Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Curr Opin Pharmacol ; 8(4): 449-57, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18619560

ABSTRACT

There is a growing body of evidence that although medical or surgical castration blocks the generation of gonadal testosterone in prostate cancer, androgens originating from other sources may continue to drive androgen receptor (AR) signaling. Recent studies have demonstrated high intratumoral levels of androgens and continued AR signaling in castration-resistant prostate cancer (CRPC), suggesting that androgens may also be synthesized de novo. Inhibiting the systemic biosynthesis of androgens in CRPC by targeting CYP17 may thus represent a rational therapeutic approach since this enzyme catalyses two key steroid reactions involving 17alpha-hydroxylase and C(17,20)-lyase in the androgen biosynthesis pathway. This review will discuss the rationale for and implications of targeting CYP17 in CRPC and focus on established and novel CYP17 inhibitors, including ketoconazole, abiraterone acetate, and VN/124-1, which are agents currently at different stages of development.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Prostatic Neoplasms/drug therapy , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Androgen Receptor Antagonists , Animals , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Enzyme Inhibitors/pharmacology , Humans , Male , Prostatic Neoplasms/enzymology , Receptors, Androgen/biosynthesis , Receptors, Androgen/drug effects , Steroid 17-alpha-Hydroxylase/biosynthesis , Steroid 17-alpha-Hydroxylase/drug effects
2.
BMC Genomics ; 9: 487, 2008 Oct 16.
Article in English | MEDLINE | ID: mdl-18925944

ABSTRACT

BACKGROUND: Dichlorodiphenyltrichloroethane (DDT) is a persistent estrogenic organochlorine pesticide that is a rodent hepatic tumor promoter, with inconclusive carcinogenicity in humans. We have previously reported that o, p'-DDT elicits primarily PXR/CAR-mediated activity, rather than ER-mediated hepatic responses, and suggested that CAR-mediated effects, as opposed to ER-mediated effects, may be more important in tumor promotion in the rat liver. To further characterize species-specific hepatic responses, gene expression analysis, with complementary histopathology and tissue level analyses were investigated in immature, ovariectomized C57BL/6 mice treated with 300 mg/kg o, p'-DDT, and compared to Sprague-Dawley rat data. RESULTS: Rats and mice exhibited negligible histopathology with rapid o, p'-DDT metabolism. Gene expression profiles were also similar, exhibiting PXR/CAR regulation with the characteristic induction of Cyp2b10 and Cyp3a11. However, PXR-specific target genes such as Apoa4 or Insig2 exhibited more pronounced induction compared to CAR-specific genes in the mouse. In addition, mouse Car mRNA levels decreased, possibly contributing to the preferential activation of mouse PXR. ER-regulated genes Cyp17a1 and Cyp7b1 were also induced, suggesting o, p'-DDT also elicits ER-mediated gene expression in the mouse, while ER-mediated effects were negligible in the rat, possibly due to the inhibitory effects of CAR on ER activities. In addition, o, p'-DDT induced Gadd45a, Gadd45b and Cdkn1, suggesting DNA damage may be an additional risk factor. Furthermore, elevated blood DHEA-S levels at 12 h after treatment in the mouse may also contribute to the endocrine-related effects of o, p'-DDT. CONCLUSION: Although DDT is known to cause rodent hepatic tumors, the marked species differences in PXR/CAR structure, expression patterns and ligand preference as well as significant species-specific differences in steroidogenesis, especially CYP17A1 expression and activity, confound the extrapolation of these results to humans. Nevertheless, the identification of potential modes of action as well as species-specific responses may assist in the selection and further development of more appropriate models for assessing the toxicity of DDT to humans and wildlife.


Subject(s)
Dichlorodiphenyl Dichloroethylene/toxicity , Insecticides/toxicity , Liver/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/metabolism , Receptors, Steroid/metabolism , Transcription Factors/metabolism , Androstenedione/blood , Animals , Cluster Analysis , Constitutive Androstane Receptor , Dehydroepiandrosterone Sulfate/blood , Dichlorodiphenyl Dichloroethylene/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Insecticides/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Pregnane X Receptor , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Receptors, Steroid/drug effects , Receptors, Steroid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Transcription Factors/drug effects , Transcription Factors/genetics
3.
Arch Toxicol ; 82(11): 851-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18449530

ABSTRACT

We recently showed that prenatal exposure to diesel exhaust (DE) disrupts spermatogenesis in mouse offspring. This study was undertaken to determine whether filtered DE in which 99.97% of diesel exhaust particles >0.3 microm in diameter were removed affects spermatogenesis in growing mice. After prenatal exposure to filtered DE for 2-16 days postcoitum, we examined daily sperm production (DSP), testicular histology, serum testosterone levels and mRNA expression of hormone synthesis process-related factors. In the filtered DE exposed group, DSP was markedly reduced at 12 weeks compared with the control group; clean air exposed group. Histological examination showed multinucleated giant cells and partial vacuolation in the seminiferous tubules of the exposed group. Testosterone was elevated significantly at 5 weeks. Moreover, luteinizing hormone receptor mRNA at 5 and 12 weeks, 17alpha-hydroxylase/C17-20-lyase and 17beta-hydroxysteroid dehydrogenase mRNAs at 12 weeks were significantly elevated. These results suggest that filtered DE retains its toxic effects on the male reproductive system following prenatal exposure.


Subject(s)
Prenatal Exposure Delayed Effects , Spermatogenesis/drug effects , Testis/drug effects , Vehicle Emissions/toxicity , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Female , Gene Expression Regulation/drug effects , Giant Cells/drug effects , Giant Cells/metabolism , Male , Mice , Mice, Inbred ICR , Particle Size , Pregnancy , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, LH/drug effects , Receptors, LH/metabolism , Seminiferous Tubules/drug effects , Sperm Count , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/metabolism , Testis/metabolism , Testosterone/blood , Time Factors , Vacuoles/drug effects , Vacuoles/metabolism
4.
Endocrinology ; 159(11): 3689-3698, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30219917

ABSTRACT

Abiraterone acetate (AA) is a potent inhibitor of steroidogenic enzyme 17α-hydroxylase/17,20-lyase (CYP17A1). AA is approved for the treatment of prostate cancer but could also be used to treat patients with Cushing syndrome (CS). Similar to humans, canine glucocorticoid synthesis requires CYP17A1, providing a useful animal model. The objective of this study was to preclinically investigate the effect of AA on adrenocortical hormone production, cell viability, and mRNA expression of steroidogenic enzymes in canine primary adrenocortical cell cultures (n = 9) from the adrenal glands of nine healthy dogs. The cells were incubated with AA (0.125 nM to 10 ĀµM) for 72 hours under basal conditions and with 100 nM ACTH(1-24). Adrenocortical hormone concentrations were measured in culture medium using liquid chromatography-mass spectrometry, RNA was isolated from cells for subsequent real-time quantitative PCR analysis, and cell viability was assessed with an alamarBlue™ assay. AA reduced cortisol (IC50, 21.4 Ā± 4.6 nM) without affecting aldosterone under basal and ACTH-stimulated conditions. AA increased progesterone under basal and ACTH-stimulated conditions but reduced corticosterone under basal conditions, suggesting concurrent inhibition of 21-hydroxylation. AA did not affect the mRNA expression of steroidogenic enzymes and did not inhibit cell viability. In summary, primary canine adrenocortical cell culture is a useful model system for drug testing. For the treatment of CS, AA may to be superior to other steroidogenesis inhibitors due to its low toxicity. For future in vivo studies, dogs with endogenous CS may provide a useful animal model.


Subject(s)
Abiraterone Acetate/pharmacology , Adrenal Cortex/cytology , Cushing Syndrome/metabolism , RNA, Messenger/drug effects , Steroid 17-alpha-Hydroxylase/drug effects , Steroid Synthesis Inhibitors/pharmacology , Aldosterone/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Corticosterone/metabolism , Dogs , Hydrocortisone/metabolism , In Vitro Techniques , Progesterone/metabolism , RNA, Messenger/metabolism , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism
5.
Clin Pharmacol Ther ; 104(1): 201-210, 2018 07.
Article in English | MEDLINE | ID: mdl-29027195

ABSTRACT

The testis-specific Y-encoded-like protein (TSPYL) gene family includes TSPYL1 to TSPYL6. We previously reported that TSPYL5 regulates cytochrome P450 (CYP) 19A1 expression. Here we show that TSPYLs, especially TSPYL 1, 2, and 4, can regulate the expression of many CYP genes, including CYP17A1, a key enzyme in androgen biosynthesis, and CYP3A4, an enzyme that catalyzes the metabolism of abiraterone, a CYP17 inhibitor. Furthermore, a common TSPYL1 single nucleotide polymorphism (SNP), rs3828743 (G/A) (Pro62Ser), abolishes TSPYL1's ability to suppress CYP3A4 expression, resulting in reduced abiraterone concentrations and increased cell proliferation. Data from a prospective clinical trial of 87 metastatic castration-resistant prostate cancer patients treated with abiraterone acetate/prednisone showed that the variant SNP genotype (A) was significantly associated with worse response and progression-free survival. In summary, TSPYL genes are novel CYP gene transcription regulators, and genetic alteration within these genes significantly influences response to drug therapy through transcriptional regulation of CYP450 genes.


Subject(s)
Abiraterone Acetate/therapeutic use , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 Enzyme Inhibitors/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Steroid 17-alpha-Hydroxylase/genetics , Abiraterone Acetate/pharmacology , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inhibitors/pharmacology , DNA-Binding Proteins , Dehydroepiandrosterone/metabolism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Hep G2 Cells , Humans , Male , Neoplasm Metastasis , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Proportional Hazards Models , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Messenger/metabolism , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/drug effects , Treatment Outcome
6.
Endocrinology ; 147(2): 927-36, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16254025

ABSTRACT

Reproductive and developmental disorders are the most sensitive toxic effects caused by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). TCDD is thought to produce many, if not all, of these toxic effects by impairing steroidogenesis and/or steroid action during the prenatal or early postnatal stages. However, the mechanism of the antisex steroid effect of TCDD is not well understood. This study revealed that steroidogenic acute-regulatory protein (StAR), a key transporter of cholesterol for steroidogenesis, in the testes of fetal rats are down-regulated by maternal exposure to TCDD. It was also shown that many mRNAs of steroidogenetic enzymes, including cytochromes P450 11A1, 17, and 11B1 and 3beta-hydroxysteroid dehydrogenase, are reduced in fetuses of TCDD-treated dams in a testis-specific manner. The same was also observed for the expression of estrogen-alpha receptors and androgen receptors. Whereas StAR expression was not affected by TCDD in cultured fetal testis, the fetal serum content of LH, a pituitary regulator of StAR, was significantly reduced by TCDD. In agreement with this, pituitary expression of LHbeta subunit mRNA in fetuses was reduced by maternal exposure to TCDD, whereas the alpha-subunit remained unchanged. The reduction in LHbeta is suggested to occur by a mechanism different from the reduction in the GnRH level. Direct supply of exogenous gonadotropin to TCDD-exposed fetuses completely abolished the reduction of StAR expression. Taken together, these results demonstrate that TCDD impairs steroidogenesis in the fetus by targeting pituitary gonadotropins.


Subject(s)
Cholesterol/metabolism , Environmental Pollutants/toxicity , Gene Expression Regulation, Developmental/drug effects , Luteinizing Hormone, beta Subunit/drug effects , Pituitary Gland/embryology , Polychlorinated Dibenzodioxins/toxicity , Prenatal Exposure Delayed Effects , Animals , Down-Regulation , Female , Gonadal Steroid Hormones/metabolism , Luteinizing Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/metabolism , Male , Maternal Exposure , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphoproteins/drug effects , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pregnancy , RNA, Messenger/analysis , Rats , Rats, Wistar , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Testis/drug effects , Testis/embryology , Testis/metabolism
7.
Expert Opin Investig Drugs ; 25(6): 697-707, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26954621

ABSTRACT

INTRODUCTION: Prostate cancer is the most common cancer in elderly males. Regardless of the initial hormonal treatment in metastatic disease, a significant proportion of patients develop castration resistant prostate cancer (CRPC). A better understanding of the molecular mechanisms behind castration resistance has led to the approval of oral medications such as abiraterone acetate and enzalutamide. Relevant research is accelerated with numerous agents being tested for the management of CRPC. AREAS COVERED: The authors present Phase I and II studies targeting the androgen receptor for the treatment of CRPC. Three groups of agents are identified according to the mechanism of action. These include the CYP-17 modulators (Orteronel, Galeterone, VT-464 and CFG-920), novel antiandrogens (Apatorsen, ARN-509, ODM-201, EZN-4176, AZD-3514) and bipolar androgen therapy. EXPERT OPINION: Further understanding of the mechanisms leading to castration resistance in prostate cancer can reveal potential targets for the development of novel anti-cancer agents. Except for the development of novel antiandrogens and CYP-17 modulators, bipolar androgen therapy is an interesting therapeutic approach. The combinations of the novel agents tested in Phase I and II studies with established agents is another field of interest. The real challenge is to distinguish a novel anti-cancer agent with acceptable tolerability and the best outcome.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Aged , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Androgen Receptor Antagonists/pharmacology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Drug Design , Humans , Male , Prostatic Neoplasms, Castration-Resistant/pathology , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/metabolism
8.
Diabetes ; 46(7): 1235-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9200662

ABSTRACT

The crucial role of glucocorticoids in obesity and insulin resistance and the actions of the OB protein leptin on the hypothalamic-pituitary-adrenal (HPA) axis suggest that there is an important interaction of leptin with the glucocorticoid system. Therefore, we designed a study to test the effect of leptin directly on adrenocortical steroidogenesis. Primary cultures of bovine adrenocortical cells were incubated with increasing concentrations (10-1,000 ng/ml) of recombinant mouse leptin for 24 h, and the effects of leptin on basal and ACTH-stimulated cortisol secretion were determined. The accumulation of P450 17alpha mRNA following incubation with ACTH (10 nmol/l) and leptin (10-1,000 ng/ml) was analyzed by Northern blot. Adrenocortical cells were characterized by immunohistochemical staining for 17alpha-hydroxyprogesterone. Leptin (10-1,000 ng/ml) inhibited basal and ACTH-stimulated cortisol release. At a concentration that occurs in obese individuals in vivo (100 ng/ml), it reduced basal cortisol secretion to 52.7 +/- 37% (mean +/- SE). The rise in cortisol secretion following maximal ACTH stimulation (10 nmol/l) was blunted to 55.2 +/- 27%. At more physiological concentrations of ACTH (0.1 nmol/l), the inhibition of cortisol release by coincubation with low doses of leptin (10 ng/ml) was even more pronounced, leading to a reduction to 32.8% (1,248 +/- 134 vs. 410 +/- 157 nmol/l). Addition of OB protein (10-1,000 ng/ml) led to a dose-dependent reduction of ACTH-stimulated cytochrome P450 17alpha mRNA accumulation (from 80 to 45%), suggesting that leptin regulates adrenal steroidogenesis at the transcriptional level. These data clearly demonstrate that leptin inhibits cortisol production in adrenocortical cells and therefore appears to be a metabolic signal that directly acts on the adrenal gland.


Subject(s)
Adrenal Glands/physiology , Adrenocorticotropic Hormone/pharmacology , Hydrocortisone/metabolism , Proteins/pharmacology , Steroid 17-alpha-Hydroxylase/drug effects , Adrenal Glands/cytology , Adrenal Glands/drug effects , Animals , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Leptin , Male , Mice , Proteins/physiology , RNA, Messenger/analysis , RNA, Messenger/genetics , Recombinant Proteins/pharmacology , Steroid 17-alpha-Hydroxylase/genetics
9.
Mol Endocrinol ; 13(1): 167-75, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892022

ABSTRACT

Cytochrome P450c17 catalyzes steroid 17alpha-hydroxylase and 17,20-lyase activities and hence is a key enzyme in the production of human glucocorticoids and sex steroids. These two activities are catalyzed in a single substrate-binding site but are regulated independently in human physiology. We have recently shown that cytochrome b5 facilitates 17,20-lyase activity by allosterically promoting the interaction of P450c17 with P450 oxidoreductase (OR) and that the human P450c17 mutations, R347H and R358Q, selectively destroy 17,20-lyase activity while sparing 17alpha-hydroxylase activity. We transfected COS-1 cells with vectors for these P450c17 mutants and found that an excess of OR and b5 restored a small amount of 17,20-lyase activity, suggesting the mutations interfere with electron donation. To determine whether these mutations selectively interfere with the interaction of P450c17 and its electron-donating system, we expressed each P450cl7 mutant in yeast with or without OR, b5, or both, and measured enzyme kinetics in yeast microsomes using pregnenolone and 17alpha-hydroxypregnenolone as substrates. The apparent Michaelis-Menten (Km) values for the R347H mutant with and without coexpressed OR were 0.2 and 0.6 microM, respectively, and for the R358Q mutant with and without OR they were 0.3 and 0.4 microM, respectively; these values did not differ significantly from the wild-type values of 0.4 and 0.8 microM with and without OR, respectively. Furthermore, coincubation with 17alpha-hydroxypregnenolone showed a competitive mechanism for interference of catalysis. The similar kinetics and the competitive inhibition prove that the mutations did not affect the active site. Coexpression of the mutants with OR yielded insignificant 17,20-lyase activity, but addition of a 30:1 molar excess cytochrome b5 to these microsomes restored partial 17,20-lyase activity, with the R358Q mutant achieving twice the activity of the R347H mutant. These data indicate that both mutations selectively interfere with 17,20-lyase activity by altering the interaction of P450c17 with OR, thus proving that the lyase activity was disrupted by interfering with electron transfer. Furthermore, the data offer the first evidence that R347 is a crucial component of the site at which b5 interacts with the P450c17 x OR complex to promote electron transfer.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Cytochromes b5/metabolism , Mutation , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , 17-alpha-Hydroxypregnenolone/metabolism , Animals , COS Cells/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytochromes b5/genetics , Cytochromes b5/pharmacology , Humans , Kinetics , Microsomes/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Steroid 17-alpha-Hydroxylase/drug effects , Structure-Activity Relationship , Yeasts/genetics
10.
Mol Endocrinol ; 13(6): 946-57, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10379893

ABSTRACT

To test the hypothesis that the hyperandrogenemia associated with polycystic ovary syndrome (PCOS) results from an intrinsic abnormality in ovarian theca cell steroidogenesis, we examined steroid hormone production, steroidogenic enzyme activity, and mRNA expression in normal and PCOS theca cells propagated in long-term culture. Progesterone (P4), 17alpha-hydroxyprogesterone (17OHP4), and testosterone (T) production per cell were markedly increased in PCOS theca cell cultures. Moreover, basal and forskolin-stimulated pregnenolone, P4, and dehydroepiandrosterone metabolism were increased dramatically in PCOS theca cells. PCOS theca cells were capable of substantial metabolism of precursors into T, reflecting expression of an androgenic 17beta-hydroxysteroid dehydrogenase. Forskolin-stimulated cholesterol side chain cleavage enzyme (CYP11A) and 17alpha-hydroxylase/17,20-desmolase (CYP17) expression were augmented in PCOS theca cells compared with normal cells, whereas no differences were found in steroidogenic acute regulatory protein mRNA expression. Collectively, these observations establish that increased CYP11A and CYP17 mRNA expression, as well as increased CYP17, 3beta-hydroxysteroid dehydrogenase, and 17beta-hydroxysteroid dehydrogenase enzyme activity per theca cell, and consequently increased production of P4, 17OHP4, and T, are stable properties of PCOS theca cells. These findings are consistent with the notion that there is an intrinsic alteration in the steroidogenic activity of PCOS thecal cells that encompasses multiple steps in the biosynthetic pathway.


Subject(s)
Androgens/biosynthesis , Polycystic Ovary Syndrome/metabolism , Theca Cells/metabolism , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Adult , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Colforsin/pharmacology , Dehydroepiandrosterone/metabolism , Female , Gene Expression Regulation , Humans , Phenotype , Phosphoproteins/drug effects , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pregnenolone/metabolism , Progesterone/metabolism , Reference Values , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Theca Cells/drug effects
11.
Clinics (Sao Paulo) ; 70(2): 144-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25789524

ABSTRACT

OBJECTIVE: To analyze steroidogenesis-related gene expression in the rat ovary exposed to melatonin supplementation. METHODS: Thirty-two virgin adult female rats were randomized to two groups as follows: the control group GI received vehicle and the experimental group GII received melatonin supplementation (10 Āµg/night per animal) for 60 consecutive days. After the treatment, animals were anesthetized and the collected ovaries were immediately placed in liquid nitrogen for complementary deoxyribonucleic acid microarray analyses. A GeneChip(Ā®) Kit Rat Genome 230 2.0 Affymetrix Array was used for gene analysis and the experiment was repeated three times for each group. The results were normalized with the GeneChip(Ā®) Operating Software program and confirmed through analysis with the secondary deoxyribonucleic acid-Chip Analyzer (dChip) software. The data were confirmed by real-time reverse transcription polymerase chain reaction analysis. Genes related to ovarian function were further confirmed by immunohistochemistry. RESULTS: We found the upregulation of the type 9 adenylate cyclase and inhibin beta B genes and the downregulation of the cyclic adenosine monophosphate response element modulator and cytochrome P450 family 17a1 genes in the ovarian tissue of GII compared to those of the control group. CONCLUSION: Our data suggest that melatonin supplementation decreases gene expression of cyclic adenosine monophosphate, which changes ovarian steroidogenesis.


Subject(s)
Adenylyl Cyclases/genetics , Gene Expression/drug effects , Inhibin-beta Subunits/genetics , Melatonin/pharmacology , Ovary/drug effects , Adenylyl Cyclases/metabolism , Animals , Cyclic AMP/metabolism , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/metabolism , Dietary Supplements , Female , Inhibin-beta Subunits/metabolism , Melatonin/metabolism , Models, Animal , Ovary/metabolism , RNA, Complementary/isolation & purification , Random Allocation , Rats, Wistar , Real-Time Polymerase Chain Reaction/methods , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Tissue Array Analysis/methods , Up-Regulation
12.
Toxicol In Vitro ; 29(1): 155-61, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25317747

ABSTRACT

Monocrotophos (MCP) pesticide, listed as a UNEP Prior Informed Consent chemical, has been proved to exert toxic effects on the reproductive system of teleost fishes by changing the balance of sex steroid hormones. To investigate the effects of MCP on steroidogenesis in vitro, the rainbow trout (Oncorhynchus mykiss) gonadal cell line RTG-2 was exposed to different MCP concentrations for 48 h. The levels of 17 Ɵ-estradiol (E(2)) and testosterone in the medium were measured by radioimmunoassay and the expression of steroidogenic acute regulatory protein and cytochrome P450 enzymes CYP11A1, CYP17, and CYP19A was detected by quantitative real-time PCR. The results showed that 1.0 and 10.0 Āµg/L MCP pesticide induced E(2) levels and promoted steroidogenic enzyme expression. The possible mechanisms of MCP steroidogenic activity were investigated using inhibitors of protein kinase A (PKA) and protein kinase C. The PKA inhibitor H-89 abrogated the 10.0 Āµg/L MCP-induced transcriptional up-regulation of steroidogenic enzymes, suggesting an involvement of PKA-dependent mechanism in the disruption of steroidogenesis by the MCP pesticide in rainbow trout RTG-2 cells.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/drug effects , Estradiol/biosynthesis , Insecticides/toxicity , Monocrotophos/toxicity , Signal Transduction/drug effects , Testosterone/biosynthesis , Animals , Aromatase/biosynthesis , Aromatase/drug effects , Cell Line , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Cholesterol Side-Chain Cleavage Enzyme/drug effects , Endocrine Disruptors/toxicity , Gonads/cytology , Gonads/drug effects , Gonads/metabolism , Oncorhynchus mykiss/metabolism , Oncorhynchus mykiss/physiology , Real-Time Polymerase Chain Reaction , Steroid 17-alpha-Hydroxylase/biosynthesis , Steroid 17-alpha-Hydroxylase/drug effects
13.
Endocrinology ; 156(7): 2530-40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25868050

ABSTRACT

Bone morphogenetic proteins (BMPs) comprise one of the largest subgroups in the TGF-Ɵ ligand superfamily. We have identified a functional BMP system equipped with the ligand (BMP4), receptors (BMP type II receptor, BMP type IA receptor, also called ALK3) and the signaling proteins, namely the mothers against decapentaplegic homologs 1, 4, and 5 in the human adrenal gland and the human adrenocortical cell line H295R. Microarray, quantitative RT-PCR, and immunohistochemistry confirmed that BMP4 expression was highest in the adrenal zona glomerulosa followed by the zona fasciculata and zona reticularis. Treatment of H295R cells with BMP4 caused phosphorylation of the mothers against decapentaplegic and a profound decrease in synthesis of the C19 steroids dehydroepiandrosterone (DHEA), DHEA sulfate, and androstenedione. Administration of BMP4 to cultures of H295R cells also caused a profound decrease in the mRNA and protein levels of 17α-hydroxylase/17,20-lyase (CYP17A1 and P450c17, respectively) but no significant effect on the mRNA levels of cholesterol side-chain cleavage cytochrome P450 (CYP11A1) or type 2 3Ɵ-hydroxysteroid dehydrogenase (HSD3B2). Furthermore, Noggin (a BMP inhibitor) was able to reverse the negative effects of BMP4 with respect to both CYP17A1 transcription and DHEA secretion in the H295R cell line. Collectively the present data suggest that BMP4 is an autocrine/paracrine negative regulator of C19 steroid synthesis in the human adrenal and works by suppressing P450c17.


Subject(s)
Adrenal Cortex/metabolism , Androstenedione/biosynthesis , Bone Morphogenetic Protein 4/genetics , Dehydroepiandrosterone Sulfate/metabolism , Dehydroepiandrosterone/biosynthesis , RNA, Messenger/metabolism , Adrenal Glands/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cell Line , Cholesterol Side-Chain Cleavage Enzyme/drug effects , Cholesterol Side-Chain Cleavage Enzyme/genetics , Gene Expression Profiling , Humans , Paracrine Communication/genetics , Progesterone Reductase/drug effects , Progesterone Reductase/genetics , RNA, Messenger/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Smad1 Protein/metabolism , Smad4 Protein/metabolism , Smad5 Protein/metabolism , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Zona Fasciculata/metabolism , Zona Glomerulosa/metabolism , Zona Reticularis/metabolism
14.
J Mol Endocrinol ; 17(1): 55-60, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8863187

ABSTRACT

The cytochrome P450 enzyme 17 alpha-hydroxylase (P450c17) is required for androgen synthesis and therefore regulates substrate supply for aromatization. In this study, changes in P450c17 activity and mRNA levels were measured during ovarian development in the normal mouse and in the hypogonadal (hpg) mouse which lacks circulating gonadotrophins. At birth, low levels of P450c17 activity and mRNA were detectable in normal ovaries. This basal level of expression did not change until after day 10 at which time both enzyme activity and mRNA levels increased by six- to eightfold. In the hpg mouse, levels of P450c17 mRNA were normal at birth but did not change significantly during subsequent development and were significantly less than normal by day 15. Results show that there is a low level of gonadotrophin-independent expression of P450c17 in the ovary at birth and that gonadotrophins are required for the subsequent increase in expression between days 10 and 15. In the ovary, P450c17 is expressed solely in the thecal/interstitial compartment and interstitial cells arise in the mouse ovary around day 11. Changes in P450c17 are likely, therefore, to be related to gonadotrophin-dependent development of the interstitial tissue in the mouse. Treatment of adult hpg mice with LH and FSH showed that both gonadotrophins can act to increase P450c17 activity. Since FSH acts only on the granulosa cell compartment of the ovary it is likely that FSH acts through a paracrine mechanism to regulate thecal/interstitial cell activity.


Subject(s)
Hypogonadism/enzymology , Ovary/enzymology , RNA, Messenger/genetics , Steroid 17-alpha-Hydroxylase/genetics , Animals , Enzyme Activation , Female , Follicle Stimulating Hormone/pharmacology , Luteinizing Hormone/pharmacology , Mice , Ovary/drug effects , Ovary/growth & development , RNA, Messenger/metabolism , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/metabolism
15.
J Steroid Biochem Mol Biol ; 92(3): 199-208, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15555913

ABSTRACT

In addition to causing MĆ¼llerian duct regression in fetal males, MĆ¼llerian inhibiting substance (MIS) inhibits the expression of the bifunctional cytochrome P450, C17 hydroxylase/C(17-20) lyase (Cyp17), the enzyme that catalyzes the committed step in sex steroid synthesis. To investigate the paracrine effects of MIS on steroidogenic activity, we have performed assays with microsomes from mouse MA-10 Leydig cells. With microsomes from untreated MA-10 cells, progesterone was largely metabolized by 5alpha-reductase and subsequently converted by 3-keto steroid reductases to allopregnanolone and epiallopregnanolone. Addition of cAMP to the cells shifted microsomal steroid production to the Cyp17 product androstenedione and its 5alpha,3beta-reduced form, epiandrosterone. Microsomes from MIS-treated cells were less active with the progesterone substrate than those of untreated cells but co-treatment of the cells with both MIS and cAMP mitigated the cAMP-induced shift of the microsomes to androstenedione production. Quantitative analyses of steroid production by Cyp17 showed that cAMP decreased the amount of 17-hydroxyprogesterone produced relative to the androstenedione, suggesting that cAMP signaling lowers the efficiency of the Cyp17 hydroxylase activity or else increases the efficiency of its lyase activity. Addition of MIS to the cAMP-treated cells partially reversed this effect, as well. These results indicate that cAMP induces MA-10 cells to switch from producing 5alpha-reduced progesterone metabolites to producing androstenedione and its metabolites by increasing Cyp17 expression and its relative lyase activity, both of which are inhibited by MIS.


Subject(s)
Cyclic AMP/pharmacology , Glycoproteins/pharmacology , Leydig Cells/drug effects , Steroids/biosynthesis , Testicular Hormones/pharmacology , 17-alpha-Hydroxyprogesterone/metabolism , Androstenedione/biosynthesis , Animals , Anti-Mullerian Hormone , Leydig Cells/metabolism , Male , Mice , Microsomes/drug effects , Microsomes/metabolism , Progesterone/metabolism , Rats , Steroid 17-alpha-Hydroxylase/drug effects
16.
J Steroid Biochem Mol Biol ; 60(1-2): 77-85, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9182861

ABSTRACT

The effects of anticonvulsants on the activities of cytochromes P-450(17alpha,lyase) (CYP17), P-450arom (CYP19), P-450C21 (CYP21), P-450SCC (CYP11A1), and P-450(11beta) (CYP11B1) mono-oxygenase systems were studied using rat testicular microsomes, human placental microsomes, bovine adrenocortical microsomes, bovine adrenocortical mitochondria and purified cytochrome P-450(11beta). Phenytoin, clonazepam and carbamazepine inhibited the steroidogenesis catalysed by these cytochrome P-450 mono-oxygenase systems and the Ki values for each anticonvulsant were determined. Neither hydantoin nor sodium valproate inhibited the activities of steroidogenic cytochromes P-450. When the activities of cytochromes P-450arom and P-450C21 were measured in the presence of anticonvulsants, the Ki values (0.15 mM) for phenytoin were close to the plasma concentration of phenytoin under therapeutic conditions. Phenytoin, clonazepam and carbamazepine directly inhibited the monooxygenase activities of cytochromes P-450, because they did not affect the activities of NADPH-cytochrome P-450 reductase, NADPH-adrenoferredoxin reductase and adrenoferredoxin.


Subject(s)
Anticonvulsants/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Steroids/metabolism , Adrenodoxin/drug effects , Adrenodoxin/metabolism , Androstenedione/metabolism , Animals , Aromatase/drug effects , Aromatase/metabolism , Cattle , Cholesterol/metabolism , Cholesterol Side-Chain Cleavage Enzyme/drug effects , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/drug effects , Desoxycorticosterone/metabolism , Estrogens/metabolism , Estrone/metabolism , Female , Ferredoxin-NADP Reductase/drug effects , Ferredoxin-NADP Reductase/metabolism , Humans , Male , Microsomes/drug effects , Microsomes/metabolism , NADH, NADPH Oxidoreductases/drug effects , NADH, NADPH Oxidoreductases/metabolism , NADPH-Ferrihemoprotein Reductase , Placenta/drug effects , Placenta/enzymology , Pregnancy , Pregnenolone/metabolism , Progesterone/metabolism , Rats , Steroid 11-beta-Hydroxylase/drug effects , Steroid 11-beta-Hydroxylase/metabolism , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/metabolism , Steroid 21-Hydroxylase , Testis/drug effects , Testis/metabolism
17.
Fertil Steril ; 67(4): 654-62, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9093190

ABSTRACT

OBJECTIVE: To evaluate whether ovarian function might have an influence on the adrenal hyperandrogenism present in patients with functional ovarian hyperandrogenism. DESIGN: Controlled clinical study. SETTING: Tertiary institutional hospital. PATIENT(S): Twenty-nine hirsute women with functional ovarian hyperandrogenism and 12 normal controls. INTERVENTION(S): The ACTH and GnRH tests were performed before and during triptorelin-induced ovarian suppression in patients. The normal women served as controls for the ACTH test. MAIN OUTCOME MEASURE(S): Basal and ACTH-stimulated steroid values. RESULT(S): All patients presented elevated T and free androgen index, which normalized after triptorelin. Patients with functional ovarian hyperandrogenism and adrenal hyperandrogenism, defined by elevated basal DHEAS (n = 10), presented enhanced delta 4-17, 20-lyase activity, which persisted during ovarian suppression. delta 4-17,20-lyase activity was normal in the functional ovarian hyperandrogenism patients without adrenal hyperandrogenism (n = 19). No correlation was observed between the any of the indexes of the adrenal enzymatic activities evaluated and plasma E2 or T. CONCLUSION(S): Increased adrenal delta 4-17,20-lyase activity is present in functional ovarian hyperandrogenism women with adrenal hyperandrogenism. No influence of the excess ovarian androgens or estrogens was found on any of the adrenal enzymatic pathways explored.


Subject(s)
Adrenocortical Hyperfunction/blood , Androgens/metabolism , Hyperandrogenism/blood , Ovarian Diseases/blood , Steroid 17-alpha-Hydroxylase/blood , Steroids/blood , Adolescent , Adrenocortical Hyperfunction/enzymology , Adult , Cohort Studies , Cosyntropin/administration & dosage , Delayed-Action Preparations , Female , Follicular Phase/drug effects , Follicular Phase/physiology , Gonadotropin-Releasing Hormone/administration & dosage , Humans , Hyperandrogenism/enzymology , Luteolytic Agents/administration & dosage , Ovarian Diseases/enzymology , Reference Values , Steroid 17-alpha-Hydroxylase/drug effects , Triptorelin Pamoate/administration & dosage
18.
Reprod Fertil Dev ; 14(1-2): 1-6, 2002.
Article in English | MEDLINE | ID: mdl-12051514

ABSTRACT

In the present study, it was hypothesized that the adrenocorticotrophin hormone receptor (ACTH-R) would be up-regulated in the adrenal gland of the sheep fetus following infusion of physiological amounts of ACTH, as shown for adrenal cortical cells in culture. In chronically catheterized sheep, an intravenous infusion of ACTH(1-24) was given to 6 fetuses for 24 h at a rate of 0.5 microg h(-1), starting on Day 126 or 127 of gestation (term approximately 147 days). Four control fetuses received an infusion of vehicle (saline). Total RNA was extracted from the fetal adrenal glands by the guanidinium thiocyanate method. Expression of specific mRNAs was determined by ribonuclease protection assay using cRNA probes directed against: ACTH-R; the steroid enzymes side-chain cleavage (P450scc), 3beta-hydroxysteroid dehydrogenase (3beta-HSD), 17apha-hydroxylase (P450c17) and 21beta-hydroxylase (P450c21); and beta-actin. Ratios of mRNA expression to beta-actin mRNA expression (arbitrary units) were calculated to correct for differences in RNA quality between samples. The concentration (mean +/- SEM) of immunoreactive cortisol in fetal plasma was greater after ACTH infusion than after vehicle infusion (47 +/- 3 v. 13 +/- 2 ng mL(-1) respectively; P<0.001). Adrenal expression of P450scc and P450c21 mRNA increased after ACTH infusion (P<0.05), whereas expression of P450c17 and 3beta-HSD mRNA was unchanged. There was no difference in ACTH-R mRNA expression between ACTH- and vehicle-infused fetuses (254 +/- 48 v. 305 +/- 76 arbitrary units respectively). It was concluded that ACTH is able to increase plasma cortisol concentrations in the sheep fetus by up-regulating cortisol synthesis in the adrenal gland, but that in vivo this does not require up-regulation of ACTH-R mRNA.


Subject(s)
Adrenal Cortex/embryology , Adrenal Cortex/physiology , Adrenocorticotropic Hormone/metabolism , Receptors, Corticotropin/genetics , 3-Hydroxysteroid Dehydrogenases/drug effects , 3-Hydroxysteroid Dehydrogenases/genetics , Acid-Base Equilibrium , Adrenal Cortex/drug effects , Adrenocorticotropic Hormone/pharmacology , Animals , Blood Gas Analysis , Blood Pressure/drug effects , Cholesterol Side-Chain Cleavage Enzyme/drug effects , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Heart Rate, Fetal/drug effects , Hydrocortisone/blood , Hydrocortisone/immunology , Infusions, Intravenous , Pregnancy , RNA, Messenger/analysis , Receptors, Corticotropin/drug effects , Receptors, Corticotropin/metabolism , Reproducibility of Results , Sheep , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Steroid 21-Hydroxylase , Up-Regulation
19.
Urol Clin North Am ; 39(4): 453-64, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23084523

ABSTRACT

Androgen receptor (AR)-mediated signaling is critical to the growth and survival of prostate cancer. Although medical castration and antiandrogen therapy can decrease AR activity and lower PSA, castration resistance eventually develops. Recent work exploring the molecular structure and evolution of AR in response to hormonal therapies has revealed novel mechanisms of progression of castration-resistant prostate cancer and yielded new targets for drug development. This review focuses on understanding the mechanisms of persistent AR signaling in the castrate environment, and highlights new therapies either currently available or in clinical trials, including androgen synthesis inhibitors and novel direct AR inhibitors.


Subject(s)
Androgen Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Prostatic Neoplasms/drug therapy , Receptors, Androgen/drug effects , Androgen Antagonists/therapeutic use , Androstadienes/pharmacology , Androstadienes/therapeutic use , Antineoplastic Agents/therapeutic use , Benzamides , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Male , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/physiopathology , Receptors, Androgen/physiology , Steroid 17-alpha-Hydroxylase/drug effects , Testosterone/metabolism , Thiohydantoins/pharmacology , Thiohydantoins/therapeutic use
20.
Urol Clin North Am ; 39(4): 583-91, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23084533

ABSTRACT

The treatment of metastatic castration-resistant prostate cancer has evolved since the approval of docetaxel-based therapy. Since docetaxel approval, three new agents have gained approval for this indication: sipuleucel-T, cabazitaxel, and abiraterone. Recent Phase III trials have also demonstrated survival benefits for MDV-3100 and radium-223 though regulatory approval ispending. Practicing physicians face the challenge of determining the optimal sequencing of these new agents. This dilemma is particularly relevant to the post-docetaxel setting, in which the indication for several of these agents overlaps. This article details the efficacy and safety of these agents to provide a framework for their clinical use.


Subject(s)
Androstenols/therapeutic use , Antineoplastic Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Taxoids/therapeutic use , Androstenes , Androstenols/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzamides , Docetaxel , Humans , Male , Neoplasms, Hormone-Dependent , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/secondary , Radium/therapeutic use , Receptors, Androgen/drug effects , Steroid 17-alpha-Hydroxylase/drug effects , Taxoids/administration & dosage , Taxoids/chemistry , Treatment Failure
SELECTION OF CITATIONS
SEARCH DETAIL