Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Endocrinology ; 163(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34902009

RESUMEN

Concordant transcriptional regulation can generate multiple gene products that collaborate to achieve a common goal. Here we report a case of concordant transcriptional regulation that instead drives a single protein to be produced in the same cell type from divergent promoters. This gene product-the RHOX5 homeobox transcription factor-is translated from 2 different mRNAs with different 5' untranslated regions (UTRs) transcribed from alternative promoters. Despite the fact that these 2 promoters-the proximal promoter (Pp) and the distal promoter (Pd)-exhibit different patterns of tissue-specific activity, share no obvious sequence identity, and depend on distinct transcription factors for expression, they exhibit a remarkably similar expression pattern in the testes. In particular, both depend on androgen signaling for expression in the testes, where they are specifically expressed in Sertoli cells and have a similar stage-specific expression pattern during the seminiferous epithelial cycle. We report evidence for 3 mechanisms that collaborate to drive concordant Pp/Pd expression. First, both promoters have an intrinsic ability to respond to androgen receptor and androgen. Second, the Pp acts as an enhancer to promote androgen-dependent transcription from the Pd. Third, Pd transcription is positively autoregulated by the RHOX5 protein, which is first produced developmentally from the Pp. Together, our data support a model in which the Rhox5 homeobox gene evolved multiple mechanisms to activate both of its promoters in Sertoli cells to produce Rhox5 in an androgen-dependent manner during different phases of spermatogenesis.


Asunto(s)
Andrógenos/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Regiones Promotoras Genéticas , Células de Sertoli/metabolismo , Factores de Transcripción/genética , Regiones no Traducidas 5' , Animales , Metilación de ADN , Genes Homeobox , Masculino , Ratones , Ratones Endogámicos C57BL , Plásmidos/metabolismo , Isoformas de Proteínas , Receptores Androgénicos/metabolismo , Túbulos Seminíferos/metabolismo , Espermatogénesis , Testículo/metabolismo , Factores de Transcripción/metabolismo
2.
Cell Metab ; 23(5): 837-51, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27133133

RESUMEN

Although men with testosterone deficiency are at increased risk for type 2 diabetes (T2D), previous studies have ignored the role of testosterone and the androgen receptor (AR) in pancreatic ß cells. We show that male mice lacking AR in ß cells (ßARKO) exhibit decreased glucose-stimulated insulin secretion (GSIS), leading to glucose intolerance. The AR agonist dihydrotestosterone (DHT) enhances GSIS in cultured male islets, an effect that is abolished in ßARKO(-/y) islets and human islets treated with an AR antagonist. In ß cells, DHT-activated AR is predominantly extranuclear and enhances GSIS by increasing islet cAMP and activating the protein kinase A. In mouse and human islets, the insulinotropic effect of DHT depends on activation of the glucagon-like peptide-1 (GLP-1) receptor, and accordingly, DHT amplifies the incretin effect of GLP-1. This study identifies AR as a novel receptor that enhances ß cell function, a finding with implications for the prevention of T2D in aging men.


Asunto(s)
Núcleo Celular/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Receptores Androgénicos/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Intolerancia a la Glucosa/patología , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Noqueados , Modelos Biológicos , Receptores Androgénicos/deficiencia , Transducción de Señal/efectos de los fármacos , Testosterona/farmacología
3.
FASEB J ; 29(4): 1540-50, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550469

RESUMEN

Androgens have important cardiometabolic actions in males, but their metabolic role in females is unclear. To determine the physiologic androgen receptor (AR)-dependent actions of androgens on atherogenesis in female mice, we generated female AR-knockout (ARKO) mice on an atherosclerosis-prone apolipoprotein E (apoE)-deficient background. After 8 weeks on a high-fat diet, but not on a normal chow diet, atherosclerosis in aorta was increased in ARKO females (+59% vs. control apoE-deficient mice with intact AR gene). They also displayed increased body weight (+18%), body fat percentage (+62%), and hepatic triglyceride levels, reduced insulin sensitivity, and a marked atherogenic dyslipidemia (serum cholesterol, +52%). Differences in atherosclerosis, body weight, and lipid levels between ARKO and control mice were abolished in mice that were ovariectomized before puberty, consistent with a protective action of ovarian androgens mediated via the AR. Furthermore, the AR agonist dihydrotestosterone reduced atherosclerosis (-41%; thoracic aorta), subcutaneous fat mass (-44%), and cholesterol levels (-35%) in ovariectomized mice, reduced hepatocyte lipid accumulation in hepatoma cells in vitro, and regulated mRNA expression of hepatic genes pivotal for lipid homeostasis. In conclusion, we demonstrate that the AR protects against diet-induced atherosclerosis in female mice and propose that this is mediated by modulation of body composition and lipid metabolism.


Asunto(s)
Aterosclerosis/prevención & control , Dislipidemias/prevención & control , Obesidad/prevención & control , Receptores Androgénicos/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Colesterol/metabolismo , Dieta/efectos adversos , Dihidrotestosterona/farmacología , Dislipidemias/etiología , Dislipidemias/metabolismo , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/prevención & control , Femenino , Resistencia a la Insulina , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Orquiectomía , Ovariectomía , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética
4.
Proc Natl Acad Sci U S A ; 111(18): E1924-32, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24753613

RESUMEN

Fetal growth plays a role in programming of adult cardiometabolic disorders, which in men, are associated with lowered testosterone levels. Fetal growth and fetal androgen exposure can also predetermine testosterone levels in men, although how is unknown, because the adult Leydig cells (ALCs) that produce testosterone do not differentiate until puberty. To explain this conundrum, we hypothesized that stem cells for ALCs must be present in the fetal testis and might be susceptible to programming by fetal androgen exposure during masculinization. To address this hypothesis, we used ALC ablation/regeneration to identify that, in rats, ALCs derive from stem/progenitor cells that express chicken ovalbumin upstream promoter transcription factor II. These stem cells are abundant in the fetal testis of humans and rodents, and lineage tracing in mice shows that they develop into ALCs. The stem cells also express androgen receptors (ARs). Reduction in fetal androgen action through AR KO in mice or dibutyl phthalate (DBP) -induced reduction in intratesticular testosterone in rats reduced ALC stem cell number by ∼40% at birth to adulthood and induced compensated ALC failure (low/normal testosterone and elevated luteinizing hormone). In DBP-exposed males, this failure was probably explained by reduced testicular steroidogenic acute regulatory protein expression, which is associated with increased histone methylation (H3K27me3) in the proximal promoter. Accordingly, ALCs and ALC stem cells immunoexpressed increased H3K27me3, a change that was also evident in ALC stem cells in fetal testes. These studies highlight how a key component of male reproductive development can fundamentally reprogram adult hormone production (through an epigenetic change), which might affect lifetime disease risk.


Asunto(s)
Células Madre Adultas/fisiología , Andrógenos/fisiología , Desarrollo Fetal/fisiología , Células Intersticiales del Testículo/fisiología , Células Madre Adultas/efectos de los fármacos , Animales , Callithrix , Linaje de la Célula/fisiología , Dibutil Ftalato/toxicidad , Femenino , Desarrollo Fetal/efectos de los fármacos , Células Madre Fetales/efectos de los fármacos , Células Madre Fetales/fisiología , Humanos , Técnicas In Vitro , Células Intersticiales del Testículo/efectos de los fármacos , Hormona Luteinizante/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Animales , Embarazo , Ratas , Ratas Transgénicas , Ratas Wistar , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Receptores Androgénicos/fisiología , Regeneración , Testículo/embriología , Testículo/fisiología , Testosterona/deficiencia , Testosterona/fisiología
5.
Mol Endocrinol ; 28(4): 575-91, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24606126

RESUMEN

An understanding of the molecular mechanisms by which androgens drive spermatogenesis has been thwarted by the fact that few consistent androgen receptor (AR) target genes have been identified. Here, we addressed this issue using next-generation sequencing coupled with the RiboTag approach, which purifies translated mRNAs expressed in cells that express cyclic recombinase (CRE). Using RiboTag mice expressing CRE in Sertoli cells (SCs), we identified genes expressed specifically in SCs in both prepubertal and adult mice. Unexpectedly, this analysis revealed that the SC-specific gene program is already largely defined at the initiation of spermatogenesis despite the subsequent dramatic maturational changes known to occur in SCs. To identify AR-regulated genes, we generated triple-mutant mice in which the SCs express the RiboTag but lack ARs. RNA sequencing analysis revealed hundreds of SC-expressed AR-regulated genes that had previously gone unnoticed, including suppressed genes involved in ovarian development. Comparison of the SC-enriched dataset with that from the whole testes allowed us to classify genes in terms of their degree of expression in SCs. This revealed that a greater fraction of AR-up-regulated genes than AR-down-regulated genes were expressed predominantly in SCs. Our results also revealed that AR signaling in SCs causes a large number of genes not detectably expressed in SCs to undergo altered expression, thereby providing genome-wide evidence for wide-scale communication between SCs and other cells. Taken together, our results identified novel classes of genes expressed in a hormone-dependent manner in different testicular cell subsets and highlight a new approach to analyze cell type-specific gene regulation.


Asunto(s)
Regulación de la Expresión Génica , Técnicas Genéticas , Genoma/genética , Biosíntesis de Proteínas/genética , Receptores Androgénicos/metabolismo , Células de Sertoli/metabolismo , Andrógenos/farmacología , Animales , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Células de Sertoli/efectos de los fármacos , Maduración Sexual/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
7.
FASEB J ; 26(10): 4360-72, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22798427

RESUMEN

The androgen receptor (AR) recognizes two types of DNA elements that are dimers of 5'-AGAACA-3'-like hexamers, either organized as inverted or direct repeats. We developed a mouse model [(specificity affecting AR knock-in (SPARKI)] in which the AR DNA-binding domain was mutated such that it lost binding to direct repeats but not to inverted elements. The impaired fertility of the male SPARKI mice correlates with the reduced motility of the spermatozoa, a characteristic that is developed during transit through the epididymis. Comparative transcriptome analyses revealed that the expression of 39 genes is changed in SPARKI epididymis. Remarkably, the expression of the steroid 5α-reductase type II (Srd5α2) gene, which metabolizes testosterone into the more potent dihydrotestosterone, is reduced 4-fold in SPARKI vs. wild type. The comparison of the SPARKI phenotype with that of Srd5α2-knockout mice shows, however, that the reduced Srd5α2 expression cannot explain all defects of the SPARKI epididymis. Moreover, we describe three new selective androgen response elements (AREs), which control the androgen responsiveness of the Srd5α2 gene. We conclude that the SPARKI model can be considered a knockout model for AR functioning via selective AREs and that this has a dramatic effect on sperm maturation in the epididymis.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Andrógenos/farmacología , Epidídimo/metabolismo , Receptores Androgénicos/metabolismo , Elementos de Respuesta/fisiología , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , Animales , Ensayo de Cambio de Movilidad Electroforética , Células HeLa , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Orquiectomía , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Androgénicos/genética , Elementos de Respuesta/genética
8.
Mol Endocrinol ; 26(4): 598-607, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22322597

RESUMEN

Androgens play a critical role in the development of the male reproductive system, including the positioning of the gonads. It is not clear, however, which developmental processes are influenced by androgens and what are the target tissues and cells mediating androgen signaling during testicular descent. Using a Cre-loxP approach, we have produced male mice (GU-ARKO) with conditional inactivation of the androgen receptor (Ar) gene in the gubernacular ligament connecting the epididymis to the caudal abdominal wall. The GU-ARKO males had normal testosterone levels but developed cryptorchidism with the testes located in a suprascrotal position. Although initially subfertile, the GU-ARKO males became sterile with age. We have shown that during development, the mutant gubernaculum failed to undergo eversion, a process giving rise to the processus vaginalis, a peritoneal outpouching inside the scrotum. As a result, the cremasteric sac did not form properly, and the testes remained in the low abdominal position. Abnormal development of the cremaster muscles in the GU-ARKO males suggested the participation of androgens in myogenic differentiation; however, males with conditional AR inactivation in the striated or smooth muscle cells had a normal testicular descent. Gene expression analysis showed that AR deficiency in GU-ARKO males led to the misexpression of genes involved in muscle differentiation, cell signaling, and extracellular space remodeling. We therefore conclude that AR signaling in gubernacular cells is required for gubernaculum eversion and outgrowth. The GU-ARKO mice provide a valuable model of isolated cryptorchidism, one of the most common birth defects in newborn boys.


Asunto(s)
Criptorquidismo/metabolismo , Receptores Androgénicos/genética , Testículo/metabolismo , Actinas/metabolismo , Animales , Criptorquidismo/genética , Criptorquidismo/patología , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Liso/metabolismo , Músculo Liso/patología , Tamaño de los Órganos , Receptores Androgénicos/metabolismo , Transducción de Señal , Recuento de Espermatozoides , Testículo/crecimiento & desarrollo , Testículo/patología , Testosterona/genética , Testosterona/metabolismo
9.
Mol Endocrinol ; 26(4): 538-49, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22322598

RESUMEN

Mammalian male fertility depends on the epididymis, a highly segmented organ that promotes sperm maturation and protects sperm from oxidative damage. Remarkably little is known about how gene expression is controlled in the epididymis. A candidate to regulate genes crucial for epididymal function is reproductive homeobox gene on X chromosome (RHOX)5, a homeobox transcription factor essential for optimal sperm motility that is expressed in the caput region of the epididymis. Here, we report the identification of factors that control Rhox5 gene expression in epididymal cells in a developmentally regulated and region-specific fashion. First, we identify GATA transcription factor-binding sites in the Rhox5 proximal promoter (Pp) necessary for Rhox5 expression in epididymal cells in vitro and in vivo. Adjacent to the GATA sites are androgen-response elements, which bind to the nuclear hormone receptor androgen receptor (AR), and are responsible for the AR-dependent expression of Rhox5 in epididymal cells. We provide evidence that AR is recruited to the Pp in a region-specific and developmentally regulated manner in the epididymis that is dictated not only by differential AR availability but differential methylation of the Pp. Site-specific methylation of the Pp cytosine and guanine separated by one phosphate, most of which overlap with androgen-response elements, inhibited both AR occupancy at the Pp and Pp-dependent transcription in caput epididymal cells. Together, our data support a model in which DNA methylation, AR, and GATA factors collaborate to dictate the unique developmental and region-specific expression pattern of the RHOX5 homeobox transcription factor in the caput epididymis, which in turn controls the expression of genes critical for promoting sperm motility and function.


Asunto(s)
Metilación de ADN , Epidídimo/metabolismo , Factores de Transcripción GATA/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Receptores Androgénicos/metabolismo , Factores de Transcripción/genética , Andrógenos/fisiología , Animales , Línea Celular , Genes Reporteros , Proteínas de Homeodominio/metabolismo , Luciferasas/biosíntesis , Luciferasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Receptores Androgénicos/genética , Elementos de Respuesta , Factores de Transcripción/metabolismo , Transcripción Genética
10.
Mol Cell Endocrinol ; 352(1-2): 13-25, 2012 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-21871526

RESUMEN

This review aims to evaluate the contribution of individual cell-selective knockout models to our current understanding of androgen action. Cre/loxP technology has allowed the generation of cell-selective knockout models targeting the androgen receptor (AR) in distinct putative target cells in a wide variety of organs and tissues including: testis, ovary, accessory sex tissues, muscle, bone, fat, liver, skin and myeloid tissue. In some androgen-regulated processes such as spermatogenesis and folliculogenesis this approach has lead to the identification of a key cellular mediator of androgen action (Sertoli and granulosa cells, respectively). In many target tissues, however, the final response to androgens appears to be more complex. Here, cell-selective knockout technology offers a platform upon which we can begin to unravel the more complex interplay and signaling pathways of androgens. A prototypic example is the analysis of mesenchymal-epithelial interactions in many accessory sex glands. Furthermore, for some actions of testosterone, in which part of the effect is mediated by the active metabolite 17ß-estradiol, conditional knockout technology offers a novel strategy to study the relative contribution of AR and estrogen receptor-mediated signaling. The latter approach has already resulted in a better understanding of androgen action in brain and bone. Finally, cell-selective knockout technology has generated valuable models to search for AR-controlled molecular mediators of androgen action, a strategy that has successfully been applied to the study of androgen action in the testis and in the epididymis. Although some conditional knockout models have provided clear answers to physiologic questions, it should be noted that others have pointed to unexpected complexities or technical limitations confounding interpretation of the results.


Asunto(s)
Especificidad de Órganos , Receptores Androgénicos/metabolismo , Animales , Ratones , Ratones Noqueados , Receptores Androgénicos/fisiología , Transducción de Señal
11.
Biol Reprod ; 85(5): 934-45, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21734264

RESUMEN

Our previous analysis of Sertoli cell androgen receptor (AR) knockout (SCARKO) mice revealed that several cytoskeletal components are a potential target of androgen action. Here, we found that one of these components, the beta-tubulin isotype Tubb3, is differentially regulated in testes from SCARKO mice (relative to littermate controls) from Postnatal Day 10 to adulthood. The Tubb3 gene is unique in this respect, as at Day 10, no other beta-tubulin genes are significantly regulated by AR. We further characterized androgen regulation of Tubb3 in vivo and in vitro and demonstrated that it is a conserved feature in both mice and rats. To investigate whether androgens directly regulate Tubb3 expression, we screened for androgen response elements (AREs) in the Tubb3 gene. In silico analysis revealed the presence of four ARE motifs in Tubb3 intron 1, two of which bind to AR in vitro. Mutation of one of these (ARE1) strongly reduced androgen-dependent reporter gene expression. These results, coupled with the finding that the AR binds to the Tubb3 ARE region in vivo, suggest that Tubb3 is a direct target of AR. Our data strengthen the contention that androgens exert their effects on spermatogenesis, in part, through modulation of the Sertoli cell cytoskeleton. Androgen regulation of beta-tubulin has also been described in neurons, fortifying the already known similarity in microtubule organization in Sertoli cell processes and neurons, the only other cell type in which Tubb3 is known to be expressed.


Asunto(s)
Andrógenos/metabolismo , Células de Sertoli/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Isoformas de Proteínas/metabolismo , Ratas , Ratas Wistar , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Células de Sertoli/citología , Espermatogénesis/fisiología , Tubulina (Proteína)/genética
12.
Spermatogenesis ; 1(4): 341-353, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22332118

RESUMEN

A mouse model with a Sertoli cell (SC)-selective ablation of the androgen receptor (AR)-the SCARKO mouse-demonstrated that the effects of androgens on spermatogenesis depend on the presence of an active AR in SC. This model has been extremely valuable in the study of the effects of androgens on the initiation of spermatogenesis. However, due to the early (prenatal) inactivation of the AR SCARKO mice develop a complete block in meiosis, making it impossible to study the effects of androgens on postmeiotic steps of germ cell development. It would therefore be of interest to develop a model in which the AR can be ablated at any chosen time point. Here we used a mouse line ubiquitously expressing a tamoxifen (TM)-inducible Cre recombinase to develop an inducible AR knockout model (iARKO). It is shown that treatment with TM (3 mg/day) for five consecutive days efficiently inactivates the AR in testicular tissue and decreases the expression of known AR-target genes in SC (Rhox5, Spinlw1) without markedly affecting testicular cell composition one day after the final injection. TM treatment did, however, decrease serum gonadotropin levels and the expression of several Leydig cell genes (StAR, Cyp17a1, Insl3), resulting in decreased testosterone levels. Nevertheless, the intratesticular testosterone concentration still far exceeds the estimated concentrations required to saturate the AR. It may be concluded that the study of androgen-responsive postmeiotic genes in SC may be feasible using a TM-inducible AR knockout model provided that appropriate controls are included correcting for off-target effects of TM.

13.
Endocrinology ; 152(2): 689-96, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21084446

RESUMEN

The epithelial lining of the epididymal duct expresses the androgen receptor (Ar) along its entire length and undergoes rapid and profound degeneration when androgenic support is withdrawn. However, experiments involving orchidectomy with systemic testosterone replacement, and testicular efferent duct ligation, have indicated that structural and functional integrity of the initial segment cannot be maintained by circulating androgen alone, leaving the role of androgen in this epididymal zone unclear. We addressed this question in a mouse model with intact testicular output and selective Ar inactivation in the proximal epididymis by creating double-transgenic males carrying a conditional Ar(loxP) allele and expressing Cre recombinase under the promoter of Rnase10, a gene specifically expressed in proximal epididymis. At 20-25 d of life, on the onset of Rnase10 expression, Ar became selectively inactivated in the principal cells of proximal epididymis, resulting in epithelial hypoplasia and hypotrophy. Upon the subsequent onset of spermiation, epididymal obstruction ensued, with the consequent development of spermatic granulomata, back pressure-induced atrophy of the seminiferous epithelium, orchitis, and fibrosis of the testicular parenchyma. Consistent with these findings, the mice were infertile. When the effect of Ar knockout on gene expression in the proximal epididymis was compared with that of efferent duct ligation and orchidectomy, we identified genes specifically regulated by androgen, testicular efferent fluid, and both. Our findings demonstrate that the development and function of the epididymal initial segment is critically dependent on direct androgen regulation. The phenotype of the produced knockout mouse provides a novel model for obstructive azoospermia.


Asunto(s)
Azoospermia/etiología , Epidídimo/metabolismo , Receptores Androgénicos/metabolismo , Animales , Azoospermia/genética , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Receptores Androgénicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
PLoS One ; 5(11): e14168, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21152390

RESUMEN

The observation that mice with a selective ablation of the androgen receptor (AR) in Sertoli cells (SC) (SCARKO mice) display a complete block in meiosis supports the contention that SC play a pivotal role in the control of germ cell development by androgens. To delineate the physiological and molecular mechanism responsible for this control, we compared tubular development in pubertal SCARKO mice and littermate controls. Particular attention was paid to differences in SC maturation, SC barrier formation and cytoskeletal organization and to the molecular mediators potentially involved. Functional analysis of SC barrier development by hypertonic perfusion and lanthanum permeation techniques and immunohistochemical analysis of junction formation showed that SCARKO mice still attempt to produce a barrier separating basal and adluminal compartment but that barrier formation is delayed and defective. Defective barrier formation was accompanied by disturbances in SC nuclear maturation (immature shape, absence of prominent, tripartite nucleoli) and SC polarization (aberrant positioning of SC nuclei and cytoskeletal elements such as vimentin). Quantitative RT-PCR was used to study the transcript levels of genes potentially related to the described phenomena between day 8 and 35. Differences in the expression of SC genes known to play a role in junction formation could be shown from day 8 for Cldn11, from day 15 for Cldn3 and Espn, from day 20 for Cdh2 and Jam3 and from day 35 for ZO-1. Marked differences were also noted in the transcript levels of several genes that are also related to cell adhesion and cytoskeletal dynamics but that have not yet been studied in SC (Actn3, Ank3, Anxa9, Scin, Emb, Mpzl2). It is concluded that absence of a functional AR in SC impedes the remodeling of testicular tubules expected at the onset of spermatogenesis and interferes with the creation of the specific environment needed for germ cell development.


Asunto(s)
Diferenciación Celular/fisiología , Citoesqueleto/metabolismo , Receptores Androgénicos/fisiología , Células de Sertoli/metabolismo , Animales , Cadherinas/genética , Diferenciación Celular/genética , Claudina-3 , Claudinas , Femenino , Expresión Génica , Inmunohistoquímica , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Proteínas del Tejido Nervioso/genética , Fosfoproteínas/genética , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/citología , Células de Sertoli/ultraestructura , Espermatogénesis/genética , Espermatogénesis/fisiología , Testículo/metabolismo , Testículo/ultraestructura , Factores de Tiempo , Vimentina/metabolismo , Proteína de la Zonula Occludens-1
15.
Endocrinology ; 151(11): 5428-37, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20861231

RESUMEN

The atheroprotective effect of testosterone is thought to require aromatization of testosterone to estradiol, but no study has adequately addressed the role of the androgen receptor (AR), the major pathway for the physiological effects of testosterone. We used AR knockout (ARKO) mice on apolipoprotein E-deficient background to study the role of the AR in testosterone atheroprotection in male mice. Because ARKO mice are testosterone deficient, we sham operated or orchiectomized (Orx) the mice before puberty, and Orx mice were supplemented with placebo or a physiological testosterone dose. From 8 to 16 wk of age, the mice consumed a high-fat diet. In the aortic root, ARKO mice showed increased atherosclerotic lesion area (+80%, P < 0.05). Compared with placebo, testosterone reduced lesion area both in Orx wild-type (WT) mice (by 50%, P < 0.001) and ARKO mice (by 24%, P < 0.05). However, lesion area was larger in testosterone-supplemented ARKO compared with testosterone-supplemented WT mice (+57%, P < 0.05). In WT mice, testosterone reduced the presence of a necrotic core in the plaque (80% among placebo-treated vs. 12% among testosterone-treated mice; P < 0.05), whereas there was no significant effect in ARKO mice (P = 0.20). In conclusion, ARKO mice on apolipoprotein E-deficient background display accelerated atherosclerosis. Testosterone treatment reduced atherosclerosis in both WT and ARKO mice. However, the effect on lesion area and complexity was more pronounced in WT than in ARKO mice, and lesion area was larger in ARKO mice even after testosterone supplementation. These results are consistent with an AR-dependent as well as an AR-independent component of testosterone atheroprotection in male mice.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Receptores Androgénicos/metabolismo , Testosterona/metabolismo , Animales , Aorta/efectos de los fármacos , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/genética , Aterosclerosis/prevención & control , Presión Sanguínea/fisiología , Citocinas/sangre , Lípidos/sangre , Masculino , Ratones , Ratones Noqueados , Orquiectomía , Receptores Androgénicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas , Testosterona/farmacología
16.
Philos Trans R Soc Lond B Biol Sci ; 365(1546): 1537-56, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20403868

RESUMEN

Transgenic mouse models have contributed considerably to our understanding of the cellular and molecular mechanisms by which androgens control spermatogenesis. Cell-selective ablation of the androgen receptor (AR) in Sertoli cells (SC) results in a complete block in meiosis and unambiguously identifies the SC as the main cellular mediator of the effects of androgens on spermatogenesis. This conclusion is corroborated by similar knockouts in other potential testicular target cells. Mutations resulting in diminished expression of the AR or in alleles with increased length of the CAG repeat mimick specific human forms of disturbed fertility that are not accompanied by defects in male sexual development. Transcriptional profiling studies in mice with cell-selective and general knockouts of the AR, searching for androgen-regulated genes relevant to the control of spermatogenesis, have identified many candidate target genes. However, with the exception of Rhox5, the identified subsets of genes show little overlap. Genes related to tubular restructuring, cell junction dynamics, the cytoskeleton, solute transportation and vitamin A metabolism are prominently present. Further research will be needed to decide which of these genes are physiologically relevant and to identify genes that can be used as diagnostic tools or targets to modulate the effects of androgens in spermatogenesis.


Asunto(s)
Andrógenos/fisiología , Espermatogénesis/fisiología , Testículo/fisiología , Andrógenos/genética , Animales , Masculino , Ratones , Ratones Transgénicos , Receptores Androgénicos/fisiología , Células de Sertoli/citología , Células de Sertoli/fisiología , Espermatogénesis/genética , Testículo/citología
17.
Mol Endocrinol ; 24(1): 60-75, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19901196

RESUMEN

Rhox5, the founding member of the reproductive homeobox on the X chromosome (Rhox) gene cluster, encodes a homeodomain-containing transcription factor that is selectively expressed in Sertoli cells, where it promotes the survival of male germ cells. To identify Rhox5-regulated genes, we generated 15P-1 Sertoli cell clones expressing physiological levels of Rhox5 from a stably transfected expression vector. Microarray analysis identified many genes altered in expression in response to Rhox5, including those encoding proteins controlling cell cycle regulation, apoptosis, metabolism, and cell-cell interactions. Fifteen of these Rhox5-regulated genes were chosen for further analysis. Analysis of Rhox5-null male mice indicated that at least nine of these are Rhox5-regulated in the testes in vivo. Many of them have distinct postnatal expression patterns and are regulated by Rhox5 at different postnatal time points. Most of them are expressed in Sertoli cells, indicating that they are candidates to be directly regulated by Rhox5. Transfection analysis with expression vectors encoding different mouse and human Rhox family members revealed that the regulatory response of a subset of these Rhox5-regulated genes is both conserved and redundant. Given that Rhox5 depends on androgen receptor (AR) for expression in Sertoli cells, we examined whether some Rhox5-regulated genes are also regulated by AR. We provide several lines of evidence that this is the case, leading us to propose that RHOX5 serves as a key intermediate transcription factor that directs some of the actions of AR in the testes.


Asunto(s)
Andrógenos/farmacología , Regulación de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Receptores Androgénicos/fisiología , Células de Sertoli/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Envejecimiento , Animales , Línea Celular , Células Clonales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Elementos de Respuesta/genética , Espermatogénesis/genética , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo , Transfección
18.
Biol Reprod ; 81(6): 1083-92, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19587329

RESUMEN

Cluster analysis at Postnatal Day 8-20 of putative androgen-regulated genes in mice with Sertoli cell-selective knockout of the androgen receptor (SCARKO) has pinpointed three genes (Spinlw1, Gpd1, Drd4) with an expression pattern strongly resembling that of Rhox5, the definitive Sertoli cell (SC) androgen-regulated gene. We used organotypic testis cultures from Day 8 mice to study control of these genes by (anti)androgens and follicle-stimulating hormone (FSH). Testis morphology and androgen induction of the studied genes were preserved for 48 h. Preincubation with ketoconazole for 24 h to block endogenous androgen production, followed by 24-h incubation with the synthetic androgen R1881, resulted in 45-, 5-, 19-, and 6-fold induction of mRNA levels of Rhox5, Spinlw1, Gpd1, and Drd4, respectively. However, noticeable differences in control of the studied genes were observed. Rhox5 and Spinlw1 were fully induced by R1881 in the continuous (48 h) presence of ketoconazole, whereas only marginal effects were observed on expression of Gpd1 and Drd4. Similarly, FSH only marginally affected expression of Rhox5 and Spinlw1, whereas it markedly increased Gpd1 and Drd4 expression. Explant cultures of SCARKO testes confirmed the differential effects of FSH on the studied genes and, for Gpd1, showed that the effect did not depend on a functional androgen receptor in SC, whereas this was essential for the effects of FSH on Drd4. In conclusion, organotypic cultures represent the first in vitro approach to preserving androgen responsiveness of putative SC-expressed genes. This approach facilitates detailed analysis of their regulation in ways not possible in vivo.


Asunto(s)
Andrógenos/metabolismo , Técnicas de Cultivo de Órganos/métodos , Células de Sertoli/metabolismo , Testículo/metabolismo , Análisis de Varianza , Andrógenos/farmacología , Animales , Apoptosis/fisiología , Medios de Cultivo Condicionados , Perfilación de la Expresión Génica , Glicerolfosfato Deshidrogenasa/genética , Glicerolfosfato Deshidrogenasa/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Masculino , Ratones , Tamaño de los Órganos/efectos de los fármacos , Proteínas Inhibidoras de Proteinasas Secretoras , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Dopamina D4/genética , Receptores de Dopamina D4/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/efectos de los fármacos , Testículo/citología , Testículo/efectos de los fármacos , Testosterona/metabolismo , Testosterona/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
J Neurosci ; 29(14): 4461-70, 2009 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-19357272

RESUMEN

Testosterone (T) profoundly influences central sexual differentiation and functions. In the brain, T signals either directly through androgen receptor (AR) or indirectly through estrogen receptor (ER) following aromatization into E2 (17-beta-estradiol). As T, through AR, also controls peripheral male sexual differentiation, the relative contribution of central AR in T-mediated regulation of behavioral and neuroendocrine responses still remains unclear. To address this question, we generated, by using Cre-loxP technology, mice selectively lacking AR expression in the nervous system. The mutant male urogenital tract was normally developed, and mice were able to produce offspring. Nonetheless, sexual motivation and performance as well as aggressive behaviors were affected. Only a low percentage of males displayed a complete sexual behavior and offensive attacks. The latency to show masculine behaviors was increased and copulation length prolonged. Erectile activity during mating was also altered. These alterations occurred despite increased levels of T and its metabolites, and an unaffected number of ERalpha-immunoreactive cells. Olfactory preference and neuronal activation, mapped by Fos immunoreactivity, following exposure to estrus female-soiled bedding were also normal. At comparable T levels, greater differences in masculine behaviors were observed between gonadectomized control and mutant males. AR invalidation in the nervous system also disrupted the somatotropic axis since mutant males exhibited growth retardation and decreased serum levels of insulin-like growth factor I. Our findings show that central AR is required in T-induced regulation of male-typical behaviors and gonadotrope and somatotropic axes. This genetic model offers a unique opportunity in the understanding of AR's role in cerebral functions of T.


Asunto(s)
Sistema Nervioso/metabolismo , Sistemas Neurosecretores/fisiología , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Conducta Sexual Animal/fisiología , Animales , Cerebro/metabolismo , Cerebro/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Sistemas Neurosecretores/metabolismo , Embarazo , Receptores Androgénicos/metabolismo
20.
Endocrinology ; 150(8): 3558-66, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19264874

RESUMEN

Muscle frailty is considered a major cause of disability in the elderly and chronically ill. However, the exact role of androgen receptor (AR) signaling in muscle remains unclear. Therefore, a postmitotic myocyte-specific AR knockout (mARKO) mouse model was created and investigated together with a mouse model with ubiquitous AR deletion. Muscles from mARKO mice displayed a marked reduction in AR protein (60-88%). Interestingly, body weights and lean body mass were lower in mARKO vs. control mice (-8%). The weight of the highly androgen-sensitive musculus levator ani was significantly reduced (-46%), whereas the weights of other peripheral skeletal muscles were not or only slightly reduced. mARKO mice had lower intra-abdominal fat but did not demonstrate a cortical or trabecular bone phenotype, indicating that selective ablation of the AR in myocytes affected male body composition but not skeletal homeostasis. Furthermore, muscle contractile performance in mARKO mice did not differ from their controls. Myocyte-specific AR ablation resulted in a conversion of fast toward slow fibers, without affecting muscle strength or fatigue. Similar results were obtained in ubiquitous AR deletion, showing lower body weight, whereas some but not all muscle weights were reduced. The percent slow fibers was increased, but no changes in muscle strength or fatigue could be detected. Together, our findings show that myocyte AR signaling contributes to the maintenance of muscle mass and fiber type regulation but not to muscle strength or fatigue. The levator ani weight remains the most sensitive and specific marker of AR-mediated anabolic action on muscle.


Asunto(s)
Células Musculares/metabolismo , Fatiga Muscular/fisiología , Fuerza Muscular/fisiología , Músculos/metabolismo , Músculos/fisiología , Receptores Androgénicos/fisiología , Absorciometría de Fotón , Animales , Western Blotting , Forma MM de la Creatina-Quinasa/genética , Femenino , Glucógeno/metabolismo , Inmunohistoquímica , Grasa Intraabdominal/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Microscopía Fluorescente , Fatiga Muscular/genética , Fibras Musculares Esqueléticas/metabolismo , Fuerza Muscular/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Succinato Deshidrogenasa/metabolismo , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...