Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.705
Filter
1.
Genes (Basel) ; 15(9)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39336758

ABSTRACT

The oxygen-sensing pathway is a crucial regulatory circuit that defines cellular conditions and is extensively exploited in cancer development. Pathogenic mutations in the von Hippel-Lindau (VHL) tumour suppressor impair its role as a master regulator of hypoxia-inducible factors (HIFs), leading to constitutive HIF activation and uncontrolled angiogenesis, increasing the risk of developing clear cell renal cell carcinoma (ccRCC). HIF hyperactivation can sequester HIF-1ß, preventing the aryl hydrocarbon receptor (AHR) from correctly activating gene expression in response to endogenous and exogenous ligands such as TCDD (dioxins). In this study, we used protein-protein interaction networks and gene expression profiling to characterize the impact of VHL loss on AHR activity. Our findings reveal specific expression patterns of AHR interactors following exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and in ccRCC. We identified several AHR interactors significantly associated with poor survival rates in ccRCC patients. Notably, the upregulation of the androgen receptor (AR) and retinoblastoma-associated protein (RB1) by TCDD, coupled with their respective downregulation in ccRCC and association with poor survival rates, suggests novel therapeutic targets. The strategic activation of the AHR via selective AHR modulators (SAhRMs) could stimulate its anticancer activity, specifically targeting RB1 and AR to reduce cell cycle progression and metastasis formation in ccRCC. Our study provides comprehensive insights into the complex interplay between the AHR and HIF pathways in ccRCC pathogenesis, offering novel strategies for targeted therapeutic interventions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Carcinoma, Renal Cell , Gene Expression Regulation, Neoplastic , Kidney Neoplasms , Polychlorinated Dibenzodioxins , Receptors, Aryl Hydrocarbon , Von Hippel-Lindau Tumor Suppressor Protein , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Polychlorinated Dibenzodioxins/pharmacology , Polychlorinated Dibenzodioxins/toxicity , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Retinoblastoma Binding Proteins/genetics , Retinoblastoma Binding Proteins/metabolism , Signal Transduction , Protein Interaction Maps , Ubiquitin-Protein Ligases , Aryl Hydrocarbon Receptor Nuclear Translocator
2.
Proc Natl Acad Sci U S A ; 121(40): e2406837121, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39312663

ABSTRACT

Cancers develop resistance to inhibitors of oncogenes mainly due to target-centric mechanisms such as mutations and splicing. While inhibitors or antagonists force targets to unnatural conformation contributing to protein instability and resistance, activating tumor suppressors may maintain the protein in an agonistic conformation to elicit sustainable growth inhibition. Due to the lack of tumor suppressor agonists, this hypothesis and the mechanisms underlying resistance are not understood. In estrogen receptor (ER)-positive breast cancer (BC), androgen receptor (AR) is a druggable tumor suppressor offering a promising avenue for this investigation. Spatial genomics suggests that the molecular portrait of AR-expressing BC cells in tumor microenvironment corresponds to better overall patient survival, clinically confirming AR's role as a tumor suppressor. Ligand activation of AR in ER-positive BC xenografts reprograms cistromes, inhibits oncogenic pathways, and promotes cellular elasticity toward a more differentiated state. Sustained AR activation results in cistrome rearrangement toward transcription factor PROP paired-like homeobox 1, transformation of AR into oncogene, and activation of the Janus kinase/signal transducer (JAK/STAT) pathway, all culminating in lineage plasticity to an aggressive resistant subtype. While the molecular profile of AR agonist-sensitive tumors corresponds to better patient survival, the profile represented in the resistant phenotype corresponds to shorter survival. Inhibition of activated oncogenes in resistant tumors reduces growth and resensitizes them to AR agonists. These findings indicate that persistent activation of a context-dependent tumor suppressor may lead to resistance through lineage plasticity-driven tumor metamorphosis. Our work provides a framework to explore the above phenomenon across multiple cancer types and underscores the importance of factoring sensitization of tumor suppressor targets while developing agonist-like drugs.


Subject(s)
Breast Neoplasms , Receptors, Androgen , Receptors, Estrogen , STAT Transcription Factors , Humans , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , STAT Transcription Factors/metabolism , STAT Transcription Factors/genetics , Animals , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Oncogenes , Janus Kinases/metabolism , Mice , Signal Transduction , Cell Line, Tumor , Tumor Microenvironment , Gene Expression Regulation, Neoplastic
3.
Int J Mol Sci ; 25(18)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39337600

ABSTRACT

Steroidogenic factor 1 (SF-1) is a nuclear receptor that regulates steroidogenesis and reproductive development. NR5A1/SF-1 variants are associated with a broad spectrum of phenotypes across individuals with disorders of sex development (DSDs). Oligogenic inheritance has been suggested as an explanation. SF-1 interacts with numerous partners. Here, we investigated a constellation of gene variants identified in a 46,XY severely undervirilized individual carrying an ACMG-categorized 'pathogenic' NR5A1/SF-1 variant in comparison to the healthy carrier father. Candidate genes were revealed by whole exome sequencing, and pathogenicity was predicted by different in silico tools. We found variants in NR1H2 and INHA associated with steroidogenesis, sex development, and reproduction. The identified variants were tested in cell models. Novel SF-1 and NR1H2 binding sites in the AR and INHA gene promoters were found. Transactivation studies showed that wild-type NR5A1/SF-1 regulates INHA and AR gene expression, while the NR5A1/SF-1 variant had decreased transcriptional activity. NR1H2 was found to regulate AR gene transcription; however, the NR1H2 variant showed normal activity. This study expands the NR5A1/SF-1 network of interacting partners, while not solving the exact interplay of different variants that might be involved in revealing the observed DSD phenotype. It also illustrates that understanding complex genetics in DSDs is challenging.


Subject(s)
Inhibins , Receptors, Androgen , Steroidogenic Factor 1 , Humans , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Inhibins/metabolism , Inhibins/genetics , Male , Disorder of Sex Development, 46,XY/genetics , Female , Exome Sequencing , Promoter Regions, Genetic
4.
Int J Mol Sci ; 25(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39337689

ABSTRACT

This study aims to investigate the effect of a supraphysiological dose of testosterone on the levels of sex steroid hormones and the expression and distribution of sex steroid receptors in the uterus during the endometrial receptivity development period. In this study, adult female Sprague-Dawley rats (n = 24) were subcutaneously administered 1 mg/kg/day of testosterone alone or in combination with the inhibitors (finasteride or anastrozole or both) from day 1 to day 3 post-coitus, while a group of six untreated rats served as a control group. The rats were sacrificed on the evening of post-coital day 4 of to measure sex steroid hormone levels by ELISA. Meanwhile, gene expression and protein distribution of sex steroid receptors were analysed by quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC), respectively. In this study, treatment with a supraphysiological dose of testosterone led to a significant reduction in oestrogen and progesterone levels compared to the control. The mRNA expression of the androgen receptor increased significantly in all treatment groups, while the mRNA expression of both the progesterone receptor and the oestrogen receptor-α decreased significantly in all treatment groups. The IHC findings of all sex steroid receptors were coherent with all mRNAs involved. This study shows that a supraphysiological dose of testosterone was able to interrupt the short period of the implantation window. This finding could serve as a basis for understanding the role of testosterone in endometrial receptivity in order to develop further therapeutic approaches targeting androgen-mediated disorders of endometrial receptivity.


Subject(s)
Endometrium , Rats, Sprague-Dawley , Testosterone , Animals , Female , Testosterone/metabolism , Endometrium/metabolism , Endometrium/drug effects , Rats , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , Embryo Implantation/drug effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Receptors, Steroid/metabolism , Receptors, Steroid/genetics , Progesterone/metabolism
5.
Nat Commun ; 15(1): 7984, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266569

ABSTRACT

Alterations in nuclear structure and function are hallmarks of cancer cells. Little is known about these changes in Cancer-Associated Fibroblasts (CAFs), crucial components of the tumor microenvironment. Loss of the androgen receptor (AR) in human dermal fibroblasts (HDFs), which triggers early steps of CAF activation, leads to nuclear membrane changes and micronuclei formation, independent of cellular senescence. Similar changes occur in established CAFs and are reversed by restoring AR activity. AR associates with nuclear lamin A/C, and its loss causes lamin A/C nucleoplasmic redistribution. AR serves as a bridge between lamin A/C and the protein phosphatase PPP1. Loss of AR decreases lamin-PPP1 association and increases lamin A/C phosphorylation at Ser 301, a characteristic of CAFs. Phosphorylated lamin A/C at Ser 301 binds to the regulatory region of CAF effector genes of the myofibroblast subtype. Expression of a lamin A/C Ser301 phosphomimetic mutant alone can transform normal fibroblasts into tumor-promoting CAFs.


Subject(s)
Cancer-Associated Fibroblasts , Cell Nucleus , Lamin Type A , Receptors, Androgen , Humans , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Lamin Type A/metabolism , Lamin Type A/genetics , Phosphorylation , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Nucleus/metabolism , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/genetics , Fibroblasts/metabolism , Nuclear Envelope/metabolism , Male , Tumor Microenvironment
6.
Pestic Biochem Physiol ; 204: 106065, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277380

ABSTRACT

Organophosphate pesticides have potent endocrine disrupting effects, hence banned in many countries. However, many organophosphates like chlorpyrifos, malathion et cetera continue to be used in some countries (Wolejko et al., 2022; Wolejko et al., 2022)including India. Fodder mediated ingestion of these substances may be harmful for livestock fertility. We have investigated the effect of the widely used organophosphate pesticide chlorpyrifos (CPF) and its metabolite, 3,5,6-trichloropyridinol (TCPy) on the expression of genes essential for spermatogenesis in goat testicular tissue. The testicular Sertoli cells (Sc) regulate germ cell division and differentiation under the influence of follicle stimulating hormone (FSH) and testosterone (T). Impaired FSH and T mediated signalling in Sc can compromise spermatogenesis leading to sub-fertility/infertility. As Sc express receptors (R) for FSH and T, they are highly susceptible to the endocrine disrupting effects of pesticides affecting fertility by dysregulating the functioning of Sc. Our results indicated that exposure to different concentrations of CPF and TCPy can compromise Sc function by downregulating the expression of FSHR and AR which was associated with a concomitant decline in the expression of genes essential for germ cell division and differentiation, like KITLG, INHBB, CLDN11 and GJA1. CPF also induced a significant reduction in the activity of acetylcholinesterase in the testes and increased the total testicular antioxidant capacity. Our results suggested that CPF and its metabolite TCPy may induce reproductive toxicity by dysregulating the expression of Sc specific genes essential for spermatogenesis.


Subject(s)
Chlorpyrifos , Goats , Spermatogenesis , Testis , Animals , Male , Spermatogenesis/drug effects , Chlorpyrifos/toxicity , Testis/drug effects , Testis/metabolism , Down-Regulation/drug effects , Insecticides/toxicity , Pyridines/pharmacology , Pyridines/toxicity , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Pyridones
7.
Diagn Pathol ; 19(1): 121, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39243111

ABSTRACT

OBJECTIVES: We explored Trichorhinophalangeal syndrome type 1 (TRPS1) expression in special types of breast carcinoma, and analyzed the correlation between TRPS1 and androgen receptor (AR) expression in triple-negative breast cancer (TNBC). METHODS: TRPS1 expression was analyzed in 801 patients with special types of breast carcinoma. A total of 969 TNBC were used to analyze the correlation between the expression of TRPS1 and AR. TRPS1 expression was evaluated in 1975 cases of breast cancer with different molecular types. RESULTS: A total of 801 special types of breast cancers were stained with TRPS1.TRPS1 was positive in 100% (63/63) of mucinous carcinoma, 100% (7/7) adenoid cystic carcinomas (4 classic adenoid cystic carcinomas and 3 solid-basaloid adenoid cystic carcinomas), 100% (4/4) tubular carcinomas, 100% (2/2) secretory carcinomas, and 99.59% (243/244) invasive lobular carcinomas, 99.26% (267/269) invasive micropapillary carcinomas, 97.44% (38/39) ER-positive neuroendocrine tumors, 94.44% (34/36) metaplastic breast carcinomas (MBCs), 63.73% (65/102) apocrine carcinomas. TRPS1 was negative in all triple-negative neuroendocrine carcinomas (0/7).TRPS1 was positive in 92.86% (26/28) of metastatic special types of breast cancer. TRPS1 and AR expression were analyzed in 969 cases of TNBC. 90.40% were positive for TRPS1, and 42.41% were positive for AR. A significant inverse correlation between TRPS1 and AR expression was shown in TNBC (p < .001). TRPS1 showed a higher positive rate (93.13%) in TNBC compared to GATA binding protein 3 (GATA3), gross cystic disease fluid protein 15 (GCDFP-15) and forkhead box transcription Factor C 1 (FOXC1). CONCLUSIONS: In conclusion, our study demonstrated that TRPS1 is a highly sensitive marker for most special types of breast carcinoma. TRPS1 was positive in 63.73% of apocrine carcinomas. TRPS1 and AR expression was inversely correlated in TNBC.


Subject(s)
Biomarkers, Tumor , DNA-Binding Proteins , Receptors, Androgen , Repressor Proteins , Transcription Factors , Triple Negative Breast Neoplasms , Humans , Female , Biomarkers, Tumor/analysis , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Repressor Proteins/analysis , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Transcription Factors/analysis , Receptors, Androgen/analysis , Receptors, Androgen/genetics , Immunohistochemistry , Middle Aged , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Adult
8.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39240718

ABSTRACT

Biological sex affects the activity of the hypothalamus-pituitary-adrenal (HPA) axis. However, how androgen deprivation affects this axis remains largely unknown. In this study, we investigated the effect of androgen status on different components of the HPA axis in male mice. Two weeks of androgen deprivation did not affect total plasma corticosterone levels but led to increased pituitary ACTH levels. Stress-induced total plasma corticosterone levels were increased, whereas the suppression of corticosterone after dexamethasone treatment under basal conditions was attenuated. Androgen-deprived mice displayed a 2-fold increase in plasma levels of corticosteroid binding globulin (CBG). A similar increase in CBG was observed in global androgen receptor knock-out animals, compared to wild-type littermates. Androgen deprivation was associated with a 6-fold increase in CBG mRNA in the liver and enhanced transcriptional activity at CBG regulatory regions, as evidenced by increased H3K27 acetylation. We propose that the induction of CBG as a consequence of androgen deprivation, together with the unaltered total corticosterone levels, results in lower free corticosterone levels in plasma. This is further supported by mRNA levels of androgen-independent GR target genes in the liver. The reduction in negative feedback on the HPA axis under basal condition would suffice to explain the enhanced stress reactivity after androgen deprivation. Overall, our data demonstrate that, in mice, tonic androgen receptor activation affects CBG levels in conjunction with effects on gene expression and HPA-axis reactivity.


Subject(s)
Androgens , Corticosterone , Hypothalamo-Hypophyseal System , Mice, Knockout , Pituitary-Adrenal System , Transcortin , Animals , Male , Transcortin/metabolism , Transcortin/genetics , Mice , Corticosterone/blood , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/drug effects , Androgens/blood , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/drug effects , Stress, Physiological/drug effects , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Mice, Inbred C57BL , Liver/metabolism , Liver/drug effects , Adrenocorticotropic Hormone/blood , Dexamethasone/pharmacology
9.
Nat Cancer ; 5(9): 1427-1447, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39242942

ABSTRACT

Prostate cancer (PCa) exhibits significant geoethnic disparities as reflected by distinct variations in the cancer genome and disease progression. Here, we perform a comprehensive proteogenomic characterization of localized high-risk PCa utilizing paired tumors and nearby tissues from 125 Chinese male patients, with the primary objectives of identifying potential biomarkers, unraveling critical oncogenic events and delineating molecular subtypes with poor prognosis. Our integrated analysis highlights the utility of GOLM1 as a noninvasive serum biomarker. Phosphoproteomics analysis reveals the crucial role of Ser331 phosphorylation on FOXA1 in regulating FOXA1-AR-dependent cistrome. Notably, our proteomic profiling identifies three distinct subtypes, with metabolic immune-desert tumors (S-III) emerging as a particularly aggressive subtype linked to poor prognosis and BCAT2 catabolism-driven PCa progression. In summary, our study provides a comprehensive resource detailing the unique proteomic and phosphoproteomic characteristics of PCa molecular pathogenesis and offering valuable insights for the development of diagnostic and therapeutic strategies.


Subject(s)
Biomarkers, Tumor , Hepatocyte Nuclear Factor 3-alpha , Prostatic Neoplasms , Proteogenomics , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Proteogenomics/methods , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , China , Prognosis , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Asian People/genetics , Middle Aged , Phosphorylation , Aged , Gene Expression Regulation, Neoplastic , East Asian People
10.
J Clin Invest ; 134(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39286979

ABSTRACT

The widespread use of potent androgen receptor signaling inhibitors (ARSIs) has led to an increasing emergence of AR-independent castration-resistant prostate cancer (CRPC), typically driven by loss of AR expression, lineage plasticity, and transformation to prostate cancers (PCs) that exhibit phenotypes of neuroendocrine or basal-like cells. The anti-apoptotic protein BCL2 is upregulated in neuroendocrine cancers and may be a therapeutic target for this aggressive PC disease subset. There is an unmet clinical need, therefore, to clinically characterize BCL2 expression in metastatic CRPC (mCRPC), determine its association with AR expression, uncover its mechanisms of regulation, and evaluate BCL2 as a therapeutic target and/or biomarker with clinical utility. Here, using multiple PC biopsy cohorts and models, we demonstrate that BCL2 expression is enriched in AR-negative mCRPC, associating with shorter overall survival and resistance to ARSIs. Moreover, high BCL2 expression associates with lineage plasticity features and neuroendocrine marker positivity. We provide evidence that BCL2 expression is regulated by DNA methylation, associated with epithelial-mesenchymal transition, and increased by the neuronal transcription factor ASCL1. Finally, BCL2 inhibition had antitumor activity in some, but not all, BCL2-positive PC models, highlighting the need for combination strategies to enhance tumor cell apoptosis and enrich response.


Subject(s)
Gene Expression Regulation, Neoplastic , Prostatic Neoplasms, Castration-Resistant , Proto-Oncogene Proteins c-bcl-2 , Male , Humans , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Animals , Cell Line, Tumor , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Mice , DNA Methylation , Epithelial-Mesenchymal Transition , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Lineage , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/biosynthesis
11.
Proc Natl Acad Sci U S A ; 121(39): e2407768121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39292748

ABSTRACT

Androgens exert their effects primarily by binding to the androgen receptor (AR), a ligand-dependent nuclear receptor. While androgens have anabolic effects on skeletal muscle, previous studies reported that AR functions in myofibers to regulate skeletal muscle quality, rather than skeletal muscle mass. Therefore, the anabolic effects of androgens are exerted via nonmyofiber cells. In this context, the cellular and molecular mechanisms of AR in mesenchymal progenitors, which play a crucial role in maintaining skeletal muscle homeostasis, remain largely unknown. In this study, we demonstrated expression of AR in mesenchymal progenitors and found that targeted AR ablation in mesenchymal progenitors reduced limb muscle mass in mature adult, but not young or aged, male mice, although fatty infiltration of muscle was not affected. The absence of AR in mesenchymal progenitors led to remarkable perineal muscle hypotrophy, regardless of age, due to abnormal regulation of transcripts associated with cell death and extracellular matrix organization. Additionally, we revealed that AR in mesenchymal progenitors regulates the expression of insulin-like growth factor 1 (Igf1) and that IGF1 administration prevents perineal muscle atrophy in a paracrine manner. These findings indicate that the anabolic effects of androgens regulate skeletal muscle mass via, at least in part, AR signaling in mesenchymal progenitors.


Subject(s)
Insulin-Like Growth Factor I , Mesenchymal Stem Cells , Muscle, Skeletal , Receptors, Androgen , Animals , Male , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/genetics , Muscle, Skeletal/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Muscular Atrophy/metabolism , Muscular Atrophy/pathology
12.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39253786

ABSTRACT

Prostate cancer progression is driven by androgen receptor (AR) activity, which is a target for therapeutic approaches. Enzalutamide is an AR inhibitor that prolongs the survival of patients with advanced prostate cancer. However, resistance mechanisms arise and impair its efficacy. One of these mechanisms is the expression of AR-V7, a constitutively active AR splice variant. The Mediator complex is a multisubunit protein that modulates gene expression on a genome-wide scale. MED12 and cyclin-dependent kinase (CDK)8, or its paralog CDK19, are components of the kinase module that regulates the proliferation of prostate cancer cells. In this study, we investigated how MED12 and CDK8/19 influence cancer-driven processes in prostate cancer cell lines, focusing on AR activity and the enzalutamide response. We inhibited MED12 expression and CDK8/19 activity in LNCaP (AR+, enzalutamide-sensitive), 22Rv1 (AR-V7+, enzalutamide-resistant), and PC3 (AR-, enzalutamide-insensitive) cells. Both MED12 and CDK8/19 inhibition reduced cell proliferation in all cell lines, and MED12 inhibition reduced proliferation in the respective 3D spheroids. MED12 knockdown significantly inhibited c-Myc protein expression and signaling pathways. In 22Rv1 cells, it consistently inhibited the AR response, prostate-specific antigen (PSA) secretion, AR target genes, and AR-V7 expression. Combined with enzalutamide, MED12 inhibition additively decreased the AR activity in both LNCaP and 22Rv1 cells. CDK8/19 inhibition significantly decreased PSA secretion in LNCaP and 22Rv1 cells and, when combined with enzalutamide, additively reduced proliferation in 22Rv1 cells. Our study revealed that MED12 and CDK8/19 regulate AR activity and that their inhibition may modulate response to enzalutamide in prostate cancer.


Subject(s)
Benzamides , Cell Proliferation , Cyclin-Dependent Kinase 8 , Cyclin-Dependent Kinases , Mediator Complex , Nitriles , Phenylthiohydantoin , Prostatic Neoplasms , Receptors, Androgen , Phenylthiohydantoin/pharmacology , Male , Humans , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Mediator Complex/metabolism , Mediator Complex/genetics , Cell Line, Tumor , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinase 8/genetics , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects
13.
Stem Cell Res Ther ; 15(1): 287, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256806

ABSTRACT

BACKGROUND: Androgenetic alopecia (AGA) is a common form of hair loss. Androgens, such as testosterone and dihydrotestosterone, are the main causes of AGA. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) can reduce AGA. However, preparing therapeutic doses of MSCs for clinical use is challenging. Induced pluripotent stem cell-derived MSCs (iMSCs) are homogenous and easily expandable, enabling scalable production of EVs. Hyaluronic acid (HA) can exert various functions including free radical scavenging, immune regulation, and cell migration. Herein, we examined whether hyaluronic acid (HA) stimulation of iMSCs could produce EVs with enhanced therapeutic outcomes for AGA. METHODS: EVs were collected from iMSCs primed with HA (HA-iMSC-EVs) or without HA (iMSC-EVs). The characteristics of EVs were examined using dynamic light scattering, cryo-transmission electron microscopy, immunoblotting, flow cytometry, and proteomic analysis. In vitro, we compared the potential of EVs in stimulating the survival of hair follicle dermal papilla cells undergoing testosterone-mediated AGA. Additionally, the expression of androgen receptor (AR) and relevant growth factors as well as key proteins of Wnt/ß-catenin signaling pathway (ß-catenin and phosphorylated GSK3ß) was analyzed. Subsequently, AGA was induced in male C57/BL6 mice by testosterone administration, followed by repeated injections of iMSC-EVs, HA-iMSC-EVs, finasteride, or vehicle. Several parameters including hair growth, anagen phase ratio, reactivation of Wnt/ß-catenin pathway, and AR expression was examined using qPCR, immunoblotting, and immunofluorescence analysis. RESULTS: Both types of EVs showed typical characteristics for EVs, such as size distribution, markers, and surface protein expression. In hair follicle dermal papilla cells, the mRNA levels of AR, TGF-ß, and IL-6 increased by testosterone was blocked by HA-iMSC-EVs, which also contributed to the augmented expression of trophic genes related to hair regrowth. However, no notable changes were observed in the iMSC-EVs. Re-activation of Wnt/ß-catenin was observed in HA-iMSC-EVs but not in iMSC-EVs, as shown by ß-catenin stabilization and an increase in phosphorylated GSK3ß. Restoration of hair growth was more significant in HA-iMSC-EVs than in iMSC-EVs, and was comparable to that in mice treated with finasteride. Consistently, the decreased anagen ratio induced by testosterone was reversed by HA-iMSC-EVs, but not by iMSC-EVs. An increased expression of hair follicular ß-catenin protein, as well as the reduction of AR was observed in the skin tissue of AGA mice receiving HA-iMSC-EVs, but not in those treated with iMSC-EVs. CONCLUSIONS: Our results suggest that HA-iMSC-EVs have potential to improve AGA by regulating growth factors/cytokines and stimulating AR-related Wnt/ß-catenin signaling.


Subject(s)
Alopecia , Extracellular Vesicles , Hair Follicle , Hyaluronic Acid , Mesenchymal Stem Cells , Extracellular Vesicles/metabolism , Alopecia/therapy , Alopecia/metabolism , Alopecia/drug therapy , Hyaluronic Acid/pharmacology , Hyaluronic Acid/metabolism , Animals , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Hair Follicle/metabolism , Hair Follicle/drug effects , Humans , Wnt Signaling Pathway/drug effects , Male , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Testosterone/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Mice, Inbred C57BL
14.
Breast Cancer Res ; 26(1): 132, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39272208

ABSTRACT

BACKGROUND: Despite evidence indicating the dominance of cell-of-origin signatures in molecular tumor patterns, translating these genome-wide patterns into actionable insights has been challenging. This study introduces breast cancer cell-of-origin signatures that offer significant prognostic value across all breast cancer subtypes and various clinical cohorts, compared to previously developed genomic signatures. METHODS: We previously reported that triple hormone receptor (THR) co-expression patterns of androgen (AR), estrogen (ER), and vitamin D (VDR) receptors are maintained at the protein level in human breast cancers. Here, we developed corresponding mRNA signatures (THR-50 and THR-70) based on these patterns to categorize breast tumors by their THR expression levels. The THR mRNA signatures were evaluated across 56 breast cancer datasets (5040 patients) using Kaplan-Meier survival analysis, Cox proportional hazard regression, and unsupervised clustering. RESULTS: The THR signatures effectively predict both overall and progression-free survival across all evaluated datasets, independent of subtype, grade, or treatment status, suggesting improvement over existing prognostic signatures. Furthermore, they delineate three distinct ER-positive breast cancer subtypes with significant survival in differences-expanding on the conventional two subtypes. Additionally, coupling THR-70 with an immune signature identifies a predominantly ER-negative breast cancer subgroup with a highly favorable prognosis, comparable to ER-positive cases, as well as an ER-negative subgroup with notably poor outcome, characterized by a 15-fold shorter survival. CONCLUSIONS: The THR cell-of-origin signature introduces a novel dimension to breast cancer biology, potentially serving as a robust foundation for integrating additional prognostic biomarkers. These signatures offer utility as a prognostic index for stratifying existing breast cancer subtypes and for de novo classification of breast cancer cases. Moreover, THR signatures may also hold promise in predicting hormone treatment responses targeting AR and/or VDR.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms , Receptors, Androgen , Receptors, Calcitriol , Receptors, Estrogen , Humans , Female , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/mortality , Breast Neoplasms/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Prognosis , Receptors, Estrogen/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Gene Expression Regulation, Neoplastic , Gene Expression Profiling , Kaplan-Meier Estimate , Transcriptome
15.
Nat Commun ; 15(1): 7688, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227386

ABSTRACT

Autoimmune diseases such as systemic lupus erythematosus (SLE) display a strong female bias. Although sex hormones have been associated with protecting males from autoimmunity, the molecular mechanisms are incompletely understood. Here we report that androgen receptor (AR) expressed in T cells regulates genes involved in T cell activation directly, or indirectly via controlling other transcription factors. T cell-specific deletion of AR in mice leads to T cell activation and enhanced autoimmunity in male mice. Mechanistically, Ptpn22, a phosphatase and negative regulator of T cell receptor signaling, is downregulated in AR-deficient T cells. Moreover, a conserved androgen-response element is found in the regulatory region of Ptpn22 gene, and the mutation of this transcription element in non-obese diabetic mice increases the incidence of spontaneous and inducible diabetes in male mice. Lastly, Ptpn22 deficiency increases the disease severity of male mice in a mouse model of SLE. Our results thus implicate AR-regulated genes such as PTPN22 as potential therapeutic targets for autoimmune diseases.


Subject(s)
Androgens , Autoimmunity , Lupus Erythematosus, Systemic , Protein Tyrosine Phosphatase, Non-Receptor Type 22 , Receptors, Androgen , T-Lymphocytes , Animals , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Male , Female , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Androgens/metabolism , Mice, Knockout , Lymphocyte Activation , Mice, Inbred NOD , Mice, Inbred C57BL , Disease Models, Animal , Signal Transduction
16.
Nat Commun ; 15(1): 7675, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227594

ABSTRACT

Most prostate cancers express the androgen receptor (AR), and tumor growth and progression are facilitated by exceptionally low levels of systemic or intratumorally produced androgens. Thus, absolute inhibition of the androgen signaling axis remains the goal of current therapeutic approaches to treat prostate cancer (PCa). Paradoxically, high dose androgens also exhibit considerable efficacy as a treatment modality in patients with late-stage metastatic PCa. Here we show that low levels of androgens, functioning through an AR monomer, facilitate a non-genomic activation of the mTOR signaling pathway to drive proliferation. Conversely, high dose androgens facilitate the formation of AR dimers/oligomers to suppress c-MYC expression, inhibit proliferation and drive a transcriptional program associated with a differentiated phenotype. These findings highlight the inherent liabilities in current approaches used to inhibit AR action in PCa and are instructive as to strategies that can be used to develop new therapeutics for this disease and other androgenopathies.


Subject(s)
Androgens , Cell Proliferation , Prostatic Neoplasms , Receptors, Androgen , Signal Transduction , TOR Serine-Threonine Kinases , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Male , Humans , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Androgens/metabolism , Androgens/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Protein Multimerization/drug effects , Animals
17.
Sci Rep ; 14(1): 22122, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333688

ABSTRACT

Androgen receptor (AR) overexpression has been identified in gliomas and its stem cells, suggesting that AR plays an important role in tumor carcinogenesis. The prognostic significance of AR overexpression in gliomas remains unknown. AR mRNA expression in gliomas and relevant clinical data were obtained from The Cancer Genome Atlas and Chinese Glioma Genome Atlas databases. AR expression levels were compared across gliomas of different histopathologic grades and molecular subtypes. Kaplan-Meier analyses in patients with different AR expression levels were investigated for the potential prognostic values of AR. Compared with normal brain tissue, gliomas show significantly higher AR mRNA expression (p < 0.01). AR mRNA expression was more prominent in higher grade disease and in worse prognostic molecular subtypes (p < 0.01). AR protein is more abundant in glioblastoma than in lower grade gliomas (LGG) (grade 2/3) (p < 0.0001). This is corroborated by a linear association between AR mRNA and protein expression (r = 0.65). In LGG, both higher AR mRNA and protein expression was associated with significantly worse overall survival (OS). Five-year OS for LGG patients with high versus low AR expression were 59.1% and 73.3%, respectively (p < 0.0001). AR expression is not prognostic for OS within glioblastoma patients. Gender was not associated with AR expression or prognosis. Higher AR expression levels are associated with higher grade disease and histopathologic features predicting poorer prognosis in lower grade gliomas. Higher gene expression in LGG patients is correlated with poor prognosis but not in the glioblastoma cohort suggesting saturated expression/functions of AR in glioblastoma.


Subject(s)
Brain Neoplasms , Glioma , Receptors, Androgen , Humans , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Glioma/mortality , Prognosis , Male , Female , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Gene Expression Regulation, Neoplastic , Middle Aged , Neoplasm Grading , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Kaplan-Meier Estimate , RNA, Messenger/genetics , RNA, Messenger/metabolism , Adult
18.
Int J Mol Sci ; 25(18)2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39337362

ABSTRACT

Prostate cancer (PC) is one of the most commonly diagnosed tumours among men. Second-generation androgen receptor axis-targeted (ARAT) agents, namely abiraterone acetate (AbA) and enzalutamide (ENZ), are currently used in the management of metastatic castration-resistant PC (mCRPC). However, the treatment is challenging due to the lack of prognostic biomarkers. Meanwhile, single-nucleotide polymorphisms (SNPs) have emerged as potential prognostic indicators of mCRPC. Thus, this study evaluated the impact of relevant SNPs on the treatment outcomes of 123 mCRPC patients enrolled in a hospital-based cohort study. The CYP17A1 rs2486758 C allele was associated with a 50% reduction in the risk of developing castration resistance (hazard ratio (HR) = 0.55; p = 0.003). Among patients without metastasis at tumour diagnosis and under AbA, a marginal association between YBX1 rs10493112 and progression-free survival was detected (log-rank test, p = 0.056). In the same subgroup, significant associations of HSD3B1 rs1047303 (CC/CA vs. AA; HR = 3.41; p = 0.025), YBX1 rs12030724 (AT vs. AA; HR = 3.54; p = 0.039) and YBX1 rs10493112 (log-rank test, p = 0.041; CC vs. AA/AC; HR = 3.22; p = 0.053) with overall survival were also observed, which were confirmed by multivariate Cox analyses. Although validation with larger cohorts is required, these findings suggest that SNPs could enhance the prognosis assessment of mCRPC patients, leading to a more personalised treatment.


Subject(s)
Abiraterone Acetate , Polymorphism, Single Nucleotide , Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Humans , Male , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/mortality , Aged , Receptors, Androgen/genetics , Abiraterone Acetate/therapeutic use , Middle Aged , Phenylthiohydantoin/therapeutic use , Treatment Outcome , Nitriles/therapeutic use , Benzamides/therapeutic use , Steroid 17-alpha-Hydroxylase/genetics , Aged, 80 and over , Prognosis , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Neoplasm Metastasis , Biomarkers, Tumor/genetics
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(10): 1206-1212, 2024 Oct 10.
Article in Chinese | MEDLINE | ID: mdl-39344615

ABSTRACT

OBJETIVE: To explore the clinical and molecular basis for a Chinese pedigree affected with Complete androgen insensitivity syndrome (CAIS). METHODS: A CAIS pedigree presented at Tianjin Medical University General Hospital between 2019 and 2021 was selected as the study subject. Clinical data of the proband was collected, along with peripheral blood samples from the proband and her family members. Chromosomal karyotyping, sex-determining region of the Y chromosome (SRY) testing, and next-generation sequencing (NGS) were carried out for the proband, and candidate variant was verified by Sanger sequencing of her family members. Prenatal diagnosis was provided for the sister of the proband. This study was approved by the Tianjin Medical University General Hospital (Ethics No. IRB2023-WZ-070). RESULTS: The 18-year-old proband, who has a social gender of female, underwent laparoscopic examination, which showed no presence of uterus and ovaries. The karyotype of peripheral blood sample was 46,XY, with SRY gene detected. NGS indicated that the proband has harbored a heterozygous c.1988C>G (p.Ser663Ter) variant of the AR gene. Sanger sequencing confirmed that her mother and sister had both harbored the same variant, whilst her father and younger sister were of the wild-type. Prenatal diagnosis revealed that her sister's first fetus had harbored carried the same variant, which had led to termination of pregnancy. Her second fetus did not carry the variant, and a healthy boy was born. Based on guidelines from the American College of Medical Genetics and Genomics (ACMG), the variant was classified as likely pathogenic (PM2_Supporting+PM4+PP3_Moderate+PP4). CONCLUSION: The c.1988C>G (p.Ser663Ter) variant of the AR gene probably underlay the CAIS in the proband. The accurate diagnosis of sex development disorders will rely on the physicians' thorough understanding of the clinical symptoms and pathogenic genes. Genetic testing and counseling can enable precise diagnosis, prenatal diagnosis, and guidance for reproduction.


Subject(s)
Androgen-Insensitivity Syndrome , Genetic Testing , Pedigree , Prenatal Diagnosis , Receptors, Androgen , Humans , Male , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/diagnosis , Female , Receptors, Androgen/genetics , Adolescent , Genetic Testing/methods , Pregnancy , Karyotyping , Asian People/genetics , Mutation , High-Throughput Nucleotide Sequencing , East Asian People
20.
Int J Mol Sci ; 25(16)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39201315

ABSTRACT

In prostate cancer (PCa), androgens upregulate tumorigenesis, whereas in benign tissue, the revival of androgen receptor (AR) signaling suppresses aggressive behaviors, suggesting therapeutic potential. Dogs, natural PCa models, often lack AR in PCa. We restored AR in dog PCa to investigate resultant characteristics. Three AR-null canine PCa lines (1508, Leo, 1258) were transfected with canine wild-type AR and treated with dihydrotestosterone (DHT). In 1508, AR restoration decreased clonogenicity (p = 0.03), viability (p = 0.004), migration (p = 0.03), invasion (p = 0.01), and increased expression of the tumor suppressor NKX3.1, an AR transcriptional target (p = 0.001). In Leo, AR decreased clonogenicity (p = 0.04) and the expression of another AR transcriptional target FOLH1 (p < 0.001) and increased the expression of NKX3.1 (p = 0.01). In 1258, AR increased migration (p = 0.006) and invasion (p = 0.03). Epithelial-mesenchymal transition (EMT) marker (Vimentin, N-cadherin, SNAIL1) expression increased with AR restoration in Leo and 1258 but not 1508; siRNA vimentin knockdown abrogated AR-induced 1258 migration only. Overall, 1508 showed AR-mediated tumor suppression; AR affected proliferation in Leo but not migration or invasion; and EMT and AR regulated migration and invasion in 1258 but not proliferation. This study highlights the heterogeneous nature of PCa in dogs and cell line-specific effects of AR abrogation on aggressive behaviors.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Prostatic Neoplasms , Receptors, Androgen , Signal Transduction , Animals , Dogs , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Male , Cell Line, Tumor , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness , Cell Proliferation
SELECTION OF CITATIONS
SEARCH DETAIL