Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
J Neurosci ; 40(49): 9455-9466, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33158965

ABSTRACT

Gonadal steroids modulate growth hormone (GH) secretion and the pubertal growth spurt via undefined central pathways. GH-releasing hormone (GHRH) neurons express estrogen receptor α (ERα) and androgen receptor (AR), suggesting changing levels of gonadal steroids during puberty directly modulate the somatotropic axis. We generated mice with deletion of ERα in GHRH cells (GHRHΔERα), which displayed reduced body length in both sexes. Timing of puberty onset was similar in both groups, but puberty completion was delayed in GHRHΔERα females. Lack of AR in GHRH cells (GHRHΔAR mice) induced no changes in body length, but puberty completion was also delayed in females. Using a mouse model with two reporter genes, we observed that, while GHRHtdTom neurons minimally colocalize with Kiss1hrGFP in prepubertal mice, ∼30% of GHRH neurons coexpressed both reporter genes in adult females, but not in males. Developmental analysis of Ghrh and Kiss1 expression suggested that a subpopulation of ERα neurons in the arcuate nucleus of female mice undergoes a shift in phenotype, from GHRH to Kiss1, during pubertal transition. Our findings demonstrate that direct actions of gonadal steroids in GHRH neurons modulate growth and puberty and indicate that GHRH/Kiss1 dual-phenotype neurons play a sex-specific role in the crosstalk between the somatotropic and gonadotropic axes during pubertal transition.SIGNIFICANCE STATEMENT Late maturing adolescents usually show delayed growth and bone age. At puberty, gonadal steroids have stimulatory effects on the activation of growth and reproductive axes, but the existence of gonadal steroid-sensitive neuronal crosstalk remains undefined. Moreover, the neural basis for the sex differences observed in the clinical arena is unknown. Lack of ERα in GHRH neurons disrupts growth in both sexes and causes pubertal delay in females. Deletion of androgen receptor in GHRH neurons only delayed female puberty. In adult females, not males, a subset of GHRH neurons shift phenotype to start producing Kiss1. Thus, direct estrogen action in GHRH/Kiss1 dual-phenotype neurons modulates growth and puberty and may orchestrate the sex differences in endocrine function observed during pubertal transition.


Subject(s)
Estrogen Receptor alpha/physiology , Growth Hormone-Releasing Hormone/physiology , Growth/physiology , Kisspeptins/physiology , Sexual Maturation/physiology , Signal Transduction/physiology , Animals , Estrogen Receptor alpha/genetics , Female , Gonadal Steroid Hormones/blood , Gonadal Steroid Hormones/physiology , Growth/genetics , Growth Hormone-Releasing Hormone/genetics , Hypothalamus/metabolism , Kisspeptins/genetics , Male , Mice , Mice, Knockout , Receptors, Androgen/physiology , Sex Characteristics , Sexual Maturation/genetics , Signal Transduction/genetics
2.
Cancer Res ; 79(20): 5260-5271, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31444154

ABSTRACT

Loss of expression of context-specific tumor suppressors is a critical event that facilitates the development of prostate cancer. Zinc finger and BTB domain containing transcriptional repressors, such as ZBTB7A and ZBTB16, have been recently identified as tumor suppressors that play important roles in preventing prostate cancer progression. In this study, we used combined ChIP-seq and RNA-seq analyses of prostate cancer cells to identify direct ZBTB7A-repressed genes, which are enriched for transcriptional targets of E2F, and identified that the androgen receptor (AR) played a critical role in the transcriptional suppression of these E2F targets. AR recruitment of the retinoblastoma protein (Rb) was required to strengthen the E2F-Rb transcriptional repression complex. In addition, ZBTB7A was rapidly recruited to the E2F-Rb binding sites by AR and negatively regulated the transcriptional activity of E2F1 on DNA replication genes. Finally, ZBTB7A suppressed the growth of castration-resistant prostate cancer (CRPC) in vitro and in vivo, and overexpression of ZBTB7A acted in synergy with high-dose testosterone treatment to effectively prevent the recurrence of CRPC. Overall, this study provides novel molecular insights of the role of ZBTB7A in CRPC cells and demonstrates globally its critical role in mediating the transcriptional repression activity of AR. SIGNIFICANCE: ZBTB7A is recruited to the E2F-Rb binding sites by AR and negatively regulates the transcriptional activity of E2F1 on DNA replication genes.


Subject(s)
Adenocarcinoma/genetics , DNA-Binding Proteins/physiology , Neoplasm Proteins/physiology , Prostatic Neoplasms/genetics , Receptors, Androgen/physiology , Transcription Factors/physiology , Transcription, Genetic , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Binding Sites , Cell Line, Tumor , DNA Replication/drug effects , E2F1 Transcription Factor/physiology , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Transport , RNA Interference , Recurrence , Retinoblastoma Protein/physiology , Testosterone/pharmacology
3.
J Pharmacol Sci ; 138(1): 1-8, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30236540

ABSTRACT

Androgen-independent prostate cancer (PCa) is a developed tumor derived from the local androgen dependent PCa, which often affects elderly men. Psoralea corylifolia L, a traditional Chinese medicine, has been widely used for PCa treatment as an important part of a common prescription, while the mechanism remains unclear. Our study was aimed to investigate the tumor-inhibitory effect of its main component bakuchiol in androgen-independent PCa cell line PC-3 cells. Bakuchiol significantly suppressed PC-3 cell proliferation and migration; the expressions of PCNA and MMP-9 were consistently down regulated as well. Meanwhile, both the constitutive and LPS-induced NF-κB activation were significantly inhibited by bakuchiol. The inhibitory effects of bakuchiol on cell proliferation, migration and invasion were recovered when LPS were added together with bakuchiol. SiRNA against androgen receptor (AR) or estrogen receptor ß (ERß) were transfected and the regulation of bakuchiol-suppressed proliferation, invasion, NF-κB signaling and MMP-9 secretion in response to LPS were blocked. Taken together, our data demonstrated that bakuchiol inactivated NF-κB signaling via AR and ERß, which contributes to inhibition of PC-3 cell proliferation and migration, indicating that bakuchiol is one of the key component from P. corylifolia L for PCa treatment and has a potential as anti-prostate cancer drug candidates.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Estrogen Receptor beta/physiology , NF-kappa B/metabolism , Phenols/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/drug effects , Estrogen Receptor beta/genetics , Gene Expression/drug effects , Gene Expression/genetics , Humans , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Phenols/isolation & purification , Phenols/therapeutic use , Phytotherapy , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Prostatic Neoplasms/drug therapy , Psoralea/chemistry , RNA, Small Interfering , Receptors, Androgen/genetics
4.
Am J Chin Med ; 45(6): 1147-1156, 2017.
Article in English | MEDLINE | ID: mdl-28830207

ABSTRACT

The first record of ginseng use dates back over two millennia, and ginseng is now popular in more than 35 countries. Ginsenosides are the pharmacological constituents responsible for the beneficial effects of ginseng. There is increasing evidence that ginseng and its bioactive ingredients are involved in the regulation of nuclear receptors, molecules that act in response to the specific binding of hormones, which link to a diverse array of signaling pathways, such as the ERK and PI3K/Akt pathways. Knowledge of the mechanism of how ginseng mediates these complexes is essential for the development of multi-target phytomedicine as possible therapy for different diseases. Here, we discuss the literature on the effects of ginseng and its constituents on estrogen, glucocorticoid, peroxisome proliferator-activated, and androgen nuclear hormone receptors, as well as how ginseng and its constituents exert their biological function in the treatment of cancer, obesity, and cardiovascular and neurological disorders. The accumulated results definitely show that the nuclear receptors are cellular targets of ginsenosides, but more rigorous data are required to establish and provide a scientific basis to confirm the suggested efficacy of ginseng or products with ginsenosides.


Subject(s)
Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Panax/chemistry , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Receptors, Cytoplasmic and Nuclear/drug effects , Animals , Cardiovascular Diseases/drug therapy , Female , Ginsenosides/isolation & purification , Humans , MAP Kinase Signaling System , Male , Neoplasms/drug therapy , Nervous System Diseases/drug therapy , Obesity/drug therapy , Peroxisome Proliferator-Activated Receptors/drug effects , Peroxisome Proliferator-Activated Receptors/physiology , Plant Extracts/isolation & purification , Receptors, Androgen/drug effects , Receptors, Androgen/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Estrogen/drug effects , Receptors, Estrogen/physiology , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/physiology
5.
Andrologia ; 49(5)2017 Jun.
Article in English | MEDLINE | ID: mdl-27546106

ABSTRACT

The aim of this study was to evaluate the effect of titanium dioxide (TiO2 ), a widely produced and consumed pigment in various food products, on the post-natal development of male albino rat seminal vesicle and thyroid hormones, as well as to evaluate the ameliorative effect of aged garlic extract (AGE) on TiO2 -induced alterations. Forty male rat pups (3 weeks old) were divided into four equal groups. The 1st group received distilled water orally (control group), 2nd group was given 2 ml kg-1 AGE, 3rd group was administered TiO2 (5 g kg-1 BW) day after day for 65 days, and the 4th group administered AGE 6 hr prior to TiO2 gavage. TiO2 -exposed rats showed nonsignificant changes in the serum testosterone, TSH, T3 and T4 , while serum glucose showed a significant decrease. Androgen receptor (AR) mRNA expression was significantly down-regulated and weak signal of AR immune labelling. Histopathologically, the epithelium cell lining of seminal vesicles showed focal areas of necrosis and fibrous tissue with the prominent fibrous stroma of the atrophied glands. Meanwhile, AGE supplementation ameliorated the deleterious effects of TiO2 intoxication through protecting the tissues from oxidative stress caused by TiO2 . In summary, oral administration of TiO2 resulted in abnormal developmental events in male rat seminal vesicle and AGE able to reduce TiO2 toxicity.


Subject(s)
Garlic/chemistry , Plant Extracts/pharmacology , Receptors, Androgen/drug effects , Seminal Vesicles/growth & development , Thyroid Hormones/blood , Titanium/toxicity , Animals , Blood Glucose/analysis , Male , Plant Extracts/administration & dosage , Rats , Rats, Wistar , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Seminal Vesicles/drug effects , Seminal Vesicles/pathology , Testosterone/blood , Thyrotropin/blood , Thyroxine/blood , Titanium/administration & dosage , Triiodothyronine/blood
6.
Toxicol Appl Pharmacol ; 296: 10-8, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26876616

ABSTRACT

Nausea and vomiting are components of a complex mechanism that signals food avoidance and protection of the body against the absorption of ingested toxins. This response can also be triggered by pharmaceuticals. Predicting clinical nausea and vomiting liability for pharmaceutical agents based on pre-clinical data can be problematic as no single animal model is a universal predictor. Moreover, efforts to improve models are hampered by the lack of translational animal and human data in the public domain. AZD3514 is a novel, orally-administered compound that inhibits androgen receptor signaling and down-regulates androgen receptor expression. Here we have explored the utility of integrating data from several pre-clinical models to predict nausea and vomiting in the clinic. Single and repeat doses of AZD3514 resulted in emesis, salivation and gastrointestinal disturbances in the dog, and inhibited gastric emptying in rats after a single dose. AZD3514, at clinically relevant exposures, induced dose-responsive "pica" behaviour in rats after single and multiple daily doses, and induced retching and vomiting behaviour in ferrets after a single dose. We compare these data with the clinical manifestation of nausea and vomiting encountered in patients with castration-resistant prostate cancer receiving AZD3514. Our data reveal a striking relationship between the pre-clinical observations described and the experience of nausea and vomiting in the clinic. In conclusion, the emetic nature of AZD3514 was predicted across a range of pre-clinical models, and the approach presented provides a valuable framework for predicition of clinical nausea and vomiting.


Subject(s)
Models, Animal , Nausea/chemically induced , Pyridazines/adverse effects , Receptors, Androgen/physiology , Vomiting/chemically induced , Animals , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , Ferrets , Humans , Male , Nausea/blood , Nausea/diagnosis , Predictive Value of Tests , Rats , Rats, Wistar , Vomiting/blood , Vomiting/diagnosis
7.
Curr Opin Clin Nutr Metab Care ; 17(3): 226-35, 2014 May.
Article in English | MEDLINE | ID: mdl-24572835

ABSTRACT

PURPOSE OF REVIEW: Significant progress has been made in the field of defining and describing the pathophysiology of wasting conditions such as cachexia. The number of new promising drugs, nutritional therapy alternatives, and exercise/rehabilitation programs is increasing. The purpose of this review is to give an overview of recent clinical findings from intervention studies investigating multimodal anabolic therapies utilizing drug, nutritional, and/or exercise interventions in order to counteract wasting. RECENT FINDINGS: Anabolic agents such as ghrelin and selective androgen receptor modulators are under late-phase clinical testing and hold promise as new therapies, and their ability to mitigate weight loss and improve muscle mass and physical function is evaluated. In the past 2 years, eight new studies investigating interventions with anabolic potential in wasting have been published, among which three of these studies were multimodal. SUMMARY: Targeted anabolic therapies aiming to prevent or reverse wasting might involve a combination of anabolic pharmacologic drugs, nutrition, and physical exercise working concurrently to enhance muscle protein synthesis and reduce breakdown. Some anabolic pharmacological interventions demonstrate the potential to improve muscle mass, but the multimodal interventions seem in greater extent to also demonstrate improvement in physical function.


Subject(s)
Anabolic Agents/therapeutic use , Nutrition Therapy/methods , Wasting Syndrome/therapy , Cachexia/physiopathology , Cachexia/therapy , Clinical Trials as Topic , Combined Modality Therapy , Exercise , Exercise Therapy , Ghrelin/therapeutic use , Humans , Receptors, Androgen/drug effects , Receptors, Androgen/physiology , Wasting Syndrome/physiopathology
8.
Int J Cancer ; 131(6): 1423-34, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22052438

ABSTRACT

Development and progression of prostate cancer are intimately associated with androgen receptor (AR) signaling. The emergence of hormone-refractory prostate cancer and consequent failure of conventional androgen deprivation therapies make it necessary to bypass hormonal resistance by targeting the same signaling pathway at new intervention points. In our study, we showed that cryptotanshinone inhibited the growth of AR-positive prostate cancer cells, suggesting that cryptotanshinone affected AR function. Cryptotanshinone also profoundly inhibited the transcriptional activity of AR and suppressed the expression of several AR-target genes at the mRNA and the protein levels. At the molecular level, cryptotanshinone disrupted the interaction between AR and lysine-specific demethylase 1 (LSD1), and inhibited the complex of AR and LSD1 to the promoter of AR target genes without affecting the protein degradation and translocation of AR. Cryptotanshinone increased the mono-methyl and di-methylation of Histone H3 lysine 9 (H3K9), a repressive histone marker which is demethylated and activated by LSD1. These data suggest that cryptotanshinone functions via inhibition of LSD1, a protein that promotes AR-dependent transcriptional activity via derepression of H3K9. In summary, we describe a novel mechanism whereby cryptotanshinone down-regulates AR signaling via functional inhibition of LSD1-mediated demethylation of H3K9 and represses the transcriptional activity of AR. Our data suggest that cryptotanshinone can be developed as a potential therapeutic agent for prostate cancer.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Histone Demethylases/physiology , Phenanthrenes/pharmacology , Receptors, Androgen/physiology , Signal Transduction/drug effects , Active Transport, Cell Nucleus/drug effects , Cell Line, Tumor , Cell Proliferation , DNA/metabolism , Down-Regulation , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Signal Transduction/physiology
9.
Reproduction ; 143(2): 211-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22065861

ABSTRACT

7α-methyl-19-nortestosterone (MENT) is an androgen with potent gonadotropin inhibitory activity and prostate-sparing effects. These attributes give MENT advantages over testosterone as a male contraceptive, but, as in the case of testosterone, a partial dose-dependent suppression of spermatogenesis has been observed. Combination of testosterone or MENT with synthetic progestins improves the rate of azoospermia; however, it is unknown whether these combinations affect hormone androgenicity or exert synergistic effects via progestational or androgenic interaction. Herein, using transactivation assays, we examined the ability of MENT alone or combined with several 19-nor-derived synthetic progestins to activate androgen receptor (AR)-dependent gene transcription. In addition, the capability of 7α-methyl-estradiol (7α-methyl-E(2)), an aromatized metabolite of MENT, to transactivate gene transcription via estrogen receptor α (ERα; ESR1) or ERß (ESR2) was also investigated. As expected, MENT induced gene transactivation through either the progesterone receptor (PGR) or the AR. MENT was as efficient as progesterone in activating PGR-mediated reporter gene expression, but it was ten times more potent than testosterone and dihydrotestoterone in activating of AR-driven gene expression. The addition of increasing concentrations of other 19-nortestosterone derivatives (norethisterone or levonorgestrel) did not affect, in a significant manner, the ability of MENT to activate AR-dependent reporter gene transcription. The same results were obtained with different cell lines. 7α-Methyl-E(2) resulted in potent estrogen activity via both ER subtypes with efficiency similar to natural E(2). These results suggest that the addition of 19-nortestosterone-derived progestins, as a hormonal adjuvant in male fertility strategies for effective spermatogenic suppression, does not display any detrimental effect that would interfere with MENT androgenic transcriptional activity.


Subject(s)
Nandrolone/analogs & derivatives , Progestins/pharmacology , Receptors, Androgen/physiology , Transcriptional Activation/drug effects , Cells, Cultured , Contraceptive Agents, Female/administration & dosage , Contraceptive Agents, Female/pharmacology , Drug Combinations , Drug Evaluation, Preclinical , HEK293 Cells , HeLa Cells , Humans , Nandrolone/administration & dosage , Nandrolone/pharmacology , Progestins/administration & dosage , Receptors, Androgen/metabolism , Receptors, Estrogen/agonists , Receptors, Estrogen/metabolism , Receptors, Estrogen/physiology , Receptors, Progesterone/agonists , Receptors, Progesterone/metabolism , Receptors, Progesterone/physiology , Transcription, Genetic/drug effects , Transfection , Treatment Outcome
10.
Biol Pharm Bull ; 34(10): 1584-8, 2011.
Article in English | MEDLINE | ID: mdl-21963499

ABSTRACT

Androgen deprivation therapy is a common treatment strategy for advanced prostate cancer. Though effective initially, the tumor often progresses to androgen independent stage in most patients eventually after a period of remission. One of the key factors of development of resistance is reflected in expression of androgen receptor (AR). In this study, we showed that two natural compounds, physalins A and B, both secosteriods from Physalisalkekengi var. franchetii, significantly inhibited the growth of two androgen-independent cell lines CWR22Rv1 and C42B, induced apoptosis via c-Jun N-terminal kinase (JNK) and/or extracellular signal-regulated kinase (ERK) activation, and decreased AR expression. In addition, physalins A and B down-regulated the expression of prostate specific antigen (PSA) in C42B cells which is a target gene of AR. Our results suggest that physalin A and B might be useful agents in preventing the growth of androgen-independent prostate cancer (AI-PCa).


Subject(s)
Androgen Antagonists/pharmacology , Androgens/physiology , Physalis , Phytotherapy , Prostatic Neoplasms/drug therapy , Receptors, Androgen/physiology , Secosteroids/pharmacology , Androgen Antagonists/isolation & purification , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/drug effects , Gene Expression Regulation, Neoplastic , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Molecular Targeted Therapy , Plant Preparations/isolation & purification , Plant Preparations/pharmacology , Prostate-Specific Antigen/antagonists & inhibitors , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , Secosteroids/isolation & purification , Signal Transduction/drug effects , Signal Transduction/genetics
11.
Environ Toxicol Chem ; 29(7): 1529-36, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20821602

ABSTRACT

The present study is the second in a series aiming at a systematic inventory of specific toxic effects of oils. By employing a recombinant yeast stably transfected with human estrogen receptor-alpha (ERalpha) or -beta (ERbeta) or androgen receptor (AR) and expressing yeast enhanced green fluorescent protein, the (anti-)estrogenicity and (anti-)androgenicity of 11 crude oils and refined products were studied. None of the oils tested had significant estrogenic effects in the ERalpha assay or androgenic effects in the AR assay. However, all oils were capable of inducing estrogenic responses in the ERbeta assay, with several responses being above even the maximal response of the standard 17beta-estradiol (E2). Based on the lowest effect concentrations, the potencies of oils in all the assays were between four and seven orders of magnitude lower than those of the standards E2 or testosterone (T). The potencies of the actual individual petrochemical agonists may, however, be relatively high, considering the complex composition of oils. Additive effects, antagonistic effects, and a synergistic effect were measured in the assays upon coexposure to a fixed concentration of standard (E2 or T) and increasing concentrations of oils. To investigate whether the observed effects were receptor-mediated, coexposures to the synthetic inhibitors ICI 182,780 (ERbeta assay) or flutamide (AR assay), a fixed concentration of standard, and various concentrations of oils were performed. The results suggested that the androgenic effects were receptor mediated, whereas the estrogenic effects may be only partially mediated via the receptor. The present study indicates that oils contain compounds with possible endocrine-disrupting potential, some of them acting via the hormone receptors.


Subject(s)
Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Petroleum/toxicity , Receptors, Androgen/physiology , Saccharomyces cerevisiae/drug effects , Androgen Antagonists/pharmacology , Flutamide/pharmacology , Recombination, Genetic , Saccharomyces cerevisiae/genetics
13.
Acta Pharm Hung ; 79(1): 3-9, 2009.
Article in Hungarian | MEDLINE | ID: mdl-19526676

ABSTRACT

Epilobium parviflorum Schreb. (willow-herb) is used for the treatment of benign prostatic hyperplasia (BPH), but its biological action is not entirely identified. This paper aims to report data on willow-herbs probable biological effect. In vitro studies have been made to investigate different aspects of its antioxidant capacity, anti-inflammatory (COX-inhibitory) action, steroid-receptor -agonistic and -antagonostic effect as well as aromatase-inhibitory effect. Based on our results, willow-herb possess remarkable antioxidant and COX-inhibitory action.


Subject(s)
Epilobium , Plant Preparations/therapeutic use , Prostatic Hyperplasia/drug therapy , Humans , Male , Phytotherapy , Plant Preparations/pharmacology , Receptors, Androgen/drug effects , Receptors, Androgen/physiology , Receptors, Estrogen/drug effects , Receptors, Estrogen/physiology
14.
Mol Endocrinol ; 23(3): 412-21, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19131511

ABSTRACT

Environmental sodium arsenite is a toxin that is associated with male infertility due to decreased and abnormal sperm production. Arsenic trioxide (ATO), another inorganic trivalent semimetal, is an effective therapy for acute promyelocytic leukemia, and there is investigation of its possible efficacy in prostate cancer. However, the mechanism of arsenic action in male urogenital tract tissues is not clear. Because the androgen receptor (AR) plays an important role in spermatogenesis and prostate cancer, we explored the possibility that trivalent arsenic regulates AR function. We found that arsenic inhibited AR transcriptional activity in prostate cancer and Sertoli cells using reporter gene assays testing several androgen response element-containing regions and by assessing native target gene expression. Arsenic inhibition of AR activity was not due to down-regulation of AR protein levels, decreased hormone binding to AR, disruption of AR nuclear translocation, or interference with AR-DNA binding in vitro. However, chromatin immunoprecipitation studies revealed that arsenic inhibited AR recruitment to an AR target gene enhancer in vivo. Consistent with a deficiency in AR-chromatin binding, arsenic disrupted AR amino and carboxyl termini interaction. Furthermore, ATO caused a significant decrease in prostate cancer cell proliferation that was more pronounced in cells expressing AR compared with cells depleted of AR. In addition, inhibition of AR activity by ATO and by the AR antagonist, bicalutamide, was additive. Thus, arsenic-induced male infertility may be due to inhibition of AR activity. Further, because AR is an important target in prostate cancer therapy, arsenic may serve as an effective therapeutic option.


Subject(s)
Androgen Receptor Antagonists , Arsenicals/pharmacology , Oxides/pharmacology , Transcriptional Activation/drug effects , Androgen Antagonists/pharmacology , Androgens/metabolism , Androgens/pharmacology , Anilides/administration & dosage , Anilides/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Arsenic Trioxide , Arsenicals/administration & dosage , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Drug Synergism , Humans , Male , Nitriles/administration & dosage , Nitriles/pharmacology , Nuclear Receptor Coactivator 2/metabolism , Oxides/administration & dosage , Prostatic Neoplasms/pathology , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Receptors, Androgen/physiology , Response Elements/physiology , Tosyl Compounds/administration & dosage , Tosyl Compounds/pharmacology , Tumor Cells, Cultured
15.
Reprod Biol Endocrinol ; 6: 57, 2008 Nov 24.
Article in English | MEDLINE | ID: mdl-19025659

ABSTRACT

BACKGROUND: High dietary intake of selenium or soybean isoflavones reduces prostate cancer risk. These components each affect androgen-regulated gene expression. The objective of this work was to determine the combined effects of selenium and isoflavones on androgen-regulated gene expression in rat prostate. METHODS: Male Noble rats were exposed from conception until 200 days of age to diets containing an adequate (0.33-0.45 mg/kg diet) or high (3.33-3.45 mg/kg) concentration of selenium as Se-methylselenocysteine and a low (10 mg/kg) or high (600 mg/kg) level of isoflavones in a 2 x 2 factorial design. Gene expression in the dorsolateral prostate was determined for the androgen receptor, for androgen-regulated genes, and for Akr1c9, whose product catalyzes the reduction of dihydrotestosterone to 5alpha-androstane-3alpha, 17beta-diol. Activity of hepatic glutathione peroxidise 1 and of prostatic 5alpha reductase were also assayed. RESULTS: There were no differences due to diet in activity of liver glutathione peroxidase activity. Total activity of 5alpha reductase in prostate was significantly lower (p = 0.007) in rats fed high selenium/high isoflavones than in rats consuming adequate selenium/low isoflavones. High selenium intake reduced expression of the androgen receptor, Dhcr24 (24-dehydrocholesterol reductase), and Abcc4 (ATP-binding cassette sub-family C member 4). High isoflavone intake decreased expression of Facl3 (fatty acid CoA ligase 3), Gucy1a3 (guanylate cyclase alpha 3), and Akr1c9. For Abcc4 the combination of high selenium/high isoflavones had a greater inhibitory effect than either treatment alone. The effects of selenium on gene expression were always in the direction of chemoprevention CONCLUSION: These results suggest that combined intake of high selenium and high isoflavones may achieve a greater chemopreventive effect than either compound supplemented individually.


Subject(s)
Androgens/pharmacology , Diet , Gene Expression Regulation/drug effects , Isoflavones/pharmacology , Prostate/drug effects , Selenium/pharmacology , Animal Feed/analysis , Animals , Diet/veterinary , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Gene Expression Profiling , Isoflavones/administration & dosage , Isoflavones/analysis , Male , Osmolar Concentration , Prostate/metabolism , Rats , Receptors, Androgen/physiology , Selenium/administration & dosage , Selenium/analysis
16.
Horm Behav ; 54(4): 557-64, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18582470

ABSTRACT

The neuronal nitric oxide synthase (nNOS) is involved in the control of male and female sexual behavior and its distribution in several regions of the limbic-hypothalamic system, as well as its coexistence with gonadal hormones' receptors, suggests that these hormones may play a significant role in controlling its expression. However, data illustrating the role of gonadal hormones in controlling the nNOS expression are, at present, contradictory, even if they strongly suggest an involvement of testosterone (T) in the regulation of nNOS. The action of T may be mediated through androgen (AR) or, after aromatization to estradiol (E(2)), through estrogen receptors. To elucidate the role of AR on nNOS expression, we compared male and female rats with a non-functional mutation of AR (Tfm, testicular feminization mutation) to their control littermates. We investigated some hypothalamic and limbic nuclei involved in the control of sexual behavior [medial preoptic area (MPA), paraventricular (PVN), arcuate (ARC), ventromedial (VMH) and stria terminalis (BST) nuclei]. In BST (posterior subdivision), VMH (ventral subdivision), and MPA we detected a significant sexual dimorphism in control animals and a decrease of nNOS positive elements in Tfm males compared to their littermate. In addition, we observed a significant increase of nNOS positive elements in BST (posterior) of Tfm females. No significant changes were observed in the other nuclei. These data indicate that, contrary to current opinions, androgens, through the action of AR may have a relevant role in the organization and modulation of the nNOS hypothalamic system.


Subject(s)
Hypothalamus/metabolism , Limbic System/metabolism , Nitric Oxide Synthase Type I/metabolism , Receptors, Androgen/physiology , Virilism/metabolism , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/metabolism , Androgens/physiology , Animals , Animals, Genetically Modified , Female , Limbic System/physiology , Male , Models, Biological , Rats , Rats, Wistar , Receptors, Androgen/metabolism , Sex Differentiation/physiology
17.
J Ethnopharmacol ; 118(3): 412-7, 2008 Aug 13.
Article in English | MEDLINE | ID: mdl-18562138

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Rosa rugosa Thunb. (Rosaceae) has been traditionally used for treatments of diabetes, chronic inflammatory diseases, pain, and anticancer in Korea. AIM OF STUDY: We investigate the inhibitory effect of histone acetyltransferase activity from the methanol extract of stems of Rosa rugosa on androgen receptor-mediated transcriptional regulation. MATERIALS AND METHODS: For the present study, Rosa rugosa methanol extract (RRME) was obtained from stem part of Rosa rugosa using methanol extraction. Histone acetyltransferase assay were performed to measure the inhibitory effect on acetylation, reporter assay, real-time PCR and ChIP assay were performed to measure androgen receptor-mediated transcriptional regulation, and MTT test were performed to measure cell viability. RESULTS: RRME inhibited both p300 and CBP (60-70% at 100 microg/ml) activity. We show RRME mediates agonist-dependent androgen receptor (AR) activation and suppresses antagonist-dependent inhibition. RRME treatment also decreased transcription of AR regulated genes and also reduced histone H3 and AR acetylation in the promoters of prostate-specific antigen (PSA) and beta-2-microglobulin (B2M). Finally, RRME treatment reduced the growth of LNCaP, a human prostate cancer cell line. CONCLUSION: These results demonstrate RRME is a potent HAT inhibitor, which reduced AR and histone acetylation leading to decreased AR-mediated transcription and reduced LNCaP cell growth.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Plant Extracts/pharmacology , Receptors, Androgen/physiology , Rosa , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Rosa/chemistry , Transcription, Genetic
18.
J Steroid Biochem Mol Biol ; 110(3-5): 236-43, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18515094

ABSTRACT

Androgen signaling, mediated by the androgen receptor (AR), is a critical factor influencing growth of normal and malignant breast cells. Given the increasing use of exogenous androgens in women, a better understanding of androgen action in the breast is essential. This study compared the effects of 5alpha-dihydrotestosterone (DHT) and a synthetic androgen, mibolerone, on estradiol (E(2))-induced proliferation of breast cancer cells. DHT modestly inhibited E(2)-induced proliferation and mibolerone significantly inhibited proliferation in T-47D cells. The effects of both androgens could be reversed by an AR antagonist, suggesting that their actions were mediated, in part, by AR. Whereas high physiological doses (10-100nM) of DHT reduced E(2)-mediated induction of the estrogen-regulated gene progesterone receptor (PR) to basal levels, mibolerone at lower doses (1nM) eliminated PR expression, suggesting that mibolerone may also act via the PR. In the AR positive, PR-negative MCF-7 cells, mibolerone had modest effects on E(2)-induced proliferation, but was a potent inhibitor of proliferation in the AR positive, PR positive MCF-7M11 PRA cells. The effects of mibolerone in breast cancer cells were similar to those of the progestin, medroxyprogesterone acetate. Our results demonstrate that mibolerone can have both androgenic and progestagenic actions in breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation/drug effects , Nandrolone/analogs & derivatives , Receptors, Androgen/physiology , Receptors, Progesterone/physiology , Androgens , Dihydrotestosterone/pharmacology , Drug Evaluation, Preclinical , Estradiol/pharmacology , Humans , Medroxyprogesterone Acetate/pharmacology , Nandrolone/pharmacology , Progestins/pharmacology , Testosterone Congeners/pharmacology , Tumor Cells, Cultured
19.
Prostate ; 67(11): 1152-62, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17503469

ABSTRACT

BACKGROUND: Dehydroepiandrosterone (DHEA) is an endogenous steroid that is metabolized to androgens and/or estrogens in the human prostate. DHEA levels decline with age, and use of DHEA supplements to retard the aging process is of unproved effectiveness and safety. LNCaP and LAPC-4 prostate cancer cells were used to determine whether DHEA-modulated proliferation and prostate specific antigen (PSA) production were mediated via the androgen receptor (AR) and/or ERbeta. METHODS: Cells were treated with DHEA, DHT, or E(2) and antagonists to AR (Casodex-bicalutamide) or ER (ICI 182,780) or siRNA to the respective receptors. Proliferation was assessed by MTT assay and PSA mRNA and protein secretion were measured by quantitative real-time PCR and ELISA. Associations of AR and ERbeta were analyzed by co-immunoprecipitation studies and fluorescent confocal microscopy. RESULTS: DHEA-, T-, and E(2)-induced proliferation of LNCaP cells was blunted by Casodex but not by ICI treatment. In LNCaP cells, Casodex and ICI suppressed hormone-induced PSA production. In LAPC-4 cells, DHT-stimulated PSA mRNA was inhibited by Casodex and ICI, and the minimal stimulation by DHEA was inhibited by ICI. Use of siRNAs confirmed involvement of AR and ERbeta in hormone-induced PSA production while AR-ERbeta co-association was suggested by immunoprecipitation and nuclear co-localization. CONCLUSIONS: These findings support involvement of both AR and ERbeta in mediating DHEA-, DHT-, and E(2)-induced PSA expression in prostate cancer cells.


Subject(s)
Androgen Receptor Antagonists , Dehydroepiandrosterone/pharmacology , Estrogen Receptor beta/antagonists & inhibitors , Prostate-Specific Antigen/biosynthesis , Prostatic Neoplasms/pathology , Testosterone/analogs & derivatives , Androgen Antagonists/pharmacology , Anilides/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor beta/physiology , Fulvestrant , Gene Expression/drug effects , Humans , Male , Nitriles/pharmacology , Prostate-Specific Antigen/genetics , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , RNA, Messenger/analysis , RNA, Small Interfering/pharmacology , Receptors, Androgen/physiology , Testosterone/pharmacology , Tosyl Compounds/pharmacology
20.
Endocr Relat Cancer ; 13(3): 751-78, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16954429

ABSTRACT

Despite advances in the understanding of prostate cancer (PCa) growth and development, it is still the leading incidence of cases and the second leading cause of mortality due to cancer in men. The problem of early diagnosis compounded with the emergence of androgen independence during commonly used anti-androgen therapy of PCa, have been discouraging for optimal therapeutic response. Recently, many chemopreventive agents, including silibinin, inositol hexaphosphate, decursin, apigenin, acacetin, grape seed extract, curcumin, and epigallocatechin-3 gallate have been identified in laboratory studies, which could be useful in the management of PCa. In vivo pre-clinical studies have indicated chemopreventive effect of many such agents in PCa xenograft and transgenic mouse models. The molecular targets of these agents include cell signaling, cell-cycle regulators, and survival/apoptotic molecules, which are implicated in uncontrolled PCa growth and progression. Furthermore, angiogenic and metastatic targets, including vascular endothelial growth factor, hypoxia-inducing factor-1alpha, matrix metalloproteinase, and urokinase-type plasminogen activator are also modulated by many chemopreventive agents to suppress the growth and invasive potential of PCa. This review focuses on novel PCa chemopreventive observations in laboratory studies, which could provide the rationale for the prospective use of chemopreventive agents in translational studies.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Prostatic Neoplasms/prevention & control , Angiogenesis Inhibitors/therapeutic use , Cell Cycle/drug effects , Cell Survival/drug effects , Chemoprevention , Humans , Male , Neoplasm Invasiveness , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Protein-Tyrosine Kinases/physiology , Receptors, Androgen/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL