Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Proc Natl Acad Sci U S A ; 111(18): E1924-32, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24753613

ABSTRACT

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.


Subject(s)
Adult Stem Cells/physiology , Androgens/physiology , Fetal Development/physiology , Leydig Cells/physiology , Adult Stem Cells/drug effects , Animals , Callithrix , Cell Lineage/physiology , Dibutyl Phthalate/toxicity , Female , Fetal Development/drug effects , Fetal Stem Cells/drug effects , Fetal Stem Cells/physiology , Humans , In Vitro Techniques , Leydig Cells/drug effects , Luteinizing Hormone/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Animal , Pregnancy , Rats , Rats, Transgenic , Rats, Wistar , Receptors, Androgen/deficiency , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Regeneration , Testis/embryology , Testis/physiology , Testosterone/deficiency , Testosterone/physiology
2.
FASEB J ; 29(4): 1540-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25550469

ABSTRACT

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.


Subject(s)
Atherosclerosis/prevention & control , Dyslipidemias/prevention & control , Obesity/prevention & control , Receptors, Androgen/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cholesterol/metabolism , Diet/adverse effects , Dihydrotestosterone/pharmacology , Dyslipidemias/etiology , Dyslipidemias/metabolism , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Female , Insulin Resistance , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Orchiectomy , Ovariectomy , Receptors, Androgen/deficiency , Receptors, Androgen/genetics
3.
FASEB J ; 26(10): 4360-72, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22798427

ABSTRACT

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.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Androgens/pharmacology , Epididymis/metabolism , Receptors, Androgen/metabolism , Response Elements/physiology , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Animals , Electrophoretic Mobility Shift Assay , HeLa Cells , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Orchiectomy , Real-Time Polymerase Chain Reaction , Receptors, Androgen/genetics , Response Elements/genetics
4.
Endocrinology ; 163(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34902009

ABSTRACT

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.


Subject(s)
Androgens/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Promoter Regions, Genetic , Sertoli Cells/metabolism , Transcription Factors/genetics , 5' Untranslated Regions , Animals , DNA Methylation , Genes, Homeobox , Male , Mice , Mice, Inbred C57BL , Plasmids/metabolism , Protein Isoforms , Receptors, Androgen/metabolism , Seminiferous Tubules/metabolism , Spermatogenesis , Testis/metabolism , Transcription Factors/metabolism
5.
Biol Reprod ; 85(5): 934-45, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21734264

ABSTRACT

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.


Subject(s)
Androgens/metabolism , Sertoli Cells/metabolism , Tubulin/metabolism , Animals , Base Sequence , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Protein Isoforms/metabolism , Rats , Rats, Wistar , Receptors, Androgen/deficiency , Receptors, Androgen/genetics , Sertoli Cells/cytology , Spermatogenesis/physiology , Tubulin/genetics
6.
J Neurosci ; 29(14): 4461-70, 2009 Apr 08.
Article in English | MEDLINE | ID: mdl-19357272

ABSTRACT

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.


Subject(s)
Nervous System/metabolism , Neurosecretory Systems/physiology , Receptors, Androgen/deficiency , Receptors, Androgen/genetics , Sexual Behavior, Animal/physiology , Animals , Cerebrum/metabolism , Cerebrum/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Genetic , Neurosecretory Systems/metabolism , Pregnancy , Receptors, Androgen/metabolism
7.
FASEB J ; 23(1): 232-40, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18809737

ABSTRACT

Osteoporosis and muscle frailty are important health problems in elderly men and may be partly related to biological androgen activity. This androgen action can be mediated directly through stimulation of the androgen receptor (AR) or indirectly through stimulation of estrogen receptor-alpha (ERalpha) following aromatization of androgens into estrogens. To assess the differential action of AR and ERalpha pathways on bone and body composition, AR-ERalpha double-knockout mice were generated and characterized. AR disruption decreased trabecular bone mass, whereas ERalpha disruption had no additional effect on the AR-dependent trabecular bone loss. In contrast, combined AR and ERalpha inactivation additionally reduced cortical bone and muscle mass compared with either AR or ERalpha disruption alone. ERalpha inactivation--in the presence or absence of AR--increased fat mass. We demonstrate that AR activation is solely responsible for the development and maintenance of male trabecular bone mass. Both AR and ERalpha activation, however, are needed to optimize the acquisition of cortical bone and muscle mass. ERalpha activation alone is sufficient for the regulation of fat mass. Our findings clearly define the relative importance of AR and ERalpha signaling on trabecular and cortical bone mass as well as body composition in male mice.


Subject(s)
Body Composition/genetics , Bone Density/genetics , Bone and Bones/metabolism , Estrogen Receptor alpha/metabolism , Receptors, Androgen/metabolism , Aging , Animals , Body Composition/physiology , Bone Density/physiology , Estrogen Receptor alpha/genetics , Gene Expression Regulation/physiology , Male , Mice , Mice, Knockout , Osteoporosis/metabolism , Receptors, Androgen/genetics
8.
Biochem Biophys Res Commun ; 378(1): 139-44, 2009 Jan 02.
Article in English | MEDLINE | ID: mdl-19013130

ABSTRACT

Disruption of the androgen receptor (AR) in male mice reduces cortical bone expansion and muscle mass during puberty and results in high bone turnover-related cancellous osteopenia. We hypothesized that voluntary wheel running during growth is able to rescue the effects of AR disruption on bone. To this end, 5-week-old AR knockout (ARKO) mice were randomized to a running group (cage with running wheel) and a sedentary group (cage without wheel) and followed-up until 16 weeks of age. Voluntary wheel running in ARKO mice did not influence body weight, muscle mass or periosteal bone expansion. Interestingly, voluntary running significantly reduced bone turnover in ARKO mice and prevented cancellous bone loss due to a preservation of trabecular number. Thus, voluntary running in ARKO mice was able to reduce cancellous bone resorption, suggesting that sustained exercise may potentially compensate the effects of androgen disruption on cancellous bone.


Subject(s)
Androgens/deficiency , Bone Development , Bone Resorption/prevention & control , Bone and Bones/physiology , Receptors, Androgen/genetics , Running , Animals , Bone Development/genetics , Bone Resorption/genetics , Bone Resorption/metabolism , Bone and Bones/anatomy & histology , Bone and Bones/metabolism , Male , Mice , Mice, Knockout , Organ Size , Physical Conditioning, Animal
9.
Biol Reprod ; 81(6): 1083-92, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19587329

ABSTRACT

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.


Subject(s)
Androgens/metabolism , Organ Culture Techniques/methods , Sertoli Cells/metabolism , Testis/metabolism , Analysis of Variance , Androgens/pharmacology , Animals , Apoptosis/physiology , Culture Media, Conditioned , Gene Expression Profiling , Glycerolphosphate Dehydrogenase/genetics , Glycerolphosphate Dehydrogenase/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Male , Mice , Organ Size/drug effects , Proteinase Inhibitory Proteins, Secretory , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Receptors, Dopamine D4/genetics , Receptors, Dopamine D4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/drug effects , Testis/cytology , Testis/drug effects , Testosterone/metabolism , Testosterone/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Methods Mol Biol ; 505: 237-61, 2009.
Article in English | MEDLINE | ID: mdl-19117149

ABSTRACT

The use of tissue-selective rather than ubiquitous knockouts of steroid receptors allows a more refined study of the mechanism of steroid action in defined target tissues and circumvents problems such as early lethality or major developmental defects precluding studies in affected organs. In this chapter, we describe the main steps involved in the development of tissue-selective steroid receptor knockouts by Cre/loxP technology. Problems in the development of a mouse strain with a floxed receptor allele, the selection of a suitable Cre expressing mouse strain, the generation of cell-selective knockouts by crossbreeding of the mentioned mouse strains, and the control of appropriate receptor inactivation are discussed taking the generation of mice with a Sertoli cell-selective ablation of the androgen receptor as an example.


Subject(s)
Mice, Knockout , Receptors, Steroid/metabolism , Animals , Base Sequence , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Integrases/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Steroid/genetics , Recombination, Genetic
11.
Endocrinology ; 148(5): 2027-36, 2007 May.
Article in English | MEDLINE | ID: mdl-17289843

ABSTRACT

This study sought to establish whether reduced androgen levels/action in the fetal rat testis induced by di(n-butyl) phthalate (DBP) contributes to dysgenetic features, namely reduced Sertoli cell number, occurrence of multinucleated gonocytes (MNG), and Leydig cell aggregation. Pregnant rats were administered treatments or cotreatments designed to manipulate testosterone levels [DBP, testosterone propionate (TP)] or action [flutamide, 7,12-dimethyl-benz[a]anthracene (DMBA)]. The aforementioned end points were analyzed and related to intratesticular testosterone (ITT) levels and peripheral androgen action (anogenital distance). Dysgenetic features were also evaluated in mice with inactivation of the androgen receptor (testicular feminized or ARKO mice). Exposure to DBP alone, or combined with flutamide, DMBA, or TP, resulted in reduced Sertoli cell number and ITT levels, as did exposure to TP alone; coadministration of DBP + TP caused the most severe reduction in both parameters. A positive correlation between ITT levels and Sertoli cell number was found (r = 0.791; P = 0.019). Similarly, exposure to DBP alone, or as a cotreatment, significantly increased occurrence of MNG and Leydig cell aggregation, and these were negatively correlated with ITT levels. Exposure to flutamide or DMBA alone had no significant effect on these dysgenetic end points. These findings suggest that reduced ITT decreases fetal Sertoli cell numbers and might be involved in Leydig cell aggregation and MNG. However, of these three end points, only Sertoli cell number was affected significantly in ARKO/testicular feminized mice with absent androgen action. Therefore, induction of MNG and Leydig cell aggregation might result from DBP-induced effects other than suppression of ITT levels.


Subject(s)
Gonadal Dysgenesis/pathology , Gonadal Dysgenesis/physiopathology , Testis/abnormalities , Testosterone/deficiency , Testosterone/physiology , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Androgen Antagonists/pharmacology , Animals , Body Weight , Carcinogens/pharmacology , Dibutyl Phthalate/pharmacology , Female , Feminization/pathology , Feminization/physiopathology , Flutamide/pharmacology , Giant Cells/pathology , Leydig Cells/pathology , Male , Mice , Mice, Knockout , Organ Size , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , Receptors, Androgen/genetics , Sertoli Cells/pathology , Testis/pathology
12.
Mol Endocrinol ; 20(2): 321-34, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16166195

ABSTRACT

To unravel the molecular mechanisms mediating the effects of androgens on spermatogenesis, testicular gene expression was compared in mice with Sertoli cell-selective androgen receptor knockout (SCARKO) and littermate controls on postnatal d 10. Microarray analysis identified 692 genes with significant differences in expression. Of these, 28 appeared to be down-regulated and 12 up-regulated at least 2-fold in SCARKOs compared with controls. For nine of the more than 2-fold down-regulated genes, androgen regulation was confirmed by treatment of wild-type mice with an antiandrogen (flutamide). Some of them were previously described to be androgen regulated or essential for spermatogenesis. Serine-type protease inhibitors were markedly overrepresented in this down-regulated subgroup. A time study (d 8-20), followed by cluster analysis, allowed identification of distinct expression patterns of differentially expressed genes. Three genes with a pattern closely resembling that of Pem, a prototypical androgen-regulated gene expressed in Sertoli cells, were selected for confirmation by quantitative RT-PCR and additional analysis. The data confirm that the SCARKO model allows identification of novel androgen-regulated genes in the testis. Moreover, they suggest that protease inhibitors and other proteins related to tubular restructuring and cell junction dynamics may be controlled in part by androgens.


Subject(s)
Androgens/metabolism , Gene Expression Regulation , Receptors, Androgen/genetics , Sertoli Cells/metabolism , Spermatogenesis/genetics , Testis/metabolism , Animals , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , Testis/cytology , Testis/growth & development , Transcription Factors/genetics
13.
J Bone Miner Res ; 21(4): 576-85, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16598378

ABSTRACT

UNLABELLED: The relative importance of AR and ER activation has been studied in pubertal male AR knockout and WT mice after orchidectomy and androgen replacement therapy, either with or without an aromatase inhibitor. AR activation dominates normal trabecular bone development and cortical bone modeling in male mice. Moreover, optimal periosteal bone expansion is only observed in the presence of both AR and ER activation. INTRODUCTION: Androgen receptor (AR)-mediated androgen action has traditionally been considered a key determinant of male skeletal growth. Increasing evidence, however, suggests that estrogens are also essential for normal male bone growth. Therefore, the relative importance of AR-mediated and estrogen receptor (ER)-mediated androgen action after aromatization remains to be clarified. MATERIALS AND METHODS: Trabecular and cortical bone was studied in intact or orchidectomized pubertal AR knockout (ARKO) and male wildtype (WT) mice, with or without replacement therapy (3-8 weeks of age). Nonaromatizable (dihydrotestosterone [DHT]) and aromatizable (testosterone [T]) androgens and T plus an aromatase inhibitor (anastrazole) were administered to orchidectomized ARKO and WT mice. Trabecular and cortical bone modeling were evaluated by static and dynamic histomorphometry, respectively. RESULTS: AR inactivation or orchidectomy induced a similar degree of trabecular bone loss (-68% and -71%, respectively). Both DHT and T prevented orchidectomy-induced bone loss in WT mice but not in ARKO mice. Administration of an aromatase inhibitor did not affect T action on trabecular bone. AR inactivation and orchidectomy had similar negative effects on cortical thickness (-13% and -8%, respectively) and periosteal bone formation (-50% and -26%, respectively). In orchidectomized WT mice, both DHT and T were found to stimulate periosteal bone formation and, as a result, to increase cortical thickness. In contrast, the periosteum of ARKO mice remained unresponsive to either DHT or T. Interestingly, administration of an aromatase inhibitor partly reduced T action on periosteal bone formation in orchidectomized WT mice (-34% versus orchidectomized WT mice on T), but not in ARKO mice. This effect was associated with a significant decrease in serum IGF-I (-21% versus orchidectomized WT mice on T). CONCLUSIONS: These findings suggest a major role for AR activation in normal development of trabecular bone and periosteal bone growth in male mice. Moreover, optimal stimulation of periosteal growth is only obtained in the presence of both AR and ER activation.


Subject(s)
Androgens/pharmacology , Bone and Bones/drug effects , Receptors, Androgen/deficiency , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Trabecular Meshwork/drug effects , Androgens/metabolism , Animals , Aromatase Inhibitors/pharmacology , Dihydrotestosterone/pharmacology , Enzyme Activation , Male , Mice , Mice, Knockout , Models, Animal , Orchiectomy , Osteogenesis/drug effects , Receptors, Androgen/genetics , Receptors, Estrogen/agonists , Testosterone/pharmacology
14.
Endocrinology ; 147(5): 2228-36, 2006 May.
Article in English | MEDLINE | ID: mdl-16484321

ABSTRACT

Inactivation of peroxisomal beta-oxidation in mice, by knocking out multifunctional protein-2 (MFP-2; also called d-bifunctional enzyme), causes male infertility. In the testis, extensive accumulations of neutral lipids were observed in Sertoli cells, beginning in prepubertal mice and evolving in complete testicular atrophy by the age of 4 months. Spermatogenesis was already severely affected at the age of 5 wk, and pre- and postmeiotic germ cells gradually disappeared from the tubuli seminiferi. Based on cytochemical stainings and biochemical analyses, the lipid droplets consisted of cholesteryl esters and neutral glycerolipids. Furthermore, peroxisomal beta-oxidation substrates, such as very-long-chain fatty acids and pristanic acid, accumulated in the testis, whereas the concentration of docosapentaenoic acid, a polyunsaturated fatty acid and peroxisomal beta-oxidation product, was reduced. The testicular defects were also present in double MFP-2/peroxisome proliferator-activated receptor-alpha knockout mice, ruling out the possibility that they were mediated through the activation of this nuclear receptor. Immunoreactivity for peroxisomal proteins, including MFP-2, was detected in Sertoli cells as well as in germ cells and Leydig cells. The pivotal role of peroxisomal metabolism in Sertoli cells was also demonstrated by generating mice with a Sertoli cell-selective elimination of peroxisomes through cell type-specific inactivation of the peroxin 5 gene. These mice also developed lipid inclusions and were infertile, and their testes fully degenerated by the age of 4 months. In conclusion, the present data demonstrate that peroxisomal beta-oxidation is essential for lipid homeostasis in the testis and for male fertility.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Enoyl-CoA Hydratase/metabolism , Lipids/chemistry , Multienzyme Complexes/metabolism , Sertoli Cells/metabolism , Animals , Cholesterol Esters/chemistry , Fatty Acids/metabolism , Fatty Acids/pharmacology , Fatty Acids, Unsaturated/metabolism , Fertility , Heterozygote , Homeostasis , Homozygote , Immunohistochemistry , Infertility, Male , Leydig Cells , Lipid Metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Oxygen/metabolism , PPAR alpha/genetics , Peroxisomal Multifunctional Protein-2 , Peroxisomes/metabolism , RNA/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Seminiferous Tubules/metabolism , Testis/metabolism
15.
Cell Metab ; 23(5): 837-51, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27133133

ABSTRACT

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.


Subject(s)
Cell Nucleus/metabolism , Glucose/pharmacology , Insulin/metabolism , Receptors, Androgen/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , Cyclic AMP/metabolism , Glucose Intolerance/pathology , Humans , Insulin-Secreting Cells/metabolism , Male , Mice, Knockout , Models, Biological , Receptors, Androgen/deficiency , Signal Transduction/drug effects , Testosterone/pharmacology
16.
Endocrinology ; 146(6): 2674-83, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15761038

ABSTRACT

The role of androgens in the proliferation and maturation of Sertoli cells (SC) and the development of their capacity to support spermatogenesis remains poorly understood. We evaluated these functions in complete androgen receptor knockout (ARKO) and SC-selective androgen receptor knockout (SCARKO) mice. Compared with controls, ARKO mice exhibited a progressive reduction in SC number/testis, whereas SCARKOs showed minor changes, suggesting that androgen effects on SC number are not mediated via direct action on SCs. Immunoexpression of anti-Mullerian hormone (AMH), p27(kip1), GATA-1, and sulfated glycoprotein-2, which changes according to SC maturational status, occurred normally in ARKOs and SCARKOs. Functional capacity of SCs to support spermatogonia was similar in SCARKOs and controls, whereas ARKOs showed reduced capacity with age. SC capacity to support total germ cells revealed major deficits in ARKO and SCARKO adults, particularly with respect to postmeiotic germ cells. Using quantitative RT-PCR, the expression of SC markers was compared in d 50 testes. In ARKOs, expression of Pem, fatty acid binding protein, platelet-derived growth factor-A, and transferrin were all significantly reduced, whereas FSH receptor and AMH were increased. In SCARKOs, there were modest reductions in expression of cystatin-related gene highly expressed in testis and epididymis (cystatin-TE) and claudin-11, whereas expression of Pem, fatty acid binding protein, and platelet-derived growth factor-A was markedly reduced, highlighting these as potentially androgen-regulated SC genes that merit further study. In conclusion, androgen action is not required for maturation-dependent changes in immunoexpression of the SC markers AMH, p27(kip1), GATA-1, and sulfated glycoprotein-2 but is essential for expression of other SC genes, the attainment of normal SC number, and the support of meiotic and postmeiotic germ cell development.


Subject(s)
Androgens/physiology , Receptors, Androgen/genetics , Sertoli Cells/pathology , Sertoli Cells/physiology , Spermatogenesis/physiology , Animals , Cell Differentiation/physiology , Cell Division/physiology , Female , Gene Expression/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Receptors, Androgen/physiology , Spermatogonia/cytology , Testis/pathology , Testis/physiology
17.
Endocrinology ; 146(9): 4117-26, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15919750

ABSTRACT

It is established that androgens and unidentified Sertoli cell (SC)-derived factors can influence the development of adult Leydig cells (LC) in rodents, but the mechanisms are unclear. We evaluated adult LC development and function in SC-selective androgen receptor (AR) knockout (SCARKO) and complete AR knockout (ARKO) mice. In controls, LC number increased 26-fold and LC size increased by approximately 2-fold between 12 and 140 d of age. LC number in SCARKOs was normal on d 12, but was reduced by more than 40% at later ages, although LC were larger and contained more lipid droplets and mitochondria than control LC by adulthood. ARKO LC number was reduced by up to 83% at all ages compared with controls, and LC size did not increase beyond d 12. Serum LH and testosterone levels and seminal vesicle weights were comparable in adult SCARKOs and controls, whereas LH levels were elevated 8-fold in ARKOs, although testosterone levels appeared normal. Immunohistochemistry and quantitative PCR for LC-specific markers indicated steroidogenic function per LC was probably increased in SCARKOs and reduced in ARKOs. In SCARKOs, insulin-like factor-3 and estrogen sulfotransferase (EST) mRNA expression were unchanged and increased 3-fold, respectively, compared with controls, whereas the expression of both was reduced more than 90% in ARKOs. Changes in EST expression, coupled with reduced platelet-derived growth factor-A expression, are potential causes of altered LC number and function in SCARKOs. These results show that loss of androgen action on SC has major consequences for LC development, and this could be mediated indirectly via platelet-derived growth factor-A and/or estrogens/EST.


Subject(s)
Leydig Cells/physiology , Leydig Cells/ultrastructure , Receptors, Androgen/genetics , Sertoli Cells/cytology , Sertoli Cells/physiology , Age Factors , Androgens/physiology , Animals , Cell Count , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Testis/cytology , Testis/physiology
18.
Mol Endocrinol ; 28(4): 575-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24606126

ABSTRACT

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.


Subject(s)
Gene Expression Regulation , Genetic Techniques , Genome/genetics , Protein Biosynthesis/genetics , Receptors, Androgen/metabolism , Sertoli Cells/metabolism , Androgens/pharmacology , Animals , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Sertoli Cells/drug effects , Sexual Maturation/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
20.
Mol Cell Endocrinol ; 352(1-2): 13-25, 2012 Apr 16.
Article in English | MEDLINE | ID: mdl-21871526

ABSTRACT

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.


Subject(s)
Organ Specificity , Receptors, Androgen/metabolism , Animals , Mice , Mice, Knockout , Receptors, Androgen/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL