Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20.483
Filtrer
1.
Sci Rep ; 14(1): 17877, 2024 08 02.
Article de Anglais | MEDLINE | ID: mdl-39095452

RÉSUMÉ

Differentially methylated CpG sites (dmCpGs) that distinguish prostate tumour from adjacent benign tissue could aid in the diagnosis and prognosis of prostate cancer. Previously, the identification of such dmCpGs has only been undertaken in radical prostatectomy (RP) samples and not primary diagnostic tumour samples (needle biopsy or transurethral resection of the prostate). We interrogated an Australian dataset comprising 125 tumour and 43 adjacent histologically benign diagnostic tissue samples, including 41 paired samples, using the Infinium Human Methylation450 BeadChip. Regression analyses of paired tumour and adjacent benign samples identified 2,386 significant dmCpGs (Bonferroni p < 0.01; delta-ß ≥ 40%), with LASSO regression selecting 16 dmCpGs that distinguished tumour samples in the full Australian diagnostic dataset (AUC = 0.99). Results were validated in independent North American (npaired = 19; AUC = 0.87) and The Cancer Genome Atlas (TCGA; npaired = 50; AUC = 0.94) RP datasets. Two of the 16 dmCpGs were in genes that were significantly down-regulated in Australian tumour samples (Bonferroni p < 0.01; GSTM2 and PRKCB). Ten additional dmCpGs distinguished low (n = 34) and high Gleason (n = 88) score tumours in the diagnostic Australian dataset (AUC = 0.95), but these performed poorly when applied to the RP datasets (North American: AUC = 0.66; TCGA: AUC = 0.62). The DNA methylation marks identified here could augment and improve current diagnostic tests and/or form the basis of future prognostic tests.


Sujet(s)
Ilots CpG , Méthylation de l'ADN , Tumeurs de la prostate , Humains , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/diagnostic , Tumeurs de la prostate/chirurgie , Tumeurs de la prostate/métabolisme , Mâle , Ilots CpG/génétique , Sujet âgé , Adulte d'âge moyen , Prostatectomie , Marqueurs biologiques tumoraux/génétique , Régulation de l'expression des gènes tumoraux , Australie , Pronostic
2.
Nat Commun ; 15(1): 6626, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39103353

RÉSUMÉ

N-Myc is a key driver of neuroblastoma and neuroendocrine prostate cancer (NEPC). One potential way to circumvent the challenge of undruggable N-Myc is to target the protein homeostasis (proteostasis) system that maintains N-Myc levels. Here, we identify heat shock protein 70 (HSP70) as a top partner of N-Myc, which binds a conserved "SELILKR" motif and prevents the access of E3 ubiquitin ligase, STIP1 homology and U-box containing protein 1 (STUB1), possibly through steric hindrance. When HSP70's dwell time on N-Myc is increased by treatment with the HSP70 allosteric inhibitor, STUB1 is in close proximity with N-Myc and becomes functional to promote N-Myc ubiquitination on the K416 and K419 sites and forms polyubiquitination chains linked by the K11 and K63 sites. Notably, HSP70 inhibition significantly suppressed NEPC tumor growth, increased the efficacy of aurora kinase A (AURKA) inhibitors, and limited the expression of neuroendocrine-related pathways.


Sujet(s)
Protéines du choc thermique HSP70 , Tumeurs de la prostate , Homéostasie protéique , Ubiquitin-protein ligases , Ubiquitination , Mâle , Humains , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Protéines du choc thermique HSP70/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitination/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Animaux , Aurora kinase A/métabolisme , Aurora kinase A/génétique , Aurora kinase A/antagonistes et inhibiteurs , Protéine du proto-oncogène N-Myc/métabolisme , Protéine du proto-oncogène N-Myc/génétique , Souris , Carcinome neuroendocrine/métabolisme , Carcinome neuroendocrine/génétique , Carcinome neuroendocrine/traitement médicamenteux , Carcinome neuroendocrine/anatomopathologie , Tumeurs neuroendocrines/métabolisme , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie
3.
Sci Rep ; 14(1): 18036, 2024 08 04.
Article de Anglais | MEDLINE | ID: mdl-39098988

RÉSUMÉ

Prostate cancer, one of the most prevalent malignancies among men worldwide, is intricately linked with androgen signaling, a key driver of its pathogenesis and progression. Understanding the diverse expression patterns of androgen-responsive genes holds paramount importance in unraveling the biological intricacies of this disease and prognosticating patient outcomes. In this study, utilizing consensus clustering analysis based on the expression profiles of androgen-responsive genes, prostate cancer patients from the TCGA database were stratified into two distinct subtypes, denoted as C1 and C2. Notably, the C1 subtype demonstrates a significant upregulation of certain genes, such as CGA and HSD17B12, along with a shorter progression-free survival duration, indicating a potentially unfavorable prognosis. Further analyses elucidated the immune infiltration disparities, mutation landscapes, and gene functional pathways characteristic of each subtype. Through integrated bioinformatics approaches and machine learning techniques, key genes such as BIRC5, CENPA, and MMP11 were identified as potential therapeutic targets, providing novel insights into tailored treatment strategies. Additionally, single-cell transcriptome analysis shed light on the heterogeneous expression patterns of these genes across different cell types within the tumor microenvironment. Furthermore, virtual screening identified candidate drugs targeting the BIRC5 receptor, offering promising avenues for drug development. Collectively, these findings deepen our understanding of prostate cancer biology, paving the way for personalized therapeutic interventions and advancing the quest for more effective treatments in prostate cancer management.


Sujet(s)
Androgènes , Régulation de l'expression des gènes tumoraux , Tumeurs de la prostate , Microenvironnement tumoral , Humains , Mâle , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/génétique , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Androgènes/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Analyse de profil d'expression de gènes , Pronostic , Transcriptome , Biologie informatique/méthodes
4.
Proc Natl Acad Sci U S A ; 121(33): e2402903121, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39102549

RÉSUMÉ

Immune checkpoint therapy has limited efficacy for patients with bone-metastatic castration-resistant prostate cancer (bmCRPC). To improve immunotherapy for bmCRPC, we aimed to identify the mechanism of bmCRPC-induced changes in the immune microenvironment. Among bmCRPC patients, higher levels of a 32-gene M2-like macrophage signature in bone metastasis samples correlated with shorter overall survival. Immunohistochemistry showed that CD206-positive (CD206+) macrophages were enriched in bmCRPC bone biopsy specimens compared with primary tumors or lymph node metastases. In preclinical osteogenic prostate cancer (Pca) xenograft models, CD206+ macrophages were recruited to areas with tumor-induced bone. RNA sequencing (RNAseq) analysis showed higher expression of an M2-like gene signature, with activated canonical and noncanonical Wnt pathways, in tumor-associated macrophages isolated from osteogenic tumors (bone-TAMs) than in TAMs isolated from nonosteogenic tumors (ctrl-TAMs). Mechanistic studies showed that endothelial cells (ECs) that had undergone EC-to-osteoblast (EC-to-OSB) transition, the precursors of tumor-induced OSBs, produced paracrine factors, including Wnts, CXCL14, and lysyl oxidase, which induced M2 polarization and recruited M2-like TAMs to the bone-tumor microenvironment (bone-TME). Bone-TAMs suppressed CD8+ T cells' proliferation and cytolytic activity, and these effects were partially reversed by treating bone-TAMs with Wnt inhibitors. Genetic or pharmacological inhibition of Pca-induced EC-to-OSB transition reduced the levels of M2-like macrophages in osteogenic tumors. Our study demonstrates that Pca-induced EC-to-OSB transition drives immunosuppression in the bone-TME, suggesting that therapies that reduce Pca-induced bone formation may improve immunotherapeutic outcomes for bmCRPC.


Sujet(s)
Tumeurs osseuses , Cellules endothéliales , Macrophages , Ostéoblastes , Microenvironnement tumoral , Voie de signalisation Wnt , Mâle , Microenvironnement tumoral/immunologie , Humains , Tumeurs osseuses/immunologie , Tumeurs osseuses/secondaire , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Animaux , Souris , Macrophages/métabolisme , Macrophages/immunologie , Cellules endothéliales/métabolisme , Cellules endothéliales/immunologie , Ostéoblastes/métabolisme , Ostéoblastes/immunologie , Tumeurs prostatiques résistantes à la castration/immunologie , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/métabolisme , Lignée cellulaire tumorale , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/métabolisme , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie
5.
Nat Commun ; 15(1): 6779, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39117665

RÉSUMÉ

Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase and emerging therapeutic target that is overexpressed in most castration-resistant prostate cancers and implicated as a driver of disease progression and resistance to hormonal therapies. Here we define the lineage-specific action and differential activity of EZH2 in both prostate adenocarcinoma and neuroendocrine prostate cancer (NEPC) subtypes of advanced prostate cancer to better understand the role of EZH2 in modulating differentiation, lineage plasticity, and to identify mediators of response and resistance to EZH2 inhibitor therapy. Mechanistically, EZH2 modulates bivalent genes that results in upregulation of NEPC-associated transcriptional drivers (e.g., ASCL1) and neuronal gene programs in NEPC, and leads to forward differentiation after targeting EZH2 in NEPC. Subtype-specific downstream effects of EZH2 inhibition on cell cycle genes support the potential rationale for co-targeting cyclin/CDK to overcome resistance to EZH2 inhibition.


Sujet(s)
Protéine-2 homologue de l'activateur de Zeste , Régulation de l'expression des gènes tumoraux , Tumeurs de la prostate , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Mâle , Humains , Lignée cellulaire tumorale , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Animaux , Adénocarcinome/génétique , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Différenciation cellulaire , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Souris , Lignage cellulaire
6.
BMC Cancer ; 24(1): 1004, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138404

RÉSUMÉ

BACKGROUND: Metastatic prostate cancer is a leading cause of cancer-related morbidity and mortality in men, yet the underlying molecular mechanisms are poorly understood. Plexins are transmembrane receptors for semaphorins with divergent roles in many forms of cancer. We recently found that a single clinically relevant specific amino acid change (Proline1597Leucine, (P1597L)), found in metastatic deposits of prostate cancer patients, converts PlexinB1 from a metastasis suppressor to a gene that drives prostate cancer metastasis in vivo. However, the mechanism by which PlexinB1(P1597L) promotes metastasis is not known. METHODS: Pull down assays using GST-RalGDS or -GSTRaf1-RBD were used to reveal the effect of mutant or wild-type PlexinB1 expression on Rap and Ras activity respectively. Protein-protein interactions were assessed in GST pulldown assays, Akt/ERK phosphorylation by immunoblotting and protein stability by treatment with cycloheximide. Rho/ROCK activity was monitored by measuring MLC2 phosphorylation and actin stress fiber formation. PlexinB1 function was measured using cell-collapse assays. RESULTS: We show here that the single clinically relevant P1597L amino acid change converts PlexinB1 from a repressor of Ras to a Ras activator. The PlexinB1(P1597L) mutation inhibits the RapGAP activity of PlexinB1, promoting a significant increase in Ras activity. The P1597L mutation also blocks PlexinB1-mediated reduction in Rho/ROCK activity, restraining the decrease in MLC2 phosphorylation and actin stress fiber formation induced by overexpression of wild-type PlexinB1. PlexinB1(P1597L) has little effect on the interaction of PlexinB1 with small GTPases or receptor tyrosine kinases and does not inhibit PlexinB1-stimulated Akt or ERK phosphorylation. These results indicate that the mutation affects Rho signalling via the Rap/Ras pathway. The PlexinB1(P1597L) mutation inhibits morphological cell collapse induced by wild-type PlexinB1 expression, suggesting that the mutation induces a loss of an inhibitory tumour suppressor function. CONCLUSION: These results suggest that the clinically relevant P1597L mutation in PlexinB1 may transform PlexinB1 from a suppressor to a driver of metastasis in mouse models of prostate cancer by reducing the RapGAP activity of PlexinB1, leading to Ras activation. These findings highlight the PlexinB1-Rap-Ras pathway for therapeutic intervention in prostate cancer.


Sujet(s)
Protéines de tissu nerveux , Tumeurs de la prostate , Récepteurs de surface cellulaire , Humains , Mâle , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Récepteurs de surface cellulaire/génétique , Récepteurs de surface cellulaire/métabolisme , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Lignée cellulaire tumorale , Mutation , Protéines G ras/génétique , Protéines G ras/métabolisme , Métastase tumorale , Animaux , Phosphorylation , Transduction du signal , Souris , Sémaphorines/métabolisme , Sémaphorines/génétique , Protéines G rho/métabolisme , Protéines G rho/génétique
7.
Radiology ; 312(2): e232544, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39136560

RÉSUMÉ

Background Intravenous prostate-specific membrane antigen (PSMA)-targeted radioligand therapy improves survival in men with metastatic castration-resistant prostate cancer. Yet, the impact of selective prostatic arterial administration on primary tumor uptake is unclear. Purpose To compare gallium 68 (68Ga)-PSMA-11 uptake using dynamic PET/CT in prostatic tumoral volumes of interest (VOIs) during intravenous and selective prostatic arterial infusions for individuals with untreated, high-risk prostate cancer. Materials and Methods In this prospective, intraindividual comparative study conducted at an academic medical center, five men aged 58, 61, 64, 66, and 68 years with treatment-naive prostate cancer were enrolled between January 2022 and February 2023 and underwent two dynamic 68Ga-PSMA-11 PET/CT examinations 1 week apart. During the first examination, the radiotracer was administered intravenously. During the second administration, the radiotracer was delivered into either the right or left prostatic artery through an angiographically placed microcatheter. The primary outcome was maximum standardized uptake value (SUVmax) in prostatic tumoral VOIs. The secondary outcomes included mean SUV (SUVmean) in prostatic tumoral VOIs and area under the SUVmean curves (AUC). Longitudinal mixed-effects models were used to compare dynamic SUVmax and SUVmean time-activity curves (TACs), and paired t tests were used for the remaining data. Results The mean SUVmax within tumoral VOIs was 14 (range, 3-43) for venous sessions and 938 (range, 460-1436) for arterial sessions (P = .008). The SUVmean within VOIs was greater during arterial sessions (P < .001) overall and 46-fold and 19-fold greater at peak uptake and final time points, respectively. The mean AUC was greater on arterial TACs than on venous TACs at 14600 SUV × min (range, 8353-20025 SUV × min) and 240 SUV × min (range, 69-622 SUV × min), respectively (P = .002). Conclusion Selective prostatic arterial infusion resulted in greater 68Ga-PSMA-11 tumoral SUV than intravenous infusion. Further study of local-regional, intra-arterial delivery of a PSMA-targeted theranostic agent is warranted in high-risk prostate cancer. ClinicalTrials.gov identifier: NCT04976257 © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Civelek in this issue.


Sujet(s)
Radio-isotopes du gallium , Tomographie par émission de positons couplée à la tomodensitométrie , Tumeurs de la prostate , Humains , Mâle , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Sujet âgé , Études prospectives , Tumeurs de la prostate/imagerie diagnostique , Tumeurs de la prostate/métabolisme , Adulte d'âge moyen , Radio-isotopes du gallium/pharmacocinétique , Prostate/imagerie diagnostique , Prostate/vascularisation , Isotopes du gallium , Radiopharmaceutiques/pharmacocinétique , Perfusions veineuses , Adénocarcinome/imagerie diagnostique , Adénocarcinome/métabolisme
8.
Eur J Med Res ; 29(1): 411, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39118157

RÉSUMÉ

BACKGROUND: Long noncoding RNAs (lncRNAs) and N6-methyladenosine (m6A) modification of RNA play pivotal roles in tumorigenesis and cancer progression. However, knowledge regarding the expression patterns of m6A-related lncRNAs and their corresponding m6A regulators in prostate cancer (PCa) is limited. This study aimed to delineate the landscape of m6A-related lncRNAs, develop a predictive model, and identify the critical m6A regulators of prognostic lncRNAs in PCa. METHODS: Clinical and transcriptome data of PCa patients were downloaded from The Cancer Genome Atlas (TCGA) database. Prognostic m6A-related lncRNAs were subsequently identified through Pearson correlation and univariate Cox regression analyses. The prognostic lncRNAs were clustered into two groups by consensus clustering analysis, and a risk signature model was constructed using least absolute shrinkage and selection operator (LASSO) regression analysis of the lncRNAs. This model was evaluated using survival, clinicopathological, and immunological analyses. Furthermore, based on the constructed lncRNA-m6A regulatory network and RT-qPCR results, RBM15 was identified as a critical regulator of m6A-related lncRNAs. The biological roles of RBM15 in PCa were explored through bioinformatics analysis and biological experiments. RESULTS: Thirty-four prognostic m6A-related lncRNAs were identified and categorized into two clusters with different expression patterns and survival outcomes in PCa patients. Seven m6A lncRNAs (AC105345.1, AL354989.1, AC138028.4, AC022211.1, AC020558.2, AC004076.2, and LINC02666) were selected to construct a risk signature with robust predictive ability for overall survival and were correlated with clinicopathological characteristics and the immune microenvironment of PCa patients. Among them, LINC02666 and AC022211.1 were regulated by RBM15. In addition, RBM15 expression correlated with PCa progression, survival, and the immune response. Patients with elevated RBM15 expression were more susceptible to the drug AMG-232. Moreover, silencing RBM15 decreased the viability of PCa cells and promoted apoptosis. CONCLUSION: RBM15 is involved in the regulation of prognostic lncRNAs in the risk signature and has a robust predictive ability for PCa, making it a promising biomarker in PCa.


Sujet(s)
Adénosine , Marqueurs biologiques tumoraux , Tumeurs de la prostate , ARN long non codant , Protéines de liaison à l'ARN , Humains , Mâle , ARN long non codant/génétique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Pronostic , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Adénosine/analogues et dérivés , Adénosine/métabolisme , Adénosine/génétique , Régulation de l'expression des gènes tumoraux
9.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39125581

RÉSUMÉ

There is a significant unmet need for clinical reflex tests that increase the specificity of prostate-specific antigen blood testing, the longstanding but imperfect tool for prostate cancer diagnosis. Towards this endpoint, we present the results from a discovery study that identifies new prostate-specific antigen reflex markers in a large-scale patient serum cohort using differentiating technologies for deep proteomic interrogation. We detect known prostate cancer blood markers as well as novel candidates. Through bioinformatic pathway enrichment and network analysis, we reveal associations of differentially abundant proteins with cytoskeletal, metabolic, and ribosomal activities, all of which have been previously associated with prostate cancer progression. Additionally, optimized machine learning classifier analysis reveals proteomic signatures capable of detecting the disease prior to biopsy, performing on par with an accepted clinical risk calculator benchmark.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs de la prostate , Protéomique , Humains , Mâle , Tumeurs de la prostate/diagnostic , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/sang , Marqueurs biologiques tumoraux/sang , Protéomique/méthodes , Spectrométrie de mobilité ionique/méthodes , Antigène spécifique de la prostate/sang , Sujet âgé , Apprentissage machine , Adulte d'âge moyen
10.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39125671

RÉSUMÉ

Late endosomal/lysosomal adaptor, MAPK and mTOR, or LAMTOR, is a scaffold protein complex that senses nutrients and integrates growth factor signaling. The role of LAMTOR4 in tumorigenesis is still unknown. However, there is a considerable possibility that LAMTOR4 is directly involved in tumor cell proliferation and metastasis. In the current study, we investigated the protein expression of LAMTOR4 in a cohort of 314 men who were undergoing transurethral resection of prostate (TURP) consisting of incidental, advanced and castration-resistant cases. We also correlated the data with ERG and PTEN genomic status and clinicopathological features including Gleason score and patients' outcome. Additionally, we performed in vitro experiments utilizing knockdown of LAMTOR4 in prostate cell lines, and we performed mRNA expression assessment using TCGA prostate adenocarcinoma (TCGA-PRAD) to explore the potential differentially expressed genes and pathways associated with LAMTOR4 overexpression in PCa patients. Our data indicate that high LAMTOR4 protein expression was significantly associated with poor overall survival (OS) (HR: 1.44, CI: 1.01-2.05, p = 0.047) and unfavorable cause-specific survival (CSS) (HR: 1.71, CI: 1.06-2.77, p = 0.028). Additionally, when high LAMTOR4 expression was combined with PTEN-negative cases (score 0), we found significantly poorer OS (HR: 2.22, CI: 1.37-3.59, p = 0.001) and CSS (HR: 3.46, CI: 1.86-6.46, p < 0.0001). Furthermore, ERG-positive cases with high LAMTOR4 exhibited lower OS (HR: 1.98, CI: 1.18-3.31, p = 0.01) and CSS (HR: 2.54, CI: 1.32-4.87, p = 0.005). In vitro assessment showed that knockdown of LAMTOR4 decreases PCa cell proliferation, migration, and invasion. Our data further showed that knockdown of LAMTOR4 in the LNCaP cell line significantly dysregulated the ß catenin/mTOR pathway and tumorigenesis associated pathways. Inhibiting components of the mTOR pathway, including LAMTOR4, might offer a strategy to inhibit tumor progression and metastasis in prostate cancer.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs de la prostate , Humains , Mâle , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Lignée cellulaire tumorale , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Sujet âgé , Invasion tumorale , Techniques de knock-down de gènes , Adulte d'âge moyen , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Pronostic
11.
Cell Death Dis ; 15(8): 559, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097593

RÉSUMÉ

Sharply increased reactive oxygen species (ROS) are thought to induce oxidative stress, damage cell structure and cause cell death; however, its role in prostate cancer remains unclear. Enzalutamide is a widely used anti-prostate cancer drug that antagonizes androgen binding with its receptor. Further exploration of the mechanism and potential application strategies of enzalutamide is crucial for the treatment of prostate cancer. Here, we confirmed PEX10 can be induced by ROS activators while reduce ROS level in prostate cancer cells, which weakened the anti-tumor effect of ROS activators. The androgen receptor (AR) can promote the expression of PEX10 by acting as an enhancer in cooperation with FOXA1. The anti-tumor drug enzalutamide inhibits PEX10 by inhibiting the function of AR, and synergize with ROS activators ML210 or RSL3 to produce a stronger anti-tumor effect, thereby sensitizing cells to ROS activators. This study reveals a previously unrecognized function of enzalutamide and AR by regulating PEX10 and suggests a new strategy of enzalutamide application in prostate cancer treatment.


Sujet(s)
Benzamides , Nitriles , 3-Phényl-2-thiohydantoïne , Tumeurs de la prostate , Espèces réactives de l'oxygène , Humains , 3-Phényl-2-thiohydantoïne/pharmacologie , 3-Phényl-2-thiohydantoïne/analogues et dérivés , Mâle , Benzamides/pharmacologie , Nitriles/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Lignée cellulaire tumorale , Récepteurs aux androgènes/métabolisme , Facteur nucléaire hépatocytaire HNF-3 alpha/métabolisme , Animaux , Souris , Protéines membranaires/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris nude
12.
PeerJ ; 12: e17823, 2024.
Article de Anglais | MEDLINE | ID: mdl-39099654

RÉSUMÉ

Background: Metabolic syndrome (MetS) has been shown to have a negative impact on prostate cancer (PCa). However, there is limited research on the effects of MetS on testosterone levels in metastatic prostate cancer (mPCa). Objective: This study aims to investigate the influence of MetS, its individual components, and composite metabolic score on the prognosis of mPCa patients, as well as the impact on testosterone levels. Additionally, it seeks to identify MetS-related risk factors that could impact the time of decline in testosterone levels among mPCa patients. Methods: A total of 212 patients with mPCa were included in the study. The study included 94 patients in the Non-MetS group and 118 patients in the combined MetS group. To analyze the relationship between MetS and testosterone levels in patients with mPCa. Additionally, the study aimed to identify independent risk factors that affect the time for testosterone levels decline through multifactor logistic regression analysis. Survival curves were plotted by the Kaplan-Meier method. Results: Compared to the Non-MetS group, the combined MetS group had a higher proportion of patients with high tumor burden, T stage ≥ 4, and Gleason score ≥ 8 points (P < 0.05). Patients in the combined MetS group also had higher lowest testosterone values and it took longer for their testosterone to reach the lowest level (P < 0.05). The median progression-free survival (PFS) time for patients in the Non-MetS group was 21 months, while for those in the combined MetS group it was 18 months (P = 0.001). Additionally, the median overall survival (OS) time for the Non-MetS group was 62 months, whereas for the combined MetS group it was 38 months (P < 0.001). The median PFS for patients with a composite metabolic score of 0-2 points was 21 months, 3 points was 18 months, and 4-5 points was 15 months (P = 0.002). The median OS was 62 months, 42 months, and 29 months respectively (P < 0.001). MetS was found to be an independent risk factor for testosterone levels falling to the lowest value for more than 6 months. The risk of testosterone levels falling to the lowest value for more than 6 months in patients with MetS was 2.157 times higher than that of patients with Non-MetS group (P = 0.031). Patients with hyperglycemia had a significantly higher lowest values of testosterone (P = 0.015). Additionally, patients with a BMI ≥ 25 kg/m2 exhibited lower initial testosterone levels (P = 0.007). Furthermore, patients with TG ≥ 1.7 mmol/L experienced a longer time for testosterone levels to drop to the nadir (P = 0.023). The lowest value of testosterone in the group with a composite metabolic score of 3 or 4-5 was higher than that in the 0-2 group, and the time required for testosterone levels to decrease to the lowest value was also longer (P < 0.05). Conclusion: When monitoring testosterone levels in mPCa patients, it is important to consider the impact of MetS and its components, and make timely adjustments to individualized treatment strategies.


Sujet(s)
Syndrome métabolique X , Tumeurs de la prostate , Testostérone , Humains , Mâle , Syndrome métabolique X/sang , Testostérone/sang , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/sang , Tumeurs de la prostate/mortalité , Tumeurs de la prostate/métabolisme , Études rétrospectives , Sujet âgé , Adulte d'âge moyen , Facteurs de risque , Pronostic , Grading des tumeurs , Métastase tumorale
13.
Zhonghua Bing Li Xue Za Zhi ; 53(8): 803-808, 2024 Aug 08.
Article de Chinois | MEDLINE | ID: mdl-39103261

RÉSUMÉ

Objective: To study the clinicopathological features, immunohistochemical phenotypes, molecular changes, differential diagnosis and prognosis of isolated intraductal carcinoma of the prostate (iIDC-P). Methods: Three iIDC-P cases were collected retrospectively from 2016 to 2022 at Ningbo Clinical Pathology Diagnosis Center, Ningbo, China. The clinicopathologic features and immunophenotypic profiles were studied using light microscopy and immunohistochemistry. A targeted next-generation sequencing panel was used to analyze cancer-associated mutations. Follow-up and literature review were also performed. Results: The patients' ages were 61, 67 and 77 years, and their preoperative prostate specific antigen (PSA) levels were 7.99, 7.99 and 4.86 µg/L, respectively. Case 1 and 2 were diagnosed on needle biopsy and radical prostatectomy (RP) specimens, and case 3 was diagnosed on a specimen of transurethral resection of the prostate (TURP). The RP specimen was entirely submitted for histologic examination. In the case 1, iIDC-P was found in one tissue core (involving two ducts) in the biopsy specimen, and in 6 sections (diameter, 0.3-1.1 cm) from the radical prostatectomy specimen, and one section had separate foci of low-grade acinar adenocarcinoma (diameter, 0.05 cm). In the case 2, 6 tissue sections from the biopsy specimens showed iIDC-P, and 13 sections from RP specimen showed iIDC-P (diameter, 0.5-1.6 cm), and the other 3 sections had separate low grade acinar adenocarcinoma (diameter, 0.6 cm). In the case 3, 5 tissue blocks from the TURP specimen showed iIDC-P. The case 1 and 2 showed solid architecture with expansile proliferation of neoplastic cells in native ducts and acini. The case 3 showed dense or loose cribriform pattern, with marked cytological atypia, and frequent mitotic figures. Comedonecrosis was found in solid or dense cribriform glands in the case 2. Immunohistochemically, surrounding basal cells were highlighted using high-molecular-weight cytokeratin (34ßE12 and CK5/6) and p63, while P504s was positive in the tumor cells. The tumor cells were also positive for AR and prostate markers (NKX3.1, PSA and PSAP), and negative for GATA3. The iIDC-P and acinar adenocarcinoma both showed weak PTEN expression and no ERG (nuclear) expression. In case 2 and 3, targeted sequencing revealed activated oncogenic driver mutations in MAPK and PI3K pathway genes (KRAS, MTOR and PTEN). In addition, pathogenic mutation in TP53 and FOXA1 mutation were found in the case 2 and 3, respectively. No case demonstrated TMPRSS2::ERG translocation. All cases were microsatellite stable and had lower tumor mutation burdens (range, 2.1-3.1 muts/Mb). The patients showed no biochemical recurrence or metastasis after follow-up of 16-91 months. Conclusions: iIDC-P is a special type of intraductal carcinoma of the prostate and differs from intraductal carcinoma within high-grade prostate cancer. iIDC-P has unique molecular characteristics and may represent as a molecularly unique in situ tumor of prostate cancer.


Sujet(s)
Antigène spécifique de la prostate , Tumeurs de la prostate , Humains , Mâle , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/chirurgie , Sujet âgé , Adulte d'âge moyen , Études rétrospectives , Antigène spécifique de la prostate/métabolisme , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Diagnostic différentiel , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Carcinome intracanalaire non infiltrant/anatomopathologie , Carcinome intracanalaire non infiltrant/génétique , Carcinome intracanalaire non infiltrant/métabolisme , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Carcinome canalaire/anatomopathologie , Carcinome canalaire/génétique , Carcinome canalaire/métabolisme , Carcinome canalaire/chirurgie , Résection transuréthrale de prostate , Racémases et épimérases/métabolisme , Racémases et épimérases/génétique , Séquençage nucléotidique à haut débit , Pronostic , Kératines , Protéines membranaires
14.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 85-91, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39097891

RÉSUMÉ

Prostate cancer (PCa) belongs to a prevailing neoplasm globally. Circular RNAs (circRNAs) are critical regulators in various tumors, but the role of circRNAs in PCa is obscure. In this research, a circRNA derived from the TADA2A gene (hsa_circ_0006220) was high-expressed in PCa tissues along with cell lines. Elevated Circ-0006220 expression was also related to PCa poor prognosis. Besides, circ-0006220 accelerated PCa cells malignant behaviors in vitro; it also promoted PCa tumor growth together with metastasis in vivo. Moreover, circ-0006220 competed with the Cell Division Cycle Associated 7 (CDCA7) for binding to miR-520f-3p. Circ-0006220 sponged miR-520f-3p to regulate CDCA7 expression, thereby promoting PCa cell proliferation, migration, invasion, along with metastasis. All above data suggested that circ-0006220 may be a worthy target for PCa therapeutics.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , microARN , Tumeurs de la prostate , ARN circulaire , Humains , Mâle , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , microARN/génétique , microARN/métabolisme , Invasion tumorale/génétique , Métastase tumorale , Pronostic , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , ARN circulaire/génétique , ARN circulaire/métabolisme
15.
J Cell Mol Med ; 28(15): e18511, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39098992

RÉSUMÉ

The aetiology of bone metastasis in prostate cancer (PCa) remains unclear. This study aims to identify hub genes involved in this process. We utilized machine learning, GO, KEGG, GSEA, Single-cell analysis, ROC methods to identify hub genes for bone metastasis in PCa using the TCGA and GEO databases. Potential drugs targeting these genes were identified. We validated these results using 16 specimens from patients with PCa and analysed the relationship between the hub genes and clinical features. The impact of APOC1 on PCa was assessed through in vitro experiments. Seven hub genes with AUC values of 0.727-0.926 were identified. APOC1, CFH, NUSAP1 and LGALS1 were highly expressed in bone metastasis tissues, while NR4A2, ADRB2 and ZNF331 exhibited an opposite trend. Immunohistochemistry further confirmed these results. The oxidative phosphorylation pathway was significantly enriched by the identified genes. Aflatoxin B1, benzo(a)pyrene, cyclosporine were identified as potential drugs. APOC1 expression was correlated with clinical features of PCa metastasis. Silencing APOC1 significantly inhibited PCa cell proliferation, clonality, and migration in vitro. This study identified 7 hub genes that potentially facilitate bone metastasis in PCa through mitochondrial metabolic reprogramming. APOC1 emerged as a promising therapeutic target and prognostic marker for PCa with bone metastasis.


Sujet(s)
Tumeurs osseuses , Prolifération cellulaire , Biologie informatique , Régulation de l'expression des gènes tumoraux , Tumeurs de la prostate , Humains , Tumeurs osseuses/secondaire , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Mâle , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Biologie informatique/méthodes , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Mouvement cellulaire/génétique , Analyse de profil d'expression de gènes , Réseaux de régulation génique , Pronostic
16.
J Extracell Vesicles ; 13(8): e12482, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39105261

RÉSUMÉ

It is known that small extracellular vesicles (sEVs) are released from cancer cells and contribute to cancer progression via crosstalk with recipient cells. We have previously reported that sEVs expressing the αVß3 integrin, a protein upregulated in aggressive neuroendocrine prostate cancer (NEPrCa), contribute to neuroendocrine differentiation (NED) in recipient cells. Here, we examine the impact of αVß3 expression on sEV protein content, density and function. sEVs used in this study were isolated by iodixanol density gradients and characterized by nanoparticle tracking analysis, immunoblotting and single vesicle analysis. Our proteomic profile of sEVs containing αVß3 shows downregulation of typical effectors involved in apoptosis and necrosis and an upregulation of tumour cell survival factors compared to control sEVs. We also show that the expression of αVß3 in sEVs causes a distinct reposition of EV markers (Alix, CD81, CD9) to a low-density sEV subpopulation. This low-density reposition is independent of extracellular matrix (ECM) protein interactions with sEVs. This sEV subset contains αVß3 and an αVß3 downstream effector, NgR2, a novel marker for NEPrCa. We show that sEVs containing αVß3 are loaded with higher amounts of NgR2 as compared to sEVs that do not express αVß3. Mechanistically, we demonstrate that sEVs containing NgR2 do not affect the sEV marker profile, but when injected in vivo intratumorally, they promote tumour growth and induce NED. We show that sEVs expressing NgR2 increase the activation of focal adhesion kinase (FAK), a known promoter of cancer cell proliferation, in recipient cells. We also show that NgR2 mimics the effect of sEVs containing αVß3 since it displays increased growth of NgR2 transfectants in vivo, as compared to control cells. Overall, our results describe the changes that occur in cargo, density and functions of cancer cell-derived sEVs containing the αVß3 integrin and its effector, NgR2, without affecting the sEV tetraspanin profiles.


Sujet(s)
Vésicules extracellulaires , Intégrine alphaVbêta3 , Tumeurs de la prostate , Mâle , Intégrine alphaVbêta3/métabolisme , Humains , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Vésicules extracellulaires/métabolisme , Animaux , Lignée cellulaire tumorale , Souris , Carcinogenèse/métabolisme
17.
Mol Biol Rep ; 51(1): 898, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39115711

RÉSUMÉ

BACKGROUND: The nuclear envelope (NE), which is composed of the outer and inner nuclear membranes, the nuclear pore complex and the nuclear lamina, regulates a plethora of cellular processes, including those that restrict cancer development (genomic stability, cell cycle regulation, and cell migration). Thus, impaired NE is functionally related to tumorigenesis, and monitoring of NE alterations is used to diagnose cancer. However, the chronology of NE changes occurring during cancer evolution and the connection between them remained to be precisely defined, due to the lack of appropriate cell models. METHODS: The expression and subcellular localization of NE proteins (lamins A/C and B1 and the inner nuclear membrane proteins emerin and ß-dystroglycan [ß-DG]) during prostate cancer progression were analyzed, using confocal microscopy and western blot assays, and a prostate cancer cell system comprising RWPE-1 epithelial prostate cells and several prostate cancer cell lines with different invasiveness. RESULTS: Deformed nuclei and the mislocalization and low expression of lamin A/C, lamin B1, and emerin became more prominent as the invasiveness of the prostate cancer lines increased. Suppression of lamin A/C expression was an early event during prostate cancer evolution, while a more extensive deregulation of NE proteins, including ß-DG, occurred in metastatic prostate cells. CONCLUSIONS: The RWPE-1 cell line-based system was found to be suitable for the correlation of NE impairment with prostate cancer invasiveness and determination of the chronology of NE alterations during prostate carcinogenesis. Further study of this cell system would help to identify biomarkers for prostate cancer prognosis and diagnosis.


Sujet(s)
Lamine A , Lamine B , Protéines membranaires , Enveloppe nucléaire , Protéines nucléaires , Tumeurs de la prostate , Mâle , Humains , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Enveloppe nucléaire/métabolisme , Lignée cellulaire tumorale , Protéines membranaires/métabolisme , Lamine B/métabolisme , Lamine A/métabolisme , Lamine A/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Dystroglycanes/métabolisme , Régulation de l'expression des gènes tumoraux , Noyau de la cellule/métabolisme
18.
BMC Med Genomics ; 17(1): 210, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138470

RÉSUMÉ

BACKGROUND: Prostate cancer (PCa) stands as the second most prevalent malignancy impacting male health, and the disease's evolutionary course presents formidable challenges in the context of patient treatment and prognostic management. Charged multivesicular body protein 4 C (CHMP4C) participates in the development of several cancers by regulating cell cycle functions. However, the role of CHMP4C in prostate cancer remains unclear. METHODS: In terms of bioinformatics, multiple PCa datasets were employed to scrutinize the expression of CHMP4C. Survival analysis coupled with a nomogram approach was employed to probe into the prognostic significance of CHMP4C. Gene set enrichment analysis (GSEA) was conducted to interrogate the functional implications of CHMP4C. In terms of cellular experimentation, the verification of RNA and protein expression levels was executed through the utilization of qRT-PCR and Western blotting. Upon the establishment of a cell line featuring stable CHMP4C knockdown, a battery of assays, including Cell Counting Kit-8 (CCK-8), wound healing, Transwell, and flow cytometry, were employed to discern the impact of CHMP4C on the proliferation, migration, invasion, and cell cycle function of PCa cells. RESULTS: The expression of CHMP4C exhibited upregulation in both PCa cells and tissues, and patients demonstrating elevated CHMP4C expression levels experienced a notably inferior prognosis. The nomogram, constructed using CHMP4C along with clinicopathological features, demonstrated a commendable capacity for prognostic prediction. CHMP4C knockdown significantly inhibited the proliferation, migration, and invasion of PCa cells (LNcaP and PC3). CHMP4C could impact the advancement of the PCa cell cycle, and its expression might be regulated by berberine. Divergent CHMP4C expression among PCa patients could induce alterations in immune cell infiltration and gene mutation frequency. CONCLUSIONS: Our findings suggest that CHMP4C might be a prognostic biomarker in PCa, potentially offering novel perspectives for the advancement of precision therapy for PCa.


Sujet(s)
Cycle cellulaire , Prolifération cellulaire , Tumeurs de la prostate , Humains , Mâle , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Pronostic , Cycle cellulaire/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Mouvement cellulaire/génétique , Complexes de tri endosomique requis pour le transport/génétique , Complexes de tri endosomique requis pour le transport/métabolisme , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Nomogrammes
19.
Biol Pharm Bull ; 47(8): 1437-1446, 2024.
Article de Anglais | MEDLINE | ID: mdl-39143009

RÉSUMÉ

Bisdemethoxycurcumin (BDMC) is one of major forms of curcuminoids found in the rhizomes of turmeric. Docetaxel (DTX) is the standard of care for men diagnosed with androgen-independent prostate cancers. Here we report for the first time that BDMC could reinforce the effect of DTX against prostate cancer in vitro and in vivo. In vitro study, PC3 and LNCaP cells were cultured and treated with BDMC and DTX alone or in combination. The effects on cell viability were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Apoptosis was assessed by annexin V/propidium iodide (PI) staining, while cell cycle was assessed by PI staining. Bax, Bcl-2, caspase, poly(ADP-ribose)polymerase (PARP), cyclin B1 and CDK1 expression were assayed by Western blot. We found that a combination treatment of BDMC (10 µM) with DTX (10 nM) was more effective in the inhibition of PC3 and LNCaP cell growth and induction of apoptosis as well as G2/M arrest, which is accompanied with the significant inhibition of Bcl-2, cyclin B1, CDK1 expression and significant increase of Bax, cleaved caspase-9, cleaved caspase-3 and cleaved PARP, than those by treatment of BDMC or DTX alone. Moreover, in vivo evaluation further demonstrated the superior anticancer efficacy of BDMC and DTX compared to DTX alone in a murine prostate cancer model. These results suggest that BDMC can be an attractive therapeutic candidate in enhancing the efficacy of DTX in prostate cancer treatment.


Sujet(s)
Antinéoplasiques , Apoptose , Diarylheptanoïdes , Docetaxel , Tumeurs de la prostate , Mâle , Diarylheptanoïdes/pharmacologie , Diarylheptanoïdes/usage thérapeutique , Humains , Animaux , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Docetaxel/pharmacologie , Docetaxel/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Synergie des médicaments , Cycline B1/métabolisme , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Souris , Curcumine/analogues et dérivés , Curcumine/pharmacologie , Curcumine/usage thérapeutique , Survie cellulaire/effets des médicaments et des substances chimiques , Taxoïdes/pharmacologie , Taxoïdes/usage thérapeutique , Protéines proto-oncogènes c-bcl-2/métabolisme , Souris de lignée BALB C , Poly(ADP-ribose) polymerases/métabolisme , Protéine-kinase CDC2/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE