Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Endocrinology ; 161(8)2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32609838

RESUMEN

Disorders of androgen imbalance, such as hyperandrogenism in females or hypoandrogenism in males, increase risk of visceral adiposity, type 2 diabetes, and infertility. Androgens act upon androgen receptors (AR) which are expressed in many tissues. In the brain, AR are abundant in hypothalamic nuclei involved in regulation of reproduction and energy homeostasis, yet the role of androgens acting via AR in specific neuronal populations has not been fully elucidated. Leptin receptor (LepRb)-expressing neurons coexpress AR predominantly in hypothalamic arcuate and ventral premammillary nuclei (ARH and PMv, respectively), with low colocalization in other LepRb neuronal populations, and very low colocalization in the pituitary gland and gonads. Deletion of AR from LepRb-expressing cells (LepRbΔAR) has no effect on body weight, energy expenditure, and glucose homeostasis in male and female mice. However, LepRbΔAR female mice show increased body length later in life, whereas male LepRbΔAR mice show an increase in spontaneous ambulatory activity. LepRbΔAR mice display typical pubertal timing, estrous cycles, and fertility, but increased testosterone levels in males. Removal of sex steroid negative feedback action induced an exaggerated rise in luteinizing hormone in LepRbΔAR males and follicle-stimulating hormone in LepRbΔAR females. Our findings show that AR can directly affect a subset of ARH and PMv neurons in a sex-specific manner and demonstrate specific androgenic actions in the neuroendocrine hypothalamus.


Asunto(s)
Sistemas Neurosecretores/fisiopatología , Receptores Androgénicos/genética , Receptores de Leptina/genética , Caminata/fisiología , Animales , Metabolismo Energético/genética , Epistasis Genética , Femenino , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Sistemas Neurosecretores/metabolismo , Sistemas Neurosecretores/patología , Receptores Androgénicos/deficiencia , Caracteres Sexuales , Transducción de Señal/genética
2.
Hypertension ; 63(6): 1345-53, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24688120

RESUMEN

The male sex has a higher risk to develop coronary artery diseases, including atherosclerosis. The androgen receptor (AR) is expressed in several atherosclerosis-associated cell types, including monocytes/macrophages, endothelial cells (ECs), and smooth muscle cells (SMCs), but its pathophysiological role in each cell type during the development of atherosclerotic lesions remains unclear. Using the Cre-loxP system, we selectively knocked out AR in these 3 cell types and the resultant AR knockout (ARKO) mice, monocyte/macrophage ARKO, EC-ARKO, and SMC-ARKO, were then crossed with the low-density lipoprotein receptor (LDLR) deficient (LDLR(-/-)) mice to develop monocyte/macrophage ARKO-LDLR(-/-), EC-ARKO-LDLR(-/-), and SMC-ARKO-LDLR(-/-) mice for the study of atherosclerosis. The results showed that the monocyte/macrophage ARKO-LDLR(-/-) mice had reduced atherosclerosis compared with the wild-type-LDLR(-/-) control mice. However, no significant difference was detected in EC-ARKO-LDLR(-/-) and SMC-ARKO-LDLR(-/-) mice compared with wild-type-LDLR(-/-) mice, suggesting that the AR in monocytes/macrophages, and not in ECs and SMCs, plays a major role to promote atherosclerosis. Molecular mechanism dissection suggested that AR in monocytes/macrophages upregulated the tumor necrosis factor-α, integrin ß2, and lectin-type oxidized LDL receptor 1 molecules that are involved in 3 major inflammation-related processes in atherosclerosis, including monocytes/macrophages migration and adhesion to human umbilical vein ECs, and subsequent foam cell formation. Targeting AR via the AR degradation enhancer, ASC-J9, in wild-type-LDLR(-/-) mice showed similar effects as seen in monocyte/macrophage ARKO-LDLR(-/-) mice with little influence on lipid profile. In conclusion, the AR in monocytes/macrophages plays key roles in atherosclerosis and targeting AR with ASC-J9 may represent a new potential therapeutic approach to battle atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Receptores Androgénicos/deficiencia , Animales , Aterosclerosis/genética , Aterosclerosis/prevención & control , Western Blotting , Antígenos CD18/metabolismo , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Células Cultivadas , Curcumina/análogos & derivados , Curcumina/farmacología , Dieta Alta en Grasa , Células Espumosas/citología , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Monocitos/citología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Receptores Androgénicos/genética , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factor de Necrosis Tumoral alfa/metabolismo
3.
Mol Cell Biol ; 27(13): 4807-14, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17470551

RESUMEN

Androgen receptor (AR) mediates diverse androgen actions, particularly reproductive processes in males and females. AR-mediated androgen signaling is considered to also control metabolic processes; however, the molecular basis remains elusive. In the present study, we explored the molecular mechanism of late-onset obesity in male AR null mutant (ARKO) mice. We determined that the obesity was caused by a hypercorticoid state. The negative feedback system regulating glucocorticoid production was impaired in ARKO mice. Male and female ARKO mice exhibited hypertrophic adrenal glands and glucocorticoid overproduction, presumably due to high levels of adrenal corticotropic hormone. The pituitary glands of the ARKO males had increased expression of proopiomelanocortin and decreased expression of the glucocorticoid receptor (GR). There were no overt structural abnormalities and no alteration in the distribution of cell types in the pituitaries of male ARKO mice. Additionally, there was normal production of the other hormones within the glucocorticoid feedback system in both the pituitary and hypothalamus. In a cell line derived from pituitary glands, GR expression was under the positive control of the activated AR. Thus, this study suggests that the activated AR supports the negative feedback regulation of glucocorticoid production via up-regulation of GR expression in the pituitary gland.


Asunto(s)
Glucocorticoides/biosíntesis , Hipófisis/metabolismo , Receptores Androgénicos/metabolismo , Glándulas Suprarrenales/patología , Hormona Adrenocorticotrópica/sangre , Animales , Apoptosis , Línea Celular , Proliferación Celular , Corticosterona/sangre , Dihidrotestosterona , Retroalimentación Fisiológica , Femenino , Regulación de la Expresión Génica , Hipertrofia , Sistema Hipotálamo-Hipofisario , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Hipófisis/patología , Sistema Hipófiso-Suprarrenal , Receptores Androgénicos/deficiencia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
4.
J Mol Endocrinol ; 35(3): 547-55, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16326839

RESUMEN

Androgens mediate their effects in target cells via the androgen receptor (AR), which acts predominantly as a ligand-dependent transcription factor. In addition, androgens induce rapid activation of second messenger signal transduction cascades, and this is thought to occur via non-genomic mechanisms. We have used the Cre/loxP system to generate an AR knockout (ARKO) mouse targeting exon 3, which encodes the second zinc finger of the DNA-binding domain. To generate universal ARKO mice, floxed AR mice were mated with CMV-Cre mice, which express Cre recombinase ubiquitously. Deletion of the floxed allele in our mice does not disrupt the reading frame, and has been designed so that the mutant AR can bind ligand but not target genes. ARKO males displayed a complete androgen insensitivity phenotype, with female external genitalia and a reduction in body weight compared with wild-type males (P < 0.001). Testes of ARKO males were smaller than control males (P < 0.0001) and were located intra-abdominally. We have demonstrated that genotypically XY mice lacking the second zinc finger of the AR have a female phenotype, and we conclude that the genomic actions of the AR (mediated by DNA binding) are indispensable for normal male sexual differentiation.


Asunto(s)
Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Diferenciación Sexual/genética , Síndrome de Resistencia Androgénica/genética , Síndrome de Resistencia Androgénica/metabolismo , Síndrome de Resistencia Androgénica/patología , Animales , Secuencia de Bases , Sitios de Unión/genética , ADN Complementario/genética , Femenino , Marcación de Gen , Genoma , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Embarazo , Receptores Androgénicos/química , Receptores Androgénicos/deficiencia , Dedos de Zinc/genética
5.
Cancer Res ; 65(15): 6640-50, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16061644

RESUMEN

Malignantly transformed stem cells represent a potential common nidus for the primary cancer and the recurrent cancer that arises after treatment failure. Putative prostate stem cells and prostate tumor stem cells in benign and malignant human prostate tissue, in primary human prostate xenografts, and in the transgenic adenocarcinoma of the mouse prostate (TRAMP) mouse model of prostate cancer, are defined by expression of breast cancer resistance protein (BCRP), a marker of pluripotent hematopoietic, muscle, and neural stem cells, and by an absence of androgen receptor (AR) protein. Inhibition of BCRP-mediated efflux of dihydrotestosterone by novobiocin or fumitremorgin C in a rat prostate progenitor cell line that expresses BCRP and AR mRNAs, but minimal AR protein, results in stabilization and nuclear translocation of AR protein, providing a mechanism for lack of AR protein in BCRP-expressing stem cells. In both benign and malignant human prostate tissue, the rare epithelial cells that express BCRP and lack AR protein are localized in the basal cell compartment, survive androgen deprivation, and maintain proliferative potential in the hypoxic, androgen-deprived prostate. Putative prostate tumor stem cells that express BCRP but not AR protein in TRAMP are the source of a BCRP-negative and AR-negative, Foxa2- and SV40Tag-expressing, transit amplifying compartment that progresses to the poorly differentiated carcinomas that arise rapidly after castration. Therefore, BCRP expression isolates prostate stem/tumor stem cells from the prostate tissue microenvironment through constitutive efflux of androgen, protecting the putative tumor stem cells from androgen deprivation, hypoxia, or adjuvant chemotherapy, and providing the nidus for recurrent prostate cancer.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/biosíntesis , Andrógenos/metabolismo , Proteínas de Neoplasias/biosíntesis , Células Madre Neoplásicas/metabolismo , Neoplasias de la Próstata/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/metabolismo , Andrógenos/deficiencia , Animales , Línea Celular , Núcleo Celular/metabolismo , Humanos , Indoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/patología , Novobiocina/farmacología , Próstata/metabolismo , Neoplasias de la Próstata/patología , Procesamiento Proteico-Postraduccional , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Trasplante Heterólogo
6.
Cancer Res ; 65(11): 4836-43, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15930304

RESUMEN

Prostate cancer is the second leading cause of death in men in western countries and is usually treated by surgery and/or radiotherapy. More recently, hyperthermia has been introduced into clinical trials investigating a possible effect in the first-line treatment of prostate cancer. However, the molecular mechanisms of hyperthermia are not completely understood. In this study, we investigated the effects of hyperthermia on proteasome function and its significance for signal transduction, cell death and androgen receptor (AR) expression in PC-3, LnCaP, and DU-145 human and TRAMP-C2 murine prostate cancer cells. Hyperthermia caused apoptosis and radiosensitization and decreased 26S proteasome activity in all three human cell lines to about 40% of untreated control cells. 20S proteasome activity was not affected by heat. Heat treatment inhibited constitutive and radiation-induced activation of nuclear factor kappaB caused by stabilization of IkappaB. Although stabilization of AR by proteasome inhibitors has been reported previously, AR protein levels in LnCaP cells decreased dramatically after heat. Our data suggest that inhibition of proteasome function and dependent signal transduction pathways might be a major molecular mechanisms of heat-induced apoptosis and radiosensitization. Hyperthermia abrogates AR expression in androgen-dependent cells and might thus promote malignant progression of prostate cancer.


Asunto(s)
Hipertermia Inducida , Neoplasias de la Próstata/terapia , Inhibidores de Proteasoma , Receptores Androgénicos/deficiencia , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Trastornos de Estrés por Calor/enzimología , Trastornos de Estrés por Calor/patología , Humanos , Masculino , Ratones , FN-kappa B/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Complejo de la Endopetidasa Proteasomal , Receptores Androgénicos/biosíntesis , Transducción de Señal/fisiología
7.
Wien Med Wochenschr Suppl ; 91: 1-15, 1985.
Artículo en Alemán | MEDLINE | ID: mdl-3895756

RESUMEN

One has to distinguish masculine sex behavior and estrogens alone or in combination with gestagens evoke feminine sex behavior. The central integrator for the induction of sex behavior is located in diencephalic nuclei. If sex hormones are lacking, the sex drive is fading off, except in women. Sex hormones are also responsible for the determination of those neutral centres controlling male or female sex behavior later in life in most species. Based on animal datas and on retrospective inquiries of homosexuals or mothers of homosexuals, a hypothesis for the etiology of homo-, bi- and hyposexuality has been developed by Dorner. Absence or deficiency of androgens in the critical phase of "brain differentiation" leads to male homo-, bi- or hyposexuality, respectively. If androgens become active in the critical phase of female differentiation, then the result will be female homo-, bi- or hyposexuality, respectively. This hypothesis will be critical evaluated.


Asunto(s)
Hormonas Esteroides Gonadales/fisiología , Conducta Sexual/fisiología , Adolescente , Adulto , Anciano , Síndrome de Resistencia Androgénica/fisiopatología , Andrógenos/deficiencia , Andrógenos/fisiología , Animales , Niño , Ciproterona/farmacología , Trastornos del Desarrollo Sexual , Femenino , Homosexualidad , Humanos , Hipotálamo/fisiología , Hormona Luteinizante/metabolismo , Masculino , Persona de Mediana Edad , Ovario/trasplante , Hipófisis/metabolismo , Receptores Androgénicos/deficiencia , Análisis para Determinación del Sexo , Testículo/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA