (Figure 2H and I), with the combined analysis showing even higher diagnostic accuracy, AUC of 0.927 (95 % CI: 0.882–0.972) (Figure 2J). Multivariate logistic regression analysis revealed that these two miRNAs independently raised the risk of both the onset and advancement of BPH (Tables 1 and 2). Additionally, the blood GLMI combined with miR-96-5p and miR-181a-5p provided more accurate predictions for the occurrence and progression of BPH compared to using only a single factor (AUC=0.927 [95 % CI: 0.892–0.962]/0.962 [95 % CI: 0.932–0.991]) (Figure 2K–L). Moreover, the correlation analysis shows that their blood levels were positively correlated with IPSS (Supplementary Figure 1), further demonstrating their clinical value in predicting the occurrence and development of BPH.

The role of miR-96-5p/miR-181a-5p in diagnosing BPH. (A–B) Levels of miR-96-5p/miR-181a-5p in the blood samples of healthy volunteers and BPH patients. ROC curves for miR-96-5p and miR-181a-5p in predicting BPH occurrence, either (C–D) independently or (E) jointly. (F–G) Levels of miR-96-5p and miR-181a-5p in the blood of BPH patients across different IPSS groups. ROC curves for miR-96-5p and miR-181a-5p predicting BPH development, either (H–I) independently or (J) jointly. ROC curves for miR-96-5p and miR-181a-5p combined with GLMI in predicting BPH incidence (K) and progression (L). ***p<0.001.
Multivariate logistic regression analysis of factors in predicting BPH.
| Factor | OR | 95 % CI | p-Value | |
|---|---|---|---|---|
| Lower | Upper | |||
| Age | 1.017 | 0.932 | 1.110 | 0.699 |
| Smoking | 1.084 | 0.440 | 2.671 | 0.861 |
| Drinking | 1.015 | 0.408 | 2.528 | 0.974 |
| BMI | 1.118 | 0.964 | 1.296 | 0.140 |
| FBG | 1.490 | 0.843 | 2.632 | 0.170 |
| HbA1c | 1.354 | 0.882 | 2.077 | 0.166 |
| TC | 1.461 | 0.926 | 2.307 | 0.103 |
| TG | 1.294 | 0.929 | 1.804 | 0.128 |
| miR-96-5p | 2.306 | 1.446 | 3.677 | <0.001 |
| miR-181a-5p | 2.513 | 1.542 | 4.097 | <0.001 |
-
BPH, benign prostatic hyperplasia; OR, odds ratio; CI, confidence interval; BMI, body mass index; FBG, fasting blood glucose; HbA1c, hemoglobin A1c; TC, total cholesterol; TG, triglyceride.
Multivariate logistic regression analysis of factors in predicting severity of BPH.
| Factor | OR | 95 % CI | p-Value | |
|---|---|---|---|---|
| Lower | Upper | |||
| Age | 1.156 | 0.972 | 1.374 | 1.102 |
| Smoking | 2.206 | 0.448 | 10.852 | 0.331 |
| Drinking | 1.017 | 0.218 | 4.736 | 0.983 |
| BMI | 1.294 | 0.613 | 2.733 | 0.499 |
| FBG | 2.397 | 0.688 | 8.354 | 0.170 |
| HbA1c | 1.575 | 0.937 | 2.649 | 0.087 |
| TC | 1.547 | 0.409 | 5.849 | 0.520 |
| TG | 1.190 | 0.490 | 2.888 | 0.701 |
| miR-96-5p | 1.700 | 1.088 | 2.656 | 0.020 |
| miR-181a-5p | 1.826 | 1.197 | 2.786 | 0.005 |
-
BPH, benign prostatic hyperplasia; OR, odds ratio; CI, confidence interval; BMI, body mass index; FBG, fasting blood glucose; HbA1c, hemoglobin A1c; TC, total cholesterol; TG, triglyceride.
Effects of miR-96-5p/miR-181a-5p on WPMY-1 and BPH-1 functions
MiR-96-5p and miR-181a-5p were highly expressed in BPH-1 cells (Figure 3A and B). Inhibition of miR-96-5p or miR-181a-5p significantly reduced their levels (Figure 3C and D). This reduction decreased the viability of WPMY-1 and BPH-1 cells (Figure 3E and F) and promoted cell apoptosis (Figure 3G and H), indicating that miR-96-5p/miR-181a-5p may promote BPH pathogenesis.

Roles of miR-96-5p and miR-181a-5p in WPMY-1 and BPH-1 cells. (A–B) The expression of miR-96-5p and miR-181a-5p. (C–D) Working efficiency of miR-96-5p and miR-181a-5p inhibitors. Effects of miR-96-5p/miR-181a-5p on the cell viability (E–F) and apoptosis (G–H). NC, negative control. *p<0.05; **p<0.01; ***p<0.001.
Regulation of FOXP2 by miR-96-5p and miR-181a-5p
To further explore the mechanisms by which miR-96-5p and miR-181a-5p affect the functions of WPMY-1 and BPH-1 cells, we used the TargetScan8.0 database to predict their target genes. Through the creation of a Venn diagram that included the targets of miR-96-5p, miR-181a-5p, the genes associated with BPH, and the genes that are differentially expressed in BPH and normal prostate tissues (GSE119195), we identified FOXP2 as a key gene (Figure 4A). The TargetScan8.0 predictions indicated that miR-181a-5p and miR-96-5p both have binding sites in FOXP2 3′UTR (Figure 4B). Transfection with inhibitor of miR-96-5p/miR-181a-5p enhanced the cell luciferase intensity with wild-type (wt) FOXP2 vector transfection, but did not affect the mutant (mut) transfection group (Figures 4C and D). Inhibition of either miR-96-5p or miR-181a-5p significantly increased FOXP2 expression (Figure 4E–H), suggesting that these miRNAs negatively regulate FOXP2 expression by targeting its mRNA.

Regulation of FOXP2 by miR-96-5p and miR-181a-5p. (A) Venn diagram showing the overlap among targets of miR-96-5p/miR-181a-5p, genes associated with BPH from the GeneCards database, and genes differentially expressed in BPH and normal prostate tissues. (B) The binding site of miR-96-5p or miR-181a-5p with FOXP2 mRNA. Effects of the miR-96-5p or miR-181a-5p inhibitor on cellular luciferase activity transfected with luciferase vector of wt-/mut-FOXP2 (C–D), and on the expression of FOXP2 (E–H). NC, negative control. ***p<0.001; ns, no significant difference.
FOXP2 inhibited the functions of miR-181a-5p and miR-96-5p
FOXP2 was found to be under-expressed in BPH-1 cells and BPH patients (Figure 5A and B). Transfection of small interfering RNA for FOXP2 (si-FOXP2) significantly reduced FOXP2 expression (Figure 5C and D). Moreover, the miR-181a-5p/miR-96-5p inhibitor decreased the viability and induced apoptosis in WPMY-1 and BPH-1 cells. This effect was reversed by co-transfection with si-FOXP2 (Figure 5E–H). These results indicate that miR-181a-5p and miR-96-5p played potential functions in BPH by regulating FOXP2.

FOXP2 inhibited the functions of miR-96-5p and miR-181a-5p. (A–B) The expression of FOXP2 in WPMY-1/BPH-1 cells and BPH patients. (C–D) The working efficiency of the small interfering RNA of FOXP2. Effects of the co-transfection of miR-96-5p or miR-181a-5p inhibitor with si-FOXP2 on the viability (E–F) and apoptosis (G–H) of WPMY-1 and BPH-1 cells. NC, negative control. *p<0.05; **p<0.01; ***p<0.001.
Discussion
It is still not fully understood what causes BPH. Currently, the reported pathogenic factors mainly include androgens and their receptors, the imbalance between proliferative and apoptotic cells, the influence of growth and inflammatory factors, the interaction between prostate stromal and glandular epithelial cells, and genetic factors [8]. However, the molecular mechanisms underlying BPH pathogenesis remain poorly understood.
In this research, we found that the levels of blood GLMI, miR-96-5p, and miR-181a-5p were significantly upregulated in patients with BPH, and they further increased with the deterioration of the disease. Correlation analysis revealed their close clinical correlation with severe progression of BPH: The levels of blood GLMI, miR-96-5p/miR-181a-5p increased with the increase of IPSS value, indicating that they are possible biomarkers of BPH. The ROC curve model showed that GLMI, miR-96-5p, and miR-181a-5p all demonstrated high predictive value for the occurrence and severe progression of BPH, and their combination showed higher predictive reliability. In the mechanism exploration, miR-181a-5p and miR-96-5p negatively regulate the expression of FOXP2 in BPH-1 cells, which is conducive to the rapid proliferation of cells.
Abnormal blood glucose metabolism is a significant factor in worsening BPH. Elevated FBG levels can speed up BPH progression. Poor and persistent blood sugar control can lead to a notable increase in PV [9]. BPH patients with abnormal HbA1c levels tend to have higher PV compared to those with normal HbA1c [10]. On a related note, abnormal lipid metabolism is closely linked to a higher risk of BPH. Studies indicate that individuals with hyperlipidemia are more prone to developing BPH than older men without this condition [11]. Following statin therapy, improvements in lipid markers among BPH patients with MS resulted in reduced PV and significant improvement in LUTS [12]. Therefore, MS, particularly abnormalities in glucose and lipid metabolism, significantly contribute to the onset and progression of BPH. Analysis using the ROC curve indicated that blood GLMI such as FBG, HbA1c, TC, and TG can effectively classify BPH occurrence and progression. Moreover, combining GLMI with miR-96-5p and miR-181a-5p showed higher diagnostic accuracy in BPH.
The role of miRNAs in BPH has been widely recorded. The expression of miR-188-3p has been reported to have a loss in BPH tissues. Its exogenous upregulation can increase the susceptibility of prostatic luminal cells to Eastin and increase ferroptosis in cells. miR-188-3p is regulated by lncRNA TUG1 in the competitive endogenous RNA network: TUG1 can promote GPX4 expression by competitively binding to miR-188-3p, thereby hindering the process of cellular ferroptosis [13]. The overexpression of miR-1199-5p in BPH can mediate the down-regulation of steroid 5 alpha-reductase 2 (SRD5A2), which leads to a decrease in the sensitivity of prostate cells to finasteride (a clinical drug for the treatment of benign prostatic hyperplasia) [14]. The level of urine miR-21-5p in patients with BPH shows a significant difference before and after tadalafil treatment, and it is regarded as a predictive indicator of tadalafil response [15]. MiR-96-5p was reported to be over-expressed in prostate cancer (PCa) tissues, promoting PCa cell to transfer by inhibiting NDRG1 [16], [17], [18]. In addition, miR-181a-5p is negatively regulated by MIIP in PCa, thereby promoting KLF17 expression to hinder the epithelial-mesenchymal transition process of cancer cells [19]. However, whether miR-96-5p and miR-181a-5p are involved in the occurrence of BPH remains unknown. Here, our findings reveal the potential functions of these two miRNAs by regulating FOXP2 in promoting BPH progression and elaborate on their clinical value in combination with GLMI as biomarkers for BPH.
FOXP2, a target of miR-96-5p/miR-181a-5p, is less prevalent in BPH-1 cells compared to WPMY-1 cells. Overexpression of FOXP2 inhibits proliferation and promotes apoptosis in liver and thyroid cancer cells by regulating the KDM5A/FBP1 axis and RPS6KA6 expression [20], 21]. Additionally, crocin can prevent breast cancer (BC) cells from proliferating by lowering miR-122-5p level, which in turn increases the expression of SPRY2 and FOXP2 [22]. These studies suggest that FOXP2 may function as an inhibitor of cell proliferation. We also demonstrated that silencing FOXP2 reversed the decreased viability and increased apoptosis rate of WPMY-1 and BPH-1 cells caused by inhibiting miR-96-5p or miR-181a-5p, indicating that FOXP2 may hinder BPH progression.
In conclusion, our fundamental research provides potential biomarkers for the diagnosis of BPH and the monitoring of disease progression: we have revealed the value of miR-96-5p, miR-181a-5p, and GLMI in diagnosing BPH and discussed the potential molecular mechanisms underlying BPH pathogenesis. However, the verification of this molecular mechanism in vivo experiments will further enhance the reliability of the conclusion. In the future, we will focus on this point. Moreover, we will further collect more clinical samples to enhance the credibility of GLMI combined with miR-96-5p/miR-181a-5p as a diagnostic model for BPH. Nevertheless, this study still provides new ideas for understanding the pathogenesis of BPH.
-
Research ethics: This study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the Ethics Committee of Zhangjiakou First Hospital, Approval Number: 2023040, Date: 2023-06-28.
-
Informed consent: Informed consent was obtained from all participants included in the study.
-
Author contributions: Y.P. Li proposed and designed the framework of this study. H.Y. Zhang completed the collation and pre-processing of the raw data required for subsequent analysis. Y.P. Li and Y.J. Yue performed a detailed analysis of the data and drafted this manuscript. All authors were involved in revising it and approved the version to be published. All authors have participated sufficiently in this work and take responsibility for appropriate portions of the content. All authors read and approved the final manuscript.
-
Use of Large Language Models, AI and Machine Learning Tools: Not applicable.
-
Conflict of interest: The author states no conflict of interest.
-
Research funding: The authors report no funding.
-
Data availability: The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.
References
1. Langan, RC. Benign prostatic hyperplasia. Prim Care 2019;46:223–32. https://doi.org/10.1016/j.pop.2019.02.003.Search in Google Scholar PubMed
2. Zi, H, Liu, MY, Luo, LS, Huang, Q, Luo, PC, Luan, HH, et al.. Global burden of benign prostatic hyperplasia, urinary tract infections, urolithiasis, bladder cancer, kidney cancer, and prostate cancer from 1990 to 2021. Mil Med Res 2024;11:64. https://doi.org/10.1186/s40779-024-00569-w.Search in Google Scholar PubMed PubMed Central
3. Miernik, A, Gratzke, C. Current treatment for benign prostatic hyperplasia. Dtsch Arztebl Int 2020;117:843–54. https://doi.org/10.3238/arztebl.2020.0843.Search in Google Scholar PubMed PubMed Central
4. Fahed, G, Aoun, L, Bou Zerdan, M, Allam, S, Bou Zerdan, M, Bouferraa, Y, et al.. Metabolic syndrome: updates on pathophysiology and management in 2021. Int J Mol Sci 2022;23. https://doi.org/10.3390/ijms23020786.Search in Google Scholar PubMed PubMed Central
5. Fu, Y, Zhou, Z, Yang, B, Zhang, K, He, L, Zhang, X. The relationship between the clinical progression of benign prostatic hyperplasia and metabolic syndrome: a prospective study. Urol Int 2016;97:330–5. https://doi.org/10.1159/000448484.Search in Google Scholar PubMed
6. De Nunzio, C, Cindolo, L, Gacci, M, Pellegrini, F, Carini, M, Lombardo, R, et al.. Metabolic syndrome and lower urinary tract symptoms in patients with benign prostatic enlargement: a possible link to storage symptoms. Urology 2014;84:1181–7. https://doi.org/10.1016/j.urology.2014.07.018.Search in Google Scholar PubMed
7. Zhang, N, Li, Z, Bai, F, Ji, N, Zheng, Y, Li, Y, et al.. MicroRNA expression profiles in benign prostatic hyperplasia. Mol Med Rep 2018;17:3853–8. https://doi.org/10.3892/mmr.2017.8318.Search in Google Scholar PubMed
8. Devlin, CM, Simms, MS, Maitland, NJ. Benign prostatic hyperplasia – what do we know? BJU Int 2021;127:389–99. https://doi.org/10.1111/bju.15229.Search in Google Scholar PubMed
9. Meng, J, Liu, Y, Guan, SY, Ma, H, Zhang, X, Fan, S, et al.. Age, height, BMI and FBG predict prostate volume in ageing benign prostatic hyperplasia: evidence from 5285 patients. Int J Clin Pract 2019:e13438. https://doi.org/10.1111/ijcp.13438.Search in Google Scholar PubMed
10. Qu, X, Huang, Z, Meng, X, Zhang, X, Dong, L, Zhao, X. Prostate volume correlates with diabetes in elderly benign prostatic hyperplasia patients. Int Urol Nephrol 2014;46:499–504. https://doi.org/10.1007/s11255-013-0555-3.Search in Google Scholar PubMed
11. Shih, HJ, Huang, CJ, Lin, JA, Kao, MC, Fan, YC, Tsai, PS. Hyperlipidemia is associated with an increased risk of clinical benign prostatic hyperplasia. Prostate 2018;78:113–20. https://doi.org/10.1002/pros.23451.Search in Google Scholar PubMed
12. Zhang, X, Zeng, X, Dong, L, Zhao, X, Qu, X. The effects of statins on benign prostatic hyperplasia in elderly patients with metabolic syndrome. World J Urol 2015;33:2071–7. https://doi.org/10.1007/s00345-015-1550-3.Search in Google Scholar PubMed
13. Zhan, M, Xu, H, Yu, G, Chen, Q, Yang, R, Chen, Y, et al.. Androgen receptor deficiency-induced TUG1 in suppressing ferroptosis to promote benign prostatic hyperplasia through the miR-188-3p/GPX4 signal pathway. Redox Biol 2024;75:103298. https://doi.org/10.1016/j.redox.2024.103298.Search in Google Scholar PubMed PubMed Central
14. Liu, Z, Lin, Z, Cao, F, Jiang, M, Jin, S, Cui, Y, et al.. Upregulation of mir-1199-5p is associated with reduced type 2 5-alpha reductase expression in benign prostatic hyperplasia. BMC Urol 2022;22:172. https://doi.org/10.1186/s12894-022-01121-5.Search in Google Scholar PubMed PubMed Central
15. Tanaka, T, Minami, A, Tashiro, K, Yoshida, N, Tohda, A, Yamakoshi, Y, et al.. Urine miR-21-5p as a potential biomarker for predicting effectiveness of tadalafil in benign prostatic hyperplasia. Future Sci OA 2018;4:FSO304. https://doi.org/10.4155/fsoa-2018-0012.Search in Google Scholar PubMed PubMed Central
16. Lian, Z, Chang, T, Ma, S, Li, J, Zhang, H, Wang, X, et al.. MiR-96-5p induced NDRG1 deficiency promotes prostate cancer migration and invasion through regulating the NF-kappaB signaling pathway. Cancer Biomark 2022;35:83–98. https://doi.org/10.3233/cbm-210072.Search in Google Scholar
17. Pelka, K, Klicka, K, Grzywa, TM, Gondek, A, Marczewska, JM, Garbicz, F, et al.. miR-96-5p, miR-134-5p, miR-181b-5p and miR-200b-3p heterogenous expression in sites of prostate cancer versus benign prostate hyperplasia-archival samples study. Histochem Cell Biol 2021;155:423–33. https://doi.org/10.1007/s00418-020-01941-2.Search in Google Scholar PubMed PubMed Central
18. Schitcu, VH, Raduly, L, Nutu, A, Zanoaga, O, Ciocan, C, Munteanu, VC, et al.. MicroRNA dysregulation in prostate cancer. Pharmgenomics Pers Med 2022;15:177–93. https://doi.org/10.2147/pgpm.s348565.Search in Google Scholar
19. Hu, W, Yan, F, Ru, Y, Xia, M, Yan, G, Zhang, M, et al.. MIIP inhibits EMT and cell invasion in prostate cancer through miR-181a/b-5p-KLF17 axis. Am J Cancer Res 2020;10:630–47.Search in Google Scholar
20. Yan, L, Sun, H, Chen, Y, Yu, X, Zhang, J, Li, P. FOXP2 suppresses the proliferation, invasion, and aerobic glycolysis of hepatocellular carcinoma cells by regulating the KDM5A/FBP1 axis. Environ Toxicol 2024;39:341–56. https://doi.org/10.1002/tox.23971.Search in Google Scholar PubMed
21. Yang, F, Xiao, Z, Zhang, S. FOXP2 regulates thyroid cancer cell proliferation and apoptosis via transcriptional activation of RPS6KA6. Exp Ther Med 2022;23:434. https://doi.org/10.3892/etm.2022.11361.Search in Google Scholar PubMed PubMed Central
22. Jia, Y, Yang, H, Yu, J, Li, Z, Jia, G, Ding, B, et al.. Crocin suppresses breast cancer cell proliferation by down-regulating tumor promoter miR-122-5p and up-regulating tumor suppressors FOXP2 and SPRY2. Environ Toxicol 2023;38:1597–608. https://doi.org/10.1002/tox.23789.Search in Google Scholar PubMed
Supplementary Material
This article contains supplementary material (https://doi.org/10.1515/tjb-2025-0043).
© 2025 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.