Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Biol Chem ; 284(52): 36367-36376, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19846549

RESUMEN

Androgen replacement therapy is a promising strategy for the treatment of frailty; however, androgens pose risks for unwanted effects including virilization and hypertrophy of reproductive organs. Selective Androgen Receptor Modulators (SARMs) retain the anabolic properties of androgens in bone and muscle while having reduced effects in other tissues. We describe two structurally similar 4-aza-steroidal androgen receptor (AR) ligands, Cl-4AS-1, a full agonist, and TFM-4AS-1, which is a SARM. TFM-4AS-1 is a potent AR ligand (IC(50), 38 nm) that partially activates an AR-dependent MMTV promoter (55% of maximal response) while antagonizing the N-terminal/C-terminal interaction within AR that is required for full receptor activation. Microarray analyses of MDA-MB-453 cells show that whereas Cl-4AS-1 behaves like 5alpha-dihydrotestosterone (DHT), TFM-4AS-1 acts as a gene-selective agonist, inducing some genes as effectively as DHT and others to a lesser extent or not at all. This gene-selective agonism manifests as tissue-selectivity: in ovariectomized rats, Cl-4AS-1 mimics DHT while TFM-4AS-1 promotes the accrual of bone and muscle mass while having reduced effects on reproductive organs and sebaceous glands. Moreover, TFM-4AS-1 does not promote prostate growth and antagonizes DHT in seminal vesicles. To confirm that the biochemical properties of TFM-4AS-1 confer tissue selectivity, we identified a structurally unrelated compound, FTBU-1, with partial agonist activity coupled with antagonism of the N-terminal/C-terminal interaction and found that it also behaves as a SARM. TFM-4AS-1 and FTBU-1 represent two new classes of SARMs and will allow for comparative studies aimed at understanding the biophysical and physiological basis of tissue-selective effects of nuclear receptor ligands.


Asunto(s)
Anabolizantes/farmacocinética , Andrógenos , Congéneres de la Testosterona/farmacología , Anabolizantes/química , Animales , Antígenos Virales de Tumores/metabolismo , Masculino , Regiones Promotoras Genéticas , Próstata/crecimiento & desarrollo , Próstata/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/metabolismo , Congéneres de la Testosterona/química , Proteínas del Núcleo Viral/metabolismo
2.
Pharmacol Biochem Behav ; 80(3): 379-85, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15740779

RESUMEN

Nuclear receptors are transcription factors that usually interact, in a ligand-dependent manner, with specific DNA sequences located within promoters of target genes. The nuclear receptors can also be controlled in a ligand-independent manner via the action of membrane receptors and cellular signaling pathways. 5-Tetradecyloxy-2-furancarboxylic acid (TOFA) was shown to stimulate transcription from the MMTV promoter via chimeric receptors that consist of the DNA binding domain of GR and the ligand binding regions of the PPARbeta or LXRbeta nuclear receptors (GR/PPARbeta and GR/LXRbeta). TOFA and hydroxycholesterols also modulate transcription from NF-kappaB- and AP-1-controlled reporter genes and induce neurite differentiation in PC12 cells. In CV-1 cells that express D(1) dopamine receptors, D(1) dopamine receptor stimulation was found to inhibit TOFA-stimulated transcription from the MMTV promoter that is under the control of chimeric GR/PPARbeta and GR/LXRbeta receptors. Treatment with the D(1) dopamine receptor antagonist, SCH23390, prevented dopamine-mediated suppression of transcription, and by itself increased transcription controlled by GR/LXRbeta. Furthermore, combined treatment of CV-1 cells with TOFA and SCH23390 increased transcription controlled by the GR/LXRbeta chimeric receptor synergistically. The significance of this in vitro synergy was demonstrated in vivo, by the observation that SCH23390 (but not haloperidol)-mediated catalepsy in rats was potentiated by TOFA, thus showing that an agent that mimics the in vitro activities of compounds that activate members of the LXR and PPAR receptor families can influence D1 dopamine receptor elicited responses.


Asunto(s)
Receptor Cross-Talk/fisiología , Receptores de Dopamina D1/fisiología , Transducción de Señal/fisiología , Factor de Transcripción AP-1/fisiología , Transcripción Genética/fisiología , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Dopamina/farmacología , Antagonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Macaca , Masculino , Virus del Tumor Mamario del Ratón/genética , Ratas , Receptor Cross-Talk/efectos de los fármacos , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
3.
Mol Endocrinol ; 18(8): 2011-23, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15131260

RESUMEN

The androgen receptor (AR) is a member of the steroid receptor superfamily that plays critical roles in the development and maintenance of the male reproductive system and in prostate cancer. Actions of AR are controlled by interaction with several classes of coregulators. In this study, we have identified LATS2/KPM as a novel AR-interacting protein. Human LATS1 and LATS2 are tumor suppressors that are homologs of Drosophila warts/lats. The interaction surface of LATS2 is mapped to the central region of the protein, whereas the AR ligand binding domain is sufficient for this interaction. LATS2 functions as a modulator of AR by inhibiting androgen-regulated gene expression. The mechanism of LATS2-mediated repression of AR activity appears to involve the inhibition of AR NH2- and COOH-terminal interaction. Chromatin immunoprecipitation assays in human prostate carcinoma cells reveal that LATS2 and AR are present in the protein complex that binds at the promoter and enhancer regions of prostate-specific antigen, and overexpression of LATS2 results in a reduction in androgen-induced expression of endogenous prostate-specific antigen mRNA. Immunohistochemistry shows that LATS2 and AR are localized within the prostate epithelium and that LATS2 expression is lower in human prostate tumor samples than in normal prostate. The results suggest that LATS2 may play a role in AR-mediated transcription and contribute to the development of prostate cancer.


Asunto(s)
Antagonistas de Receptores Androgénicos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Elementos de Facilitación Genéticos/genética , Humanos , Ligandos , Masculino , Regiones Promotoras Genéticas/genética , Próstata/metabolismo , Próstata/patología , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Unión Proteica , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Estructura Terciaria de Proteína , Receptores Androgénicos/química , Receptores Androgénicos/metabolismo , Activación Transcripcional , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/aislamiento & purificación
4.
Maturitas ; 48(4): 463-71, 2004 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-15283940

RESUMEN

OBJECTIVE: Develop a rat model for the evaluation of estrogenic agents on estrogen deficiency-induced changes in thermoregulation. METHODS: OVX rats are impaired in thermoregulation which manifests itself as an elevation in basal tail skin temperature (TST) and are less able to respond to temperature changes than intact rats. RESULTS: Administration of estrogen subcutaneously to estrogen-depleted rats either as depot formulation, biodegradable pellets, or daily injections, suppressed the increased TST. OVX rats maintained on a diet devoid of phytoestrogens had a higher TST by several degrees than OVX rats fed normal chow, offering greater ability to test estrogenic agents on thermoregulation. Depletion of estrogen in intact rats via chronic administration of leuprolide acetate, a GnRH agonist, also increased TST, which was in turn suppressed by estrogen. In intact rats, tamoxifen exhibited estrogen antagonistic activity elevating TST, while in OVX rats, tamoxifen acted as an agonist by suppressing TST. CONCLUSION: OVX rats kept on a diet devoid of phytoestrogens are a sensitive model for estrogen-dependent thermoregulation.


Asunto(s)
Estrógenos/farmacología , Fitoestrógenos/farmacología , Temperatura Cutánea/efectos de los fármacos , Cola (estructura animal)/fisiología , Tamoxifeno/farmacología , Animales , Antagonistas de Estrógenos/farmacología , Femenino , Modelos Animales , Ovariectomía , Ratas , Ratas Sprague-Dawley , Cola (estructura animal)/efectos de los fármacos , Factores de Tiempo
5.
J Biol Chem ; 280(47): 38898-901, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16166078

RESUMEN

Signaling by androgens and interferons (IFN) plays an important role in prostate cancer initiation and progression. Using microarray analysis, we describe here a functional cross-talk between dihydrotestosterone and interferon signaling. Glutathione S-transferase pull-down and co-immunoprecipitation experiments reveal that the androgen receptor and the interferon-activated RNase L interact with each other in a ligand-dependent manner. Furthermore, overexpression of wild type RNase L confers IFN sensitivity to a dihydrotestosterone-inducible reporter gene, whereas R462Q-mutated RNase L does not. Based on our data we hypothesize that in 22RV1 cells, activated androgen receptor (AR) contributes to the insensitivity to IFN of the cell. Accordingly, we show that AR knockdown restores responsiveness to IFNgamma. Our findings support a model in which both the activation of AR and the down-regulation of IFN signaling can synergize to promote cell survival and suppress apoptosis. This model provides the molecular basis to understand how mutated RNase L can lead to early onset PCa and illustrates how inflammatory cytokines and nuclear hormone signaling contribute to tumor development.


Asunto(s)
Endorribonucleasas/metabolismo , Interferones/metabolismo , Receptores Androgénicos/metabolismo , Andrógenos/metabolismo , Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular , Dihidrotestosterona/farmacología , Endorribonucleasas/genética , Activación Enzimática , Femenino , Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Interferón gamma/farmacología , Ligandos , Masculino , Modelos Biológicos , Mutación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptor Cross-Talk , Receptores Androgénicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA