Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.610
Filtrer
1.
J Immunother Cancer ; 12(7)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39060021

RÉSUMÉ

BACKGROUND: Androgen deprivation therapy (ADT) is a front-line treatment for prostate cancer. In some men, their tumors can become refractory leading to the development of castration-resistant prostate cancer (CRPC). This causes tumors to regrow and metastasize, despite ongoing treatment, and impacts negatively on patient survival. ADT is known to stimulate the accumulation of immunosuppressive cells like protumoral tumor-associated macrophages (TAMs), myeloid-derived suppressor cells and regulatory T cells in prostate tumors, as well as hypofunctional T cells. Protumoral TAMs have been shown to accumulate around tumor blood vessels during chemotherapy and radiotherapy in other forms of cancer, where they drive tumor relapse. Our aim was to see whether such perivascular (PV) TAMs also accumulate in ADT-treated prostate tumors prior to CRPC, and, if so, whether selectively inducing them to express a potent immunostimulant, interferon beta (IFNß), would stimulate antitumor immunity and delay CRPC. METHODS: We used multiplex immunofluorescence to assess the effects of ADT on the distribution and activation status of TAMs, CD8+T cells, CD4+T cells and NK cells in mouse and/or human prostate tumors. We then used antibody-coated, lipid nanoparticles (LNPs) to selectively target a STING agonist, 2'3'-cGAMP (cGAMP), to PV TAMs in mouse prostate tumors during ADT. RESULTS: TAMs accumulated at high density around blood vessels in response to ADT and expressed markers of a protumoral phenotype including folate receptor-beta (FR-ß), MRC1 (CD206), CD169 and VISTA. Additionally, higher numbers of inactive (PD-1-) CD8+T cells and reduced numbers of active (CD69+) NK cells were present in these PV tumor areas. LNPs coated with an antibody to FR-ß selectively delivered cGAMP to PV TAMs in ADT-treated tumors, where they activated STING and upregulated the expression of IFNß. This resulted in a marked increase in the density of active CD8+T cells (along with CD4+T cells and NK cells) in PV tumor areas, and significantly delayed the onset of CRPC. Antibody depletion of CD8+T cells during LNP administration demonstrated the essential role of these cells in delay in CRPC induced by LNPs. CONCLUSION: Together, our data indicate that targeting a STING agonist to PV TAMs could be used to extend the treatment window for ADT in prostate cancer.


Sujet(s)
Macrophages , Protéines membranaires , Mâle , Animaux , Souris , Protéines membranaires/métabolisme , Protéines membranaires/agonistes , Macrophages/métabolisme , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Humains , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Antagonistes des androgènes/usage thérapeutique , Antagonistes des androgènes/pharmacologie , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/immunologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Résistance aux médicaments antinéoplasiques
2.
Sci Adv ; 10(25): eadn8709, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38905345

RÉSUMÉ

Androgen deprivation therapy (ADT) for prostate cancer is associated with an increased risk of dementia, including Alzheimer's disease (AD). The mechanistic connection between ADT and AD-related cognitive impairment in patients with prostate cancer remains elusive. We established a clinically relevant prostate cancer-bearing AD mouse model to explore this. Both tumor-bearing and ADT induce complex changes in immune and inflammatory responses in peripheral blood and in the brain. ADT disrupts the integrity of the blood-brain barrier (BBB) and promotes immune cell infiltration into the brain, enhancing neuroinflammation and gliosis without affecting the amyloid plaque load. Moreover, treatment with natalizumab, an FDA-approved drug targeting peripheral immune cell infiltration, reduces neuroinflammation and improves cognitive function in this model. Our study uncovers an inflammatory mechanism, extending beyond amyloid pathology, that underlies ADT-exacerbated cognitive deficits, and suggests natalizumab as a potentially effective treatment in alleviating the detrimental effects of ADT on cognition.


Sujet(s)
Maladie d'Alzheimer , Antagonistes des androgènes , Barrière hémato-encéphalique , Encéphale , Dysfonctionnement cognitif , Modèles animaux de maladie humaine , Animaux , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Mâle , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/induit chimiquement , Dysfonctionnement cognitif/anatomopathologie , Dysfonctionnement cognitif/étiologie , Souris , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Encéphale/métabolisme , Humains , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Antagonistes des androgènes/effets indésirables , Antagonistes des androgènes/pharmacologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Natalizumab/effets indésirables , Natalizumab/pharmacologie , Natalizumab/usage thérapeutique , Plaque amyloïde/anatomopathologie , Plaque amyloïde/traitement médicamenteux
3.
Biochem Pharmacol ; 226: 116385, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38909784

RÉSUMÉ

We have previously demonstrated that androgen-dependent prostate cancer (PCa) cell lines enter a state of senescence following exposure to androgen deprivation therapies (ADT). ADT-induced senescence was found to be transient, as senescent cells develop castration resistance and re-emerge into a proliferative state even under continuous androgen deprivation in vitro. Moreover, the BCL-XL/BCL-2 inhibitor, ABT-263 (navitoclax), an established senolytic agent, promoted apoptosis of senescent PCa cells, suppressing proliferative recovery and subsequent tumor cell outgrowth. As this strategy has not previously been validated in vivo, we used a clinically relevant, syngeneic murine model of PCa, where mice were either castrated or castrated followed by the administration of ABT-263. Our results largely confirm the outcomes previously reported in vitro; specifically, castration alone results in a transient tumor growth suppression with characteristics of senescence, which is prolonged by exposure to ABT-263. Most critically, mice that underwent castration followed by ABT-263 experienced a statistically significant prolongation in survival, with an increase of 14.5 days in median survival time (56 days castration alone vs. 70.5 days castration + ABT-263). However, as is often the case in studies combining the promotion of senescence with a senolytic (the "one-two" punch approach), the suppression of tumor growth by the inclusion of the senolytic agent was transient, allowing for tumor regrowth once the drug treatment was terminated. Nevertheless, the results of this work suggest that the "one-two" punch senolytic strategy in PCa may effectively interfere with, diminish, or delay the development of the lethal castration-resistant phenotype.


Sujet(s)
Dérivés de l'aniline , Vieillissement de la cellule , Tumeurs de la prostate , Sulfonamides , Mâle , Animaux , Souris , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Vieillissement de la cellule/physiologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Sulfonamides/pharmacologie , Humains , Lignée cellulaire tumorale , Sénothérapie/pharmacologie , Sénothérapie/usage thérapeutique , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Androgènes/métabolisme , Androgènes/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Souris de lignée C57BL
4.
Reproduction ; 168(2)2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38833564

RÉSUMÉ

In brief: Atrazine, like oestrogen, disorganises laminin formation and reduces the number of germ cells and Sertoli cells in the developing testes of the tammar wallaby. This study suggests that interfering with the balance of androgen and oestrogen affects the integrity of laminin structure and testis differentiation. Abstract: The herbicide atrazine was banned in Europe in 2003 due to its endocrine disrupting activity but remains widely used. The integrity of the laminin structure in fetal testis cords requires oestrogen signalling but overexposure to xenoestrogens in the adult can cause testicular dysgenesis. However, whether xenoestrogens affect laminin formation in developing testes has not been investigated. Here we examined the effects of atrazine in the marsupial tammar wallaby during early development and compare it with the effects of the anti-androgen flutamide, oestrogen, and the oestrogen degrader fulvestrant. The tammar, like all marsupials, gives birth to altricial young, allowing direct treatment of the developing young during the male programming window (day 20-40 post partum (pp)). Male pouch young were treated orally with atrazine (5 mg/kg), flutamide (10 mg/kg), 17ß-oestradiol (2.5 mg/kg) and fulvestrant (1 mg/kg) daily from day 20 to 40 pp. Distribution of laminin, vimentin, SOX9 and DDX4, cell proliferation and mRNA expression of SRY, SOX9, AMH, and SF1 were examined in testes at day 50 post partum after the treatment. Direct exposure to atrazine, flutamide, 17ß-oestradiol, and fulvestrant all disorganised laminin but had no effect on vimentin distribution in testes. Atrazine reduced the number of germ cells and Sertoli cells when examined at day 40-50 pp and day 20 to 40 pp, respectively. Both flutamide and fulvestrant reduced the number of germ cells and Sertoli cells. Atrazine also downregulated SRY expression and impaired SOX9 nuclear translocation. Our results demonstrate that atrazine can compromise normal testicular differentiation during the critical male programming window.


Sujet(s)
Atrazine , Différenciation cellulaire , Herbicides , Laminine , Testicule , Mâle , Animaux , Testicule/effets des médicaments et des substances chimiques , Testicule/métabolisme , Testicule/cytologie , Atrazine/pharmacologie , Laminine/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Herbicides/pharmacologie , Macropodidae/métabolisme , Cellules de Sertoli/effets des médicaments et des substances chimiques , Cellules de Sertoli/métabolisme , Cellules de Sertoli/cytologie , Oestrogènes/pharmacologie , Oestrogènes/métabolisme , Perturbateurs endocriniens/pharmacologie , Numération cellulaire , Antagonistes des androgènes/pharmacologie , Flutamide/pharmacologie
5.
Cancer Lett ; 597: 217068, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-38901665

RÉSUMÉ

With the widespread use of anti-androgen therapy, such as abiraterone and enzalutamide, the incidence of neuroendocrine prostate cancer (NEPC) is increasing. NEPC is a lethal form of prostate cancer (PCa), with a median overall survival of less than one year after diagnosis. In addition to the common bone metastases seen in PCa, NEPC exhibits characteristics of visceral metastases, notably liver metastasis, which serves as an indicator of a poor prognosis clinically. Key factors driving the neuroendocrine plasticity of PCa have been identified, yet the underlying mechanism behind liver metastasis remains unclear. In this study, we identified PROX1 as a driver of neuroendocrine plasticity in PCa, responsible for promoting liver metastases. Mechanistically, anti-androgen therapy alleviates transcriptional inhibition of PROX1. Subsequently, elevated PROX1 levels drive both neuroendocrine plasticity and liver-specific transcriptional reprogramming, promoting liver metastases. Moreover, liver metastases in PCa induced by PROX1 depend on reprogrammed lipid metabolism, a disruption that effectively reduces the formation of liver metastases.


Sujet(s)
Protéines à homéodomaine , Tumeurs du foie , Tumeurs de la prostate , Protéines suppresseurs de tumeurs , Mâle , Humains , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Tumeurs du foie/secondaire , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme , Animaux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Souris , Métabolisme lipidique/effets des médicaments et des substances chimiques , Plasticité cellulaire/effets des médicaments et des substances chimiques , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/métabolisme , Tumeurs neuroendocrines/traitement médicamenteux , Carcinome neuroendocrine/génétique , Carcinome neuroendocrine/anatomopathologie , Carcinome neuroendocrine/métabolisme , Carcinome neuroendocrine/traitement médicamenteux , Carcinome neuroendocrine/secondaire
6.
BMC Cancer ; 24(1): 713, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858662

RÉSUMÉ

BACKGROUND: This paper attempted to clarify the role and mechanism of vacuolar protein sorting-associated protein 72 homolog (VPS72) in the progression of prostate cancer (PCa). METHODS: Clinical information and gene expression profiles of patients with prostate cancer were obtained from The Cancer Genome Atlas (TCGA). VPS72 expression in PCa and the potential mechanism by which VPS72 affects PCa progression was investigated. Next, we performed COX regression analysis to identify the independent prognostic factors of PCa, and constructed a nomogram. The sensitivity of chemotherapeutic medications was anticipated using "pRRophetic". Subsequently, in vitro assays to validate the effect of VPS72 on PCa cell proliferation, migration and susceptibility to anti-androgen therapy. RESULTS: The expression of VPS72 was considerably higher in PCa tissues compared to normal tissues. Significant correlations were found between high VPS72 expression and a poor prognosis and adverse clinicopathological factors. The nomogram model constructed based on VPS72 expression has good predictive performance. According to GSEA, VPS72-related genes were enriched in the NF-kB pathways, cytokine-cytokine receptor interaction and chemokine signaling pathway in PCa. Although PCa with low VPS72 expression was more adaptable to chemotherapeutic medications, our in vitro experiment showed that VPS72 knockdown significantly decreased the PCa cell migration, proliferation, and resistance to anti-androgen therapy. CONCLUSIONS: In summary our findings suggests that VPS72 could play a crucial role in the malignant progression of PCa, and its expression level can be employed as a possible biomarker of PCa prognosis.


Sujet(s)
Marqueurs biologiques tumoraux , Prolifération cellulaire , Évolution de la maladie , Tumeurs de la prostate , Sujet âgé , Humains , Mâle , Adulte d'âge moyen , Antagonistes des androgènes/usage thérapeutique , Antagonistes des androgènes/pharmacologie , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Nomogrammes , Pronostic , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Protéines du transport vésiculaire/génétique , Protéines du transport vésiculaire/métabolisme
7.
Talanta ; 275: 126174, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38705021

RÉSUMÉ

To analyze a complex sample for endocrine activity, different tests must be performed to clarify androgen/estrogen agonism, antagonism, cytotoxicity, anti-cytotoxicity, and corresponding false-positive reactions. This means a large amount of work. Therefore, a six-fold planar multiplex bioassay concept was developed to evaluate up to the mentioned six endpoints or mechanisms simultaneously in the same sample analysis. Separation of active constituents from interfering matrix via high-performance thin-layer chromatography and effect differentiation via four vertical stripes (of agonists and end-products of the respective enzyme-substrate reaction) applied along each separated sample track were key to success. First, duplex endocrine bioassay versions were established. For the androgen/anti-androgen bioassay applied via piezoelectric spraying, the mean limit of biological detection of bisphenol A was 14 ng/band and its mean half maximal inhibitory concentration IC50 was 116 ng/band. Applied to trace analysis of six migrate samples from food packaging materials, 19 compound zones with agonistic or antagonistic estrogen/androgen activities were detected, with up to seven active compound zones within one migrate. For the first time, the S9 metabolism of endocrine effective compounds was studied on the same surface and revealed partial deactivation. Coupled to high-resolution mass spectrometry, molecular formulas were tentatively assigned to compounds, known to be present in packaging materials or endocrine active or previously unknown. Finally, the detection of cytotoxicity/anti-cytotoxicity and false-positives was integrated into the duplex androgen/anti-androgen bioassay. The resulting six-fold multiplex planar bioassay was evaluated with positive control standards and successfully applied to one migrate sample. The streamlined stripe concept for multiplex planar bioassays made it possible to assign different mechanisms to individual active compounds in a complex sample. The concept is generic and can be transferred to other assays.


Sujet(s)
Dosage biologique , Dosage biologique/méthodes , Humains , Perturbateurs endocriniens/analyse , Perturbateurs endocriniens/pharmacologie , Faux positifs , Phénols/analyse , Phénols/composition chimique , Phénols/pharmacologie , Composés benzhydryliques/analyse , Composés benzhydryliques/pharmacologie , Composés benzhydryliques/composition chimique , Androgènes/analyse , Androgènes/métabolisme , Antagonistes des androgènes/analyse , Antagonistes des androgènes/pharmacologie
8.
Expert Opin Drug Metab Toxicol ; 20(6): 491-502, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38778707

RÉSUMÉ

INTRODUCTION: The therapeutic scenario of metastatic hormone-sensitive prostate cancer (mHSPC) has dramatically changed in recent years, with the approval of new-generation Androgen Receptor Signaling Inhibitors (ARSIs), in combination with the androgen deprivation therapy (ADT), which was the previous standard of care. Despite showing a similar clinical efficacy, ARSIs, all of which are administered orally, are different in terms of pharmacokinetic and drug-drug interactions (DDIs). AREAS COVERED: This review covers the main pharmacokinetic characteristics of ARSIs that have been approved for the first-line therapy of mHSPC patients, underlying the differences among these molecules and focusing on the known or possible interactions with other drugs. Full-text articles and abstracts were searched in PubMed. EXPERT OPINION: Since prostate cancer occurs mainly in older age, comorbidities and the consequent polypharmacy increase the DDI risk in mHSPC patients who are candidates for ARSI. Waiting for new therapeutic options, in the absence of direct comparisons, pharmacokinetic knowledge is essential to guide clinicians in prescribing ARSI in this setting.


Sujet(s)
Antagonistes du récepteur des androgènes , Interactions médicamenteuses , Métastase tumorale , Tumeurs de la prostate , Transduction du signal , Humains , Mâle , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Antagonistes du récepteur des androgènes/pharmacocinétique , Antagonistes du récepteur des androgènes/administration et posologie , Antagonistes du récepteur des androgènes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Polypharmacie , Sujet âgé , Antagonistes des androgènes/administration et posologie , Antagonistes des androgènes/pharmacocinétique , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/effets indésirables , Administration par voie orale , Antinéoplasiques hormonaux/pharmacocinétique , Antinéoplasiques hormonaux/administration et posologie , Antinéoplasiques hormonaux/pharmacologie , Animaux
9.
J Immunother Cancer ; 12(5)2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38772685

RÉSUMÉ

RATIONALE: Androgen deprivation therapy (ADT) is the primary treatment for recurrent and metastatic prostate cancer. In addition to direct antitumor effects, ADT has immunomodulatory effects such as promoting T-cell infiltration and enhancing antigen processing/presentation. Previous studies in our laboratory have demonstrated that ADT also leads to increased expression of the androgen receptor (AR) and increased recognition of prostate tumor cells by AR-specific CD8+T cells. We have also demonstrated that ADT combined with a DNA vaccine encoding the AR significantly slowed tumor growth and improved the survival of prostate tumor-bearing mice. The current study aimed to investigate the impact of the timing and sequencing of ADT with vaccination on the tumor immune microenvironment in murine prostate cancer models to further increase the antitumor efficacy of vaccines. METHODS: Male FVB mice implanted with Myc-CaP tumor cells, or male C57BL/6 mice implanted with TRAMP-C1 prostate tumor cells, were treated with a DNA vaccine encoding AR (pTVG-AR) and ADT. The sequence of administration was evaluated for its effect on tumor growth, and tumor-infiltrating immune populations were characterized. RESULTS: Vaccination prior to ADT (pTVG-AR → ADT) significantly enhanced antitumor responses and survival. This was associated with increased tumor infiltration by CD4+ and CD8+ T cells, including AR-specific CD8+T cells. Depletion of CD8+T cells prior to ADT significantly worsened overall survival. Following ADT treatment, however, Gr1+ myeloid-derived suppressor cells (MDSCs) increased, and this was associated with fewer infiltrating T cells and reduced tumor growth. Inhibiting Gr1+MDSCs recruitment, either by using a CXCR2 antagonist or by cycling androgen deprivation with testosterone replacement, improved antitumor responses and overall survival. CONCLUSION: Vaccination prior to ADT significantly improved antitumor responses, mediated in part by increased infiltration of CD8+T cells following ADT. Targeting MDSC recruitment following ADT further enhanced antitumor responses. These findings suggest logical directions for future clinical trials to improve the efficacy of prostate cancer vaccines.


Sujet(s)
Vaccins anticancéreux , Tumeurs de la prostate , Récepteurs aux androgènes , Mâle , Animaux , Souris , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/anatomopathologie , Récepteurs aux androgènes/métabolisme , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/pharmacologie , Vaccins anticancéreux/immunologie , Vaccins à ADN/usage thérapeutique , Vaccins à ADN/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Antagonistes des androgènes/pharmacologie , Lignée cellulaire tumorale , Souris de lignée C57BL , Vaccination , Humains , Microenvironnement tumoral , Modèles animaux de maladie humaine , Lymphocytes T CD8+/immunologie
10.
Biosensors (Basel) ; 14(4)2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38667168

RÉSUMÉ

Prostate cancer (PCa) displays diverse intra-tumoral traits, impacting its progression and treatment outcomes. This study aimed to refine PCa cell culture conditions for dynamic monitoring of androgen receptor (AR) activity at the single-cell level. We introduced an extracellular matrix-Matrigel (ECM-M) culture model, enhancing cellular tracking during bioluminescence single-cell imaging while improving cell viability. ECM-M notably tripled the traceability of poorly adherent PCa cells, facilitating robust single-cell tracking, without impeding substrate permeability or AR response. This model effectively monitored AR modulation by antiandrogens across various PCa cell lines. Single-cell imaging unveiled heterogeneous antiandrogen responses within populations, correlating non-responsive cell proportions with drug IC50 values. Integrating ECM-M culture with the PSEBC-TSTA biosensor enabled precise characterization of ARi responsiveness within diverse cell populations. Our ECM-M model stands as a promising tool to assess heterogeneous single-cell treatment responses in cancer, offering insights to link drug responses to intracellular signaling dynamics. This approach enhances our comprehension of the nuanced and dynamic nature of PCa treatment responses.


Sujet(s)
Matrice extracellulaire , Tumeurs de la prostate , Humains , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Matrice extracellulaire/métabolisme , Mâle , Lignée cellulaire tumorale , Antagonistes des androgènes/pharmacologie , Récepteurs aux androgènes/métabolisme , Analyse sur cellule unique , Microscopie , Techniques de biocapteur , Mesures de luminescence
11.
Oncogene ; 43(21): 1631-1643, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38589675

RÉSUMÉ

Androgen deprivation therapy (ADT) is the first line of treatment for metastatic prostate cancer (PCa) that effectively delays the tumor progression. However, it also increases the risk of venous thrombosis event (VTE) in patients, a leading cause of mortality. How a pro-thrombotic cascade is induced by ADT remains poorly understood. Here, we report that protein disulfide isomerase A2 (PDIA2) is upregulated in PCa cells to promote VTE formation and enhance PCa cells resistant to ADT. Using various in vitro and in vivo models, we demonstrated a dual function of PDIA2 that enhances tumor-mediated pro-coagulation activity via tumor-derived extracellular vehicles (EVs). It also stimulates PCa cell proliferation, colony formation, and xenograft growth androgen-independently. Mechanistically, PDIA2 activates the tissue factor (TF) on EVs through its isomerase activity, which subsequently triggers a pro-thrombotic cascade in the blood. Additionally, TF-containing EVs can activate the Src kinase inside PCa cells to enhance the AR signaling ligand independently. Androgen deprivation does not alter PDIA2 expression in PCa cells but enhances PDIA2 translocation to the cell membrane and EVs via suppressing the clathrin-dependent endocytic process. Co-recruitment of AR and FOXA1 to the PDIA2 promoter is required for PDIA2 transcription under androgen-deprived conditions. Importantly, blocking PDIA2 isomerase activity suppresses the pro-coagulation activity of patient plasma, PCa cell, and xenograft samples as well as castrate-resistant PCa xenograft growth. These results demonstrate that PDIA2 promotes VTE and tumor progression via activating TF from tumor-derived EVs. They rationalize pharmacological inhibition of PDIA2 to suppress ADT-induced VTE and castrate-resistant tumor progression.


Sujet(s)
Évolution de la maladie , Tumeurs prostatiques résistantes à la castration , Protein Disulfide-Isomerases , Thrombose veineuse , Animaux , Humains , Mâle , Souris , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/effets indésirables , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/génétique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Protein Disulfide-Isomerases/métabolisme , Protein Disulfide-Isomerases/génétique , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Thromboplastine/métabolisme , Thromboplastine/génétique , Thrombose veineuse/métabolisme , Thrombose veineuse/induit chimiquement , Thrombose veineuse/anatomopathologie , Thrombose veineuse/génétique , Thrombose veineuse/étiologie , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Prostate ; 84(9): 814-822, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38558458

RÉSUMÉ

BACKGROUND: Tumor initiation and progression necessitate a metabolic shift in cancer cells. Consequently, the progression of prostate cancer (PCa), a leading cause of cancer-related deaths in males globally, involves a shift from lipogenic to glycolytic metabolism. Androgen deprivation therapy (ADT) serves as the standard treatment for advanced-stage PCa. However, despite initial patient responses, castrate resistance emerges ultimately, necessitating novel therapeutic approaches. Therefore, in this study, we aimed to investigate the role of monocarboxylate transporters (MCTs) in PCa post-ADT and evaluate their potential as therapeutic targets. METHODS: PCa cells (LNCaP and C4-2 cell line), which has high prostate-specific membrane antigen (PSMA) and androgen receptor (AR) expression among PCa cell lines, was used in this study. We assessed the expression of MCT1 in PCa cells subjected to ADT using charcoal-stripped bovine serum (CSS)-containing medium or enzalutamide (ENZ). Furthermore, we evaluated the synergistic anticancer effects of combined treatment with ENZ and SR13800, an MCT1 inhibitor. RESULTS: Short-term ADT led to a significant upregulation in folate hydrolase 1 (FOLH1) and solute carrier family 16 member 1 (SLC16A1) gene levels, with elevated PSMA and MCT1 protein levels. Long-term ADT induced notable changes in cell morphology with further upregulation of FOLH1/PSMA and SLC16A1/MCT1 levels. Treatment with ENZ, a nonsteroidal anti-androgen, also increased PSMA and MCT1 expression. However, combined therapy with ENZ and SR13800 led to reduced PSMA level, decreased cell viability, and suppressed expression of cancer stem cell markers and migration indicators. Additionally, analysis of human PCa tissues revealed a positive correlation between PSMA and MCT1 expression in tumor regions. CONCLUSIONS: Our results demonstrate that ADT led to a significant upregulation in MCT1 levels. However, the combination of ENZ and SR13800 demonstrated a promising synergistic anticancer effect, highlighting a potential therapeutic significance for patients with PCa undergoing ADT.


Sujet(s)
Antagonistes des androgènes , Benzamides , Transporteurs d'acides monocarboxyliques , Nitriles , 3-Phényl-2-thiohydantoïne , Tumeurs de la prostate , Symporteurs , Mâle , Humains , Transporteurs d'acides monocarboxyliques/métabolisme , Transporteurs d'acides monocarboxyliques/antagonistes et inhibiteurs , Transporteurs d'acides monocarboxyliques/génétique , Lignée cellulaire tumorale , 3-Phényl-2-thiohydantoïne/pharmacologie , 3-Phényl-2-thiohydantoïne/analogues et dérivés , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Nitriles/pharmacologie , Symporteurs/métabolisme , Symporteurs/antagonistes et inhibiteurs , Symporteurs/génétique , Benzamides/pharmacologie
13.
Aging (Albany NY) ; 16(8): 7249-7266, 2024 04 19.
Article de Anglais | MEDLINE | ID: mdl-38643469

RÉSUMÉ

OBJECTIVE: Prostate cancer (PCa) is the second disease threatening men's health, and anti-androgen therapy (AAT) is a primary approach for treating this condition. Increasing evidence suggests that long non-coding RNAs (lncRNAs) play crucial roles in the development of PCa and the process of AAT resistance. The objective of this study is to utilize bioinformatics methods to excavate lncRNAs association with AAT resistance and investigate their biological functions. METHODS: AAT resistance-related risk score model (ARR-RSM) was established by multivariate Cox analysis. Paired clinical tissue samples of 36 PCa patients and 42 blood samples from patients with PSA over 4 ng/ml were collected to verify the ARR-RSM. In vitro, RT-qPCR, CCK-8 and clone formation assays were displayed to verify the expression and function of AL354989.1 and AC007405.2. RESULTS: Pearson correlation analysis identified 996 lncRNAs were associated with AAT resistance (ARR-LncRs). ARR-RSM was established using multivariate Cox regression analysis, and PCa patients were divided into high-risk and low-risk groups. High-risk patients showed increased expression of AL354989.1 and AC007405.2 had poorer prognoses. The high-risk score correlated with advanced T-stage and N-stage. The AUC of ARR-RSM outperformed tPSA in diagnosing PCa. Silencing of AC007405.2 and AL354989.1 inhibited PCa cells proliferation and AAT resistance. CONCLUSIONS: In this study, we have discovered the clinical significance of AC007405.2 and AL354989.1 in predicting the prognosis and diagnosing PCa patients. Furthermore, we have confirmed their correlation with various clinical features. These findings provide potential targets for PCa treatment and a novel diagnostic and predictive indicator for precise PCa diagnosis.


Sujet(s)
Antagonistes des androgènes , Marqueurs biologiques tumoraux , Résistance aux médicaments antinéoplasiques , Tumeurs de la prostate , ARN long non codant , Sujet âgé , Humains , Mâle , Antagonistes des androgènes/usage thérapeutique , Antagonistes des androgènes/pharmacologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Régulation de l'expression des gènes tumoraux , Pronostic , Tumeurs de la prostate/génétique , Tumeurs de la prostate/diagnostic , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , ARN long non codant/génétique , ARN long non codant/métabolisme
14.
J Nanobiotechnology ; 22(1): 192, 2024 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-38637848

RÉSUMÉ

Androgen deprivation therapy (ADT) is a crucial and effective strategy for prostate cancer, while systemic administration may cause profound side effects on normal tissues. More importantly, the ADT can easily lead to resistance by involving the activation of NF-κB signaling pathway and high infiltration of M2 macrophages in tumor microenvironment (TME). Herein, we developed a biomimetic nanotherapeutic platform by deriving cell membrane nanovesicles from cancer cells and probiotics to yield the hybrid cellular nanovesicles (hNVs), loading flutamide (Flu) into the resulting hNVs, and finally modifying the hNVs@Flu with Epigallocatechin-3-gallate (EGCG). In this nanotherapeutic platform, the hNVs significantly improved the accumulation of hNVs@Flu-EGCG in tumor sites and reprogramed immunosuppressive M2 macrophages into antitumorigenic M1 macrophages, the Flu acted on androgen receptors and inhibited tumor proliferation, and the EGCG promoted apoptosis of prostate cancer cells by inhibiting the NF-κB pathway, thus synergistically stimulating the antitumor immunity and reducing the side effects and resistance of ADT. In a prostate cancer mouse model, the hNVs@Flu-EGCG significantly extended the lifespan of mice with tumors and led to an 81.78% reduction in tumor growth compared with the untreated group. Overall, the hNVs@Flu-EGCG are safe, modifiable, and effective, thus offering a promising platform for effective therapeutics of prostate cancer.


Sujet(s)
Facteur de transcription NF-kappa B , Tumeurs de la prostate , Humains , Mâle , Animaux , Souris , Facteur de transcription NF-kappa B/métabolisme , Androgènes/usage thérapeutique , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Immunothérapie/méthodes , Thé , Lignée cellulaire tumorale , Microenvironnement tumoral
15.
Toxicon ; 243: 107722, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38653393

RÉSUMÉ

Flutamide is frequently used in the management of prostate cancer, hirsutism, and acne. It is a non-steroidal anti-androgenic drug and causes hepatotoxicity. The current study's objective is to evaluate sophorin's hepatoprotective effectiveness against flutamide-induced hepatotoxicity in Wistar rats. Sophorin is a citrus flavonoid glycoside, also known as rutin, which is a low molecular weight polyphenolic compound with natural antioxidant properties and reported to have promising hepatoprotective efficacy. In this study, sophorin was used at a dose of 100 mg/kg body weight in purified water via oral route for 4 week daily whereas, flutamide was used at a dose of 100 mg kg/b.wt for 4 weeks daily in 0.5% carboxy methyl cellulose (CMC) through the oral route for the induction of hepatotoxicity. Flutamide administration leads to enhanced reactive oxygen species (ROS) generation, an imbalance in redox homeostasis and peroxidation of lipid resulted in reduced natural antioxidant level in liver tissue. Our result demonstrated that sophorin significantly abrogate flutamide induced lipid peroxidation, protein carbonyl (PC), and also significantly increasesed in enzymatic activity/level of tissue natural antioxidant such as reduced glutathione(GSH), glutathione reductase(GR), catalase, and superoxide dismutase(SOD). Additionally, sophorin reduced the activity of cytochrome P450 3A1 in liver tissue which was elevated due to flutamide treatment. Furthermore, sophorin treatment significantly decreased the pro-inflammatory cytokines (TNF-α and IL-6) level. Immunohistochemical analysis for the expression of inflammatory proteins (iNOS and COX-2) in hepatic tissue was decreased after sophorin treatment against flutamide-induced hepatotoxicity. Moreover, sophorin suppressed the infiltration of mast cells in liver tissue which further showed anti-inflammatory potential of sophorin. Our histological investigation further demonstrated sophorin's hepatoprotective function by restoring the typical histology of the liver. Based on the aforementioned information, we are able to come to the conclusion that sophorin supplementation might benefit wistar rats with flutamide-induced hepatic damage by reducing oxidative stress and hepatocellular inflammation.


Sujet(s)
Lésions hépatiques dues aux substances , Flutamide , Foie , Rat Wistar , Animaux , Flutamide/pharmacologie , Rats , Lésions hépatiques dues aux substances/traitement médicamenteux , Lésions hépatiques dues aux substances/prévention et contrôle , Mâle , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Peroxydation lipidique/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Antagonistes des androgènes/pharmacologie
16.
J Immunother Cancer ; 12(4)2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38663936

RÉSUMÉ

RATIONALE: Androgen deprivation therapy (ADT) is pivotal in treating recurrent prostate cancer and is often combined with external beam radiation therapy (EBRT) for localized disease. However, for metastatic castration-resistant prostate cancer, EBRT is typically only used in the palliative setting, because of the inability to radiate all sites of disease. Systemic radiation treatments that preferentially irradiate cancer cells, known as radiopharmaceutical therapy or targeted radionuclide therapy (TRT), have demonstrable benefits for treating metastatic prostate cancer. Here, we explored the use of a novel TRT, 90Y-NM600, specifically in combination with ADT, in murine prostate tumor models. METHODS: 6-week-old male FVB mice were implanted subcutaneously with Myc-CaP tumor cells and given a single intravenous injection of 90Y-NM600, in combination with ADT (degarelix). The combination and sequence of administration were evaluated for effect on tumor growth and infiltrating immune populations were analyzed by flow cytometry. Sera were assessed to determine treatment effects on cytokine profiles. RESULTS: ADT delivered prior to TRT (ADT→TRT) resulted in significantly greater antitumor response and overall survival than if delivered after TRT (TRT→ADT). Studies conducted in immunodeficient NRG mice failed to show a difference in treatment sequence, suggesting an immunological mechanism. Myeloid-derived suppressor cells (MDSCs) significantly accumulated in tumors following TRT→ADT treatment and retained immune suppressive function. However, CD4+ and CD8+ T cells with an activated and memory phenotype were more prevalent in the ADT→TRT group. Depletion of Gr1+MDSCs led to greater antitumor response following either treatment sequence. Chemotaxis assays suggested that tumor cells secreted chemokines that recruited MDSCs, notably CXCL1 and CXCL2. The use of a selective CXCR2 antagonist, reparixin, further improved antitumor responses and overall survival when used in tumor-bearing mice treated with TRT→ADT. CONCLUSION: The combination of ADT and TRT improved antitumor responses in murine models of prostate cancer, however, this was dependent on the order of administration. This was found to be associated with one treatment sequence leading to an increase in infiltrating MDSCs. Combining treatment with a CXCR2 antagonist improved the antitumor effect of this combination, suggesting a possible approach for treating advanced human prostate cancer.


Sujet(s)
Cellules myéloïdes suppressives , Tumeurs de la prostate , Animaux , Mâle , Cellules myéloïdes suppressives/effets des médicaments et des substances chimiques , Cellules myéloïdes suppressives/métabolisme , Cellules myéloïdes suppressives/immunologie , Souris , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/radiothérapie , Radiopharmaceutiques/usage thérapeutique , Radiopharmaceutiques/pharmacologie , Humains , Lignée cellulaire tumorale , Radio-isotopes de l'yttrium/usage thérapeutique , Radio-isotopes de l'yttrium/pharmacologie , Modèles animaux de maladie humaine , Antagonistes des androgènes/usage thérapeutique , Antagonistes des androgènes/pharmacologie , Association thérapeutique
18.
J Med Virol ; 96(3): e29540, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38529542

RÉSUMÉ

The sex disparity in COVID-19 outcomes with males generally faring worse than females has been associated with the androgen-regulated expression of the protease TMPRSS2 and the cell receptor ACE2 in the lung and fueled interest in antiandrogens as potential antivirals. In this study, we explored enzalutamide, an antiandrogen used commonly to treat prostate cancer, as a potential antiviral against the human coronaviruses which cause seasonal respiratory infections (HCoV-NL63, -229E, and -OC43). Using lentivirus-pseudotyped and authentic HCoV, we report that enzalutamide reduced 229E and NL63 entry and infection in both TMPRSS2- and nonexpressing immortalized cells, suggesting a TMPRSS2-independent mechanism. However, no effect was observed against OC43. To decipher this distinction, we performed RNA-sequencing analysis on 229E- and OC43-infected primary human airway cells. Our results show a significant induction of androgen-responsive genes by 229E compared to OC43 at 24 and 72 h postinfection. The virus-mediated effect on AR-signaling was further confirmed with a consensus androgen response element-driven luciferase assay in androgen-depleted MRC-5 cells. Specifically, 229E induced luciferase-reporter activity in the presence and absence of the synthetic androgen mibolerone, while OC43 inhibited induction. These findings highlight a complex interplay between viral infections and androgen-signaling, offering insights for disparities in viral outcomes and antiviral interventions.


Sujet(s)
Androgènes , Benzamides , Coronavirus humain 229E , Nitriles , 3-Phényl-2-thiohydantoïne , Mâle , Femelle , Humains , Androgènes/métabolisme , Androgènes/pharmacologie , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/métabolisme , Saisons , Antiviraux/pharmacologie , Antiviraux/métabolisme , Luciferases
19.
Mol Carcinog ; 63(6): 1051-1063, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38482990

RÉSUMÉ

Men with prostate cancer are at increased risk of developing cognitive decline by the use of second-generation androgen signaling inhibitors. To date, reliable and sensitive biomarkers that could distinguish men at high risk of cognitive dysfunction under androgen deprivation therapy (ADT) have not been characterized. We used high-throughput transcriptional profiling utilizing human prostate cancer cell culture models mimicking ADT, biomarker selection using minimal common oncology data elements-cytoscape, and bioinformatic analyses employing Advaita® iPathwayGuide and DisGeNET for identification of disease-related gene associations. Validation analysis of genes was performed on brain neuronal and glial cells by quantitative real-time polymerase chain reaction assay. Our systematic analysis of androgen deprivation-associated genes involved multiple biological processes, including neuroactive ligand-receptor interaction, axon guidance, cytokine-cytokine receptor interaction, and metabolic and cancer signaling pathways. Genes associated with neuroreceptor ligand interaction, including gamma-aminobutyric acid (GABA) A and B receptors and nuclear core proteins, were identified as top upstream regulators. Functional enrichment and protein-protein interaction network analysis highlighted the role of ligand-gated ion channels (LGICs) and their receptors in cognitive dysfunction. Gene-disease association assigned forgetfulness, intellectual disability, visuospatial deficit, bipolar disorder, and other neurocognitive impairment with upregulation of type-1 angiotensin II receptor, brain-derived neurotrophic factor, GABA type B receptor subunit 2 (GABBR2), GABRA3, GABRA5, GABRB1, glycine receptor beta, glutamate ionotropic receptor N-methyl-D-aspartate receptor (NMDA) type subunit 1, glutamate ionotropic receptor NMDA type subunit 2D, 5-hydroxytryptamine receptor 1D, interferon beta 1, and nuclear receptor subfamily 3 group C member 1 as top differentially expressed genes. Validation studies of brain glial cells, neurons, and patients on ADT demonstrated the association of these genes with cognitive decline. Our findings highlight LGICs as potential biomarkers for ADT-mediated cognitive decline. Further validation of these biomarkers may lead to future practical clinical use.


Sujet(s)
Dysfonctionnement cognitif , Tumeurs de la prostate , Humains , Mâle , Dysfonctionnement cognitif/induit chimiquement , Dysfonctionnement cognitif/génétique , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Antagonistes des androgènes/effets indésirables , Antagonistes des androgènes/pharmacologie , Lignée cellulaire tumorale , Canaux ioniques/génétique , Canaux ioniques/métabolisme , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Cartes d'interactions protéiques
20.
BMC Cancer ; 24(1): 346, 2024 Mar 19.
Article de Anglais | MEDLINE | ID: mdl-38500100

RÉSUMÉ

BACKGROUND: The androgen/androgen receptor (AR)-signaling axis plays a central role in prostate cancer (PCa). Upon androgen-binding the AR dimerizes with another AR, and translocates into the nucleus where the AR-dimer activates/inactivates androgen-dependent genes. Consequently, treatments for PCa are commonly based on androgen deprivation therapy (ADT). The clinical benefits of ADT are only transitory and most tumors develop mechanisms allowing the AR to bypass its need for physiological levels of circulating androgens. Clinical failure of ADT is often characterized by the synthesis of a constitutively active AR splice variant, termed AR-V7. AR-V7 mRNA expression is considered as a resistance mechanism following ADT. AR-V7 no longer needs androgenic stimuli for nuclear entry and/or dimerization. METHODS: Our goal was to mechanistically decipher the interaction between full-length AR (AR-FL) and AR-V7 in AR-null HEK-293 cells using the NanoLuc Binary Technology under androgen stimulation and deprivation conditions. RESULTS: Our data point toward a hypothesis that AR-FL/AR-FL homodimers form in the cytoplasm, whereas AR-V7/AR-V7 homodimers localize in the nucleus. However, after androgen stimulation, all the AR-FL/AR-FL, AR-FL/AR-V7 and AR-V7/AR-V7 dimers were localized in the nucleus. CONCLUSIONS: We showed that AR-FL and AR-V7 form heterodimers that localize to the nucleus, whereas AR-V7/AR-V7 dimers were found to localize in the absence of androgens in the nucleus.


Sujet(s)
Luciferases , Tumeurs prostatiques résistantes à la castration , Tumeurs de la prostate , Mâle , Humains , Récepteurs aux androgènes/génétique , Récepteurs aux androgènes/métabolisme , Androgènes , Tumeurs de la prostate/anatomopathologie , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Cellules HEK293 , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Isoformes de protéines/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE