Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.076
Filter
1.
BMC Cancer ; 24(1): 1231, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39369238

ABSTRACT

BACKGROUND: The characteristics of a tumor are largely determined by its interaction with the surrounding micro-environment (TME). TME consists of both cellular and non-cellular components. Cancer-associated fibroblasts (CAFs) are a major component of the TME. They are a source of many secreted factors that influence the survival and progression of tumors as well as their response to drugs. Identification of markers either overexpressed in CAFs or unique to CAFs would pave the way for novel therapeutic strategies that in combination with conventional chemotherapy are likely to have better patient outcome. METHODS: Fibroblasts have been derived from Benign Prostatic Hyperplasia (BPH) and prostate cancer. RNA from these has been used to perform a transcriptome analysis in order to get a comparative profile of normal and cancer-associated fibroblasts. RESULTS: The study has identified 818 differentially expressed mRNAs and 17 lincRNAs between normal and cancer-associated fibroblasts. Also, 15 potential lincRNA-miRNA-mRNA combinations have been identified which may be potential biomarkers. CONCLUSIONS: This study identified differentially expressed markers between normal and cancer-associated fibroblasts that would help in targeted therapy against CAFs/derived factors, in combination with conventional therapy. However, this would in future need more experimental validation.


Subject(s)
Cancer-Associated Fibroblasts , Gene Expression Profiling , Prostatic Neoplasms , Transcriptome , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Fibroblasts/metabolism , Tumor Microenvironment/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/metabolism
2.
Sci Rep ; 14(1): 23120, 2024 10 04.
Article in English | MEDLINE | ID: mdl-39367121

ABSTRACT

Benign prostatic hyperplasia (BPH) as a common geriatric disease in urology, the incidence and prevalence are rapidly increasing with the aging society, prompting an urgent need for effective prevention and treatment of BPH. However, limited therapeutic efficacy and higher risk of complications result in the treatment of BPH remaining challenging. The unclear pathogenic mechanism also hampers further exploration of therapeutic approaches for BPH. In this study, we used multi-omics methods to integrate genomics, transcriptomics, immunomics, and metabolomics data and identify biomolecules associated with BPH. We performed transcriptomic imputation, summary data-based Mendelian randomization (SMR), joint/conditional analysis, colocalization analysis, and FOCUS to explore high-confidence genes associated with BPH in blood and prostate tissue. Subsequently, three-step SMR was used to identify the DNA methylation sites regulating high-confidence genes to improve the pathogenic pathways of BPH. We also used cis-instruments of druggable genes to conduct SMR analysis to find potential drug targets for BPH. Finally, we used MR analysis to explore the immune pathways and metabolomics related to BPH. Multiple analytical methods identified BTN3A2 (Blood: TWAS Z score = 5.02912, TWAS P = 4.93 × 10-7; Prostate: TWAS Z score = 4.89, TWAS P = 1.01 × 10-6) and C4A (Blood: TWAS Z score = 4.90754, TWAS P = 9.22 × 10-7; Prostate: TWAS Z score = 5.084, TWAS P = 3.70 × 10-7) as high-confidence genes for BPH and identified the cg14345882-BTN3A2-BPH pathogenic pathway. We also used druggable gene data to identify 30 promising therapeutic target genes, including BTN3A2 and C4A. For MR analysis of immune pathways, we identified immune cell surface molecules as well as the inflammatory factor IL-17 (OR = 1.25, 95% CI = 1.09-1.43, PFDR = 0.12, Maximum likelihood) as risk factors for BPH. In addition, we found that disulfide levels of cysteinylglycine (OR = 1.11, 95% CI = 1.05-1.18, P = 5.18 × 10-4, Weighted median), oxidation levels of cysteinylglycine (OR = 1.09, 95% CI = 1.04-1.14, P = 3.87 × 10-4, Weighted median), and sebacate levels (OR = 1.05, 95% CI = 1.02-1.08, P = 3.0 × 10-4, Maximum likelihood) increase the risk of BPH. This multi-omics study explored biomolecules associated with BPH, improved the pathogenic pathways of BPH, and identified promising therapeutic targets. Our results provide evidence for future studies aimed at developing appropriate therapeutic interventions.


Subject(s)
Mendelian Randomization Analysis , Prostatic Hyperplasia , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/drug therapy , Humans , Male , Metabolomics/methods , DNA Methylation , Transcriptome , Genomics/methods , Polymorphism, Single Nucleotide , Genetic Predisposition to Disease , Multiomics
3.
BMC Cancer ; 24(1): 1242, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379919

ABSTRACT

BACKGROUND: Prostate cancer (PCa) usually manifests atypical symptoms in the early stage, and once symptoms appear, most PCa patients have developed to the advanced stage, failing to undergo radical surgery. In this study, PCa occurrence-related biomarkers were explored based on single-cell RNA sequencing (scRNA-seq) data. METHODS: scRNA-seq data of prostate normal (Normal), benign prostatic hyperplasia (BPH), and PCa (Tumor) samples were acquired from the Gene Expression Omnibus (GEO). Cellular subsets associated with PCa occurrence were obtained using cell annotation. Additionally, the mRNA expression of nuclear enriched abundant transcript 1 (NEAT1) was detected by quantitative real-time PCR (qRT-PCR). The effects of NEAT1 on cell proliferation and apoptosis were analyzed by 5-ethynyl-2-deoxyuridine (EdU) and flow cytometry. Subsequently, cell-derived xenograft (CDX) models were constructed and divided into the LV-NC and LV-shNEAT1 groups. After the tumor tissues of CDX model mice in each group were extracted, the cell growth and Ki67 expression were observed separately using hematoxylin-eosin (H&E) staining and immunohistochemistry (IHC). RESULTS: Ten cellular subsets were obtained via cell annotation, and significantly differential changes were observed between Basal intermediate and Luminal during the course of BPH to PCa. NEAT1-Luminal was highly recruited in the Tumor group with low stemness and high malignancy scores. Matrix metallopeptidase 7 (MMP7)- keratin 17 (KRT17)-Basal intermediate had high ratios in the Tumor group with low stemness and high malignancy scores. The results of pseudotime analysis revealed that NEAT1-Luminal in the Tumor group were consistently distributed with tumor stage cells. In vitro assays showed that NEAT1 expression was elevated in PCa cells, and NEAT1 knockdown could inhibit cell proliferation and induce apoptosis. CDX assays indicated that silencing NEAT1 could reduce the growth rate of PCa tumor volume in CDX model mice. H&E staining results showed that nuclei of tumor cells were reduced and exhibited lighter color in the LV-shNEAT1 group compared with the LV-NC group. IHC results showed that Ki67 positivity was significantly lower in the LV-shNEAT1 group than in the LV-NC group. CONCLUSION: NEAT1 expression is increased in PCa, and NEAT1 can be a potential biomarker and therapeutic target for PCa.


Subject(s)
Apoptosis , Biomarkers, Tumor , Cell Proliferation , Prostatic Neoplasms , RNA, Long Noncoding , Single-Cell Analysis , Male , Humans , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Animals , Mice , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/metabolism , Xenograft Model Antitumor Assays
4.
Sci Rep ; 14(1): 23667, 2024 10 10.
Article in English | MEDLINE | ID: mdl-39390078

ABSTRACT

Previous research has suggested that circulating inflammatory proteins are associated with benign prostatic disease (BPD). This Mendelian randomization (MR) study was conducted to further investigate the causal relationship between 91 inflammatory proteins and BPD. Genome-wide association study (GWAS) summarized data of benign prostatic hyperplasia (BPH) and prostatitis were obtained from the FinnGen Biobank. The latest study offered the GWAS data on 91 proteins related to inflammation. We performed a bidirectional MR to investigate the causal association between inflammatory proteins and BPD. The outcomes of the IVW method indicated that decreased levels of circulating interleukin-17 C (IL-17 C) (OR = 0.92, 95%CI = 0.85-0.99, p-value = 0.0344) were suggestively associated with a higher risk of BPH and elevated levels of interleukin-10 receptor subunit alpha (IL-10RA) (OR = 1.24, 95%CI = 1.05-1.47, p-value = 0.0132) and urokinase-type plasminogen activator (uPA) (OR = 1.13, 95%CI = 1.00-1.28, p-value = 0.0421) were suggestively related to a higher risk of prostatitis. Furthermore, reverse MR revealed that BPH may promote the expression of circulating factors, including natural killer cell receptor 2B4 (CD244) (OR = 1.07, 95%CI = 1.01-1.13, p-value = 0.0192), T-cell surface glycoprotein CD6 isoform (CD6) (OR = 1.07, 95%CI = 1.01-1.13, p-value = 0.0192), and leukemia inhibitory factor receptor (LIF-R) (OR = 1.07, 95%CI = 1.01-1.15, p-value = 0.0163). Moreover, the results of sensitivity analyses indicate that heterogeneity and horizontal pleiotropy are unlikely to distort the findings. The results of this study indicate a potential association between circulating inflammatory proteins and BPD, which may become new diagnostic indicators or drug targets for clinical application in the prevention and treatment of BPD. However, further investigation is required.


Subject(s)
Genome-Wide Association Study , Mendelian Randomization Analysis , Prostatic Hyperplasia , Humans , Male , Prostatic Hyperplasia/blood , Prostatic Hyperplasia/genetics , Polymorphism, Single Nucleotide , Genetic Predisposition to Disease , Prostatitis/blood , Prostatitis/genetics , Inflammation/blood , Inflammation/genetics , Biomarkers/blood
5.
Int J Mol Sci ; 25(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39273277

ABSTRACT

Our study highlights the apoptosis, cell cycle, DNA ploidy, and autophagy molecular mechanisms network to identify prostate pathogenesis and its prognostic role. Caspase 3/7 expressions, cell cycle, adhesion glycoproteins, autophagy, nuclear shrinkage, and oxidative stress by flow-cytometry analysis are used to study the BPH microenvironment's heterogeneity. A high late apoptosis expression by caspases 3/7 activity represents an unfavorable prognostic biomarker, a dependent predictor factor for cell adhesion, growth inhibition by arrest in the G2/M phase, and oxidative stress processes network. The heterogeneous aggressive phenotype prostate adenoma primary cell cultures present a high S-phase category (>12%), with an increased risk of death or recurrence due to aneuploid status presence, representing an unfavorable prognostic biomarker, a dependent predictor factor for caspase 3/7 activity (late apoptosis and necrosis), and cell growth inhibition (G2/M arrest)-linked mechanisms. Increased integrin levels in heterogenous BPH cultures suggest epithelial-mesenchymal transition (EMT) that maintains an aggressive phenotype by escaping cell apoptosis, leading to the cell proliferation necessary in prostate cancer (PCa) development. As predictor biomarkers, the biological mechanisms network involved in apoptosis, the cell cycle, and autophagy help to establish patient prognostic survival or target cancer therapy development.


Subject(s)
Apoptosis , Autophagy , Cell Cycle , Prostatic Hyperplasia , Humans , Male , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/genetics , Prognosis , Primary Cell Culture , Epithelial-Mesenchymal Transition/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Phenotype , Aged , Caspase 3/metabolism , Cell Proliferation , Caspase 7/metabolism , Middle Aged , Oxidative Stress
6.
Int J Mol Sci ; 25(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39337566

ABSTRACT

RNAs, especially non-coding RNAs (ncRNAs), are crucial players in regulating cellular mechanisms due to their ability to interact with and regulate other molecules. Altered expression patterns of ncRNAs have been observed in prostate cancer (PCa), contributing to the disease's initiation, progression, and treatment response. This study aimed to evaluate the ability of a specific set of RNAs, including long ncRNAs (lncRNAs), microRNAs (miRNAs), and mRNAs, to discriminate between PCa and the non-neoplastic condition benign prostatic hyperplasia (BPH). After selecting by literature mining the most relevant RNAs differentially expressed in biofluids from PCa patients, we evaluated their discriminatory power in samples of unfiltered urine from 50 PCa and 50 BPH patients using both real-time PCR and droplet digital PCR (ddPCR). Additionally, we also optimized a protocol for urine sample manipulation and RNA extraction. This two-way validation study allowed us to establish that miRNAs (i.e., miR-27b-3p, miR-574-3p, miR-30a-5p, and miR-125b-5p) are more efficient biomarkers for PCa compared to long RNAs (mRNAs and lncRNAs) (e.g., PCA3, PCAT18, and KLK3), as their dysregulation was consistently reported in the whole urine of patients with PCa compared to those with BPH in a statistically significant manner regardless of the quantification methodology performed. Moreover, a significant increase in diagnostic performance was observed when molecular signatures composed of different miRNAs were considered. Hence, the abovementioned circulating ncRNAs represent excellent potential non-invasive biomarkers in urine capable of effectively distinguishing individuals with PCa from those with BPH, potentially reducing cancer overdiagnosis.


Subject(s)
Biomarkers, Tumor , MicroRNAs , Prostatic Hyperplasia , Prostatic Neoplasms , Humans , Male , Prostatic Hyperplasia/urine , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/diagnosis , Prostatic Neoplasms/urine , Prostatic Neoplasms/genetics , Prostatic Neoplasms/diagnosis , Aged , MicroRNAs/urine , MicroRNAs/genetics , Biomarkers, Tumor/urine , Biomarkers, Tumor/genetics , Middle Aged , Diagnosis, Differential , RNA, Long Noncoding/urine , RNA, Long Noncoding/genetics , RNA, Messenger/urine , RNA, Messenger/genetics , Gene Expression Regulation, Neoplastic , Aged, 80 and over
7.
Biol Direct ; 19(1): 61, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095835

ABSTRACT

Myofibroblast buildup and prostatic fibrosis play a crucial role in the development of benign prostatic hyperplasia (BPH). Treatments specifically targeting myofibroblasts could be a promising approach for treating BPH. Tadalafil, a phosphodiesterase type 5 (PDE5) inhibitor, holds the potential to intervene in this biological process. This study employs prostatic stromal fibroblasts to induce myofibroblast differentiation through TGFß1 stimulation. As a result, tadalafil significantly inhibited prostatic stromal fibroblast proliferation and fibrosis process, compared to the control group. Furthermore, our transcriptome sequencing results revealed that tadalafil inhibited FGF9 secretion and simultaneously improved miR-3126-3p expression via TGFß1 suppression. Overall, TGFß1 can trigger pro-fibrotic signaling through miR-3126-3p in the prostatic stroma, and the use of tadalafil can inhibit this process.


Subject(s)
Fibroblast Growth Factor 9 , Fibrosis , MicroRNAs , Phosphodiesterase 5 Inhibitors , Prostatic Hyperplasia , Tadalafil , Male , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Tadalafil/pharmacology , Phosphodiesterase 5 Inhibitors/pharmacology , Humans , Fibroblast Growth Factor 9/metabolism , Fibroblast Growth Factor 9/genetics , Prostate/drug effects , Prostate/metabolism , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Cell Proliferation/drug effects
8.
Redox Biol ; 75: 103298, 2024 09.
Article in English | MEDLINE | ID: mdl-39121689

ABSTRACT

Benign prostatic hyperplasia (BPH), characterized by the non-malignant enlargement of the prostate, exhibits a pronounced association with inflammation resulting from androgen receptor (AR) deficiency. Ferroptosis, a cell death mechanism triggered by iron-dependent lipid peroxidation and closely linked to inflammation, has yet to be fully understood in the context of BPH. Using RNA sequencing, we observed a significant elevation of taurine-upregulated gene 1 (TUG1) long noncoding RNA (lncRNA) in BPH tissues compared to normal prostate tissue. High levels of TUG1 exhibited a discernible correlation with both prostate volume and the extent of inflammatory infiltration in BPH patients. The suppression of TUG1 not only led to a reduction in prostate size but also ameliorated AR-deficiency-induced prostatic hyperplasia. Mechanistically, a decrease in AR in prostate luminal cells prompted macrophage aggregation and the release of IL-1ß, subsequently fostering the transcription of TUG1 via MYC. Induced TUG1, through competitive binding with miR-188-3p, facilitated the expression of GPX4, thereby diminishing intracellular ROS levels and impeding ferroptosis in prostate luminal cells. Notably, the ferroptosis inducer JKE-1674 alleviated inflammation-induced prostatic hyperplasia in vivo. Together, these findings suggest that AR deficiency crucially inhibits ferroptosis, promoting BPH via the TUG1/miR-188-3p/GPX4 signaling axis, and making ferroptosis induction a promising therapeutic strategy for BPH patients with AR deficiency.


Subject(s)
Ferroptosis , MicroRNAs , Phospholipid Hydroperoxide Glutathione Peroxidase , Prostatic Hyperplasia , RNA, Long Noncoding , Receptors, Androgen , Signal Transduction , Animals , Humans , Male , Mice , Disease Susceptibility , Ferroptosis/genetics , MicroRNAs/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , RNA, Long Noncoding/genetics
9.
World J Urol ; 42(1): 439, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39046536

ABSTRACT

PURPOSE: To confirm if the CYP17A1 gene regulates the ratio of T/E leading to MetS-BPH. METHODS: 824 men, aged 47-88 years, were recruited into this study through consecutive routine physical examination programs and long-term outpatient screening. Several parameters, including SNPs of CYP17A1 gene, total testosterone, estradiol, and the ratio of total testosterone to estradiol (T/E) were obtained for each participant. Based on the diagnosis of BPH, MetS, and MetS-BPH, the participants were divided into BPH and non-BPH groups, MetS and non-MetS groups, and MetS-BPH and non-MetS-BPH groups. Values of the obtained parameters were evaluated using one-way analysis of variance, Student's t-test, Chi-squared test, and logistic regression analysis. RESULTS: SNPs of the CYP17A1 gene, including the rs743572 genotypes (GG, GA, and AA), rs3781287 genotypes (GG, GT, TT), and rs4919686 genotypes (CC, CA, and AA), were present in every group. Only the GG genotype of rs743572 was independently associated with BPH (OR = 5.868, 95% CI: 3.363-7.974, P < 0.001), MetS (OR = 7.228, 95% CI: 3.925-11.331, P < 0.001), and MetS-BPH (OR = 3.417, 95% CI: 1.783-5.266, P < 0.001) after adjusting for age. In the population of genotype GG of rs743572, the decrease in T/E ratio was an independent risk factor for BPH (OR = 839.756, 95% CI: 36.978-1334.263, P = 0.001), MetS (OR = 376.988, 95% CI: 12.980-488.976, P < 0.003), and MetS-BPH (OR = 388.236, 95% CI: 24.869-495.363, P = 0.003). CONCLUSION: The GG genotype of rs743572 in CYP17A1 gene regulating the decrease of T/E ratio can be an independent risk factor for MetS-BPH populations. TRIAL REGISTRATION NUMBER: ChiCTR2200057632 "retrospectively registered". DATE OF REGISTRATION: March 15, 2022 "retrospectively registered".


Subject(s)
Genotype , Prostatic Hyperplasia , Steroid 17-alpha-Hydroxylase , Testosterone , Humans , Steroid 17-alpha-Hydroxylase/genetics , Male , Middle Aged , Aged , Prostatic Hyperplasia/genetics , Retrospective Studies , Aged, 80 and over , Risk Factors , Testosterone/blood , Estradiol/blood , Polymorphism, Single Nucleotide , Cohort Studies
10.
Front Biosci (Landmark Ed) ; 29(7): 256, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39082359

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is one of the most common malignant tumors of the male urinary system, and its incidence and mortality rates have been increasing worldwide. Benign prostatic hyperplasia (BPH) represents stromal and epithelial cell proliferation in the prostate in elderly males. Abnormal activation of inflammation-related signalling molecules, such as toll-like receptor 4 (TLR4) and Janus kinase/signal transducers and activators of transcription (JAK/STAT) has been linked to the initiation and progression of various human diseases including PCa and BPH. Cylindromatosis (CYLD) gene alterations are associated with PCa progression. In this study, the contribution of CYLD, JAK2, and TLR4 gene variants to PCa and BPH risks and their associations with prostate-specific antigen (PSA) levels, immunophenotype, and clinical features in Vietnamese men were determined. METHODS: A total of 102 patients with PCa, 65 with BPH, and 114 healthy controls were enrolled. The immunophenotype was analyzed by flow cytometry, cytokine secretion by enzyme-linked immunosorbent assay (ELISA), and gene variants by DNA sequencing. RESULTS: Lower levels of transforming growth factor ß (TGF-ß) and higher numbers of CD13+CD117- and CD56+CD25+ cells were observed in the PCa group than in the BPH group. Genetic analysis of the CYLD gene identified five single nucleotide polymorphisms (SNPs), of which c.2351-47 C>T, c.2351-46A>T, and rs1971432171 T>G had significantly higher frequencies in PCa patients than in the control and BPH groups. Sequencing of the TLR4 gene revealed five nucleotide changes, in which the rs2149356 SNP showed an increased risk for both PCa and BPH and the c.331-206 SNP had a reduced risk for PCa. Importantly, the expansion of activated natural killer (NK) cells and higher levels of PSA were found in PCa patients carrying the CT genotype of the CYLD c.2351-47 compared to those with the wild-type genotype. CONCLUSION: Activation of NK cells in CYLD-sensitive PCa patients was associated with serum PSA release and the CYLD c.2351-47 variant may be a significant risk factor for prostatitis in PCa patients.


Subject(s)
Deubiquitinating Enzyme CYLD , Janus Kinase 2 , Prostate-Specific Antigen , Prostatic Hyperplasia , Prostatic Neoplasms , Toll-Like Receptor 4 , Humans , Male , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/blood , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/blood , Toll-Like Receptor 4/genetics , Deubiquitinating Enzyme CYLD/genetics , Deubiquitinating Enzyme CYLD/metabolism , Prostate-Specific Antigen/blood , Aged , Janus Kinase 2/genetics , Middle Aged , Immunophenotyping , Genotype , Polymorphism, Single Nucleotide , Case-Control Studies
11.
Prostate ; 84(12): 1128-1137, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38824441

ABSTRACT

BACKGROUND: The unmet challenge in prostate cancer (PCa) management is to discriminate it from benign prostate hyperplasia (BPH) due to the lack of specific diagnostic biomarkers. Contemporary research on potential PCa biomarkers is directed toward methylated cell-free DNA (cfDNA) from liquid biopsies since epigenetic mechanisms are strongly involved in PCa development. METHODS: In the present research, cfDNA methylation of the LGALS3 gene in blood and seminal plasma of PCa and BPH patients was assessed using pyrosequencing, as well as LGALS3 DNA methylation in tissue biopsies. Liquid biopsy samples were taken from patients with clinical suspicion of PCa, who were subsequently divided into two groups, that is, 42 with PCa and 55 with BPH, according to the histopathological analysis. RESULTS: Statistically significant higher cfDNA methylation of LGALS3 in seminal plasma of BPH than in PCa patients was detected by pyrosequencing. ROC curve analysis showed that it could distinguish PCa and BPH patients with 56.4% sensitivity and 70.4% specificity, while PSA did not differ between the two patient groups. In contrast, there was no statistically significant difference in LGALS3 cfDNA methylation in blood plasma between the two patient groups. In prostate tumor tissue, there was a statistically significant DNA hypermethylation of LGALS3 compared to surrounding nontumor tissue and BPH tissue. CONCLUSIONS: The DNA hypermethylation of the LGALS3 gene represents an event specific to PCa development. In conclusion, LGALS3 cfDNA methylation in seminal fluid discriminates early PCa and BPH presenting itself as a powerful novel PCa biomarker highly outperforming PSA.


Subject(s)
Biomarkers, Tumor , DNA Methylation , Prostate-Specific Antigen , Prostatic Hyperplasia , Prostatic Neoplasms , Semen , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/blood , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Aged , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/genetics , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/diagnosis , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/blood , Prostatic Hyperplasia/pathology , Semen/metabolism , Semen/chemistry , Middle Aged , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/blood , Galectin 3/genetics , Galectin 3/metabolism , Galectin 3/blood , Sensitivity and Specificity , Blood Proteins , Galectins
12.
Front Endocrinol (Lausanne) ; 15: 1348310, 2024.
Article in English | MEDLINE | ID: mdl-38904040

ABSTRACT

Objectives: The relationship between cathepsins and prostate cancer (PCa) has been reported. However, there is a lack of research on cathepsins and benign prostate diseases (BPDs). This study investigated the potential genetic link between cathepsins and BPDs through the utilization of Mendelian randomization (MR) analysis to determine if a causal relationship exists. Methods: Publicly accessible summary statistics on BPDs were obtained from FinnGen Biobank. The data comprised 149,363 individuals, with 30,066 cases and 119,297 controls for BPH, and 123,057 individuals, with 3,760 cases and 119,297 controls for prostatitis. The IEU OpenGWAS provided the Genome-wide association data on ten cathepsins. To evaluate the causal relationship between BPDs and cathepsins, five distinct MR analyses were employed, with the primary method being the inverse variance weighted (IVW) approach. Additionally, sensitivity analyses were conducted to examine the horizontal pleiotropy and heterogeneity of the findings. Results: The examination of IVW MR findings showed that cathepsin O had a beneficial effect on BPH (IVW OR=0.94, 95% CI 0.89-0.98, P=0.0055), while cathepsin X posed a threat to prostatitis (IVW OR=1.08, 95% CI 1.00-1.16, P=0.047). Through reverse MR analysis, it was revealed that prostatitis had an adverse impact on cathepsin V (IVW OR=0.89, 95% CI 0.80-0.99, P=0.035), while no favorable association was observed between BPH and cathepsins. The results obtained from MR-Egger, weighted median, simple mode, and weighted mode methods were consistent with the findings of the IVW approach. Based on sensitivity analyses, heterogeneity, and horizontal pleiotropy are unlikely to distort the results. Conclusion: This study offers the initial evidence of a genetic causal link between cathepsins and BPDs. Our findings revealed that cathepsin O was beneficial in preventing BPH, whereas cathepsin X posed a potential threat to prostatitis. Additionally, prostatitis negatively affected cathepsin V level. These three cathepsins could be targets of diagnosis and treatment for BPDs, which need further research.


Subject(s)
Cathepsins , Genome-Wide Association Study , Mendelian Randomization Analysis , Prostatic Hyperplasia , Humans , Male , Cathepsins/genetics , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/epidemiology , Polymorphism, Single Nucleotide , Case-Control Studies , Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , Prostatic Neoplasms/epidemiology , Prostatitis/genetics , Prostatitis/epidemiology , Prostatic Diseases/genetics , Prostatic Diseases/epidemiology
13.
Med Oncol ; 41(7): 169, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839666

ABSTRACT

To investigate extracellular vesicles (EVs), biomarkers for predicting lymph node invasion (LNI) in patients with high-risk prostate cancer (HRPCa), plasma, and/or urine samples were prospectively collected from 45 patients with prostate cancer (PCa) and five with benign prostatic hyperplasia (BPH). Small RNA sequencing was performed to identify miRNAs in the EVs. All patients with PCa underwent radical prostatectomy and extended pelvic lymph node dissection. Differentially expressed miRNAs were identified in patients with and without pathologically-verified LNI. The candidate miRNAs were validated in low-risk prostate cancer (LRPCa) and BPH. Four miRNA species (e.g., miR-126-3p) and three miRNA species (e.g., miR-27a-3p) were more abundant in urinary and plasma EVs, respectively, of patients with PCa. None of these miRNA species were shared between urinary and plasma EVs. miR-126-3p was significantly more abundant in patients with HR PCa with LNI than in those without (P = 0.018). miR-126-3p was significantly more abundant in the urinary EVs of patients with HRPCa than in those with LRPCa (P = 0.017) and BPH (P = 0.011). In conclusion, urinary EVs-derived miR-126-3p may serve as a good biomarker for predicting LNI in patients with HRPCa.


Subject(s)
Biomarkers, Tumor , Extracellular Vesicles , Lymphatic Metastasis , MicroRNAs , Prostatic Neoplasms , Humans , Male , MicroRNAs/urine , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/urine , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Aged , Middle Aged , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/urine , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/urine , Lymph Nodes/pathology , Prostatectomy , Prospective Studies
14.
BMC Cancer ; 24(1): 774, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937672

ABSTRACT

BACKGROUND: Although it is thought that prostatitis or benign prostatic hyperplasia (BPH) is related to prostate cancer (PCa), the underlying causal effects of these diseases are unclear. METHODS: We assessed the causal relationship between prostatitis or BPH and PCa using a two-sample Mendelian randomization (MR) approach. The data utilized in this study were sourced from genome-wide association study. The association of genetic variants from cohorts of prostatitis or BPH and PCa patients was determined using inverse-variance weighted and MR Egger regression techniques. The direction of chance was determined using independent genetic variants with genome-wide significance (P < 5 × 10-6). The accuracy of the results was confirmed using sensitivity analyses. RESULTS: MR analysis showed that BPH had a significant causal effect on PCa (Odds Ratio = 1.209, 95% Confidence Interval: 0.098-0.281, P = 5.079 × 10- 5) while prostatitis had no significant causal effect on PCa (P > 0.05). Additionally, the pleiotropic test and leave-one-out analysis showed the two-sample MR analyses were valid and reliable. CONCLUSIONS: This MR study supports that BPH has a positive causal effect on PCa, while genetically predicted prostatitis has no causal effect on PCa. Nonetheless, further studies should explore the underlying biochemical mechanism and potential therapeutic targets for the prevention of these diseases.


Subject(s)
Genome-Wide Association Study , Mendelian Randomization Analysis , Prostatic Hyperplasia , Prostatic Neoplasms , Prostatitis , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Hyperplasia/genetics , Prostatitis/genetics , Prostatitis/complications , Polymorphism, Single Nucleotide , Genetic Predisposition to Disease
15.
Genes (Basel) ; 15(5)2024 05 04.
Article in English | MEDLINE | ID: mdl-38790213

ABSTRACT

MicroRNAs (miRNAs) are a class of small non-coding RNAs that may function as tumor suppressors or oncogenes. Alteration of their expression levels has been linked to a range of human malignancies, including cancer. The objective of this investigation is to assess the relative expression levels of certain miRNAs to distinguish between prostate cancer (PCa) from benign prostatic hyperplasia (BPH). Blood plasma was collected from 66 patients diagnosed with BPH and 58 patients with PCa. Real-time PCR technology was used to evaluate the relative expression among the two groups for miR-106a-5p and miR-148a-3p. The significant downregulation of both miRNAs in plasma from PCa versus BPH patients suggests their potential utility as diagnostic biomarkers for distinguishing between these conditions. The concurrent utilization of these two miRNAs slightly enhanced the sensitivity for discrimination among the two analyzed groups, as shown in ROC curve analysis. Further validation of these miRNAs in larger patient cohorts and across different stages of PCa may strengthen their candidacy as clinically relevant biomarkers for diagnosis and prognosis.


Subject(s)
Biomarkers, Tumor , MicroRNAs , Prostatic Hyperplasia , Prostatic Neoplasms , Humans , Male , MicroRNAs/genetics , MicroRNAs/blood , Prostatic Neoplasms/genetics , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/blood , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Aged , Middle Aged , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/blood , Prostatic Hyperplasia/diagnosis , Pilot Projects , Gene Expression Regulation, Neoplastic
16.
Hum Genomics ; 18(1): 49, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778357

ABSTRACT

BACKGROUND: Given the high prevalence of BPH among elderly men, pinpointing those at elevated risk can aid in early intervention and effective management. This study aimed to explore that polygenic risk score (PRS) is effective in predicting benign prostatic hyperplasia (BPH) incidence, prognosis and risk of operation in Han Chinese. METHODS: A retrospective cohort study included 12,474 male participants (6,237 with BPH and 6,237 non-BPH controls) from the Taiwan Precision Medicine Initiative (TPMI). Genotyping was performed using the Affymetrix Genome-Wide TWB 2.0 SNP Array. PRS was calculated using PGS001865, comprising 1,712 single nucleotide polymorphisms. Logistic regression models assessed the association between PRS and BPH incidence, adjusting for age and prostate-specific antigen (PSA) levels. The study also examined the relationship between PSA, prostate volume, and response to 5-α-reductase inhibitor (5ARI) treatment, as well as the association between PRS and the risk of TURP. RESULTS: Individuals in the highest PRS quartile (Q4) had a significantly higher risk of BPH compared to the lowest quartile (Q1) (OR = 1.51, 95% CI = 1.274-1.783, p < 0.0001), after adjusting for PSA level. The Q4 group exhibited larger prostate volumes and a smaller volume reduction after 5ARI treatment. The Q1 group had a lower cumulative TURP probability at 3, 5, and 10 years compared to the Q4 group. PRS Q4 was an independent risk factor for TURP. CONCLUSIONS: In this Han Chinese cohort, higher PRS was associated with an increased susceptibility to BPH, larger prostate volumes, poorer response to 5ARI treatment, and a higher risk of TURP. Larger prospective studies with longer follow-up are warranted to further validate these findings.


Subject(s)
Genetic Predisposition to Disease , Multifactorial Inheritance , Polymorphism, Single Nucleotide , Prostatic Hyperplasia , Aged , Humans , Male , Middle Aged , 5-alpha Reductase Inhibitors/therapeutic use , Asian People/genetics , East Asian People , Genetic Risk Score , Multifactorial Inheritance/genetics , Polymorphism, Single Nucleotide/genetics , Prognosis , Prostate/pathology , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/genetics , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Retrospective Studies , Risk Factors , Taiwan/epidemiology
18.
World J Urol ; 42(1): 216, 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38581575

ABSTRACT

BACKGROUND: Previous research has focused on the association between immune cells and the development of benign prostatic hyperplasia (BPH). Nevertheless, the causal relationships in this context remain uncertain. METHODS: This study employed a comprehensive and systematic two-sample Mendelian randomization (MR) analysis to determine the causal relationships between immunophenotypes and BPH. We examined the causal associations between 731 immunophenotypes and the risk of BPH by utilizing publicly available genetic data. Integrated sensitivity analyses were performed to validate the robustness, assess heterogeneity, and examine horizontal pleiotropy in the results. RESULTS: We discovered that 38 immunophenotypes have a causal effect on BPH. Subsequently, four of these immunophenotypes underwent verification using weighted median, weighted mode, and inverse variance weighted (IVW) algorithms, which included CD19 on CD24+ CD27+, CD19 on naive-mature B cell, HLA DR on CD14- CD16+ and HLA DR+ T cell%lymphocyte. Furthermore, BPH exhibited a significant association with three immunophenotypes: CD19 on IgD+ CD38dim (ß = -0.152, 95% CI = 0.746-0.989, P = 0.034), CD19 on IgD+ (ß = -0.167, 95% CI = 0.737-0.973, P = 0.019), and CD19 on naive-mature B cell (ß = -0.166, 95% CI = 0.737-0.972, P = 0.018). CONCLUSIONS: Our study provides valuable insights for future clinical investigations by establishing a significant association between immune cells and BPH.


Subject(s)
Prostatic Hyperplasia , Humans , Male , Prostatic Hyperplasia/genetics , Mendelian Randomization Analysis , Adaptor Proteins, Signal Transducing , Algorithms , HLA-DR Antigens
19.
BMC Urol ; 24(1): 91, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38643096

ABSTRACT

BACKGROUND: Sleep quality may be related to benign prostatic hyperplasia (BPH), however causal associations have not been established. This study aimed to evaluate causal relationships between six sleep traits ([i] day time napping, [ii] daytime sleepiness, [iii] insomnia, [iv] long sleep duration, [v] short sleep duration, and [vi] sleep duration per hour) and BPH through a bidirectional Mendelian randomization (MR) study. METHODS: Genome-wide association summary statistics of sleep traits and BPH were downloaded from public databases. Inverse variance weighting (IVW) was used as the main approach for causal inference. For causal estimates identified by IVW, various sensitivity analyses were performed to assess the reliability of the results: (i) four additional MR methods to complement IVW; (ii) Cochran's Q test to assess heterogeneity; (iii) MR-Egger intercept test and MR-PRESSO global test to assess horizontal pleiotropy; and (iv) leave-one-out method to assess stability. RESULTS: Forward MR analyses indicated that genetically predicted insomnia symptom significantly increased BPH risk (OR = 1.267, 95% CI: 1.003-1.601, P = 0.048), while reverse MR analyses identified that genetically predicted liability to BPH significantly increased the incidence of insomnia (OR = 1.026, 95% CI: 1.000-1.052, P = 0.048). In a replicate MR analysis based on summary statistics including exclusively male participants, the finding of increased risk of BPH due to genetically predicted insomnia symptom was further validated (OR = 1.488, 95% CI: 1.096-2.022, P = 0.011). No further causal links were identified. In addition, sensitivity tests demonstrated the reliability of the MR results. CONCLUSION: This study identified that a higher prevalence of genetically predicted insomnia symptoms may significantly increase the risk of BPH, while genetically predicted liability to BPH may in turn increase the incidence of insomnia symptom. Therefore, improving sleep quality and reducing the risk of insomnia could be a crucial approach for the prevention of BPH.


Subject(s)
Prostatic Hyperplasia , Sleep Initiation and Maintenance Disorders , Humans , Male , Sleep Initiation and Maintenance Disorders/genetics , Prostatic Hyperplasia/complications , Prostatic Hyperplasia/genetics , Genome-Wide Association Study , Mendelian Randomization Analysis , Reproducibility of Results
20.
J Transl Med ; 22(1): 380, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654277

ABSTRACT

OBJECTIVE: Proliferative nodular formation represents a characteristic pathological feature of benign prostatic hyperplasia (BPH) and serves as the primary cause for prostate volume enlargement and consequent lower urinary tract symptoms (LUTS). Its specific mechanism is largely unknown, although several cellular processes have been reported to be involved in BPH initiation and development and highlighted the crucial role of epithelial cells in proliferative nodular formation. However, the technological limitations hinder the in vivo investigation of BPH patients. METHODS: The robust cell type decomposition (RCTD) method was employed to integrate spatial transcriptomics and single cell RNA sequencing profiles, enabling the elucidation of epithelial cell alterations during nodular formation. Immunofluorescent and immunohistochemical staining was performed for verification. RESULTS: The alterations of epithelial cells during the formation of nodules in BPH was observed, and a distinct subgroup of basal epithelial (BE) cells, referred to as BE5, was identified to play a crucial role in driving this progression through the hypoxia-induced epithelial-mesenchymal transition (EMT) signaling pathway. BE5 served as both the initiating cell during nodular formation and the transitional cell during the transformation from luminal epithelial (LE) to BE cells. A distinguishing characteristic of the BE5 cell subgroup in patients with BPH was its heightened hypoxia and upregulated expression of FOS. Histological verification results confirmed a significant association between c-Fos expression and key biological processes such as hypoxia and cell proliferation, as well as the close relationship between hypoxia and EMT in BPH tissues. Furthermore, a strong link between c-Fos expression and the progression of BPH was also been validated. Additionally, notable functional differences were observed in glandular and stromal nodules regarding BE5 cells, with BE5 in glandular nodules exhibiting enhanced capacities for EMT and cell proliferation characterized by club-like cell markers. CONCLUSIONS: This study elucidated the comprehensive landscape of epithelial cells during in vivo nodular formation in patients, thereby offering novel insights into the initiation and progression of BPH.


Subject(s)
Epithelial Cells , Epithelial-Mesenchymal Transition , Prostatic Hyperplasia , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome , Humans , Male , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Transcriptome/genetics , Gene Expression Profiling , Aged , Middle Aged , Cell Proliferation , Spatial Analysis
SELECTION OF CITATIONS
SEARCH DETAIL