Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
J Steroid Biochem Mol Biol ; 183: 192-201, 2018 10.
Article in English | MEDLINE | ID: mdl-29936123

ABSTRACT

The progression of castration resistant prostate cancer (CRPC) is driven by the intratumoral conversion of adrenal androgen precursors to potent androgens. The expression of aldo-keto reductase 1C3 (AKR1C3), which catalyses the reduction of weak androgens to more potent androgens, is significantly increased in CRPC tumours. The oxidation of androgens to their inactive form is catalysed by 17ß-hydroxysteroid dehydrogenase type 2 (17ßHSD2), but little attention is given to the expression levels of this enzyme. In this study, we show that the 11-oxygenated androgen precursors of adrenal origin are the preferred substrate for AKR1C3. In particular we show that the enzymatic efficiency of AKR1C3 is 8- and 24-fold greater for 11-ketoandrostenedione than for the classic substrates androstenedione and 5α-androstanedione, respectively. Using three independent experimental systems and a computational model we subsequently show that increased ratios of AKR1C3:17ßHSD2 significantly favours the flux through the 11-oxygenated androgen pathway as compared to the classical or 5α-androstanedione pathways. Our findings reveal that the flux through the classical and 5α-androstanedione pathways are limited by the low catalytic efficiently of AKR1C3 towards classical androgens combined with the high catalytic efficiency of 17ßHSD2, and that the expression of the oxidative enzyme therefore plays a vital role in determining the steady state concentration of active androgens. Using microarray data from prostate tissue we confirm that the AKR1C3:17ßHSD2 ratio is significantly increased in patients undergoing androgen deprivation therapy as compared to benign tissue, and further increased in patients with CRPC. Taken together this study therefore demonstrates that the ratio of AKR1C3:17ßHSD2 is more important than AKR1C3 expression alone in determining intratumoral androgen levels and that 11-oxygenated androgens may play a bigger role in CRPC than previously anticipated.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/metabolism , Androgens/chemistry , Androgens/metabolism , Computational Biology/methods , Estradiol Dehydrogenases/metabolism , Oxygen/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Humans , Male , Prostatic Neoplasms, Castration-Resistant/pathology , Steroids/metabolism , Tumor Cells, Cultured
2.
Article in English | MEDLINE | ID: mdl-29627638

ABSTRACT

Liquid chromatography tandem mass spectrometry (LC-MS/MS) assays are considered the reference standard for serum steroid hormone analyses, while full urinary steroid profiles are only achievable by gas chromatography (GC-MS). Both LC-MS/MS and GC-MS have well documented strengths and limitations. Recently, commercial ultra-high performance supercritical fluid chromatography-tandem mass spectrometry (UHPSFC-MS/MS) systems have been developed. These systems combine the resolution of GC with the high-throughput capabilities of UHPLC. Uptake of this new technology into research and clinical labs has been slow, possibly due to the perceived increase in complexity. Here we therefore present fundamental principles of UHPSFC-MS/MS and the likely applications for this technology in the clinical research setting, while commenting on potential hurdles based on our experience to date.


Subject(s)
Chromatography, Supercritical Fluid/methods , Steroids , Gas Chromatography-Mass Spectrometry , Humans , Hydrophobic and Hydrophilic Interactions , Steroids/blood , Steroids/chemistry , Tandem Mass Spectrometry/methods
3.
Endocrinology ; 158(12): 4165-4173, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28938470

ABSTRACT

Congenital adrenal hyperplasia is a group of common inherited disorders leading to glucocorticoid deficiency. Most cases are caused by 21-hydroxylase deficiency (21OHD). The systemic consequences of imbalanced steroid hormone biosynthesis due to severe 21OHD remains poorly understood. Therefore, we developed a zebrafish model for 21OHD, which focuses on the impairment of glucocorticoid biosynthesis. A single 21-hydroxylase gene (cyp21a2) is annotated in the zebrafish genome based on sequence homology. Our in silico analysis of the 21-hydroxylase (Cyp21a2) protein sequence suggests a sufficient degree of similarity for the usage of zebrafish cyp21a2 to model aspects of human 21OHD in vivo. We determined the spatiotemporal expression patterns of cyp21a2 by whole-mount in situ hybridization and reverse transcription polymerase chain reaction throughout early development. Early cyp21a2 expression is restricted to the interrenal gland (zebrafish adrenal counterpart) and the brain. To further explore the in vivo consequences of 21OHD we created several cyp21a2 null-allele zebrafish lines by using a transcription activator-like effector nuclease genomic engineering strategy. Homozygous mutant zebrafish larvae showed an upregulation of the hypothalamic-pituitary-interrenal (HPI) axis and interrenal hyperplasia. Furthermore, Cyp21a2-deficient larvae had a typical steroid profile, with reduced concentrations of cortisol and increased concentrations of 17-hydroxyprogesterone and 21-deoxycortisol. Affected larvae showed an upregulation of the HPI axis and interrenal hyperplasia. Downregulation of the glucocorticoid-responsive genes pck1 and fkbp5 indicated systemic glucocorticoid deficiency. Our work demonstrates the crucial role of Cyp21a2 in glucocorticoid biosynthesis in zebrafish larvae and establishes an in vivo model allowing studies of systemic consequences of altered steroid hormone synthesis.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Interrenal Gland/metabolism , Steroid 21-Hydroxylase/genetics , Zebrafish Proteins/genetics , Adrenal Hyperplasia, Congenital/embryology , Adrenal Hyperplasia, Congenital/enzymology , Animals , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/metabolism , Fish Diseases/embryology , Fish Diseases/enzymology , Fish Diseases/genetics , Gene Expression Regulation, Developmental , Glucocorticoids/biosynthesis , Hyperplasia/enzymology , Hyperplasia/genetics , In Situ Hybridization , Interrenal Gland/embryology , Interrenal Gland/pathology , Larva/enzymology , Larva/genetics , Larva/metabolism , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Steroid 21-Hydroxylase/metabolism , Zebrafish , Zebrafish Proteins/metabolism
4.
J Steroid Biochem Mol Biol ; 166: 54-67, 2017 02.
Article in English | MEDLINE | ID: mdl-27345701

ABSTRACT

Adrenal C19 steroids serve as precursors to active androgens in the prostate. Androstenedione (A4), 11ß-hydroxyandrostenedione (11OHA4) and 11ß-hydroxytestosterone (11OHT) are metabolised to potent androgen receptor (AR) agonists, dihydrotestosterone (DHT), 11-ketotestosterone (11KT) and 11-ketodihydrotestosterone (11KDHT). The identification of 11OHA4 metabolites, 11KT and 11KDHT, as active androgens has placed a new perspective on adrenal C11-oxy C19 steroids and their contribution to prostate cancer (PCa). We investigated adrenal androgen metabolism in normal epithelial prostate (PNT2) cells and in androgen-dependent prostate cancer (LNCaP) cells. We also analysed steroid profiles in PCa tissue and plasma, determining the presence of the C19 steroids and their derivatives using ultra-performance liquid chromatography (UHPLC)- and ultra-performance convergence chromatography tandem mass spectrometry (UPC2-MS/MS). In PNT2 cells, sixty percent A4 (60%) was primarily metabolised to 5α-androstanedione (5αDIONE) (40%), testosterone (T) (10%), and androsterone (AST) (10%). T (30%) was primarily metabolised to DHT (10%) while low levels of A4, 5αDIONE and 3αADIOL (≈20%) were detected. Conjugated steroids were not detected and downstream products were present at <0.05µM. Only 20% of 11OHA4 and 11OHT were metabolised with the former yielding 11keto-androstenedione (11KA4), 11KDHT and 11ß-hydroxy-5α-androstanedione (11OH-5αDIONE) and the latter yielding 11OHA4, 11KT and 11KDHT with downstream products <0.03µM. In LNCaP cells, A4 (90%) was metabolised to AST-glucuronide via the alternative pathway while T was detected as T-glucuronide with negligible conversion to downstream products. 11OHA4 (80%) and 11OHT (60%) were predominantly metabolised to 11KA4 and 11KT and in both assays more than 50% of 11KT was detected in the unconjugated form. In tissue, we detected C11-oxy C19 metabolites at significantly higher levels than the C19 steroids, with unconjugated 11KDHT, 11KT and 11OHA4 levels ranging between 13 and 37.5ng/g. Analyses of total steroid levels in plasma showed significant levels of 11OHA4 (≈230-440nM), 11KT (≈250-390nM) and 11KDHT (≈19nM). DHT levels (<0.14nM) were significantly lower. In summary, 11ß-hydroxysteroid dehydrogenase type 2 activity in PNT2 cells was substantially lower than in LNCaP cells, reflected in the conversion of 11OHA4 and 11OHT. Enzyme substrate preferences suggest that the alternate pathway is dominant in normal prostate cells. Glucuronidation activity was not detected in PNT2 cells and while all T derivatives were efficiently conjugated in LNCaP cells, 11KT was not. Substantial 11KT levels were also detected in both PCa tissue and plasma. 11OHA4 therefore presents a significant androgen precursor and its downstream metabolism to 11KT and 11KDHT as well as its presence in PCa tissue and plasma substantiate the importance of this adrenal androgen.


Subject(s)
Adrenal Glands/metabolism , Androstenedione/analogs & derivatives , Prostatic Neoplasms/metabolism , Testosterone/analogs & derivatives , Aged , Androgens/metabolism , Androstenedione/metabolism , Cell Line, Tumor , Dihydrotestosterone/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Glucuronic Acid/chemistry , Humans , Hydroxytestosterones/metabolism , Male , Steroids/chemistry , Steroids/metabolism , Tandem Mass Spectrometry , Testosterone/metabolism
5.
J Clin Endocrinol Metab ; 102(3): 840-848, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27901631

ABSTRACT

Context: Androgen excess is a defining feature of polycystic ovary syndrome (PCOS), but the exact origin of hyperandrogenemia remains a matter of debate. Recent studies have highlighted the importance of the 11-oxygenated C19 steroid pathway to androgen metabolism in humans. In this study, we analyzed the contribution of 11-oxygenated androgens to androgen excess in women with PCOS. Methods: One hundred fourteen women with PCOS and 49 healthy control subjects underwent measurement of serum androgens by liquid chromatography-tandem mass spectrometry. Twenty-four-hour urinary androgen excretion was analyzed by gas chromatography-mass spectrometry. Fasting plasma insulin and glucose were measured for homeostatic model assessment of insulin resistance. Baseline demographic data, including body mass index, were recorded. Results: As expected, serum concentrations of the classic androgens testosterone (P < 0.001), androstenedione (P < 0.001), and dehydroepiandrosterone (P < 0.01) were significantly increased in PCOS. Mirroring this, serum 11-oxygenated androgens 11ß-hydroxyandrostenedione, 11-ketoandrostenedione, 11ß-hydroxytestosterone, and 11-ketotestosterone were significantly higher in PCOS than in control subjects, as was the urinary 11-oxygenated androgen metabolite 11ß-hydroxyandrosterone. The proportionate contribution of 11-oxygenated to total serum androgens was significantly higher in patients with PCOS compared with control subjects [53.0% (interquartile range, 48.7 to 60.3) vs 44.0% (interquartile range, 32.9 to 54.9); P < 0.0001]. Obese (n = 51) and nonobese (n = 63) patients with PCOS had significantly increased 11-oxygenated androgens. Serum 11ß-hydroxyandrostenedione and 11-ketoandrostenedione correlated significantly with markers of insulin resistance. Conclusions: We show that 11-oxygenated androgens represent the majority of circulating androgens in women with PCOS, with close correlation to markers of metabolic risk.


Subject(s)
Androgens/metabolism , Blood Glucose/metabolism , Hyperandrogenism/metabolism , Insulin/metabolism , Obesity/metabolism , Polycystic Ovary Syndrome/metabolism , Adult , Androstenedione/analogs & derivatives , Androstenedione/metabolism , Androstenes/metabolism , Case-Control Studies , Chromatography, Liquid , Dehydroepiandrosterone/metabolism , Female , Gas Chromatography-Mass Spectrometry , Humans , Hydroxytestosterones/metabolism , Hyperandrogenism/complications , Insulin Resistance , Obesity/complications , Polycystic Ovary Syndrome/complications , Tandem Mass Spectrometry , Testosterone/analogs & derivatives , Testosterone/metabolism , Young Adult
6.
Article in English | MEDLINE | ID: mdl-27479683

ABSTRACT

11-Oxygenated steroids such as 11-ketotestosterone and 11-ketodihydrotestosterone have recently been shown to play a putative role in the development and progression of castration resistant prostate cancer. In this study we report on the development of a high throughput ultra-performance convergence chromatography tandem mass spectrometry (UPC(2)-MS/MS) method for the analysis of thirteen 11-oxygenated and six canonical C19 steroids isolated from a cell culture matrix. Using an Acquity UPC(2) BEH 2-EP column we found that UPC(2) resulted in superior selectivity, increased chromatographic efficiency and a scattered elution order when compared to conventional reverse phase ultra-performance liquid chromatography (UPLC). Furthermore, there was a significant improvement in sensitivity (5-50 times). The lower limits of quantification ranged between 0.01-10ngmL(-1), while the upper limit of quantification was 100ngmL(-1) for all steroids. Accuracy, precision, intra-day variation, recovery, matrix effects and process efficiency were all evaluated and found to be within acceptable limits. Taken together we show that the increased power of UPC(2)-MS/MS allows the analyst to complete in vitro assays at biologically relevant concentrations for the first time and in so doing determine the routes of steroid metabolism which is vital for studies of androgen responsive cancers, such as prostate cancer, and could highlight new mechanisms of disease progression and new targets for cancer therapy.


Subject(s)
Androgens/analysis , Chromatography, Liquid/methods , Chromatography, Supercritical Fluid/methods , High-Throughput Screening Assays , Tandem Mass Spectrometry/methods , Humans , Reference Standards , Reproducibility of Results
7.
J Steroid Biochem Mol Biol ; 138: 132-42, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23685396

ABSTRACT

11ß-Hydroxyandrostenedione (11OHA4), which is unique to the adrenal, was first isolated from human adrenal tissue in the fifties. It was later shown in the sixties that 11ß-hydroxytestosterone (11OHT) was also produced by the human adrenal. Attention has shifted back to these adrenal androgens once more, as improved analytical techniques have enabled more accurate detection of steroid hormones. In this paper, we investigated the origin of these metabolites as well as their subsequent metabolism and examined a possible physiological role for 11OHA4 in prostate cancer cells. In H295R cells treated with forskolin and trilostane, etomidate, a reported cytochrome P450 11ß-hydroxylase (CYP11B1) inhibitor, blocked the production of corticosterone, cortisol, 11OHA4 and 11OHT. The metabolism of androstenedione and testosterone by CYP11B1 and aldosterone synthase (CYP11B2) was assayed. Androstenedione was converted by CYP11B1, while the conversion by CYP11B2 was negligible. Both enzymes readily converted testosterone. The metabolism of these 11ß-hydroxylated metabolites by 11ß-hydroxysteroid dehydrogenase (11ßHSD) types 1 and 2 was subsequently investigated. 11ßHSD2 catalyzed the conversion of both 11OHA4 and 11OHT to their respective keto-steroids, while 11ßHSD1 catalyzed the conversion of 11-ketoandrostenedione and 11-ketotestosterone to their respective hydroxy-steroids in Chinese hamster ovary cells. Investigating a functional role, steroid 5α-reductase types 1 and 2 converted 11OHA4 to 11ß-hydroxy-5α-androstanedione (11OH-5α-dione), identified by accurate mass detection. UPLC-MS/MS analyses of 11OHA4 metabolism in LNCaP androgen-dependent prostate cancer cells, identified the 5α-reduced metabolite as well as 11-ketoandrostenedione and 11-ketotestosterone, with the latter indicating conversion by 17ß-hydroxysteroid dehydrogenase. Downstream metabolism by 11ßHSD2 and by 5α-reductase may therefore indicate a physiological role for 11OHA4 and/or 11OH-5α-dione in normal and prostate cancer cells.


Subject(s)
Adrenal Glands/metabolism , Androstenedione/analogs & derivatives , Androstenedione/metabolism , Cholestenone 5 alpha-Reductase/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Humans , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL