Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
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 µM) 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 ± 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
2.
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
3.
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
4.
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-ß 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 3ß-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
5.
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 µg/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(®) 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(®) 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
6.
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 ß-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 µg/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 µg/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
7.
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
8.
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
9.
Toxicol Lett ; 192(3): 271-7, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19913079

ABSTRACT

Cytochrome P450c17 (CYP17) has been linked to various hormone-related diseases, including breast cancer, thus being a potential target for cancer chemoprevention. We studied the naturally occurring phytochemical enterolactone (ENL) and 13 VIOXX-related lactone derivatives (CRI-1 to CRI-13) for their effects on CYP17 activity and expression and on cell cycle status in the human H295R adrenocorticocarcinoma cell line. Of the tested compounds, only CRI-3, -7, -10 and -12 showed to be inhibitors of CYP17 activity in H295R cells. This inhibition was not due to decreased mRNA expression, but was apparently caused by post-translational modification of the CYP17 enzyme. The MAPK kinase (MEK) inhibitor PD98059 induced CYP17 activity by 24%, while co-incubation of the CRI-s with PD98059, reduced CYP17 activity even further than the reduction caused by the CRI-s alone. In addition, CRI-3, -7, -10 and -12 arrested the cell cycle in the G(2)/M phase. The structure-activity similarities of the CRI-s with known micro-tubule binding agents strongly suggest that cell cycle arrest is a result of interaction with tubulin. We conclude that the proposed cancer chemopreventive actions of ENL are not mediated through interaction with CYP17 or cell cycle status. Of the VIOXX-related lactone derivatives, CRI-7 could prove useful in the prevention of hormone-dependent cancers, such as breast cancer, since in vitro it shows low cytotoxicity, it is a potent inhibitor of CYP17 activity and strong inducer of cell cycle arrest.


Subject(s)
4-Butyrolactone/analogs & derivatives , Adrenal Cortex Neoplasms/enzymology , Adrenocortical Carcinoma/enzymology , Lactones/pharmacology , Lignans/pharmacology , Phytoestrogens/pharmacology , Steroid 17-alpha-Hydroxylase/drug effects , Sulfones/pharmacology , 4-Butyrolactone/pharmacology , Adrenal Cortex Neoplasms/physiopathology , Adrenocortical Carcinoma/physiopathology , Cell Cycle/drug effects , Cell Line, Tumor , Enzyme Induction/drug effects , Flavonoids/pharmacology , Gene Expression/drug effects , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplasms, Hormone-Dependent/prevention & control , Protein Processing, Post-Translational/drug effects , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/biosynthesis , Structure-Activity Relationship
10.
Fertil Steril ; 91(5 Suppl): 1990-7, 2009 May.
Article in English | MEDLINE | ID: mdl-18672234

ABSTRACT

OBJECTIVE: To investigate whether insulin resistance (IR) within theca cells may directly contribute to their hyperandrogenism, a heritable trait of polycystic ovary syndrome (PCOS). DESIGN: In vitro cell model. SETTING: University-affiliated laboratory. ANIMAL(S): Porcine ovaries. INTERVENTION(S): Ovarian theca cells from porcine follicles were isolated and cultured. Insulin resistance was induced in theca cells without (Con) or with dexamethasone (Dex); cells were further treated by troglitazone (Tro) and metformin (Met) in IR cells or by vehicle only in IR and Con cells. MAIN OUTCOME MEASURE(S): Medium glucose and T levels; reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot for insulin signal molecules and androgenic enzyme. RESULT(S): As compared with Con cells, Dex-treated cells had significantly lower [(3)H]-glucose uptake (565 +/- 58 cpm/10(6) vs. 1077 +/- 78 cpm/10(6)) but higher medium glucose levels (16.31 +/- 0.39 nmol/L vs. 10.62 +/- 1.02 nmol/L) and had approximately twofold T levels (0.82 +/- 0.20 microg/L vs. 0.38+/-0.08 microg/L). Troglitazone and Met significantly reduced the medium glucose and testosterone concentrations to levels comparable to those in Con cells. The RT-PCR and Western blot showed that the two sensitizers in different ways reversed the altered messenger RNA and protein expression of insulin receptor substrate-1, glucose transporter-4, peroxisome proliferator-activated receptor-gamma, and 17 alpha-hydroxylase in Dex-induced IR cells. CONCLUSION(S): Insulin resistance induced by Dex could directly exaggerate androgenic potential within theca cells, suggesting the possible involvement of this ovarian metabolic phenotype in PCOS hyperandrogenism.


Subject(s)
Androgens/pharmacology , Chromans/pharmacology , Glucose Transporter Type 4/genetics , Insulin Resistance/physiology , Metformin/pharmacology , Theca Cells/physiology , Thiazolidinediones/pharmacology , Animals , Cell Culture Techniques/methods , Female , Glucose Transporter Type 4/drug effects , Insulin Receptor Substrate Proteins/drug effects , Insulin Receptor Substrate Proteins/genetics , PPAR gamma/drug effects , PPAR gamma/genetics , RNA, Messenger/drug effects , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Swine , Theca Cells/cytology , Theca Cells/drug effects , Troglitazone
11.
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
12.
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
13.
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
14.
Toxicol Appl Pharmacol ; 225(2): 142-53, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17822730

ABSTRACT

The H295R cell bioassay was used to evaluate the potential endocrine disrupting effects of 18 of the most commonly used pharmaceuticals in the United States. Exposures for 48 h with single pharmaceuticals and binary mixtures were conducted; the expression of five steroidogenic genes, 3betaHSD2, CYP11beta1, CYP11beta2, CYP17 and CYP19, was quantified by Q-RT-PCR. Production of the steroid hormones estradiol (E2), testosterone (T) and progesterone (P) was also evaluated. Antibiotics were shown to modulate gene expression and hormone production. Amoxicillin up-regulated the expression of CYP11beta2 and CYP19 by more than 2-fold and induced estradiol production up to almost 3-fold. Erythromycin significantly increased CYP11beta2 expression and the production of P and E2 by 3.5- and 2.4-fold, respectively, while production of T was significantly decreased. The beta-blocker salbutamol caused the greatest induction of CYP17, more than 13-fold, and significantly decreased E2 production. The binary mixture of cyproterone and salbutamol significantly down-regulated expression of CYP19, while a mixture of ethynylestradiol and trenbolone, increased E2 production 3.7-fold. Estradiol production was significantly affected by changes in concentrations of trenbolone, cyproterone, and ethynylestradiol. Exposures with individual pharmaceuticals showed the possible secondary effects that drugs may exert on steroid production. Results from binary mixture exposures suggested the possible type of interactions that may occur between drugs and the joint effects product of such interactions. Dose-response results indicated that although two chemicals may share a common mechanism of action the concentration effects observed may be significantly different.


Subject(s)
Adrenocortical Carcinoma/metabolism , Cytochrome P-450 Enzyme System/drug effects , Endocrine Disruptors/pharmacology , Gene Expression Regulation/drug effects , Gonadal Steroid Hormones/biosynthesis , 3-Hydroxysteroid Dehydrogenases/drug effects , 3-Hydroxysteroid Dehydrogenases/genetics , Aromatase/drug effects , Aromatase/genetics , Cell Line, Tumor , Cytochrome P-450 CYP11B2/drug effects , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 Enzyme System/genetics , Dose-Response Relationship, Drug , Drug Interactions , Estradiol/biosynthesis , Humans , Progesterone/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Steroid 11-beta-Hydroxylase/drug effects , Steroid 11-beta-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Testosterone/biosynthesis
15.
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
16.
Biol Reprod ; 73(5): 908-17, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15987825

ABSTRACT

The phthalate ester di(n-butyl) phthalate (DBP) causes feminization of male rats upon in utero exposure by repressing expression of genes required for testicular steroidogenesis. Previous work in our laboratory has shown that repression of gene expression and steroidogenesis in the fetal testis is apparent within a few hours of DBP exposure. The purpose of this study was to determine the precise timing of DBP-associated gene expression changes in the fetal testis using transcriptional profiling and to determine whether DBP exerts similar effects on steroidogenesis in the fetal adrenal. A DBP time-course experiment showed that testicular steroidogenesis was decreased within 1 h of DBP exposure and that this decrease preceded the repressed transcription of Star (steroidogenic acute regulatory protein); Scarb1 (scavenger receptor class B, member 1; also know as Sr-b1); Cyp11a1 (cytochrome P450, family 11, subfamily a, polypeptide 1; also known as P450SCC); and Cyp17a1 (cytochrome P450 family 17, subfamily a, polypeptide 1; also known as Cyp17). Gene expression profiling demonstrated rapid (within 1 to 3 h) and transient induction of immediate early genes in the fetal testis after administration of DBP to the pregnant dam. There was a statistically insignificant decrease in corticosterone production by the fetal adrenal after in utero exposure to DBP from Gestation Day 12 to Gestation Day 19. The extent of steroidogenesis diminution was much less in the adrenal than in the testis (approximately 45% decrease in the adrenal versus 87% decrease in the testis) and expression of genes required for steroidogenesis in the adrenal was unaffected by DBP. Together, these studies demonstrate that DBP initiates a rapid and dynamic change in gene expression in the fetal testis that likely plays a role in the reduction in steroidogenesis that is unique to the fetal testis relative to the steroidogenically active fetal adrenal.


Subject(s)
Adrenal Glands/embryology , Adrenal Glands/metabolism , Dibutyl Phthalate/adverse effects , Gene Expression Regulation, Developmental/drug effects , Testis/embryology , Testis/metabolism , Adrenal Glands/drug effects , Animals , Cholesterol Side-Chain Cleavage Enzyme/drug effects , Cholesterol Side-Chain Cleavage Enzyme/genetics , Corticosterone/metabolism , Female , Male , Mice , Organ Specificity , Phosphoproteins/drug effects , Phosphoproteins/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Scavenger Receptors, Class B/drug effects , Scavenger Receptors, Class B/genetics , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/genetics , Testis/drug effects , Testosterone/metabolism
17.
Epilepsia ; 46(3): 444-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15730543

ABSTRACT

PURPOSE: Women with epilepsy apparently have a higher incidence of polycystic ovary syndrome (PCOS) than do women without epilepsy. Whether the underlying disease or the antiepileptic drug (AED) treatment is responsible for this increased risk is unknown, although clinical reports implicate valproic acid (VPA) as a potential cause. The steroidogenic enzymes 3beta HSDII (3beta-hydroxysteroid dehydrogenase) and P450c17 (17alpha-hydroxylase/17,20 lyase) are essential for C19 steroid biosynthesis, which is enhanced during adrenarche and in PCOS. METHODS: To determine whether the AEDs VPA, carbamazepine (CBZ), topiramate (TPM), or lamotrigine (LYG) directly affect the activities of human 3beta HSDII and P450c17, we added them to yeast expressing human P450c17 or 3beta HSDII and assayed enzymatic activities in the microsomal fraction. RESULTS: Concentrations of VPA < or = 10 mM had no effect on activities of P450c17; however, VPA inhibited 3beta HSDII activity starting at 0.3 mM (reference serum unbound concentration, 0.035-0.1 mM) with an IC50 of 10.1 mM. CBZ, TPM, and LTG did not influence 3beta HSDII or P450c17 activities at typical reference serum unbound concentrations, but did inhibit 3beta HSDII and P450c17 at concentrations >10-fold higher. CONCLUSIONS: None of the tested AEDs influenced 3beta HSDII or P450c17 activities at concentrations normally used in AED therapy. However, VPA started to inhibit 3beta HSDII activity at concentrations 3 times above the typical reference serum unbound concentration. Because inhibition of 3beta HSDII activity will shift steroidogenesis toward C19 steroid production when P450c17 activities are unchanged, very high doses of VPA may promote C19 steroid biosynthesis, thus resembling PCOS. CBZ, TPM, and LTG influenced 3beta HSDII and P450c17 only at toxic concentrations.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Anticonvulsants/pharmacokinetics , Fructose/analogs & derivatives , Steroid 17-alpha-Hydroxylase/metabolism , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/drug effects , Adrenal Cortex Hormones/biosynthesis , Androgens/biosynthesis , Carbamazepine/pharmacokinetics , Female , Fructose/pharmacokinetics , Humans , Kinetics , Lamotrigine , Microsomes/enzymology , Mineralocorticoids/biosynthesis , Polycystic Ovary Syndrome/enzymology , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/drug effects , Substrate Specificity , Topiramate , Triazines/pharmacokinetics , Valproic Acid/pharmacokinetics
18.
J Steroid Biochem Mol Biol ; 92(3): 199-208, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15555913

ABSTRACT

In addition to causing Müllerian duct regression in fetal males, Müllerian 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
19.
Mol Carcinog ; 34(3): 151-63, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12112309

ABSTRACT

We previously observed that the serum testosterone level was greatly reduced in the course of diethylnitrosamine-nodularin-induced hepatocarcinogenesis in Fischer 344 male rats (Lim et al., Gastroenterological Carcinogenesis, 1999). As an extension of this observation, this study was undertaken to investigate the molecular mechanism of downregulation of testosterone and its effect on target organs in Fischer 344 male rats treated with the hepatotoxin nodularin. After treating the rats with nodularin, a marked reduction of the testosterone level was noted in both serum and testis, with an accompanying accumulation of cholesterol in serum. Reduction of serum testosterone was not due to increased degradation of testosterone in the liver but to impaired biosynthesis in the testes, reduced activities of the cholesterol side chain cleavage enzyme and 17alpha-hydroxylase, and decreased expression of the steroidogenic acute regulatory protein gene, all of which constitute rate-limiting steps for testosterone biosynthesis in the testes. Intraperitoneal injection of nodularin into rats induced cuboidal changes of glandular epithelium in ventral prostates and apoptotic changes of spermatogonium, for example, nuclear chromatin condensation, shrinkage, and detachment from Sertoli cells, which included many lysosomal granules. Leydig cells also showed evidence of chromatin condensation and significant induction of peroxisome proliferation. In conclusion, the potential causes of impaired testosterone biosynthesis might have been apoptosis of Leydig cells induced by direct toxicity of the hepatotoxin on testes or hypothalamopituitary dysfunction.


Subject(s)
Apoptosis/drug effects , Bacterial Toxins/pharmacology , Leydig Cells/metabolism , Peptides, Cyclic/pharmacology , Testosterone/biosynthesis , Animals , Cholesterol/blood , Diethylnitrosamine/pharmacology , Leydig Cells/drug effects , Leydig Cells/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Luteinizing Hormone/blood , Male , Prostate/drug effects , Prostate/pathology , Rats , Rats, Inbred F344 , Steroid 17-alpha-Hydroxylase/drug effects , Steroid 17-alpha-Hydroxylase/metabolism , Testis/drug effects , Testis/metabolism , Testosterone/blood
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...