Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
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.
Proc Natl Acad Sci U S A ; 108(19): 7962-7, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21518863

ABSTRACT

The steroid hormone signaling axis is thought to play a central role in initiation and progression of many hormonally regulated epithelial tumors. It is unclear whether all cancer-initiating signals depend on an intact hormone receptor signaling machinery. To ascertain whether cell autonomous androgen receptor (AR) is essential for initiation of prostate intraepithelial neoplasia (PIN), the response of AR-null prostate epithelia to paracrine and cell autonomous oncogenic signals was assessed in vivo by using the prostate regeneration model system. Epithelial-specific loss of AR blocked paracrine FGF10-induced PIN, whereas the add back of exogenous AR restored this response. In contrast, PIN initiated by cell-autonomous, chronic-activated AKT developed independent of epithelial AR signaling. Our findings demonstrate a selective role for AR in the initiation of PIN, dependent on the signaling pathways driving tumor formation. Insights into the role of hormone receptor signaling in the initiation of epithelial tumors may help define this axis as a target for chemoprevention of carcinomas.


Subject(s)
Neoplasms, Hormone-Dependent/etiology , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Animals , Base Sequence , Carcinogens/metabolism , DNA Primers/genetics , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Paracrine Communication , Prostatic Intraepithelial Neoplasia/etiology , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Receptors, Androgen/deficiency , Receptors, Androgen/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
4.
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
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
8.
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
9.
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
10.
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
11.
Reprod Biol Endocrinol ; 7: 88, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19712470

ABSTRACT

BACKGROUND: Testicular microliths are calcifications found within the seminiferous tubules. In humans, testicular microlithiasis (TM) has an unknown etiology but may be significantly associated with testicular germ cell tumors. Factors inducing microlith development may also, therefore, act as susceptibility factors for malignant testicular conditions. Studies to identify the mechanisms of microlith development have been hampered by the lack of suitable animal models for TM. METHODS: This was an observational study of the testicular phenotype of different mouse models. The mouse models were: cryptorchid mice, mice lacking androgen receptors (ARs) on the Sertoli cells (SCARKO), mice with a ubiquitous loss of androgen ARs (ARKO), hypogonadal (hpg) mice which lack circulating gonadotrophins, and hpg mice crossed with SCARKO (hpg.SCARKO) and ARKO (hpg.ARKO) mice. RESULTS: Microscopic TM was seen in 94% of hpg.ARKO mice (n=16) and the mean number of microliths per testis was 81+/-54. Occasional small microliths were seen in 36% (n=11) of hpg testes (mean 2+/-0.5 per testis) and 30% (n=10) of hpg.SCARKO testes (mean 8+/-6 per testis). No microliths were seen in cryptorchid, ARKO or SCARKO mice. There was no significant effect of FSH or androgen on TM in hpg.ARKO mice. CONCLUSION: We have identified a mouse model of TM and show that lack of endocrine stimulation is a cause of TM. Importantly, this model will provide a means with which to identify the mechanisms of TM development and the underlying changes in protein and gene expression.


Subject(s)
Androgens/pharmacology , Hypogonadism/physiopathology , Lithiasis/pathology , Testicular Diseases/pathology , Androgens/administration & dosage , Animals , Cryptorchidism/genetics , Cryptorchidism/physiopathology , Dihydrotestosterone/administration & dosage , Dihydrotestosterone/pharmacology , Disease Models, Animal , Female , Gonadotropins/genetics , Gonadotropins/physiology , Humans , Hypogonadism/genetics , Lithiasis/genetics , Male , Mice , Mice, Knockout , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Sertoli Cells/metabolism , Testicular Diseases/genetics , Testis/drug effects , Testis/metabolism , Testis/pathology , Testosterone/administration & dosage , Testosterone/pharmacology
12.
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
13.
Cancer Res ; 67(17): 8180-7, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804731

ABSTRACT

Development and progression of cancer is accompanied by marked changes in the expression and activity of enzymes involved in the cellular homeostasis of fatty acids. One class of enzymes that play a particularly important role in this process are the acetyl-CoA carboxylases (ACC). ACCs produce malonyl-CoA, an intermediate metabolite that functions as substrate for fatty acid synthesis and as negative regulator of fatty acid oxidation. Here, using the potent ACC inhibitor soraphen A, a macrocyclic polyketide from myxobacteria, we show that ACC activity in cancer cells is essential for proliferation and survival. Even at nanomolar concentrations, soraphen A can block fatty acid synthesis and stimulate fatty acid oxidation in LNCaP and PC-3M prostate cancer cells. As a result, the phospholipid content of cancer cells decreased, and cells stopped proliferating and ultimately died. LNCaP cells predominantly died through apoptosis, whereas PC-3M cells showed signs of autophagy. Supplementation of the culture medium with exogenous palmitic acid completely abolished the effects of soraphen A and rescued the cells from cell death. Interestingly, when added to cultures of premalignant BPH-1 cells, soraphen A only slightly affected cell proliferation and did not induce cell death. Together, these findings indicate that cancer cells have become dependent on ACC activity to provide the cell with a sufficient supply of fatty acids to permit proliferation and survival, introducing the concept of using small-molecule ACC inhibitors as therapeutic agents for cancer.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Cell Proliferation/drug effects , Macrolides/pharmacology , Neoplasms/pathology , Apoptosis/drug effects , Autophagy/drug effects , Cell Death/drug effects , Drug Evaluation, Preclinical , Fatty Acids/biosynthesis , Fatty Acids/metabolism , Humans , Male , Palmitic Acid/pharmacology , Phospholipids/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
14.
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
15.
Mol Endocrinol ; 20(10): 2265-77, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16455816

ABSTRACT

The cellular effects of androgens are mediated by a cognate receptor, the androgen receptor. Typically, the androgen receptor is viewed to exert its activity by binding to androgen response elements located in or near the promoter region of target genes, thereby directly affecting the expression of these genes. However, increasing evidence indicates that androgens may also indirectly influence the expression of genes that do not contain androgen response elements by modulating the activity of secondary transcription factors, mediating the expression of growth factors acting in a paracrine or autocrine fashion, or by inducing changes in the production of other hormones. These indirect effects of androgens can induce cascade-like actions and may play an important role in more complex processes involving coordinated responses of genes, cells, and organs. Previously, our laboratory has identified and characterized a novel indirect mechanism of androgen action involving proteolytical activation of the key lipogenic transcription factor sterol regulatory element-binding protein (SREBP), resulting in the coordinate up-regulation of entire cellular lipogenic pathways. Interestingly, activation of SREBPs by androgens occurs not only under normal physiological conditions but has also been observed in a growing number of pathologies, and more in particular in the setting of steroid-regulated cancers, where increased lipogenesis has been shown to have remarkable diagnostic and prognostic potential and is considered a prime target for novel therapeutic approaches. This review aims to analyze current insights into the molecular mechanism(s) underlying androgen activation of the SREBP pathway and to ascertain the extent to which this phenomenon can be generalized to androgen-responsive cell systems.


Subject(s)
Androgens/metabolism , Gene Expression Regulation/physiology , Lipogenesis/physiology , Signal Transduction/physiology , Sterol Regulatory Element Binding Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism
16.
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
17.
Cancer Res ; 65(15): 6719-25, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061653

ABSTRACT

Overexpression of lipogenic enzymes is a common characteristic of many cancers. Thus far, studies aimed at the exploration of lipogenic enzymes as targets for cancer intervention have focused on fatty acid synthase (FAS), the enzyme catalyzing the terminal steps in fatty acid synthesis. Chemical inhibition or RNA interference (RNAi)-mediated knockdown of FAS consistently inhibits the growth and induces death of cancer cells. Accumulation of the FAS substrate malonyl-CoA has been implicated in the mechanism of cytotoxicity of FAS inhibition. Here, using RNAi technology, we have knocked down the expression of acetyl-CoA carboxylase-alpha (ACC-alpha), the enzyme providing the malonyl-CoA substrate. Silencing of the ACC-alpha gene resulted in a similar inhibition of cell proliferation and induction of caspase-mediated apoptosis of highly lipogenic LNCaP prostate cancer cells as observed after FAS RNAi. In nonmalignant cells with low lipogenic activity, no cytotoxic effects of knockdown of ACC-alpha or FAS were observed. These findings indicate that accumulation of malonyl-CoA is not a prerequisite for cytotoxicity induced by inhibition of tumor-associated lipogenesis and suggest that in addition to FAS, ACC-alpha is a potential target for cancer intervention.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/genetics , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Acetyl-CoA Carboxylase/biosynthesis , Acetyl-CoA Carboxylase/metabolism , Apoptosis/genetics , Cell Growth Processes/genetics , Cell Line, Tumor , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Male , Malonyl Coenzyme A/metabolism , Prostatic Neoplasms/genetics , RNA Interference , RNA, Small Interfering/genetics
18.
Cancer Res ; 65(6): 2441-8, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15781660

ABSTRACT

Aggressive cancer cells typically show a high rate of energy-consuming anabolic processes driving the synthesis of lipids, proteins, and DNA. Here, we took advantage of the ability of the cell-permeable nucleoside 5-aminoimidazole-4-carboxamide (AICA) riboside to increase the intracellular levels of AICA ribotide, an AMP analogue, mimicking a low energy status of the cell. Treatment of cancer cells with AICA riboside impeded lipogenesis, decreased protein translation, and blocked DNA synthesis. Cells treated with AICA riboside stopped proliferating and lost their invasive properties and their ability to form colonies. When administered in vivo, AICA riboside attenuated the growth of MDA-MB-231 tumors in nude mice. These findings point toward a central tie between energy, anabolism, and cancer and suggest that the cellular energy sensing machinery in cancer cells is an exploitable target for cancer prevention and/or therapy.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Breast Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Ribonucleosides/pharmacology , Adenosine Monophosphate/metabolism , Adenylate Kinase/metabolism , Aminoimidazole Carboxamide/metabolism , Animals , Biomimetic Materials/metabolism , Biomimetic Materials/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Energy Metabolism , Female , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Ribonucleosides/metabolism , Ribonucleotides/metabolism , Xenograft Model Antitumor Assays
19.
Mol Cancer Ther ; 5(9): 2211-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16985054

ABSTRACT

Because of its ability to mimic a low energy status of the cell, the cell-permeable nucleoside 5-aminoimidazole-4-carboxamide (AICA) riboside was proposed as an antineoplastic agent switching off major energy-consuming processes associated with the malignant phenotype (lipid production, DNA synthesis, cell proliferation, cell migration, etc.). Key to the antineoplastic action of AICA riboside is its conversion to ZMP, an AMP mimetic that at high concentrations activates the AMP-activated protein kinase (AMPK). Here, in an attempt to increase the efficacy of AICA riboside, we pretreated cancer cells with methotrexate, an antimetabolite blocking the metabolism of ZMP. Methotrexate enhanced the AICA riboside-induced accumulation of ZMP and led to a decrease in the levels of ATP, which functions as an intrasteric inhibitor of AMPK. Consequently, methotrexate markedly sensitized AMPK for activation by AICA riboside and potentiated the inhibitory effects of AICA riboside on tumor-associated processes. As cotreatment elicited antiproliferative effects already at concentrations of compounds that were only marginally effective when used alone, our findings on the cooperation between methotrexate and AICA riboside provide new opportunities both for the application of classic antimetabolic chemotherapeutics, such as methotrexate, and for the exploitation of the energy-sensing machinery as a target for cancer intervention.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Breast Neoplasms/drug therapy , Carcinoma, Squamous Cell/drug therapy , Methotrexate/pharmacology , Ribonucleosides/pharmacology , AMP-Activated Protein Kinases , Adenosine Triphosphate/metabolism , Aminoimidazole Carboxamide/antagonists & inhibitors , Aminoimidazole Carboxamide/metabolism , Aminoimidazole Carboxamide/pharmacokinetics , Aminoimidazole Carboxamide/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , DNA, Neoplasm/antagonists & inhibitors , DNA, Neoplasm/biosynthesis , Drug Synergism , Enzyme Activation/drug effects , Humans , Lipids/biosynthesis , Multienzyme Complexes/metabolism , Nucleotide Deaminases/antagonists & inhibitors , Nucleotide Deaminases/genetics , Nucleotide Deaminases/metabolism , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/antagonists & inhibitors , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/genetics , Phosphoribosylaminoimidazolecarboxamide Formyltransferase/metabolism , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/genetics , Phosphoribosylglycinamide Formyltransferase/metabolism , Protein Serine-Threonine Kinases/metabolism , Purines/antagonists & inhibitors , Purines/biosynthesis , RNA Interference , Ribonucleosides/pharmacokinetics , Ribonucleotides/antagonists & inhibitors , Ribonucleotides/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL