Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 45
Filtrer
1.
iScience ; 27(8): 110562, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39175775

RÉSUMÉ

The interaction between prostate cancer (PCa) cells and prostate stromal cells fosters an immunosuppressive tumor microenvironment (TME) that promotes tumor growth and immune evasion. However, the specific signaling pathways involved remain unclear. We identified a key mechanism involving the CXCL5/CXCR2 and LIF/LIFR pathways, which create a feedforward loop that enhances neuroendocrine differentiation (NED) in PCa cells and upregulates WNT1-inducible signaling pathway protein 1 (WISP1) in both cell types. WISP1 upregulation is essential for inducing immune checkpoints and immunosuppressive cytokines via LIF/LIFR signaling and STAT3 phosphorylation. This process leads to increased neuroendocrine markers, immune checkpoints, cell proliferation, and migration. Notably, WISP1 levels in patient sera correlate with PCa progression, suggesting its potential as a biomarker. Our findings elucidate the mechanisms by which reciprocal communication between PCa cells and stromal cells contributes to the formation of an immunosuppressive TME, driving the malignant progression of PCa and highlighting potential targets for therapeutic intervention.

2.
Sci Signal ; 17(840): eadc9142, 2024 06 11.
Article de Anglais | MEDLINE | ID: mdl-38861615

RÉSUMÉ

Neuroendocrine prostate cancer (PCa) (NEPC), an aggressive subtype that is associated with poor prognosis, may arise after androgen deprivation therapy (ADT). We investigated the molecular mechanisms by which ADT induces neuroendocrine differentiation in advanced PCa. We found that transmembrane protein 1 (MCTP1), which has putative Ca2+ sensing function and multiple Ca2+-binding C2 domains, was abundant in samples from patients with advanced PCa. MCTP1 was associated with the expression of the EMT-associated transcription factors ZBTB46, FOXA2, and HIF1A. The increased abundance of MCTP1 promoted PC3 prostate cancer cell migration and neuroendocrine differentiation and was associated with SNAI1-dependent EMT in C4-2 PCa cells after ADT. ZBTB46 interacted with FOXA2 and HIF1A and increased the abundance of MCTP1 in a hypoxia-dependent manner. MCTP1 stimulated Ca2+ signaling and AKT activation to promote EMT and neuroendocrine differentiation by increasing the SNAI1-dependent expression of EMT and neuroendocrine markers, effects that were blocked by knockdown of MCTP1. These data suggest an oncogenic role for MCTP1 in the maintenance of a rare and aggressive prostate cancer subtype through its response to Ca2+ and suggest its potential as a therapeutic target.


Sujet(s)
Différenciation cellulaire , Transition épithélio-mésenchymateuse , Tumeurs de la prostate , Animaux , Humains , Mâle , Souris , Androgènes/métabolisme , Androgènes/pharmacologie , Signalisation calcique/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux , Facteur nucléaire hépatocytaire HNF-3 bêta/métabolisme , Facteur nucléaire hépatocytaire HNF-3 bêta/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Cellules neuroendocrines/métabolisme , Cellules neuroendocrines/anatomopathologie , Cellules PC-3 , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Facteurs de transcription de la famille Snail/métabolisme , Facteurs de transcription de la famille Snail/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique
3.
Cancer Lett ; 598: 217090, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-38945201

RÉSUMÉ

The tumor microenvironment (TME) of prostate cancer (PCa) is characterized by high levels of immunosuppressive molecules, including cytokines and chemokines. This creates a hostile immune landscape that impedes effective immune responses. The interleukin-1 (IL-1) receptor antagonist (IL1RN), a key anti-inflammatory molecule, plays a significant role in suppressing IL-1-related immune and inflammatory responses. Our research investigates the oncogenic role of IL1RN in PCa, particularly its interactions with muscarinic acetylcholine receptor 4 (CHRM4), and its involvement in driving immunosuppressive pathways and M2-like macrophage polarization within the PCa TME. We demonstrate that following androgen deprivation therapy (ADT), the IL1RN-CHRM4 interaction in PCa activates the MAPK/AKT signaling pathway. This activation upregulates the transcription factors E2F1 and MYCN, stimulating IL1RN production and creating a positive feedback loop that increases CHRM4 abundance in both PCa cells and M2-like macrophages. This ADT-driven IL1RN/CHRM4 axis significantly enhances immune checkpoint markers associated with neuroendocrine differentiation and treatment-resistant outcomes. Higher serum IL1RN levels are associated with increased disease aggressiveness and M2-like macrophage markers in advanced PCa patients. Additionally, elevated IL1RN levels correlate with better clinical outcomes following immunotherapy. Clinical correlations between IL1RN and CHRM4 expression in advanced PCa patients and neuroendocrine PCa organoid models highlight their potential as therapeutic targets. Our data suggest that targeting the IL1RN/CHRM4 signaling could be a promising strategy for managing PCa progression and enhancing treatment responses.


Sujet(s)
Différenciation cellulaire , Antagoniste du récepteur à l'interleukine-1 , Tumeurs de la prostate , Microenvironnement tumoral , Mâle , Humains , Antagoniste du récepteur à l'interleukine-1/pharmacologie , Antagoniste du récepteur à l'interleukine-1/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Macrophages/métabolisme , Macrophages/immunologie , Facteur de transcription E2F1/métabolisme , Facteur de transcription E2F1/génétique
4.
Mol Oncol ; 18(6): 1665-1686, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38381121

RÉSUMÉ

Prostate stromal cells play a crucial role in the promotion of tumor growth and immune evasion in the tumor microenvironment (TME) through intricate molecular alterations in their interaction with prostate cancer (PCa) cells. While the impact of these cells on establishing an immunosuppressive response and influencing PCa aggressiveness remains incompletely understood. Our study shows that the activation of the leukemia inhibitory factor (LIF)/LIF receptor (LIFR) pathway in both prostate tumor and stromal cells, following androgen deprivation therapy (ADT), leads to the development of an immunosuppressive TME. Activation of LIF/LIFR signaling in PCa cells induces neuroendocrine differentiation (NED) and upregulates immune checkpoint expression. Inhibition of LIF/LIFR attenuates these effects, underscoring the crucial role of LIF/LIFR in linking NED to immunosuppression. Prostate stromal cells expressing LIFR contribute to NED and immunosuppressive marker abundance in PCa cells, while LIFR knockdown in prostate stromal cells reverses these effects. ADT-driven LIF/LIFR signaling induces brain-derived neurotrophic factor (BDNF) expression, which, in turn, promotes NED, aggressiveness, and immune evasion in PCa cells. Clinical analyses demonstrate elevated BDNF levels in metastatic castration-resistant PCa (CRPC) and a positive correlation with programmed death-ligand 1 (PDL1) and immunosuppressive signatures. This study shows that the crosstalk between PCa cells and prostate stromal cells enhances LIF/LIFR signaling, contributing to an immunosuppressive TME and NED in PCa cells through the upregulation of BDNF.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Tumeurs de la prostate , Microenvironnement tumoral , Mâle , Humains , Facteur neurotrophique dérivé du cerveau/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/immunologie , Lignée cellulaire tumorale , Microenvironnement tumoral/immunologie , Transduction du signal/effets des médicaments et des substances chimiques , Facteur inhibiteur de la leucémie/métabolisme , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie , Sous-unité alpha du récepteur au facteur d'inhibition de la leucémie/métabolisme , Sous-unité alpha du récepteur au facteur d'inhibition de la leucémie/génétique , Animaux , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/métabolisme , Tumeurs neuroendocrines/immunologie , Différenciation cellulaire
6.
Int J Mol Sci ; 24(15)2023 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-37569304

RÉSUMÉ

Prostate cancer (PCa) is one of the most prevalent cancers in men, yet its pathogenic pathways remain poorly understood. Transcriptomics and high-throughput sequencing can help uncover cancer diagnostic targets and understand biological circuits. Using prostate adenocarcinoma (PRAD) datasets of various web-based applications (GEPIA, UALCAN, cBioPortal, SR Plot, hTFtarget, Genome Browser, and MetaCore), we found that upregulated dysbindin domain-containing 1 (DBNDD1) expression in primary prostate tumors was strongly correlated with pathways involving the cell cycle, mitotic in KEGG, WIKI, and REACTOME database, and transcription factor-binding sites with the DBNDD1 gene in prostate samples. DBNDD1 gene expression was influenced by sample type, cancer stage, and promoter methylation levels of different cancers, such as PRAD, liver hepatocellular carcinoma (LIHC), and lung adenocarcinoma (LUAD). Regulation of glycogen synthase kinase (GSK)-3ß in bipolar disorder and ATP/ITP/GTP/XTP/TTP/CTP/UTP metabolic pathways was closely correlated with the DBNDD1 gene and its co-expressed genes in PCa. DBNDD1 gene expression was positively associated with immune infiltration of B cells, Myeloid-derived suppressor cell (MDSC), M2 macrophages, andneutrophil, whereas negatively correlated with CD8+ T cells, T follicular helper cells, M1 macrophages, and NK cells in PCa. These findings suggest that DBNDD1 may serve as a viable prognostic marker not only for early-stage PCa but also for immunotherapies.

7.
Cell Death Dis ; 14(5): 304, 2023 05 04.
Article de Anglais | MEDLINE | ID: mdl-37142586

RÉSUMÉ

Current treatment options for prostate cancer focus on targeting androgen receptor (AR) signaling. Inhibiting effects of AR may activate neuroendocrine differentiation and lineage plasticity pathways, thereby promoting the development of neuroendocrine prostate cancer (NEPC). Understanding the regulatory mechanisms of AR has important clinical implications for this most aggressive type of prostate cancer. Here, we demonstrated the tumor-suppressive role of the AR and found that activated AR could directly bind to the regulatory sequence of muscarinic acetylcholine receptor 4 (CHRM4) and downregulate its expression. CHRM4 was highly expressed in prostate cancer cells after androgen-deprivation therapy (ADT). CHRM4 overexpression may drive neuroendocrine differentiation of prostate cancer cells and is associated with immunosuppressive cytokine responses in the tumor microenvironment (TME) of prostate cancer. Mechanistically, CHRM4-driven AKT/MYCN signaling upregulated the interferon alpha 17 (IFNA17) cytokine in the prostate cancer TME after ADT. IFNA17 mediates a feedback mechanism in the TME by activating the CHRM4/AKT/MYCN signaling-driven immune checkpoint pathway and neuroendocrine differentiation of prostate cancer cells. We explored the therapeutic efficacy of targeting CHRM4 as a potential treatment for NEPC and evaluated IFNA17 secretion in the TME as a possible predictive prognostic biomarker for NEPC.


Sujet(s)
Tumeurs de la prostate , Mâle , Humains , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Protéine du proto-oncogène N-Myc/métabolisme , Protéines proto-oncogènes c-akt , Antagonistes des androgènes/usage thérapeutique , Interféron alpha/usage thérapeutique , Microenvironnement tumoral , Lignée cellulaire tumorale , Différenciation cellulaire , Récepteurs aux androgènes/métabolisme , Récepteur muscarinique de type M4/usage thérapeutique
8.
Redox Biol ; 62: 102686, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36963289

RÉSUMÉ

Conventional treatment of prostate cancer (PCa) uses androgen-deprivation therapy (ADT) to inhibit androgen receptor (AR) signaling-driven tumor progression. ADT-induced PCa recurrence may progress to an AR-negative phenotype with neuroendocrine (NE) histologic features, which are associated with metabolic disturbances and poor prognoses. However, the metabolic pathways that regulate NE differentiation (NED) in PCa remain unclear. Herein, we show a regulatory mechanism in NED-associated metabolism dysfunction induced by ADT, whereby overexpression of pyruvate kinase L/R (PKLR) mediates oxidative stress through upregulation of reactive oxygen species modulator 1 (ROMO1), thereby promoting NED and aggressiveness. ADT mediates the nuclear translocation of PKLR, which binds to the MYCN/MAX complex to upregulate ROMO1 and NE-related genes, leading to altered mitochondrial function and NED of PCa. Targeting nuclear PKLR/MYCN using bromodomain and extra-terminal motif (BET) inhibitors has the potential to reduce PKLR/MYCN-driven NED. Abundant ROMO1 in serum samples may provide prognostic information in patients with ADT. Our results suggest that ADT resistance leads to upregulation of PKLR/MYCN/ROMO1 signaling, which may drive metabolic reprogramming and NED in PCa. We further show that increased abundance of serum ROMO1 may be associated with the development of NE-like PCa.


Sujet(s)
Tumeurs prostatiques résistantes à la castration , Tumeurs de la prostate , Humains , Mâle , Antagonistes des androgènes/pharmacologie , Lignée cellulaire tumorale , Protéines membranaires , Protéines mitochondriales/métabolisme , Protéine du proto-oncogène N-Myc/génétique , Protéine du proto-oncogène N-Myc/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/métabolisme , Pyruvate kinase/métabolisme , Transduction du signal
9.
Transl Oncol ; 28: 101613, 2023 Feb.
Article de Anglais | MEDLINE | ID: mdl-36608541

RÉSUMÉ

Prostate cancer (PCa) is one of the most commonly diagnosed cancers among men worldwide. Despite the presence of accumulated clinical strategies for PCa management, limited prognostic/sensitive biomarkers are available to follow up on disease occurrence and progression. MicroRNAs (miRNAs) are small non-coding RNAs that control gene expression through post-transcriptional regulation of their complementary target messenger RNA (mRNA). MiRNAs modulate fundamental biological processes and play crucial roles in the pathology of various diseases, including PCa. Multiple evidence proved an aberrant miRNA expression profile in PCa, which is actively involved in the carcinogenic process. The robust and pleiotropic impact of miRNAs on PCa suggests them as potential candidates to help more understand the molecular landscape of the disease, which is likely to provide tools for early diagnosis and prognosis as well as additional therapeutic strategies to manage prostate tumors. Here, we emphasize the most consistently reported dysregulated miRNAs and highlight the contribution of their altered downstream targets with PCa hallmarks. Also, we report the potential effectiveness of using miRNAs as diagnostic/prognostic biomarkers in PCa and the high-throughput profiling technologies that are being used in their detection. Another key aspect to be discussed in this review is the promising implication of miRNAs molecules as therapeutic tools and targets for fighting PCa.

10.
Cell Death Dis ; 13(3): 252, 2022 03 19.
Article de Anglais | MEDLINE | ID: mdl-35306527

RÉSUMÉ

Neuroendocrine differentiation (NED) frequently occurs in androgen-deprivation therapy (ADT)-resistant prostate cancer (PCa) and is typically associated with metabolic pathway alterations, acquisition of lineage plasticity, and malignancy. There is no conventional therapeutic approach for PCa patients with NED pathologic features because the molecular targets are unknown. Here, we evaluated the regulatory mechanism of NED-associated metabolic reprogramming induced by ADT. We detected that the loss of the androgen-responsive transcription factor, zinc finger, and BTB domain containing 10 (ZBTB10), can activate pyruvate kinase L/R (PKLR) to enhance a NED response that is associated with glucose uptake by PCa cells. PKLR exhibits a tumor-promoting effect in PCa after ADT, but ZBTB10 can compensate for the glucose metabolism and NED capacity of PKLR through the direct transcriptional downregulation of PKLR. Targeting PKLR by drug repurposing with FDA-approved compounds can reduce the aggressiveness and NED of ADT-resistant PCa. We demonstrated that PKLR acts as a modulator to activate NED in PCa enhancement by loss of ZBTB10, thereby enabling PCa cells to mount a glycolysis response essential for therapeutic resistance. Our findings highlight the broad relation between NED and metabolic dysfunction to provide gene expression-based biomarkers for NEPC treatment.


Sujet(s)
Tumeurs prostatiques résistantes à la castration , Tumeurs de la prostate , Pyruvate kinase/métabolisme , Antagonistes des androgènes/pharmacologie , Androgènes/pharmacologie , Régulation négative , Humains , Mâle , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Pyruvate kinase/génétique , Protéines de répression/métabolisme
11.
Br J Cancer ; 126(5): 778-790, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34815524

RÉSUMÉ

BACKGROUND: Castration-resistant prostate cancer (CRPC) patients frequently develop neuroendocrine differentiation, with high mortality and no effective treatment. However, the regulatory mechanism that connects neuroendocrine differentiation and metabolic adaptation in response to therapeutic resistance of prostate cancer remain to be unravelled. METHODS: By unbiased cross-correlation between RNA-sequencing, database signatures, and ChIP analysis, combining in vitro cell lines and in vivo animal models, we identified that PCK1 is a pivotal regulator in therapy-induced neuroendocrine differentiation of prostate cancer through a LIF/ZBTB46-driven glucose metabolism pathway. RESULTS: Upregulation of PCK1 supports cell proliferation and reciprocally increases ZBTB46 levels to promote the expression of neuroendocrine markers that are conducive to the development of neuroendocrine characteristic CRPC. PCK1 and neuroendocrine marker expressions are regulated by the ZBTB46 transcription factor upon activation of LIF signalling. Targeting PCK1 can reduce the neuroendocrine phenotype and decrease the growth of prostate cancer cells in vitro and in vivo. CONCLUSION: Our study uncovers LIF/ZBTB46 signalling activation as a key mechanism for upregulating PCK1-driven glucose metabolism and neuroendocrine differentiation of CRPC, which may yield significant improvements in prostate cancer treatment after ADT using PCK1 inhibitors.


Sujet(s)
Glucose/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Facteur inhibiteur de la leucémie/génétique , Phosphoenolpyruvate carboxykinase (GTP)/génétique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Facteurs de transcription/génétique , Régulation positive , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Rétrocontrôle physiologique , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Souris , Cellules PC-3 , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/métabolisme , Analyse de séquence d'ARN
12.
Oncogenesis ; 10(11): 81, 2021 Nov 20.
Article de Anglais | MEDLINE | ID: mdl-34799554

RÉSUMÉ

Neuroendocrine differentiation (NED) is associated with WNT signaling activation and can be significantly observed after failure of androgen-deprivation therapy (ADT) for prostatic adenocarcinomas. Cytokine signaling is stimulated in NED prostate cancer; however, how ADT-upregulated WNT signaling promotes activation of cytokine signaling and contributes to NED of prostate cancer is poorly understood. In this study, we identified ADT-mediated upregulation of transcription factor 7 like 1 (TCF7L1), which increases the cytokine response and enhances NED of prostate cancer through interleukin (IL)-8/C-X-C motif chemokine receptor type 2 (CXCR2) signaling activation. ADT induced the secretion of WNT4 which upon engagement of TCF7L1 in prostate cancer cells, enhanced IL-8 and CXCR2 expressions. TCF7L1 directly binds to the regulatory sequence region of IL-8 and CXCR2 through WNT4 activation, thus upregulating IL-8/CXCR2 signaling-driven NED and cell motility. Analysis of prostate tissue samples collected from small-cell neuroendocrine prostate cancer (SCPC) and castration-resistant prostate cancer (CRPC) tumors showed an increased intensity of nuclear TCF7L1 associated with CXCR2. Our results suggest that induction of WNT4/TCF7L1 results in increased NED and malignancy in prostate cancer that is linked to dysregulation of androgen receptor signaling and activation of the IL-8/CXCR2 pathway.

13.
Cancers (Basel) ; 13(14)2021 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-34298665

RÉSUMÉ

Prostate cancer (PCa) is one of the most common cancers in the world and causes thousands of deaths every year. Conventional therapy for PCa includes surgery and androgen deprivation therapy (ADT). However, about 10-20% of all PCa cases relapse; there is also the further development of castration resistant adenocarcinoma (CRPC-Adeno) or neuroendocrine (NE) PCa (CRPC-NE). Due to their androgen-insensitive properties, both CRPC-Adeno and CRPC-NE have limited therapeutic options. Accordingly, this study reveals the inductive mechanisms of CRPC (for both CRPC-Adeno and CRPC-NE) and fulfils an urgent need for the treatment of PCa patients. Although previous studies have illustrated the emerging roles of epidermal growth factor receptors (EGFR), signal transducer, and activator of transcription 3 (STAT3) signaling in the development of CRPC, the regulatory mechanisms of this interaction between EGFR and STAT3 is still unclear. Our recent studies have shown that crosstalk between EGFR and STAT3 is critical for NE differentiation of PCa. In this review, we have collected recent findings with regard to the involvement of EGFR and STAT3 in malignancy progression and discussed their interactions during the development of therapeutic resistance for PCa.

14.
Commun Biol ; 4(1): 22, 2021 01 04.
Article de Anglais | MEDLINE | ID: mdl-33398073

RÉSUMÉ

Nerve growth factor (NGF) contributes to the progression of malignancy. However, the functional role and regulatory mechanisms of NGF in the development of neuroendocrine prostate cancer (NEPC) are unclear. Here, we show that an androgen-deprivation therapy (ADT)-stimulated transcription factor, ZBTB46, upregulated NGF via ZBTB46 mediated-transcriptional activation of NGF. NGF regulates NEPC differentiation by physically interacting with a G-protein-coupled receptor, cholinergic receptor muscarinic 4 (CHRM4), after ADT. Pharmacologic NGF blockade and NGF knockdown markedly inhibited CHRM4-mediated NEPC differentiation and AKT-MYCN signaling activation. CHRM4 stimulation was associated with ADT resistance and was significantly correlated with increased NGF in high-grade and small-cell neuroendocrine prostate cancer (SCNC) patient samples. Our results reveal a role of the NGF in the development of NEPC that is linked to ZBTB46 upregulation and CHRM4 accumulation. Our study provides evidence that the NGF-CHRM4 axis has potential to be considered as a therapeutic target to impair NEPC progression.


Sujet(s)
Adénocarcinome/métabolisme , Carcinome neuroendocrine/étiologie , Facteur de croissance nerveuse/métabolisme , Tumeurs de la prostate/métabolisme , Facteurs de transcription/métabolisme , Adénocarcinome/traitement médicamenteux , Antagonistes des androgènes/effets indésirables , Carcinome neuroendocrine/métabolisme , Carcinome neuroendocrine/anatomopathologie , Études cas-témoins , Résistance aux médicaments antinéoplasiques , Humains , Mâle , Cellules PC-3 , Prostate/anatomopathologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Récepteur muscarinique de type M4/métabolisme
15.
Oncogene ; 39(44): 6757-6775, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32963351

RÉSUMÉ

Neuroendocrine (NE) differentiation is a well-recognized phenotypic change of prostate cancer after androgen deprivation therapy (ADT), and it ultimately develops into an aggressive subset of this disease. However, the contribution of signaling pathways that lead to metabolic disorders and NE differentiation of prostate cancer remains unclear. In this study, we identified that ADT induced upregulation of the succinate-CoA ligase GDP-forming beta subunit (SUCLG2), which regulates succinate metabolism and NE differentiation of prostate cancer. We demonstrated a connection that upregulation of epidermal growth factor receptor (EGFR)-leukemia inhibitory factor receptor (LIFR) signaling induced SUCLG2 expression in prostate cancer cells. The LIFR is upregulated by nuclear EGFR, which acts as a transcriptional regulator, directly binds to the LIFR promoter, and drives NE differentiation and glycolysis of prostate cancer. LIFR upregulation is associated with SUCLG2, which increased succinate synthesis and enzymatic activities of mitochondrial nucleoside diphosphate kinase (NDPK) in prostate cancer cells. Knockdown of SUCLG2 suppressed NE differentiation in cultured cells and reduced prostate tumor growth in a xenograft model. Analysis of prostate tissue samples showed increased intensity of nuclear EGFR associated with the LIFR and SUCLG2 in castration-resistant prostate cancer tumors. Our study provides a mechanism whereby ADT upregulates EGFR-LIFR signaling that activates SUCLG2, which subsequently stimulates the metabolic changes associated with NE differentiation and aggressive prostate cancer phenotype.


Sujet(s)
Antagonistes des androgènes/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Sous-unité alpha du récepteur au facteur d'inhibition de la leucémie/génétique , Tumeurs neuroendocrines/génétique , Tumeurs prostatiques résistantes à la castration/génétique , Succinate-coA ligases/métabolisme , Antagonistes des androgènes/usage thérapeutique , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/génétique , Lignée cellulaire tumorale , Noyau de la cellule/anatomopathologie , Transdifférenciation cellulaire/effets des médicaments et des substances chimiques , Transdifférenciation cellulaire/génétique , Récepteurs ErbB/métabolisme , Techniques de knock-down de gènes , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/génétique , Humains , Sous-unité alpha du récepteur au facteur d'inhibition de la leucémie/métabolisme , Mâle , Souris , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs neuroendocrines/anatomopathologie , Régions promotrices (génétique) , Prostate/effets des médicaments et des substances chimiques , Prostate/anatomopathologie , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Récepteurs aux androgènes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Succinate-coA ligases/génétique , Régulation positive/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Molecules ; 25(10)2020 May 20.
Article de Anglais | MEDLINE | ID: mdl-32443915

RÉSUMÉ

Prostate cancer (PCa) is one of the most prevalent and malignant cancer types in men, which causes more than three-hundred thousand cancer death each year. At late stage of PCa progression, bone marrow is the most often metastatic site that constitutes almost 70% of metastatic cases of the PCa population. However, the characteristic for the osteo-philic property of PCa is still puzzling. Recent studies reported that the Wnt and Ras signaling pathways are pivotal in bone metastasis and that take parts in different cytological changes, but their crosstalk is not well studied. In this review, we focused on interactions between the Wnt and Ras signaling pathways during each stage of bone metastasis and present the fate of those interactions. This review contributes insights that can guide other researchers by unveiling more details with regard to bone metastasis and might also help in finding potential therapeutic regimens for preventing PCa bone metastasis.


Sujet(s)
Tumeurs osseuses/génétique , Prostate/métabolisme , Tumeurs de la prostate/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/secondaire , Évolution de la maladie , Régulation de l'expression des gènes tumoraux/génétique , Humains , Mâle , Métastase tumorale , Prostate/anatomopathologie , Tumeurs de la prostate/anatomopathologie , Transduction du signal/génétique
19.
Int J Mol Sci ; 20(11)2019 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-31181727

RÉSUMÉ

Prostate cancer (PCa) is the second most common killer among men in Western countries. Targeting androgen receptor (AR) signaling by androgen deprivation therapy (ADT) is the current therapeutic regime for patients newly diagnosed with metastatic PCa. However, most patients relapse and become resistant to ADT, leading to metastatic castration-resistant PCa (CRPC) and eventually death. Several proposed mechanisms have been proposed for CRPC; however, the exact mechanism through which CRPC develops is still unclear. One possible pathway is that the AR remains active in CRPC cases. Therefore, understanding AR signaling networks as primary PCa changes into metastatic CRPC is key to developing future biomarkers and therapeutic strategies for PCa and CRPC. In the current review, we focused on three novel biomarkers (ZBTB46, SPDEF, and ETV6) that were demonstrated to play critical roles in CRPC progression, epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKI) drug resistance, and the epithelial-to-mesenchymal transition (EMT) for patients treated with ADT or AR inhibition. In addition, we summarize how these potential biomarkers can be used in the clinic for diagnosis and as therapeutic targets of PCa.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Tumeurs prostatiques résistantes à la castration/métabolisme , Protéines proto-oncogènes c-ets/métabolisme , Protéines de répression/métabolisme , Facteurs de transcription/métabolisme , Animaux , Marqueurs biologiques tumoraux/génétique , Humains , Mâle , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Protéines proto-oncogènes c-ets/génétique , Protéines de répression/génétique , Transduction du signal , Facteurs de transcription/génétique , ETS Translocation Variant 6 Protein
20.
Clin Cancer Res ; 25(13): 4128-4140, 2019 07 01.
Article de Anglais | MEDLINE | ID: mdl-30962287

RÉSUMÉ

PURPOSE: The molecular targets for castration-resistant prostate cancer (CRPC) are unknown because the disease inevitably recurs, and therapeutic approaches for patients with CRPC remain less well understood. We sought to investigate regulatory mechanisms that result in increased therapeutic resistance, which is associated with neuroendocrine differentiation of prostate cancer and linked to dysregulation of the androgen-responsive pathway. EXPERIMENTAL DESIGN: The underlying intracellular mechanism that sustains the oncogenic network involved in neuroendocrine differentiation and therapeutic resistance of prostate cancer was evaluated to investigate and identify effectors. Multiple sets of samples with prostate adenocarcinomas and CRPC were assessed via IHC and other assays. RESULTS: We demonstrated that leukemia inhibitory factor (LIF) was induced by androgen deprivation therapy (ADT) and was upregulated by ZBTB46 in prostate cancer to promote CRPC and neuroendocrine differentiation. LIF was found to be induced in patients with prostate cancer after ADT and was associated with enriched nuclear ZBTB46 staining in high-grade prostate tumors. In prostate cancer cells, high ZBTB46 output was responsible for the activation of LIF-STAT3 signaling and neuroendocrine-like features. The abundance of LIF was mediated by ADT-induced ZBTB46 through a physical interaction with the regulatory sequence of LIF. Analysis of serum from patients showed that cases of higher tumor grade and metastatic prostate cancer exhibited higher LIF titers. CONCLUSIONS: Our findings suggest that LIF is a potent serum biomarker for diagnosing advanced prostate cancer and that targeting the ZBTB46-LIF axis may therefore inhibit CRPC development and neuroendocrine differentiation after ADT.


Sujet(s)
Facteur inhibiteur de la leucémie/métabolisme , Tumeurs prostatiques résistantes à la castration/métabolisme , Facteurs de transcription/métabolisme , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Animaux , Lignée cellulaire tumorale , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Humains , Immunohistochimie , Mâle , Souris , Modèles biologiques , Grading des tumeurs , Pronostic , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Liaison aux protéines , Récepteurs aux androgènes/métabolisme , Facteur de transcription STAT-3/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE