Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.430
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38731981

RESUMO

We aimed to analyze the association between CYP7B1 and prostate cancer, along with its association with proteins involved in cancer and metabolic processes. A retrospective analysis was performed on 390 patients with prostate cancer (PC) or benign prostatic hyperplasia (BPH). We investigated the interactions between CYP7B1 expression and proteins associated with PC and metabolic processes, followed by an analysis of the risk of biochemical recurrence based on CYP7B1 expression. Of the 139 patients with elevated CYP7B1 expression, 92.8% had prostate cancer. Overall, no increased risk of biochemical recurrence was associated with CYP7B1 expression. However, in a non-diabetic subgroup analysis, higher CYP7B1 expression indicated a higher risk of biochemical recurrence, with an HR of 1.78 (CI: 1.0-3.2, p = 0.05). PC is associated with elevated CYP7B1 expression. In a subgroup analysis of non-diabetic patients, elevated CYP7B1 expression was associated with an increased risk of biochemical recurrence, suggesting increased cancer aggressiveness.


Assuntos
Biomarcadores Tumorais , Família 7 do Citocromo P450 , Neoplasias da Próstata , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/genética , Biomarcadores Tumorais/metabolismo , Idoso , Família 7 do Citocromo P450/metabolismo , Família 7 do Citocromo P450/genética , Pessoa de Meia-Idade , Progressão da Doença , Estudos Retrospectivos , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patologia , Imuno-Histoquímica , Análise Serial de Tecidos , Recidiva Local de Neoplasia/metabolismo , Esteroide Hidroxilases
2.
Cell Commun Signal ; 22(1): 257, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38711089

RESUMO

Benign prostatic hyperplasia (BPH) is a multifactorial disease in which abnormal growth factor activation and embryonic reawakening are considered important factors. Here we demonstrated that the aberrant activation of transforming growth factor ß (TGF-ß)/Rho kinase 1 (ROCK1) increased the stemness of BPH tissue by recruiting mesenchymal stem cells (MSCs), indicating the important role of embryonic reawakening in BPH. When TGF-ß/ROCK1 is abnormally activated, MSCs are recruited and differentiate into fibroblasts/myofibroblasts, leading to prostate stromal hyperplasia. Further research showed that inhibition of ROCK1 activation suppressed MSC migration and their potential for stromal differentiation. Collectively, our findings suggest that abnormal activation of TGF-ß/ROCK1 regulates stem cell lineage specificity, and the small molecule inhibitor GSK269962A could target ROCK1 and may be a potential treatment for BPH.


Assuntos
Células-Tronco Mesenquimais , Hiperplasia Prostática , Fator de Crescimento Transformador beta , Quinases Associadas a rho , Quinases Associadas a rho/metabolismo , Masculino , Hiperplasia Prostática/patologia , Hiperplasia Prostática/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular , Próstata/patologia , Próstata/metabolismo , Movimento Celular , Camundongos , Células Estromais/metabolismo , Células Estromais/patologia
3.
Reprod Domest Anim ; 59(5): e14580, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38698701

RESUMO

The pathogenesis of benign prostatic hyperplasia (BPH) in dogs is still not fully understood. Some studies suggest that oxidative stress may have a potential role in the pathogenesis of BPH in dogs. Age-related hormonal changes may activate a chronic inflammatory response in the prostate. This causes the generation of reactive oxygen species (ROS) and results in oxidative stress. Excessive production of ROS results in DNA damage and hyperplastic transformation of prostatic cells. The use of antioxidants for improvement of treatment outcomes for canine PBH has been discussed. Further research is needed on the importance of oxidative stress in the development of BPH in dogs and the usefulness of antioxidants in the supportive treatment of this condition.


Assuntos
Antioxidantes , Doenças do Cão , Estresse Oxidativo , Hiperplasia Prostática , Espécies Reativas de Oxigênio , Animais , Cães , Masculino , Hiperplasia Prostática/veterinária , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patologia , Espécies Reativas de Oxigênio/metabolismo , Dano ao DNA , Próstata/patologia , Próstata/metabolismo
4.
J Transl Med ; 22(1): 380, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654277

RESUMO

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.


Assuntos
Células Epiteliais , Transição Epitelial-Mesenquimal , Hiperplasia Prostática , Análise de Sequência de RNA , Análise de Célula Única , Transcriptoma , Humanos , Masculino , Hiperplasia Prostática/patologia , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/genética , Transcriptoma/genética , Perfilação da Expressão Gênica , Idoso , Pessoa de Meia-Idade , Proliferação de Células , Análise Espacial
5.
Mol Med ; 30(1): 41, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38519941

RESUMO

BACKGROUND: Benign prostatic hyperplasia (BPH) is a prevalent disease affecting elderly men, with chronic inflammation being a critical factor in its development. Omentin-1, also known as intelectin-1 (ITLN-1), is an anti-inflammatory protein primarily found in the epithelial cells of the small intestine. This study aimed to investigate the potential of ITLN-1 in mitigating BPH by modulating local inflammation in the prostate gland. METHODS: Our investigation involved two in vivo experimental models. Firstly, ITLN-1 knockout mice (Itln-1-/-) were used to study the absence of ITLN-1 in BPH development. Secondly, a testosterone propionate (TP)-induced BPH mouse model was treated with an ITLN-1 overexpressing adenovirus. We assessed BPH severity using prostate weight index and histological analysis, including H&E staining, immunohistochemistry, and enzyme-linked immunosorbent assay. In vitro, the impact of ITLN-1 on BPH-1 cell proliferation and inflammatory response was evaluated using cell proliferation assays and enzyme-linked immunosorbent assay. RESULTS: In vivo, Itln-1-/- mice exhibited elevated prostate weight index, enlarged lumen area, and higher TNF-α levels compared to wild-type littermates. In contrast, ITLN-1 overexpression in TP-induced BPH mice resulted in reduced prostate weight index, lumen area, and TNF-α levels. In vitro studies indicated that ITLN-1 suppressed the proliferation of prostate epithelial cells and reduced TNF-α production in macrophages, suggesting a mechanism involving the inhibition of macrophage-mediated inflammation. CONCLUSION: The study demonstrates that ITLN-1 plays a significant role in inhibiting the development of BPH by reducing local inflammation in the prostate gland. These findings highlight the potential of ITLN-1 as a therapeutic target in the management of BPH.


Assuntos
Hiperplasia Prostática , Humanos , Masculino , Camundongos , Animais , Idoso , Hiperplasia Prostática/genética , Hiperplasia Prostática/tratamento farmacológico , Hiperplasia Prostática/metabolismo , Fator de Necrose Tumoral alfa , Extratos Vegetais/farmacologia , Próstata/metabolismo , Próstata/patologia , Inflamação/patologia
6.
Int J Mol Sci ; 25(6)2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38542507

RESUMO

Prostate-specific membrane antigen (PSMA) and caveolin-1 are membrane proteins that are overexpressed in prostate cancer (PCa) and are involved in tumor growth and increase in aggressiveness. The aim of the present study is therefore to evaluate PSMA and caveolin-1 proteins from plasma exosomes as effective liquid biopsy biomarkers for PCa. This study included 39 patients with PCa and 33 with benign prostatic hyperplasia (BPH). The shape and size of the exosomes were confirmed by transmission electron microscopy (TEM) and scanning electron microscopy (SEM) analysis. Immunogold analysis showed that PSMA is localized to the membrane of exosomes isolated from the plasma of both groups of participants. The relative protein levels of PSMA and caveolin-1 in the plasma exosomes of PCa and BPH patients were determined by Western blot analysis. The relative level of the analyzed plasma exosomal proteins was compared between PCa and BPH patients and the relevance of the exosomal PSMA and caveoin-1 level to the clinicopathological parameters in PCa was investigated. The analysis performed showed an enrichment of exosomal PSMA in the plasma of PCa patients compared to the exosomes of men with BPH. The level of exosomal caveolin-1 in plasma was significantly higher in PCa patients with high PSA levels, clinical-stage T3 or T4 and in the group of PCa patients with aggressive PCa compared to favorable clinicopathological features or tumor aggressiveness. Plasma exosomes may serve as a suitable object for the identification of potential biomarkers for the early diagnosis and prognosis of PCa as well as carriers of therapeutic agents in precision medicine of PCa treatment.


Assuntos
Hiperplasia Prostática , Neoplasias da Próstata , Masculino , Humanos , Hiperplasia Prostática/metabolismo , Próstata/patologia , Caveolina 1/metabolismo , Sérvia , Biomarcadores Tumorais/metabolismo , Neoplasias da Próstata/metabolismo , Antígeno Prostático Específico/metabolismo
7.
J Ethnopharmacol ; 328: 118068, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38513777

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Prostatitis and benign prostatic hyperplasia (BPH) are inflammations of the prostate gland, which surrounds the urethra in males. Jinqiancao granules are a traditional Chinese medicine used to treat kidney stones and this medicine consists of four herbs: Desmodium styracifolium (Osbeck) Merr., Pyrrosia calvata (Baker) Ching, Plantago asiatica L. and stigma of Zea mays L. AIM OF THE STUDY: We hypothesized that Jinqiancao granules could be a potential therapy for prostatitis and BPH, and this work aimed to elucidate active compounds in Jinqiancao granules and their target mechanisms for the potential treatment of the two diseases. MATERIALS AND METHODS: Jinqiancao granules were commercially available and purchased. Database-driven data mining and networking were utilized to establish a general correlation between Jinqiancao granules and the two diseases above. Ultra-performance liquid chromatography-mass spectrometry was used for compound separation and characterization. The characterized compounds were evaluated on four G-protein coupled receptors (GPCRs: GPR35, muscarinic acetylcholine receptor M3, alpha-1A adrenergic receptor α1A and cannabinoid receptor CB2). A dynamic mass redistribution technique was applied to evaluate compounds on four GPCRs. Nitric acid (NO) inhibition was tested on the macrophage cell line RAW264.7. Molecular docking was conducted on GPR35-active compounds and GPR35 crystal structure. Statistical analysis using GEO datasets was conducted. RESULTS: Seventy compounds were isolated and twelve showed GPCR activity. Three compounds showed potent GPR35 agonistic activity (EC50 < 10 µM) and the GPR35 agonism action of PAL-21 (Scutellarein) was reported for the first time. Docking results revealed that the GPR35-targeting compounds interacted at the key residues for the agonist-initiated activation of GPR35. Five compounds showed weak antagonistic activity on M3, which was confirmed to be a disease target by statistical analysis. Seventeen compounds showed NO inhibitory activity. Several compounds showed multi-target properties. An experiment-based network reflected a pharmacological relationship between Jinqiancao granules and the two diseases. CONCLUSIONS: This study identified active compounds in Jinqiancao granules that have synergistic mechanisms, contributing to anti-inflammatory effects. The findings provide scientific evidence for the potential use of Jinqiancao granules as a treatment for prostatitis and BPH.


Assuntos
Hiperplasia Prostática , Prostatite , Masculino , Humanos , Prostatite/tratamento farmacológico , Prostatite/metabolismo , Hiperplasia Prostática/tratamento farmacológico , Hiperplasia Prostática/metabolismo , Simulação de Acoplamento Molecular , Próstata , Receptores Acoplados a Proteínas G/metabolismo
8.
Cell Mol Life Sci ; 81(1): 155, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38538986

RESUMO

The prostate is a vital accessory gonad in the mammalian male reproductive system. With the ever-increasing proportion of the population over 60 years of age worldwide, the incidence of prostate diseases, such as benign prostatic hyperplasia (BPH) and prostate cancer (PCa), is on the rise and is gradually becoming a significant medical problem globally. The notch signaling pathway is essential in regulating prostate early development. However, the potential regulatory mechanism of Notch signaling in prostatic enlargement and hyperplasia remains unclear. In this study, we proved that overactivation of Notch1 signaling in mouse prostatic epithelial cells (OEx) led to prostatic enlargement via enhancing proliferation and inhibiting apoptosis of prostatic epithelial cells. Further study showed that N1ICD/RBPJ directly up-regulated the androgen receptor (AR) and enhanced prostatic sensitivity to androgens. Hyper-proliferation was not found in orchidectomized OEx mice without androgen supply but was observed after Dihydrotestosterone (DHT) supplementation. Our data showed that the number of mitochondrion in prostatic epithelial cells of OEx mice was increased, but the mitochondrial function was impaired, and the essential activity of the mitochondrial respiratory electron transport chain was significantly weakened. Disordered mitochondrial number and metabolic function further resulted in excessive accumulation of reactive oxygen species (ROS). Importantly, anti-oxidant N-Acetyl-L-Cysteine (NAC) therapy could alleviate prostatic hyperplasia caused by the over-activation of Notch1 signaling. Furthermore, we observed the incremental Notch signaling activity in progenitor-like club cells in the scRNA-seq data set of human BPH patients. Moreover, the increased number of TROP2+ progenitors and Club cells was also confirmed in our OEx mice. In conclusion, our study revealed that over-activated Notch1 signaling induces prostatic enlargement by increasing androgen receptor sensitivity, disrupting cellular mitochondrial metabolism, increasing ROS, and a higher number of progenitor cells, all of which can be effectively rescued by NAC treatment.


Assuntos
Hiperplasia Prostática , Animais , Humanos , Masculino , Camundongos , Androgênios/metabolismo , Mamíferos/metabolismo , Mitocôndrias/metabolismo , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transdução de Sinais
9.
J Transl Med ; 22(1): 255, 2024 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-38459501

RESUMO

OBJECTIVE: Benign prostatic hyperplasia (BPH) is common in elder men. The current study aims to identify differentially expressed genes (DEGs) in hyperplastic prostate and to explore the role of Nik related kinase (NRK) in BPH. METHODS: Four datasets including three bulk and one single cell RNA-seq (scRNA-seq) were obtained to perform integrated bioinformatics. Cell clusters and specific metabolism pathways were analyzed. The localization, expression and functional activity of NRK was investigated via RT-PCR, western-blot, immunohistochemical staining, flow cytometry, wound healing assay, transwell assay and CCK-8 assay. RESULTS: A total of 17 DEGs were identified by merging three bulk RNA-seq datasets. The findings of integrated single-cell analysis showed that NRK remarkably upregulated in fibroblasts and SM cells of hyperplasia prostate. Meanwhile, NRK was upregulated in BPH samples and localized almost in stroma. The expression level of NRK was significantly correlated with IPSS and Qmax of BPH patients. Silencing of NRK inhibited stromal cell proliferation, migration, fibrosis and EMT process, promoted apoptosis and induced cell cycle arrest, while overexpression of NRK in prostate epithelial cells showed opposite results. Meanwhile, induced fibrosis and EMT process were rescued by knockdown of NRK. Furthermore, expression level of NRK was positively correlated with that of α-SMA, collagen-I and N-cadherin, negatively correlated with that of E-cadherin. CONCLUSION: Our novel data identified NRK was upregulated in hyperplastic prostate and associated with prostatic stromal cell proliferation, apoptosis, cell cycle, migration, fibrosis and EMT process. NRK may play important roles in the development of BPH and may be a promising therapeutic target for BPH/LUTS.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Próstata , Hiperplasia Prostática , Proteínas Serina-Treonina Quinases , Masculino , Humanos , Idoso , Próstata/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patologia , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patologia , Fibrose
10.
Clin Exp Pharmacol Physiol ; 51(4): e13851, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38452757

RESUMO

Benign prostatic hyperplasia (BPH) is characterised by increases in prostate volume and contraction. Downregulation of the nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) signalling pathway contributes to prostate dysfunctions. Previous studies in cancer cells or vessels have shown that the epigenetic mechanisms control the gene and protein expression of the enzymes involved in the production of NO and cGMP. This study is aimed to evaluate the effect of a 2-week treatment of 5-azacytidine (5-AZA), a DNA-methyltransferase inhibitor, in the prostate function of mice fed with a high-fat diet. Functional, histological, biochemical and molecular assays were carried out. Obese mice presented greater prostate weight, α-actin expression and contractile response induced by the α-1adrenoceptors agonist. The relaxation induced by the NO-donor and the protein expression of endothelial nitric oxide synthase (eNOS) and soluble guanylate cyclase (sGC) were significantly decreased in the prostate of obese mice. The treatment with 5-AZA reverted the higher expression of α-actin, reduced the hypercontractility state of the prostate and increased the expression of eNOS and sGC and intraprostatic levels of cGMP. When prostates from obese mice treated with 5-AZA were incubated in vitro with inhibitors of the NOS or sGC, the inhibitory effect of 5-AZA was reverted, therefore, showing the involvement of NO and cGMP. In conclusion, our study paves the way to develop or repurpose therapies that recover the expression of eNOS and sGC and, hence, to improve prostate function in BPH.


Assuntos
Óxido Nítrico , Hiperplasia Prostática , Masculino , Humanos , Camundongos , Animais , Óxido Nítrico/metabolismo , Guanilato Ciclase/metabolismo , Próstata/metabolismo , Camundongos Obesos , Guanosina Monofosfato/metabolismo , Azacitidina/metabolismo , Hiperplasia Prostática/metabolismo , Actinas/metabolismo , GMP Cíclico/metabolismo
11.
Int J Biochem Cell Biol ; 169: 106551, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38360265

RESUMO

Benign prostatic hyperplasia (BPH) is one of the most common diseases in elderly men worldwide that may result in lower urinary tract symptoms (LUTS). At present, the specific pathophysiological mechanism for BPH/LUTS LUTS remains unclear. S100 calcium binding protein A4 (S100A4), a member of the calcium binding protein family, regulates a variety of biological processes including cell proliferation, apoptosis and fibrosis. The aim of the current study was to explore and clarify the possible role of S100A4 in BPH/LUTS. The human prostate stromal cell line (WPMY-1), rat prostate epithelial cells, human prostate tissues and two BPH rat models were employed in this study. The expression and localization of S100A4 were detected by quantitative real time PCR (qRT-PCR), immunofluorescence microscopy, Western blotting and immunohistochemistry analysis. Also, S100A4 knockdown or overexpression cell models were constructed and a BPH rat model was induced with testosterone propionate (T) or phenylephrine (PE). The BPH animals were treated with Niclosamide, a S100A4 transcription inhibitor. Results demonstrated that S100A4 was mainly localized in human prostatic stroma and rat prostatic epithelium, and showed a higher expression in BPH. Knockdown of S100A4 induced cell apoptosis, cell proliferation arrest and a reduction of tissue fibrosis markers. Overexpression of S100A4 reversed the aforementioned changes. We also demonstrated that S100A4 regulated proliferation and apoptosis mainly through the ERK pathway and modulated fibrosis via Wnt/ß-catenin signaling. In conclusion, our novel data demonstrate that S100A4 could play a crucial role in BPH development and may be explored as a new therapeutic target of BPH.


Assuntos
Próstata , Hiperplasia Prostática , Proteína A4 de Ligação a Cálcio da Família S100 , Idoso , Animais , Humanos , Masculino , Ratos , Apoptose , Proliferação de Células , Fibrose , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/genética , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo
12.
Food Chem Toxicol ; 185: 114450, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38215961

RESUMO

Fibrosis is the pathological basis for the clinical progression of benign prostatic hyperplasia (BPH). Prostatic fibrosis is an important risk factor in patients with BPH who experience lower urinary tract symptoms. Bisphenol A (BPA) is an environmental endocrine disruptor (EED) that causes prostate defects. The effects of BPA on the prostate were investigated in this study using mouse and human prostate cell models. BPA-induced mouse prostatic fibrosis is characterized by collagen deposition and an increase in hydroxyproline concentration. Furthermore, BPA-exposed prostatic stromal fibroblasts exosomes promote the epithelial-mesenchymal transition of epithelial cells. High-throughput RNA sequencing and functional enrichment analyses show that substantially altered mRNAs, lncRNAs and circRNAs play roles in cellular interactions and the hypoxia-inducible factor-1 signaling pathway. The results showed that exosomes participated in the pro-fibrogenic effects of BPA on the prostate by mediating communication between stromal and epithelial cells and triggering epithelial changes.


Assuntos
Compostos Benzidrílicos , Exossomos , Fenóis , Hiperplasia Prostática , Masculino , Humanos , Camundongos , Animais , Próstata , Hiperplasia Prostática/induzido quimicamente , Hiperplasia Prostática/metabolismo , Exossomos/metabolismo , Epitélio/metabolismo , Epitélio/patologia , Fibrose
13.
Artigo em Inglês | MEDLINE | ID: mdl-38198648

RESUMO

BACKGROUND: Older men frequently develop lower urinary tract symptoms attributed to benign prostatic hyperplasia (LUTS/BPH). Risk factors for LUTS/BPH include sedentary lifestyle, anxiety/depression, obesity, and frailty, which all increase with age. Although physical exercise may reduce the progression and/or severity of LUTS/BPH, the age-related mechanisms responsible remain unknown. METHODS: Voiding symptoms, body mass, and frailty were assessed after 4-weeks of voluntary wheel running in 2-month (n = 10) and 24-month (n = 8) old C57Bl/6J male mice. In addition, various social and individual behaviors were examined in these cohorts. Finally, cellular and molecular markers of inflammation and mitochondrial protein expression were assessed in prostate tissue and systemically. RESULTS: Despite running less (aged vs young X¯ = 12.3 vs 30.6 km/week; p = .04), aged mice had reduced voiding symptoms (X¯ = 67.3 vs 23.7; p < .0001) after 1 week of exercise, which was sustained through week 4 (X¯ = 67.3 vs 21.5; p < .0001). Exercise did not affect voiding symptoms in young mice. Exercise also increased mobility and decreased anxiety in both young and aged mice (p < .05). Exercise decreased expression of a key mitochondrial protein (PINK1; p < .05) and inflammation within the prostate (CD68; p < .05 and plasminogen activator inhibitor-1; p < .05) and in the serum (p < .05). However, a frailty index (X¯ = 0.17 vs 0.15; p = .46) and grip strength (X¯ = 1.10 vs 1.19; p = .24) were unchanged after 4 weeks of exercise in aged mice. CONCLUSIONS: Voluntary aerobic exercise improves voiding behavior and mobility, and decreases prostatic mitochondrial protein expression and inflammation in aged mice. This promising model could be used to evaluate molecular mechanisms of aerobic exercise as a novel lifestyle intervention for older men with LUTS/BPH.


Assuntos
Envelhecimento , Sintomas do Trato Urinário Inferior , Camundongos Endogâmicos C57BL , Condicionamento Físico Animal , Animais , Masculino , Camundongos , Condicionamento Físico Animal/fisiologia , Envelhecimento/fisiologia , Sintomas do Trato Urinário Inferior/fisiopatologia , Sintomas do Trato Urinário Inferior/metabolismo , Micção/fisiologia , Hiperplasia Prostática/metabolismo , Fragilidade/metabolismo , Fatores Etários , Próstata/metabolismo , Comportamento Animal/fisiologia
14.
Microbiol Res ; 281: 127596, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38215640

RESUMO

The research of the human microbiome in the preceding decade has yielded novel perspectives on human health and diseases. Benign prostatic hyperplasia (BPH) is a common disease in middle-aged and elderly males, which negatively affects the life quality. Existing evidence has indicated that the human microbiome, including urinary, intra-prostate, gut, oral and blood microbiome may exert a significant impact on the natural progression of BPH. The dysbiosis of the microbiome may induce inflammation at either a local or systemic level, thereby affecting the BPH. Moreover, metabolic syndrome (MetS) caused by the microbiome can also be involved in the development of BPH. Additionally, alterations in the microbiome composition during the senility process may serve as another cause of the BPH. Here, we summarize the influence of human microbiome on BPH and explore how the microbiome is linked to BPH through inflammation, MetS, and senility. In addition, we propose promising areas of investigation and discuss the implications for advancing therapeutic approaches.


Assuntos
Síndrome Metabólica , Microbiota , Hiperplasia Prostática , Masculino , Idoso , Pessoa de Meia-Idade , Humanos , Hiperplasia Prostática/etiologia , Hiperplasia Prostática/metabolismo , Inflamação , Síndrome Metabólica/complicações
15.
Tissue Cell ; 86: 102231, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37931534

RESUMO

BACKGROUND: Gremlin 1 (GREM1) has been reported to be highly expressed in prostate hyperplasia tissues. However, the role and molecular mechanism of GREM1 in benign prostatic hyperplasia (BPH) is still unclear. METHODS: In this study, expression of GREM1 in BPH-1 cells was detected by western blot assay. Cell counting kit-8 assay was performed to assess cell proliferation. Flow cytometry and western blot were used to assess cell apoptosis and cell cycle. The EMT process was detected by western blot assay and immunofluorescence staining. In addition, colivelin was used as a STAT3 activator and the expressions of STAT3/c-Myc signaling were assessed by western blot assay. RESULTS: The data showed that GREM1 silencing inhibited BPH-1 cell proliferation and promoted cell apoptosis. Moreover, GREM1 silencing repressed the cell cycle progression and the development of EMT. In addition, knockdown of GREM1 suppressed the expression of the STAT3/c-Myc signaling in BPH-1 cells and colivelin treatment rehabilitated this signaling. Moreover, c-Myc overexpression or colivelin reversed the effects of GREM1 silencing on BPH-1 cell proliferation, cell apoptosis, cell cycle, as well as EMT. CONCLUSION: To sum up, GREM1 silencing may alleviate the BPH progress by inhibiting the STAT3/c-Myc signaling.


Assuntos
Hiperplasia Prostática , Masculino , Humanos , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais/genética , Proliferação de Células/genética , Apoptose/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
16.
Adv Sci (Weinh) ; 11(5): e2304274, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38050650

RESUMO

Chronic prostatic inflammation promotes cell survival and fibrosis, leading to benign prostatic hyperplasia (BPH) with aggravated urinary symptoms. It is investigated whether yes-associated protein 1 (YAP1), an organ size controller and mechanical transductor, is implicated in inflammation-induced BPH. The correlation between YAP1 expression and fibrosis in human and rat BPH specimens is analyzed. Furthermore, the effects of YAP1 activation on prostatic cell survival and fibrosis, as well as the underlying mechanism, are also studied. As a result, total and nuclear YAP1 expression, along with downstream genes are significantly upregulated in inflammation-associated human and rat specimens. There is a significant positive correlation between YAP1 expression and the severity of fibrosis or clinical performance. YAP1 silencing suppresses cell survival by decreasing cell proliferation and increasing apoptosis, and alleviates fibrosis by reversing epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) deposition in prostatic BPH-1 and WPMY-1 cells. Mechanistically, inflammatory stimulus and rigid matrix stiffness synergistically activate the RhoA/ROCK1 pathway to provoke cytoskeleton remodeling, thereby promoting YAP1 activation to exacerbate BPH development. Overall, inflammation-triggered mechanical stiffness reinforcement activates the RhoA/ROCK1/F-actin/YAP1 axis, thereby promoting prostatic cell survival and fibrosis to accelerate BPH progression.


Assuntos
Hiperplasia Prostática , Animais , Humanos , Masculino , Ratos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Sobrevivência Celular , Fibrose , Inflamação , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patologia , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/farmacologia , Fatores de Transcrição/metabolismo
17.
Pathol Res Pract ; 253: 154970, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38056136

RESUMO

The role of epigenetic alteration in prostate cancer pathogenesis was reported. We aimed to analyze dysregulation of DNA methylase (DNA methyl transferase/DNMT) and demethylase (ten eleven translocase/TET) and the associated interplay between them during prostate tumorigenesis. Promoter methylation and RNA/protein expression of selected DNMT and TETs were analysed in normal prostate, benign prostatic hyperplasia (BPH), and prostate cancer (PCa). Genomic 5-hydroxymethylcytosine (5hmC) level was detected and correlated with DNMT and TET proteins. Clinicopathological association of molecular data was done. Our data revealed a very low frequency of promoter methylation for DNMT1 (5-3% and high frequency for TET1 (22-38%), TET2 (68-90 %), and TET3 (43-32 %) in BPH and PCa. The promoter methylation of DNMT1 (p = 0.019) showed a significantly decreasing trend, while that of TET1 (p = 0.0005) and TET2 (p < 0.0001) showed an increasing trend from normal prostate to BPH to PCa, indicating their epigenetic dysregulation during prostate tumorigenesis. RNA/protein overexpression of DNMT1 and reduced expression of TET1 and TET2 in PCa compared to BPH were associated with the promoter methylation status of genes. The 5hmC level was significantly lower in PCa than in BPH and correlated negatively with DNMT1 but positively with TET1 and TET2 proteins, suggesting dysregulation of DNA methylase and de-methylase activities during prostate tumorigenesis. Lastly, tumors having methylated TET1 and TET2 promoters showed advanced clinicopathological features (a higher PSA level/Gleason score) and increased risk of bone metastasis. In conclusion, DNMT1 upregulation and epigenetic silencing of TET1 and TET2 was seen during PCa development. TET1 and TET2 promoter methylation has prognostic importance.


Assuntos
Hiperplasia Prostática , Neoplasias da Próstata , Masculino , Humanos , Próstata/metabolismo , Prognóstico , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Metilação de DNA/genética , Epigênese Genética , Carcinogênese/genética , Transformação Celular Neoplásica/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , DNA , RNA/metabolismo , Oxigenases de Função Mista/metabolismo
18.
Am J Pathol ; 194(1): 30-51, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37827216

RESUMO

Benign prostate hyperplasia (BPH) is caused by the nonmalignant enlargement of the transition zone of the prostate gland, leading to lower urinary tract symptoms. Although current medical treatments are unsatisfactory in many patients, the limited understanding of the mechanisms driving disease progression prevents the development of alternative therapeutic strategies. The probasin-prolactin (Pb-PRL) transgenic mouse recapitulates many histopathological features of human BPH. Herein, these alterations parallel urodynamic disturbance reminiscent of lower urinary tract symptoms. Single-cell RNA-sequencing analysis of Pb-PRL mouse prostates revealed that their epithelium mainly includes low-androgen signaling cell populations analogous to Club/Hillock cells enriched in the aged human prostate. These intermediate cells are predicted to result from the reprogramming of androgen-dependent luminal cells. Pb-PRL mouse prostates exhibited increased vulnerability to oxidative stress due to reduction of antioxidant enzyme expression. One-month treatment of Pb-PRL mice with anethole trithione (ATT), a specific inhibitor of mitochondrial ROS production, reduced prostate weight and voiding frequency. In human BPH-1 epithelial cells, ATT decreased mitochondrial metabolism, cell proliferation, and stemness features. ATT prevented the growth of organoids generated by sorted Pb-PRL basal and LSCmed cells, the two major BPH-associated, androgen-independent epithelial cell compartments. Taken together, these results support cell plasticity as a driver of BPH progression and therapeutic resistance to androgen signaling inhibition, and identify antioxidant therapy as a promising treatment of BPH.


Assuntos
Sintomas do Trato Urinário Inferior , Hiperplasia Prostática , Masculino , Humanos , Camundongos , Animais , Idoso , Androgênios/farmacologia , Androgênios/metabolismo , Próstata/patologia , Hiperplasia Prostática/metabolismo , Antioxidantes/farmacologia , Plasticidade Celular , Hiperplasia/patologia , Chumbo/metabolismo , Chumbo/uso terapêutico , Camundongos Transgênicos , Prolactina/metabolismo , Prolactina/uso terapêutico , Células Epiteliais/metabolismo , Sintomas do Trato Urinário Inferior/metabolismo , Sintomas do Trato Urinário Inferior/patologia
19.
JCI Insight ; 9(2)2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-37971878

RESUMO

Benign prostatic hyperplasia (BPH) is the nodular proliferation of the prostate transition zone in older men, leading to urinary storage and voiding problems that can be recalcitrant to therapy. Decades ago, John McNeal proposed that BPH originates with the "reawakening" of embryonic inductive activity by adult prostate stroma, which spurs new ductal proliferation and branching morphogenesis. Here, by laser microdissection and transcriptional profiling of the BPH stroma adjacent to hyperplastic branching ducts, we identified secreted factors likely mediating stromal induction of prostate glandular epithelium and coinciding processes. The top stromal factors were insulin-like growth factor 1 (IGF1) and CXC chemokine ligand 13 (CXCL13), which we verified by RNA in situ hybridization to be coexpressed in BPH fibroblasts, along with their cognate receptors (IGF1R and CXCR5) on adjacent epithelium. In contrast, IGF1 but not CXCL13 was expressed in human embryonic prostate stroma. Finally, we demonstrated that IGF1 is necessary for the generation of BPH-1 cell spheroids and patient-derived BPH cell organoids in 3D culture. Our findings partially support historic speculations on the etiology of BPH and provide what we believe to be new molecular targets for rational therapies directed against the underlying processes driving BPH.


Assuntos
Hiperplasia Prostática , Masculino , Adulto , Humanos , Idoso , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Próstata/metabolismo , Epitélio/metabolismo , Fibroblastos/metabolismo , Perfilação da Expressão Gênica
20.
Cell Signal ; 113: 110933, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37866665

RESUMO

Benign prostatic hyperplasia (BPH) is a quite common chronic disease plagued elderly men and its etiology remains unclear. It was reported that the six-transmembrane epithelial antigen of prostate 4 (STEAP4) could modulate cell proliferation/apoptosis ratio and oxidative stress in cancers. Our current study aimed to explore the expression, biological function, and underlying mechanism of STEAP4 in BPH progress. Human prostate tissues and cell lines were utilized. qRT-PCR and immunofluorescence staining were employed. STEAP4 knockdown (STEAP4-KD) or STEAP4 overexpression (STEAP4-OE) cell models were established. Cell proliferation, cell cycle, apoptosis, and reactive oxygen species (ROS) were determined by cell counting kit-8 (CCK-8) assay and flow cytometry. Apoptosis-related proteins and antioxidant enzymes were identified by Western Blot. In addition, the epithelial-mesenchymal transition (EMT) process and fibrosis biomarker (collagen I and α-SMA) were analyzed. It was indicated that STEAP4 was mainly located in the prostate epithelium and upregulated in BPH tissues. STEAP4 deficiency induced apoptosis and inhibited cell survival, but had no effect on the cell cycle, fibrosis, and EMT process. In addition, ROS changes were observed in the STEAP4-KD model. Consistently, overproduction of STEAP4 suppressed apoptosis and promoted cell proliferation, as well as facilitated ROS production. We further examined AKT / mTOR, p38MAPK / p-p38MAPK, and WNT/ ß-Catenin signaling pathway and demonstrated that STEAP4 regulated the proliferation and apoptosis of prostate cells through AKT / mTOR signaling, rather than p38MAPK / p-p38MAPK and WNT/ ß-Catenin pathways. Furthermore, activating AKT / mTOR signaling with SC79 significantly reversed apoptosis triggered by STEAP4 deficiency, whereas suppressing AKT / mTOR signaling with MK2206 reduced the increase of cell viability triggered by STEAP4 overproduction. Our original data demonstrated that STEAP4 is crucial in the onset and progression of prostate hyperplasia and may become a new target for the treatment of BPH.


Assuntos
Hiperplasia Prostática , Masculino , Humanos , Idoso , Hiperplasia Prostática/metabolismo , beta Catenina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio , Serina-Treonina Quinases TOR/metabolismo , Proliferação de Células , Apoptose , Estresse Oxidativo , Fibrose , Proteínas de Membrana/metabolismo , Oxirredutases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA