Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.211
Filtrer
1.
Chem Biol Drug Des ; 104(1): e14583, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38991995

RÉSUMÉ

In this work, a series of curcumin derivatives (1a-1h, 2a-2g, and 3a-3c) were synthesized for the suppression of castration-resistant prostate cancer cells. All synthesized compounds were characterized by 1H NMR, 13C NMR, HRMS, and melting point. The in vitro cytotoxicity study shows that compounds 1a, 1e, 1f, 1h, 2g, 3a, and 3c display similar or enhanced cytotoxicity against 22Rv1 and C4-2 cells as compared to ASC-J9, other synthesized compounds display reduced cytotoxicity against 22Rv1 and C4-2 cells as compared to ASC-J9. Molecular docking simulation was performed to study the binding affinity and probable binding modes of the synthesized compounds with androgen receptor. The results show that all synthesized compounds exhibit higher cdocker interaction energies as compared to ASC-J9. Compounds 1h, 2g, and 3c not only show strong cytotoxicity against 22Rv1 and C4-2 cells but also exhibit high binding affinity with androgen receptor. In androgen receptor suppression study, compounds 1f and 2g show similar androgen receptor suppression effect as compared to ASC-J9 on C4-2 cells, compound 3c displays significantly enhanced AR suppression effect as compared to ASC-J9, 1f and 2g. Compounds 1a, 1e, 1f, 1h, 2g, 3a and 3c prepared in this work have significant potential for castration-resistant prostate cancer therapy.


Sujet(s)
Curcumine , Simulation de docking moléculaire , Tumeurs prostatiques résistantes à la castration , Récepteurs aux androgènes , Curcumine/pharmacologie , Curcumine/composition chimique , Curcumine/synthèse chimique , Curcumine/métabolisme , Mâle , Humains , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/composition chimique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Antagonistes du récepteur des androgènes/pharmacologie , Antagonistes du récepteur des androgènes/composition chimique , Antagonistes du récepteur des androgènes/synthèse chimique , Antagonistes du récepteur des androgènes/métabolisme , Sites de fixation , Liaison aux protéines
2.
Rom J Morphol Embryol ; 65(2): 209-216, 2024.
Article de Anglais | MEDLINE | ID: mdl-39020535

RÉSUMÉ

Triple-negative breast cancer (BC) represents an extensively analyzed entity to establish the overall framework of clinicopathological characteristics, with an impact on defining prognostic and predictive factors. The relationship between triple-negative BC and androgen receptor (AR) is far from being clarified. We aimed to evaluate the classical clinicopathological spectrum that characterized a triple-negative BC, focusing on AR expression. The study group comprised 124 cases of triple-negative BC. The main clinicopathological parameters were extracted from medical records. The immunohistochemical (IHC) exam was run using the following antibodies: anti-estrogen receptor (ER), anti-progesterone receptor (PR), anti-human epidermal growth factor receptor (HER2∕neu), anti-Ki67 and anti-AR. AR immunoexpression was assessed as absent (completely negative) or present (unrelated to percentages and intensity). Data were statistically analyzed. AR expression was positive in 78 (63%) cases and negative in 46 (37%) cases. Among the study group, 28 cases exhibited an AR percentage ranging from 1% to 10%, 15 cases showed a percentage between 11% and 50%, while 12 cases had AR values between 51% and 75% and 23 cases fell within the AR range of 76% to 100%. No significant differences between AR immunoexpression (negative versus positive), clinicopathological characteristics and survival parameters were found. Statistically significant differences were registered between histological type, tumor stage, distant metastasis, tumor-infiltrating lymphocytes (TILs), treatment and residual cancer burden (RCB), and survival parameters. Thus, our results sustain that AR does not affect the biological behavior of triple-negative BC.


Sujet(s)
Récepteurs aux androgènes , Tumeurs du sein triple-négatives , Humains , Récepteurs aux androgènes/métabolisme , Femelle , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Adulte d'âge moyen , Adulte , Sujet âgé , Immunohistochimie/méthodes
4.
Sci Rep ; 14(1): 16302, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009627

RÉSUMÉ

Androgen insensitivity syndrome (AIS) is a difference of sex development (DSD) characterized by different degrees of undervirilization in individuals with a 46,XY karyotype despite normal to high gonadal testosterone production. Classically, AIS is explained by hemizygous mutations in the X-chromosomal androgen receptor (AR) gene. Nevertheless, the majority of individuals with clinically diagnosed AIS do not carry an AR gene mutation. Here, we present a patient with a 46,XY karyotype, born with undervirilized genitalia, age-appropriate testosterone levels and no uterus, characteristic for AIS. Diagnostic whole exome sequencing (WES) showed a maternally inherited LINE1 (L1) retrotransposon insertion in the 5' untranslated region (5'UTR) of the AR gene. Long-read nanopore sequencing confirmed this as an insertion of a truncated L1 element of ≈ 2.7 kb and showed an increased DNA methylation at the L1 insertion site in patient-derived genital skin fibroblasts (GSFs) compared to healthy controls. The insertion coincided with reduced AR transcript and protein levels in patient-derived GSFs confirming the clinical diagnosis AIS. Our results underline the relevance of retrotransposons in human disease, and expand the growing list of human diseases associated with them.


Sujet(s)
Syndrome d'insensibilité aux androgènes , Méthylation de l'ADN , Épigenèse génétique , Éléments LINE , Récepteurs aux androgènes , Humains , Syndrome d'insensibilité aux androgènes/génétique , Syndrome d'insensibilité aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Récepteurs aux androgènes/métabolisme , Mâle , Éléments LINE/génétique , Femelle , , Transcription génétique
5.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-39000396

RÉSUMÉ

Latrophilins (LPHNs), a group of the G-protein-coupled receptor to which a spider venom latrotoxin (LTX) is known to bind, remain largely uncharacterized in neoplastic diseases. In the present study, we aimed to determine the role of LPHNs in the progression of prostate cancer. We assessed the actions of LPHNs, including LPHN1, LPHN2, and LPHN3, in human prostate cancer lines via their ligand (e.g., α-LTX, FLRT3) treatment or shRNA infection, as well as in surgical specimens. In androgen receptor (AR)-positive LNCaP/C4-2/22Rv1 cells, dihydrotestosterone considerably increased the expression levels of LPHNs, while chromatin immunoprecipitation assay revealed the binding of endogenous ARs, including AR-V7, to the promoter region of each LPHN. Treatment with α-LTX or FLRT3 resulted in induction in the cell viability and migration of both AR-positive and AR-negative lines. α-LTX and FLRT3 also enhanced the expression of Bcl-2 and phosphorylated forms of JAK2 and STAT3. Meanwhile, the knockdown of each LPHN showed opposite effects on all of those mediated by ligand treatment. Immunohistochemistry in radical prostatectomy specimens further showed the significantly elevated expression of each LPHN in prostate cancer, compared with adjacent normal-appearing prostate, which was associated with a significantly higher risk of postoperative biochemical recurrence in both univariate and multivariable settings. These findings indicate that LPHNs function as downstream effectors of ARs and promote the growth of androgen-sensitive, castration-resistant, or even AR-negative prostate cancer.


Sujet(s)
Évolution de la maladie , Tumeurs de la prostate , Récepteurs aux androgènes , Mâle , Humains , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Lignée cellulaire tumorale , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Mouvement cellulaire/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux , Kinase Janus-2/métabolisme , Kinase Janus-2/génétique , Récepteurs peptidiques/métabolisme , Récepteurs peptidiques/génétique , Isoformes de protéines/métabolisme , Isoformes de protéines/génétique , Transduction du signal , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Épissage alternatif
6.
Nat Commun ; 15(1): 5629, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965223

RÉSUMÉ

Mutations that decrease or increase the activity of the tyrosine phosphatase, SHP2 (encoded by PTPN11), promotes developmental disorders and several malignancies by varying phosphatase activity. We uncovered that SHP2 is a distinct class of an epigenetic enzyme; upon phosphorylation by the kinase ACK1/TNK2, pSHP2 was escorted by androgen receptor (AR) to chromatin, erasing hitherto unidentified pY54-H3 (phosphorylation of histones H3 at Tyr54) epigenetic marks to trigger a transcriptional program of AR. Noonan Syndrome with Multiple Lentigines (NSML) patients, SHP2 knock-in mice, and ACK1 knockout mice presented dramatic increase in pY54-H3, leading to loss of AR transcriptome. In contrast, prostate tumors with high pSHP2 and pACK1 activity exhibited progressive downregulation of pY54-H3 levels and higher AR expression that correlated with disease severity. Overall, pSHP2/pY54-H3 signaling acts as a sentinel of AR homeostasis, explaining not only growth retardation, genital abnormalities and infertility among NSML patients, but also significant AR upregulation in prostate cancer patients.


Sujet(s)
Épigenèse génétique , Histone , Homéostasie , Souris knockout , Tumeurs de la prostate , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Récepteurs aux androgènes , Animaux , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Histone/métabolisme , Mâle , Humains , Souris , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Phosphorylation , Syndrome de Noonan/génétique , Syndrome de Noonan/métabolisme , Transduction du signal , Chromatine/métabolisme
7.
Breast Cancer Res ; 26(1): 111, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965614

RÉSUMÉ

BACKGROUND: Endocrine therapy is the most important treatment modality of breast cancer patients whose tumors express the estrogen receptor α (ERα). The androgen receptor (AR) is also expressed in the vast majority (80-90%) of ERα-positive tumors. AR-targeting drugs are not used in clinical practice, but have been evaluated in multiple trials and preclinical studies. METHODS: We performed a genome-wide study to identify hormone/drug-induced single nucleotide polymorphism (SNP) genotype - dependent gene-expression, known as PGx-eQTL, mediated by either an AR agonist (dihydrotestosterone) or a partial antagonist (enzalutamide), utilizing a previously well characterized lymphoblastic cell line panel. The association of the identified SNPs-gene pairs with breast cancer phenotypes were then examined using three genome-wide association (GWAS) studies that we have published and other studies from the GWAS catalog. RESULTS: We identified 13 DHT-mediated PGx-eQTL loci and 23 Enz-mediated PGx-eQTL loci that were associated with breast cancer outcomes post ER antagonist or aromatase inhibitors (AI) treatment, or with pharmacodynamic (PD) effects of AIs. An additional 30 loci were found to be associated with cancer risk and sex-hormone binding globulin levels. The top loci involved the genes IDH2 and TMEM9, the expression of which were suppressed by DHT in a PGx-eQTL SNP genotype-dependent manner. Both of these genes were overexpressed in breast cancer and were associated with a poorer prognosis. Therefore, suppression of these genes by AR agonists may benefit patients with minor allele genotypes for these SNPs. CONCLUSIONS: We identified AR-related PGx-eQTL SNP-gene pairs that were associated with risks, outcomes and PD effects of endocrine therapy that may provide potential biomarkers for individualized treatment of breast cancer.


Sujet(s)
Tumeurs du sein , Étude d'association pangénomique , Polymorphisme de nucléotide simple , Locus de caractère quantitatif , Récepteurs aux androgènes , Humains , Tumeurs du sein/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Récepteurs aux androgènes/génétique , Récepteurs aux androgènes/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , 5alpha-Dihydrotestostérone/pharmacologie , 3-Phényl-2-thiohydantoïne/pharmacologie , 3-Phényl-2-thiohydantoïne/usage thérapeutique , Nitriles/usage thérapeutique , Génotype , Pharmacogénétique/méthodes , Variants pharmacogénomiques , Antinéoplasiques hormonaux/usage thérapeutique , Antinéoplasiques hormonaux/pharmacologie , Benzamides
8.
Invest Ophthalmol Vis Sci ; 65(8): 5, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38958971

RÉSUMÉ

Purpose: The purpose of this study was to investigate the presence of sex-steroid receptors in human choroidal tissue across different ages and sex, aiming to better understand the pronounced sex difference in central serous chorioretinopathy (CSC) occurrence. Methods: Paraffin-embedded enucleated eyes of 14 premenopausal women, 15 postmenopausal women, 10 young men (<45 years), and 10 older men (>60 years) were used. A clinically certified immunostaining was performed to detect the presence of the androgen receptor (AR), progesterone receptor (PR; isoform A and B), and estrogen receptor (ERα). The stained slides were scored in a blinded manner for positive endothelial cells and stromal cells in consecutive sections of the same choroidal region. Results: Our analysis revealed the presence of AR, PR, and ERα in endothelial cells and stromal cells of choroidal tissue. The mean proportion of AR-positive endothelial cells was higher in young men (46% ± 0.15) compared to aged-matched women (29% ± 0.12; P < 0.05, 95% confidence interval [CI]). Premenopausal women showed markedly lower mean proportion of ERα (5% ± 0.02) and PR-positive endothelial cells (2% ± 0.01) compared to postmenopausal women (15% ± 0.07 and 19% ± 0.13; both P < 0.05, 95% CI), young men (13% ± 0.04 and 21% ± 0.10; both P < 0.05, 95% CI), and older men (18% ± 0.09 and 27% ± 0.14; both P < 0.05, 95% CI). Mean PR-positive stromal cells were also less present in premenopausal women (12% ± 0.07) than in other groups. Conclusions: The number of sex-steroid receptors in the choroidal tissue differs between men and women across different ages, which aligns with the prevalence patterns of CSC in men and postmenopausal women.


Sujet(s)
Choriorétinopathie séreuse centrale , Choroïde , Récepteurs aux androgènes , Récepteurs à la progestérone , Humains , Femelle , Mâle , Choroïde/métabolisme , Choroïde/anatomopathologie , Adulte d'âge moyen , Adulte , Choriorétinopathie séreuse centrale/métabolisme , Choriorétinopathie séreuse centrale/épidémiologie , Choriorétinopathie séreuse centrale/diagnostic , Récepteurs à la progestérone/métabolisme , Récepteurs aux androgènes/métabolisme , Sujet âgé , Facteurs sexuels , Prévalence , Récepteur alpha des oestrogènes/métabolisme
9.
Radiat Oncol ; 19(1): 85, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956684

RÉSUMÉ

BACKGROUND: Radiotherapy is essential in the treatment of prostate cancer. An alternative to conventional photon radiotherapy is the application of carbon ions, which provide a superior intratumoral dose distribution and less induced damage to adjacent healthy tissue. A common characteristic of prostate cancer cells is their dependence on androgens which is exploited therapeutically by androgen deprivation therapy in the advanced prostate cancer stage. Here, we aimed to analyze the transcriptomic response of prostate cancer cells to irradiation by photons in comparison to carbon ions, focusing on DNA damage, DNA repair and androgen receptor signaling. METHODS: Prostate cancer cell lines LNCaP (functional TP53 and androgen receptor signaling) and DU145 (dysfunctional TP53 and androgen receptor signaling) were irradiated by photons or carbon ions and the subsequent DNA damage was assessed by immuno-cytofluorescence. Furthermore, the cells were treated with an androgen-receptor agonist. The effects of irradiation and androgen treatment on the gene regulation and the transcriptome were investigated by RT-qPCR and RNA sequencing, followed by bioinformatic analysis. RESULTS: Following photon or carbon ion irradiation, both LNCaP and DU145 cells showed a dose-dependent amount of visible DNA damage that decreased over time, indicating occurring DNA repair. In terms of gene regulation, mRNAs involved in the TP53-dependent DNA damage response were significantly upregulated by photons and carbon ions in LNCaP but not in DU145 cells, which generally showed low levels of gene regulation after irradiation. Both LNCaP and DU145 cells responded to photons and carbon ions by downregulation of genes involved in DNA repair and cell cycle, partially resembling the transcriptome response to the applied androgen receptor agonist. Neither photons nor carbon ions significantly affected canonical androgen receptor-dependent gene regulation. Furthermore, certain genes that were specifically regulated by either photon or carbon ion irradiation were identified. CONCLUSION: Photon and carbon ion irradiation showed a significant congruence in terms of induced signaling pathways and transcriptomic responses. These responses were strongly impacted by the TP53 status. Nevertheless, irradiation mode-dependent distinct gene regulations with undefined implication for radiotherapy outcome were revealed. Androgen receptor signaling and irradiations shared regulation of certain genes with respect to DNA-repair and cell-cycle.


Sujet(s)
Photons , Tumeurs de la prostate , Récepteurs aux androgènes , Transduction du signal , Transcriptome , Protéine p53 suppresseur de tumeur , Humains , Mâle , Carbone , Lignée cellulaire tumorale , Altération de l'ADN/effets des radiations , Réparation de l'ADN , Régulation de l'expression des gènes tumoraux/effets des radiations , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Radiothérapie par ions lourds , Tumeurs de la prostate/radiothérapie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Transduction du signal/effets des radiations , Transcriptome/effets des radiations , Protéine p53 suppresseur de tumeur/métabolisme
10.
Sci Adv ; 10(29): eadm9577, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39028815

RÉSUMÉ

Pausing of RNA polymerase II (Pol II) at transcription start sites (TSSs) primes target genes for productive elongation. Coincidentally, DNA double-strand breaks (DSBs) enrich at highly transcribed and Pol II-paused genes, although their interplay remains undefined. Using androgen receptor (AR) signaling as a model, we have uncovered AR-interacting protein 4 (ARIP4) helicase as a driver of androgen-dependent transcription induction. Chromatin immunoprecipitation sequencing analysis revealed that ARIP4 preferentially co-occupies TSSs with paused Pol II. Moreover, we found that ARIP4 complexes with topoisomerase II beta and mediates transient DSB formation upon hormone stimulation. Accordingly, ARIP4 deficiency compromised release of paused Pol II and resulted in R-loop accumulation at a panel of highly transcribed AR target genes. Last, we showed that ARIP4 binds and unwinds R-loops in vitro and that its expression positively correlates with prostate cancer progression. We propose that androgen stimulation triggers ARIP4-mediated unwinding of R-loops at TSSs, enforcing Pol II pause release to effectively drive an androgen-dependent expression program.


Sujet(s)
Androgènes , Tumeurs de la prostate , Structures en boucle R , RNA polymerase II , Récepteurs aux androgènes , Humains , Androgènes/métabolisme , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Mâle , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , RNA polymerase II/métabolisme , RNA polymerase II/génétique , Lignée cellulaire tumorale , ADN topoisomérases de type II/métabolisme , ADN topoisomérases de type II/génétique , Transcription génétique , Cassures double-brin de l'ADN , Site d'initiation de la transcription , Régulation de l'expression des gènes tumoraux , Liaison aux protéines , Activation de la transcription
11.
Ann Afr Med ; 23(3): 452-458, 2024 Jul 01.
Article de Français, Anglais | MEDLINE | ID: mdl-39034572

RÉSUMÉ

BACKGROUND: Despite the advancement in therapy, breast cancer still remains the most common malignancy in women globally due in part to its heterogeneity. Triple-negative breast cancer (TNBC) represents up to 20% of all breast cancer variants, an aggressive disease with poorer outcomes compared to other breast cancer subtypes. No targeted therapies are currently approved for TNBC, and newer treatment approaches are seriously needed. Androgen receptor (AR), another hormonal receptor, is often expressed in breast cancer, and its role depends on the relative levels of circulating estrogens and androgens. This study aimed to assess the expression of AR in breast cancer in a tertiary hospital in Ghana. METHODOLOGY: Immunohistochemical staining for AR was performed on tissue microarray (TMA) blocks, of which estrogen receptor, progesterone receptor, and Her-2/neu had already been done. 197 cases were suitable for the study. Results from the immunostaining were analyzed using the SPSS version 23 for descriptive statistics and correlations (χ2 and Pearson tests). RESULTS: 197 TMA cases were used. TNBCs constitute 61.9% of the cancers. The majority of these tumors were grade III, ductal carcinoma NST. The mean age was 49.86 ± 14.09, and the modal age group was 40-49 years. Our cases showed 23% AR expression in triple-negative cancers. The study also established that AR is more frequently expressed in low-grade tumors compared to high-grade ones. CONCLUSION: There is an appreciable level of AR expression in our cases; however, most are quadruple negative. However, AR is more frequently expressed in low-grade tumors than high-grade ones.


Résumé Contexte:Malgré les progrès thérapeutiques, le cancer du sein reste la tumeur maligne la plus courante chez les femmes dans le monde, en partie à cause de son hétérogénéité. Le cancer du sein triple négatif (CSTN) représente jusqu'à 20 % de toutes les variantes du cancer du sein, une maladie agressive dont les résultats sont moins bons que les autres sous-types de cancer du sein. Aucun traitement ciblé n'est actuellement approuvé pour le TNBC et de nouvelles approches thérapeutiques sont sérieusement nécessaires. Le récepteur aux androgènes (AR), un autre récepteur hormonal, est souvent exprimé dans le cancer du sein et son rôle dépend des niveaux relatifs d'œstrogènes et d'androgènes en circulation. Cette étude vise à évaluer l'expression de la RA dans le cancer du sein dans un hôpital tertiaire du Ghana.Méthodes:La coloration immunohistochimique pour AR a été réalisée sur des blocs de micropuces tissulaires (TMA) dont ER, PR, Her-2/neu avaient déjà été réalisés. 197 cas convenaient à l'étude. Les résultats de l'immunocoloration ont été analysés à l'aide de SPSS version 23 pour les statistiques descriptives et les corrélations (tests X2 et Pearson).Résultats:197 cas de TMA ont été utilisés. Les TNBC constituent 61,9 % des cancers. La majorité de ces tumeurs étaient des carcinomes canalaires NST de grade III. L'âge moyen était de 49,86 ± 14,09 et la tranche d'âge modale était celle des 40-49 ans. Nos cas ont montré une expression de 23 % de AR dans les cancers triples négatifs. L'étude a également établi que la RA est plus fréquemment exprimée dans les tumeurs de bas grade que dans celles de haut grade.Conclusion:Il existe un niveau appréciable d'expression des AR dans nos cas mais la plupart sont quadruple négatifs. Cependant, la RA est plus fréquemment exprimée dans les tumeurs de bas grade que dans celles de haut grade.


Sujet(s)
Marqueurs biologiques tumoraux , Immunohistochimie , Récepteurs aux androgènes , Analyse sur puce à tissus , Tumeurs du sein triple-négatives , Humains , Récepteurs aux androgènes/métabolisme , Femelle , Ghana/épidémiologie , Adulte d'âge moyen , Adulte , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Récepteurs à la progestérone/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Récepteurs des oestrogènes/métabolisme , Sujet âgé , Études de cohortes , Récepteur ErbB-2/métabolisme , Grading des tumeurs
12.
J Exp Clin Cancer Res ; 43(1): 194, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39014480

RÉSUMÉ

Continued exploration of the androgen receptor (AR) is crucial, as it plays pivotal roles in diverse diseases such as prostate cancer (PCa), serving as a significant therapeutic focus. Therefore, the Department of Urology Dresden hosted an international meeting for scientists and clinical oncologists to discuss the newest advances in AR research. The 2nd International Androgen Receptor Symposium was held in Dresden, Saxony, Germany, from 26-27.04.2024, organised by Dr. Holger H.H. Erb. Following the format of the first meeting, more than 35 scientists from 8 countries attended the event to discuss recent developments, research challenges, and identification of venues in AR research. An important new feature was the involvement of PhD students and young investigators, acknowledging the high scientific quality of their work. The symposium included three covers: new advances from clinical research, basic and translational research, and novel strategies to target AR. Moreover, based on its increasing clinical relevance, a PSMA theranostic mini-symposium was added at the end of the AR symposium to allow the audience to discuss the newest advances in PSMA theranostic. This report focuses on the highlights and discussions of the meeting.


Sujet(s)
Tumeurs de la prostate , Récepteurs aux androgènes , Humains , Mâle , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/thérapie , Tumeurs de la prostate/génétique , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique
13.
Front Endocrinol (Lausanne) ; 15: 1437179, 2024.
Article de Anglais | MEDLINE | ID: mdl-39027480

RÉSUMÉ

Prostate cancer is one of the most prevalent malignancies and is primarily driven by aberrant androgen receptor (AR) signaling. While AR-targeted therapies form the cornerstone of prostate cancer treatment, they often inadvertently activate compensatory pathways, leading to therapy resistance. This resistance is frequently mediated through changes in transcription factor (TF) crosstalk, reshaping gene regulatory programs and ultimately weakening treatment efficacy. Consequently, investigating TF interactions has become crucial for understanding the mechanisms driving therapy-resistant cancers. Recent evidence has highlighted the crosstalk between the glucocorticoid receptor (GR) and AR, demonstrating that GR can induce prostate cancer therapy resistance by replacing the inactivated AR, thereby becoming a driver of the disease. In addition to this oncogenic role, GR has also been shown to act as a tumor suppressor in prostate cancer. Owing to this dual role and the widespread use of glucocorticoids as adjuvant therapy, it is essential to understand GR's actions across different stages of prostate cancer development. In this review, we explore the current knowledge of GR in prostate cancer, with a specific focus on its crosstalk with other TFs. GR can directly and indirectly interact with a variety of TFs, and these interactions vary significantly depending on the type of prostate cancer cells. By highlighting these crosstalk interactions, we aim to provide insights that can guide the research and development of new GR-targeted therapies to mitigate its harmful effects in prostate cancer.


Sujet(s)
Tumeurs de la prostate , Récepteurs aux glucocorticoïdes , Facteurs de transcription , Humains , Récepteurs aux glucocorticoïdes/métabolisme , Récepteurs aux glucocorticoïdes/génétique , Mâle , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/génétique , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Transduction du signal , Animaux , Interactions entre récepteurs/physiologie , Régulation de l'expression des gènes tumoraux
14.
Biol Sex Differ ; 15(1): 53, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987854

RÉSUMÉ

BACKGROUND: Polycystic ovary syndrome (PCOS), the most common endocrine disorder in premenopausal women, is associated with increased obesity, hyperandrogenism, and altered brown adipose tissue (BAT) thermogenesis. MicroRNAs play critical functions in brown adipocyte differentiation and maintenance. We aim to study the role of microRNA-21 (miR-21) in altered energy homeostasis and BAT thermogenesis in a PCOS mouse model of peripubertal androgen exposure. METHODS: Three-week-old miR-21 knockout (miR21KO) or wild-type (WT) female mice were treated with dihydrotestosterone (DHT) or vehicle for 90 days. Body composition was determined by EchoMRI. Energy expenditure (EE), oxygen consumption (VO2), carbon dioxide production (VCO2), and respiratory exchange ratio (RER) were measured by indirect calorimetry. Androgen receptor (AR), and markers of adipogenesis, de novo lipogenesis, angiogenesis, extracellular matrix remodeling, and thermogenesis were quantified by RT-qPCR and/or Western-blot. RESULTS: MiR-21 ablation attenuated DHT-mediated increase in body weight while having no effect on fat or BAT mass. MiR-21 ablation attenuated DHT-mediated BAT AR upregulation. MiR-21 ablation did not alter EE; however, miR21KO DHT-treated mice have reduced VO2, VCO2, and RER. MiR-21 ablation reversed DHT-mediated decrease in food intake and increase in sleep time. MiR-21 ablation decreased some adipogenesis (Adipoq, Pparγ, and Cebpß) and extracellular matrix remodeling (Mmp-9 and Timp-1) markers expression in DHT-treated mice. MiR-21 ablation abolished DHT-mediated increases in thermogenesis markers Cpt1a and Cpt1b, while decreasing CIDE-A expression. CONCLUSIONS: Our findings suggest that BAT miR-21 may play a role in regulating DHT-mediated thermogenic dysfunction in PCOS. Modulation of BAT miR-21 levels could be a novel therapeutic approach for the treatment of PCOS-associated metabolic derangements.


Polycystic ovary syndrome (PCOS) is a common hormone disorder in premenopausal women, often linked to obesity and abnormal brown fat tissue activity. Women with PCOS have elevated male hormones, which are responsible for many metabolic problems. Our study focuses on understanding the role of microRNA-21 (miR-21) in the energy balance and brown fat tissue activity in a PCOS mouse model. We studied female mice with and without miR-21, treating them with a male hormone. We measured body composition and energy expenditure. We also analyzed the levels of specific genes and proteins related to fat tissue and energy production. Our findings showed that mice lacking miR-21 had less weight gain in response to male hormones, without fat or brown fat tissue mass changes. They also had reduced energy production, changed eating habits, and altered expression of genes related to fat tissue and energy production. In conclusion, our study suggests that miR-21 in brown fat tissue may regulate the energy imbalance caused by male hormones in PCOS. Adjusting miR-21 levels in brown fat tissue could be a new way to address the metabolic issues associated with PCOS.


Sujet(s)
Adipogenèse , Tissu adipeux brun , Modèles animaux de maladie humaine , Souris knockout , microARN , Syndrome des ovaires polykystiques , Thermogenèse , Animaux , Syndrome des ovaires polykystiques/métabolisme , Femelle , microARN/métabolisme , microARN/génétique , Tissu adipeux brun/métabolisme , 5alpha-Dihydrotestostérone/pharmacologie , Souris , Souris de lignée C57BL , Récepteurs aux androgènes/métabolisme
15.
Proc Natl Acad Sci U S A ; 121(29): e2321408121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38976730

RÉSUMÉ

Spinal and bulbar muscular atrophy (SBMA) is a slowly progressing neuromuscular disease caused by a polyglutamine (polyQ)-encoding CAG trinucleotide repeat expansion in the androgen receptor (AR) gene, leading to AR aggregation, lower motor neuron death, and muscle atrophy. AR is a ligand-activated transcription factor that regulates neuronal architecture and promotes axon regeneration; however, whether AR transcriptional functions contribute to disease pathogenesis is not fully understood. Using a differentiated PC12 cell model of SBMA, we identified dysfunction of polyQ-expanded AR in its regulation of neurite growth and maintenance. Specifically, we found that in the presence of androgens, polyQ-expanded AR inhibited neurite outgrowth, induced neurite retraction, and inhibited neurite regrowth. This dysfunction was independent of polyQ-expanded AR transcriptional activity at androgen response elements (ARE). We further showed that the formation of polyQ-expanded AR intranuclear inclusions promoted neurite retraction, which coincided with reduced expression of the neuronal differentiation marker ß-III-Tubulin. Finally, we revealed that cell death is not the primary outcome for cells undergoing neurite retraction; rather, these cells become senescent. Our findings reveal that mechanisms independent of AR canonical transcriptional activity underly neurite defects in a cell model of SBMA and identify senescence as a pathway implicated in this pathology. These findings suggest that in the absence of a role for AR canonical transcriptional activity in the SBMA pathologies described here, the development of SBMA therapeutics that preserve this activity may be desirable. This approach may be broadly applicable to other polyglutamine diseases such as Huntington's disease and spinocerebellar ataxias.


Sujet(s)
Neurites , Récepteurs aux androgènes , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Animaux , Neurites/métabolisme , Rats , Cellules PC12 , Vieillissement de la cellule , Peptides/métabolisme , Humains , Amyotrophies/métabolisme , Amyotrophies/génétique , Amyotrophies/anatomopathologie , Mutation , Amyotrophie spinale/métabolisme , Amyotrophie spinale/génétique , Amyotrophie spinale/anatomopathologie
16.
Adv Cancer Res ; 161: 403-429, 2024.
Article de Anglais | MEDLINE | ID: mdl-39032955

RÉSUMÉ

Worldwide, prostate cancer (PCa) remains a leading cause of death in men. Histologically, the majority of PCa cases are classified as adenocarcinomas, which are mainly composed of androgen receptor-positive luminal cells. PCa is initially driven by the androgen receptor axis, where androgen-mediated activation of the receptor is one of the primary culprits for disease progression. Therefore, in advanced stage PCa, patients are generally treated with androgen deprivation therapies alone or in combination with androgen receptor pathway inhibitors. However, after an initial decrease, the cancer recurs for majority patients. At this stage, cancer is known as castration-resistant prostate cancer (CRPC). Majority of CRPC tumors still depend on androgen receptor axis for its progression to metastasis. However, in around 20-30% of cases, CRPC progresses via an androgen receptor-independent pathway and is often presented as neuroendocrine cancer (NE). This NE phenotype is highly aggressive with poor overall survival as compared to CRPC adenocarcinoma. NE cancers are resistant to standard taxane chemotherapies, which are often used to treat metastatic disease. Pathologically and morphologically, NE cancers are highly diverse and often co-exist with adenocarcinoma. Due to the lack of proper biomarkers, it is often difficult to make an early diagnosis of this lethal disease. Moreover, increased tumor heterogeneity and admixtures of adeno and NE subtypes in the same tumor make early detection of NE tumors very difficult. With the advancement of our knowledge and sequencing technology, we are now able to better understand the molecular mediators of this transformation pathway. This current study will give an update on how various molecular regulators are involved in these lineage transformation processes and what challenges we are still facing to detect and treat this cancer.


Sujet(s)
Tumeurs neuroendocrines , Humains , Mâle , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/métabolisme , Tumeurs neuroendocrines/génétique , Récepteurs aux androgènes/métabolisme , 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 , Animaux , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/génétique , Régulation de l'expression des gènes tumoraux
17.
Genome Med ; 16(1): 92, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39044302

RÉSUMÉ

BACKGROUND: Black men are at a higher risk of prostate cancer (PC) diagnosis and present with more high-grade PC than White men in an equal access setting. This study aimed to identify differential transcriptional regulation between Black and White men with PC. METHODS: We performed microarray of radical prostatectomy tissue blocks from 305 Black and 238 White men treated at the Durham Veterans Affairs Medical Center. Differential expression, gene set enrichment analysis, master regulator analysis, and network modeling were conducted to compare gene expression by race. Findings were validated using external datasets that are available in the Gene Expression Omnibus (GEO) database. The first was a multi-institutional cohort of 1152 prostate cancer patients (596 Black, 556 White) with microarray data (GEO ID: GSE169038). The second was an Emory cohort of 106 patients (22 Black, 48 White, 36 men of unknown race) with RNA-seq data (GEO ID: GSE54460). Additionally, we analyzed androgen receptor (AR) chromatin binding profiles using paired AR ChIP-Seq datasets from Black and White men (GEO IDs: GSE18440 and GSE18441). RESULTS: We identified 871 differentially expressed genes between Black and White men. White men had higher activity of MYC-related pathways, while Black men showed increased activity of inflammation, steroid hormone responses, and cancer progression-related pathways. We further identified the top 10 transcription factors (TFs) in Black patients, which formed a transcriptional regulatory network centered on the AR. The activities of this network and the pathways were significantly different in Black vs. White men across multiple cohorts and PC molecular subtypes. CONCLUSIONS: These findings suggest PC in Black and White men have distinct tumor transcriptional profiles. Furthermore, a highly interactive TF network centered on AR drives differential gene expression in Black men. Additional study is needed to understand the degree to which these differences in transcriptional regulatory elements contribute to PC health disparities.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Tumeurs de la prostate , Sujet âgé , Humains , Mâle , Adulte d'âge moyen , /génétique , Analyse de profil d'expression de gènes , Réseaux de régulation génique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Récepteurs aux androgènes/génétique , Récepteurs aux androgènes/métabolisme , Transcriptome , Blanc/génétique
18.
J Med Chem ; 67(14): 11732-11750, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38991141

RÉSUMÉ

Androgen receptor (AR) signaling plays a key role in the progression of prostate cancer. This study describes the discovery and optimization of a novel series of AR PROTAC degraders that recruit the Cereblon (CRBN) E3 ligase. Having identified a series of AR ligands based on 4-(4-phenyl-1-piperidyl)-2-(trifluoromethyl)benzonitrile, our PROTAC optimization strategy focused on linker connectivity and CRBN ligand SAR to deliver potent degradation of AR in LNCaP cells. This work culminated in compounds 11 and 16 which demonstrated good rodent oral bioavailability. Subsequent SAR around the AR binding region brought in an additional desirable feature, degradation of the important treatment resistance mutation L702H. Compound 22 (AZ'3137) possessed an attractive profile showing degradation of AR and L702H mutant AR with good oral bioavailability across species. The compound also inhibited AR signaling in vitro and tumor growth in vivo in a mouse prostate cancer xenograft model.


Sujet(s)
Biodisponibilité , Tumeurs de la prostate , Récepteurs aux androgènes , Mâle , Animaux , Humains , Récepteurs aux androgènes/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Administration par voie orale , Souris , Lignée cellulaire tumorale , Relation structure-activité , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacocinétique , Antagonistes du récepteur des androgènes/pharmacologie , Antagonistes du récepteur des androgènes/composition chimique , Antagonistes du récepteur des androgènes/usage thérapeutique , Antagonistes du récepteur des androgènes/pharmacocinétique , Découverte de médicament , Tests d'activité antitumorale sur modèle de xénogreffe , Rats
19.
EBioMedicine ; 105: 105212, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38954976

RÉSUMÉ

BACKGROUND: The E1A-associated protein p300 (p300) has emerged as a promising target for cancer therapy due to its crucial role in promoting oncogenic signaling pathways in various cancers, including prostate cancer. This need is particularly significant in prostate cancer. While androgen deprivation therapy (ADT) has demonstrated promising efficacy in prostate cancer, its long-term use can eventually lead to the development of castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC). Notably, p300 has been identified as an important co-activator of the androgen receptor (AR), highlighting its significance in prostate cancer progression. Moreover, recent studies have revealed the involvement of p300 in AR-independent oncogenes associated with NEPC. Therefore, the blockade of p300 may emerge as an effective therapeutic strategy to address the challenges posed by both CRPC and NEPC. METHODS: We employed AI-assisted design to develop a peptide-based PROTAC (proteolysis-targeting chimera) drug that targets p300, effectively degrading p300 in vitro and in vivo utilizing nano-selenium as a peptide drug delivery system. FINDINGS: Our p300-targeting peptide PROTAC drug demonstrated effective p300 degradation and cancer cell-killing capabilities in both CRPC, AR-negative, and NEPC cells. This study demonstrated the efficacy of a p300-targeting drug in NEPC cells. In both AR-positive and AR-negative mouse models, the p300 PROTAC drug showed potent p300 degradation and tumor suppression. INTERPRETATION: The design of peptide PROTAC drug targeting p300 is feasible and represents an efficient therapeutic strategy for CRPC, AR-negative prostate cancer, and NEPC. FUNDING: The funding details can be found in the Acknowledgements section.


Sujet(s)
Protéine p300-E1A , Peptides , Tumeurs de la prostate , Protéolyse , Tests d'activité antitumorale sur modèle de xénogreffe , Mâle , Humains , Protéolyse/effets des médicaments et des substances chimiques , Animaux , Souris , Lignée cellulaire tumorale , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Protéine p300-E1A/métabolisme , Peptides/pharmacologie , Peptides/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Modèles animaux de maladie humaine , Récepteurs aux androgènes/métabolisme , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Tumeurs prostatiques résistantes à la castration/métabolisme , Tumeurs prostatiques résistantes à la castration/anatomopathologie , Découverte de médicament
20.
Front Immunol ; 15: 1416941, 2024.
Article de Anglais | MEDLINE | ID: mdl-38863718

RÉSUMÉ

Across the wide range of clinical conditions, there exists a sex imbalance where biological females are more prone to autoimmune diseases and males to some cancers. These discrepancies are the combinatory consequence of lifestyle and environmental factors such as smoking, alcohol consumption, obesity, and oncogenic viruses, as well as other intrinsic biological traits including sex chromosomes and sex hormones. While the emergence of immuno-oncology (I/O) has revolutionised cancer care, the efficacy across multiple cancers may be limited because of a complex, dynamic interplay between the tumour and its microenvironment (TME). Indeed, sex and gender can also influence the varying effectiveness of I/O. Androgen receptor (AR) plays an important role in tumorigenesis and in shaping the TME. Here, we lay out the epidemiological context of sex disparity in cancer and then review the current literature on how AR signalling contributes to such observation via altered tumour development and immunology. We offer insights into AR-mediated immunosuppressive mechanisms, with the hope of translating preclinical and clinical evidence in gender oncology into improved outcomes in personalised, I/O-based cancer care.


Sujet(s)
Immunothérapie , Tumeurs , Récepteurs aux androgènes , Microenvironnement tumoral , Animaux , Femelle , Humains , Mâle , Immunothérapie/méthodes , Tumeurs/immunologie , Tumeurs/thérapie , Tumeurs/métabolisme , Récepteurs aux androgènes/métabolisme , Facteurs sexuels , Transduction du signal , Résultat thérapeutique , Microenvironnement tumoral/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE