Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(1)2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-38203272

RESUMO

C11-oxy C19 and C11-oxy C21 steroids have been identified as novel steroids but their function remains unclear. This study aimed to investigate the pre-receptor regulation of C11-oxy steroids by 11ß-hydroxysteroid dehydrogenase (11ßHSD) interconversion and potential agonist and antagonist activity associated with the androgen (AR) and progesterone receptors (PRA and PRB). Steroid conversions were investigated in transiently transfected HEK293 cells expressing 11ßHSD1 and 11ßHSD2, while CV1 cells were utilised for agonist and antagonist assays. The conversion of C11-hydroxy steroids to C11-oxo steroids by 11ßHSD2 occurred more readily than the reverse reaction catalysed by 11ßHSD1, while the interconversion of C11-oxy C19 steroids was more efficient than C11-oxy C21 steroids. Furthermore, 11-ketodihydrotestosterone (11KDHT), 11-ketotestosterone (11KT) and 11ß-hydroxydihydrotestosterone (11OHDHT) were AR agonists, while only progestogens, 11ß-hydroxyprogesterone (11ßOHP4), 11ß-hydroxydihydroprogesterone (11ßOHDHP4), 11α-hydroxyprogesterone (11αOHP4), 11α-hydroxydihydroprogesterone (11αOHDHP4), 11-ketoprogesterone (11KP4), 5α-pregnan-17α-diol-3,11,20-trione (11KPdione) and 21-deoxycortisone (21dE) exhibited antagonist activity. C11-hydroxy C21 steroids, 11ßOHP4, 11ßOHDHP4 and 11αOHP4 exhibited PRA and PRB agonistic activity, while only C11-oxo steroids, 11KP4 and 11-ketoandrostanediol (11K3αdiol) demonstrated PRB agonism. While no steroids antagonised the PRA, 11OHA4, 11ß-hydroxytestosterone (11OHT), 11KT and 11KDHT exhibited PRB antagonism. The regulatory role of 11ßHSD isozymes impacting receptor activation is clear-C11-oxo androgens exhibit AR agonist activity; only C11-hydroxy progestogens exhibit PRA and PRB agonist activity. Regulation by the downstream metabolites of active C11-oxy steroids at the receptor level is apparent-C11-hydroxy and C11-oxo metabolites antagonize the AR and PRB, progestogens the former, androgens the latter. The findings highlight the intricate interplay between receptors and active as well as "inactive" C11-oxy steroids, suggesting novel regulatory tiers.


Assuntos
Progesterona , Receptores de Esteroides , Humanos , Receptores de Progesterona , Androgênios , Progestinas , Células HEK293 , Esteroides , 11-beta-Hidroxiesteroide Desidrogenases
2.
Mol Cell Endocrinol ; 525: 111189, 2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33539964

RESUMO

Adrenal steroidogenesis has, for decades, been depicted as three biosynthesis pathways -the mineralocorticoid, glucocorticoid and androgen pathways with aldosterone, cortisol and androstenedione as the respective end products. 11ß-hydroxyandrostenedione was not included as an adrenal steroid despite the adrenal output of this steroid being twice that of androstenedione. While it is the end of the line for aldosterone and cortisol, as it is in these forms that they exhibit their most potent receptor activities prior to inactivation and conjugation, 11ß-hydroxyandrostenedione is another matter entirely. The steroid, which is weakly androgenic, has its own designated pathway yielding 11-ketoandrostenedione, 11ß-hydroxytestosterone and the potent androgens, 11-ketotestosterone and 11-ketodihydrotestosterone, primarily in the periphery. Over the last decade, these C11-oxy C19 steroids have once again come to the fore with the rising number of studies contradicting the generally accepted notion that testosterone and it's 5α-reduced product, dihydrotestosterone, are the principal potent androgens in humans. These C11-oxy androgens have been shown to contribute to the androgen milieu in adrenal disorders associated with androgen excess and in androgen dependant disease progression. In this review, we will highlight these overlooked C11-oxy C19 steroids as well as the C11-oxy C21 steroids and their contribution to congenital adrenal hyperplasia, polycystic ovarian syndrome and prostate cancer. The focus is on new findings over the past decade which are slowly but surely reshaping our current outlook on human sex steroid biology.


Assuntos
Androgênios/metabolismo , Androstenodiona/análogos & derivados , Esteroides/biossíntese , Androstenodiona/química , Androstenodiona/metabolismo , Animais , Doença , Humanos , Esteroides/química
3.
Pediatr Res ; 89(1): 118-126, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32247282

RESUMO

BACKGROUND: We recently identified 35 women with polycystic ovarian syndrome (PCOS) who exhibited features of micronodular adrenocortical hyperplasia. Steroid hormone analysis can be more accurate using state-of-the-art ultra-performance convergence chromatography-tandem mass spectrometry (UPC2-MS/MS). We hypothesized that UPC2-MS/MS may be used to better define hormonally this distinct subgroup of patients with PCOS. METHODS: Plasma from PCOS patients (n = 35) and healthy volunteers (HVs, n = 19) who all received dexamethasone testing was analyzed. Samples were grouped per dexamethasone responses and followed by UPC2-MS/MS analysis. When insufficient, samples were pooled from patients with similar responses to allow quantification over the low end of the assay. RESULTS: The C11-oxy C19 (11ß-hydroxyandrostenedione, 11keto-androstenedione, 11ß-hydroxytestosterone, 11keto-testosterone):C19 (androstenedione, testosterone) steroid ratio was decreased by 1.75-fold in PCOS patients compared to HVs. Downstream steroid metabolites 11ß-hydroxyandrosterone and 11keto-androsterone were also measurable. The C11-oxy C21 steroids, 11-hydroxyprogesterone and 11keto-dihydroprogesterone levels, were 1.2- and 1.7-fold higher in PCOS patients compared to HVs, respectively. CONCLUSIONS: We hypothesized that UPC2-MS/MS may accurately quantify steroids, in vivo, and identify novel metabolites in a subgroup of patients with PCOS and adrenal abnormalities. Indeed, it appears that adrenal C11-oxy steroids have the potential of being used diagnostically to identify younger women and adolescents with PCOS who also have some evidence of micronodular adrenocortical hyperplasia. IMPACT: Adrenal C11-oxy steroids may be clinically important in identifying young patients with PCOS and adrenal abnormalities. The steroids presented in our manuscript have not yet been considered in the clinical setting so far, and we believe that this study could represent a first focused step towards the characterization of a distinct subgroup of women with PCOS who may in fact be treated differently than the average patient with PCOS. This paper can change the understanding of PCOS as one disorder: it is in fact a heterogeneous condition. In addition, for the subgroup of patients with PCOS associated with adrenocortical dysfunction, our paper provides novel hormonal markers that can be used diagnostically. Finally, the paper also adds to the basic pathophysiological understanding of adrenocortical-ovarian interactions in steroidogenesis of young women and adolescent girls with PCOS.


Assuntos
Córtex Suprarrenal/metabolismo , Cromatografia Líquida , Hiperandrogenismo/sangue , Síndrome do Ovário Policístico/sangue , Esteroides/sangue , Espectrometria de Massas em Tandem , Adolescente , Córtex Suprarrenal/fisiopatologia , Adulto , Biomarcadores/sangue , Estudos de Casos e Controles , Feminino , Humanos , Hiperandrogenismo/diagnóstico , Hiperandrogenismo/fisiopatologia , Síndrome do Ovário Policístico/diagnóstico , Síndrome do Ovário Policístico/fisiopatologia , Estudos Prospectivos , Adulto Jovem
4.
Artigo em Inglês | MEDLINE | ID: mdl-32593151

RESUMO

The C11-oxy androgens have been implicated in the progression of many diseases and endocrine-linked disorders, such as polycystic ovarian syndrome (PCOS), congenital adrenal hyperplasia, specifically 21-hydroxylase deficiency (21OHD), castration resistant prostate cancer (CRPC), as well as premature adrenarche. While the C11-oxy C19 steroids have been firmly established in the steroid arena, the C11-oxy C21 steroids are now also of significance. The current study reports on a high-throughput ultra-performance convergence chromatography tandem mass spectrometry (UPC2-MS/MS) method for the separation and quantification of 52 steroids in peripheral serum, which include the C11-oxy C19 and C11-oxy C21 steroids. Fifteen deuterium-labelled steroids were included for absolute quantification, which incorporates steroid extraction efficiency, together with one steroid and four non-steroidal compounds serving as quality controls (QC). The 15 min run-time per sample (16 min injection-to-injection time with an 8-step gradient) quantifies 68 analytes in a 2 µL injection volume. A single chromatographic step simultaneously identifies steroids in the mineralocorticoid, glucocorticoid and androgen pathways in adrenal steroidogenesis, together with steroid metabolites produced in the periphery, presenting an analytical method for the application of screening in vivo clinical samples. This study highlights cross-talk between the C11-oxy steroids, and describes the optimisation of multiple reaction monitoring required to measure steroids accurately. The limit of detection for the steroid metabolites ranged from 0.002 to 20 ng/mL and the limit of quantification from 0.02 to 100 ng/mL. The calibration range for the steroids ranged from 0.002 to 1000 ng/mL and for the QC compounds from 0.075 to 750 ng/mL. The method is fully validated in terms of accuracy (%RSD, <13%), precision (including inter-day variability across a three-day period) (%RSD, <16%), recovery (average 102.42%), matrix effect (ranging from -15.25 to 14.25%) and process efficiency (average 101.79%). The dilution protocol for the steroids, internal standards and QC compounds were validated, while the ion ratios of the steroid metabolites (%RSD, <16%) and QC compounds were monitored and the accuracy bias values (%RSD, <9%) were within acceptable limits. The method was subsequently used to quantify steroid levels in a cohort of healthy women. C11-oxy steroid metabolites produced as intermediates in steroidogenic pathways, together with end-products included in the method can potentially characterise the 11ß-hydroxyandrostenedione-, C21- and C11-oxy backdoor pathways in vivo. The identification of these C11-oxy C19 and C11-oxy C21 intermediates would allow insight into active pathways, while steroid metabolism could be traced in patients and reference ranges established in both normal and abnormal conditions. Furthermore, conditions currently undefined in terms of the C11-oxy steroids would benefit from the analysis provided by this method, while the C11-oxy steroids could be further explored in PCOS, 21OHD, CRPC and adrenarche.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Esteroides/sangue , Espectrometria de Massas em Tandem/métodos , Adulto , Feminino , Humanos , Limite de Detecção , Modelos Lineares , Reprodutibilidade dos Testes , Esteroides/química , Adulto Jovem
5.
J Steroid Biochem Mol Biol ; 199: 105614, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32007561

RESUMO

Cytochrome P450 17α-hydroxylase/17,20-lyase (CYP17A1) plays a pivotal role in the regulation of adrenal and gonadal steroid hormone biosynthesis. More recent studies highlighted the enzyme's role in the backdoor pathway leading to androgen production. Increased CYP17A1 activity in endocrine disorders and diseases are associated with elevated C21 and C19 steroids which include 17α-hydroxyprogesterone and androgens, as well as C11-oxy C21 and C11-oxy C19 steroids. We previously reported that 11ß-hydroxyprogesterone (11OHP4), 21-deoxycortisol (21dF) and their keto derivatives are converted by 5α-reductases and hydroxysteroid dehydrogenases yielding C19 steroids in the backdoor pathway. In this study the 17α-hydroxylase and 17,20-lyase activity of CYP17A1 towards the unconventional C11-oxy C21 steroid substrates and their 5α- and 3α,5α-reduced metabolites was investigated in transfected HEK-293 cells. CYP17A1 catalysed the 17α-hydroxylation of 11OHP4 to 21dF and 11-ketoprogesterone (11KP4) to 21-deoxycortisone (21dE) with negligible hydroxylation of their 5α-reduced metabolites while no lyase activity was detected. The 3α,5α-reduced C11-oxy C21 steroids-5α-pregnan-3α,11ß-diol-20-one (3,11diOH-DHP4) and 5α-pregnan-3α-ol-11,20-dione (alfaxalone) were rapidly hydroxylated to 5α-pregnan-3α,11ß,17α-triol-20-one (11OH-Pdiol) and 5α-pregnan-3α,17α-diol-11,20-dione (11K-Pdiol), with the lyase activity subsequently catalysing to conversion to the C11-oxy C19 steroids, 11ß-hydroxyandrosterone and 11-ketoandrosterone, respectively. Docking of 11OHP4, 11KP4 and the 5α-reduced metabolites, 5α-pregnan-11ß-ol-3,20-dione (11OH-DHP4) and 5α-pregnan-3,11,20-trione (11K-DHP4) with human CYP17A1 showed minimal changes in the orientation of these C11-oxy C21 steroids in the active pocket when compared with the binding of progesterone suggesting the 17,20-lyase is impaired by the C11-hydroxyl and keto moieties. The structurally similar 3,11diOH-DHP4 and alfaxalone showed a greater distance between C17 and the heme group compared to the natural substrate, 17α-hydroxypregnenolone potentially allowing more orientational freedom and facilitating the conversion of the C11-oxy C21 to C11-oxy C19 steroids. In summary, our in vitro assays showed that while CYP17A1 readily hydroxylated 11OHP4 and 11KP4, the enzyme was unable to catalyse the 17,20-lyase reaction of these C11-oxy C21 steroid products. Although CYP17A1 exhibited no catalytic activity towards the 5α-reduced intermediates, once the C4-C5 double bond and the keto group at C3 were reduced, both the hydroxylation and lyase reactions proceeded efficiently. These findings show that the C11-oxy C21 steroids could potentially contribute to the androgen pool in tissue expressing steroidogenic enzymes in the backdoor pathway.


Assuntos
Hidroxiprogesteronas/metabolismo , Progesterona/análogos & derivados , Esteroide 17-alfa-Hidroxilase/genética , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/genética , Androgênios/biossíntese , Androgênios/genética , Linhagem Celular Tumoral , Hormônios Esteroides Gonadais/biossíntese , Hormônios Esteroides Gonadais/genética , Células HEK293 , Humanos , Masculino , Progesterona/biossíntese , Progesterona/genética , Progesterona/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Testosterona/biossíntese
6.
J Steroid Biochem Mol Biol ; 196: 105497, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31626910

RESUMO

In clinical approaches to benign prostatic hyperplasia (BPH) and prostate cancer (PCa), steroidogenesis or the disruption thereof is the main thrust in treatments restricting active androgen production. Extensive studies have been undertaken focusing on testosterone and dihydrotestosterone (DHT). However, the adrenal C11-oxy C19 steroid, 11ß-hydroxyandrostenedione (11OHA4), also contributes to the active androgen pool in the prostate microenvironment, and while it has been shown to impact castration resistant prostate cancer, the C11-oxy C19 steroids together with the C11-oxy C21 steroids have not been studied in BPH. The study firstly investigated the metabolism of these adrenal steroids in the BPH-1 model. Comprehensive profiles identified 11keto-testosterone as the predominant active androgen in the metabolism of the C11-oxy C19 steroids, and we identified, for the first time, 11ß-hydroxy-5α-androstane-3α,17ß-diol, a novel steroid in the 11OHA4-pathway. Analysis of the inactivation and reactivation of the metabolites showed that DHT is more readily inactivated than 11keto-dihydrotestosterone (11KDHT). The conversion of 11ß-hydroxyprogesterone (11ßOHPROG) yielded 11keto-progesterone (11KPROG), while the latter yielded 11keto-dihydroprogesterone (11KDHPROG). BPH tissue analysis identified high levels of 11ß-hydroxyandrosterone (4-14 ng/g) and 11keto-androsterone (9-160 ng/g), together with androstenedione (A4; ∼7.5 ng/g). The major C11-oxy C21 steroids detected were 11ßOHPROG (∼46 ng/g), 11KPROG (∼130 ng/g) as well as 11KDHPROG (∼282 ng/g). While circulatory 11ßOHPROG was detected below the limit of quantification, 11KPROG and 11KDHPROG were detected at 6 and 8.5 nmol/L, respectively. Glucuronide derivatives of both 11KPROG and pregnanetriol were also detected. 11OHA4 was the major free androgen in circulation at 85.9 nmol/L, ±12-fold higher than A4, together with 5α-androstane-3α,17ß-diol quantified at 69.3 nmol/L. Circulatory C11-oxy C19 steroids levels were also significantly higher (8-fold) than the C11-oxy C21 steroid levels, while the former were similar to the C19 steroid levels, in contrast to levels in PCa. The study highlights the contribution of adrenal C11-oxy steroids to the androgen pool in BPH underscoring their limited reactivation and elimination, and significant inter-individual variations regarding steroid levels and conjugation. Targeted steroid metabolome analysis is critical to understanding prostate steroidogenesis and disease progression, and analysis of circulatory C11-oxy C19 and C11-oxy C21 steroids, together with intraprostatic levels, add to our current understanding of BPH.


Assuntos
Androstenodiona/análogos & derivados , Progesterona/análogos & derivados , Hiperplasia Prostática/metabolismo , Testosterona/análogos & derivados , Androstenodiona/química , Androstenodiona/metabolismo , Androstenodiona/farmacologia , Células Cultivadas , Humanos , Masculino , Redes e Vias Metabólicas/efeitos dos fármacos , Progesterona/metabolismo , Hiperplasia Prostática/patologia , Esteroides/química , Esteroides/metabolismo , Testosterona/metabolismo
7.
J Steroid Biochem Mol Biol ; 191: 105369, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31039398

RESUMO

11α-Hydroxyprogesterone (11αOHP4) and 11ß-hydroxyprogesterone (11ßOHP4) have been reported to be inhibitors of 11ß-hydroxysteroid dehydrogenase (11ßHSD) type 2, together with 11ß-hydroxytestosterone and 11ß-hydroxyandrostenedione, and their C11-keto derivatives being inhibitors of 11ßHSD1. Our in vitro assays in transiently transfected HEK293 cells, however, show that 11αOHP4 is a potent inhibitor of 11ßHSD2 and while this steroid does not serve as a substrate for the enzyme, the aforementioned C11-oxy steroids are indeed substrates for both 11ßHSD isozymes. 11ßOHP4 is metabolised by 11ßHSD2 yielding 11-ketoprogesterone with 11ßHSD1 catalysing the reverse reaction, similar to the reduction of the other C11-oxy steroids. In the same model system, novel 11αOHP4 metabolites were detected in its conversion by steroid-5α-reductase (SRD5A) types 1 and 2 yielding 11α-hydroxydihydroprogesterone and its conversion by cytochrome P450 17A1 (CYP17A1) yielding the hydroxylase product, 11α,17α-dihydroxyprogesterone, and the 17,20 lyase product, 11α-hydroxyandrostenedione. We also detected both 11αOHP4 and 11ßOHP4 in prostate cancer tissue- ∼23 and ∼32 ng/g respectively with 11KP4 levels >300 ng/g. In vitro assays in PC3 and LNCaP prostate cancer cell models, showed that the metabolism of 11αOHP4 and 11ßOHP4 was comparable. In LNCaP cells expressing CYP17A1, 11αOHP4 and 11ßOHP4 were metabolised with negligible substrate, 4%, remaining after 48 h, while the steroid substrate 11ß,17α-dihydroxyprogesterone (21dF) was metabolised to C11-keto C19 steroids yielding 11-ketotestosterone. Despite the fact that 11αOHP4 is not metabolised by 11ßHSD2, it is a substrate for SRD5A and CYP17A1, yielding C11α-hydroxy C19 steroids as well as the C11α-hydroxy derivative of 21dF-the latter associated with clinical conditions characterised by androgen excess. With our data showing that 11αOHP4 is present at high levels in prostate cancer tissue, the steroid may serve as a precursor to unique C11α-hydroxy C19 steroids. The potential impact of 11αOHP4 and its metabolites on human pathophysiology can however only be fully assessed once C11α-hydroxyl metabolite levels are comprehensively analysed.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/metabolismo , Androstenodiona/análogos & derivados , Hidroxiprogesteronas/metabolismo , Esteroide 17-alfa-Hidroxilase/metabolismo , Idoso , Androstenodiona/metabolismo , Linhagem Celular Tumoral , Cortodoxona/metabolismo , Células HEK293 , Humanos , Masculino , Neoplasias da Próstata/metabolismo
8.
Artigo em Inglês | MEDLINE | ID: mdl-31031706

RESUMO

Objective: This study is a post-hoc analysis of steroid hormones before and after administration of pharmacological doses of soy isoflavones in a large cohort of men and women from two independent studies. Isoflavones are reported to inhibit mineralo- and glucocorticoid hormone production as well as reproductive steroids in vivo and in vitro. We focused on cytochrome P450 17α-hydroxylase (CYP17A1) which catalyses the production of dehydroepiandrosterone (DHEA), in the androgen biosynthesis pathway to elucidate effects on sex steroids in vitro. Design and Setting: Effects of soy isoflavones on steroid levels in two studies comprising 400 patients were examined: 200 men (study 1; 3 months duration) and 200 postmenopausal women (study 2; 6 months duration), randomized to consume 15 g soy protein with 66 mg isoflavones (SPI) or 15 g soy protein alone without isoflavones (SP) daily. Effects of genistein and daidzein on steroid metabolism were determined in vitro, in HEK293 cells expressing CYP17A1 and in the human adrenocortical carcinoma H295R cell model. Results: SPI decreased serum dehydroepiandrosterone sulfate (DHEAS) levels in both men and women (P < 0.01), with decreased androstenedione (A4) (P < 0.01) in women not observed in men (P < 0.86). Cortisol, cortisone, 11-deoxycortisol, aldosterone, testosterone (T), or estradiol (E2) levels were unchanged. The dual hydroxylase and lyase activity of CYP17A1, which catalyses the biosynthesis of androgen precursors, and 3ß-hydroxysteroid dehydrogenase (3ßHSD2) were investigated in vitro. In transiently transfected HEK293 cells, only the lyase activity was inhibited by both genistein, 20% (P < 0.001) and daidzein, 58% (P < 0.0001). In forskolin-stimulated H295R cells DHEA production was decreased by daidzein (P < 0.05) and genistein, confirming inhibition of the lyase activity by the isoflavones. Conclusion: In Vivo clinical data suggested inhibition of CYP17A1 17,20 lyase within the adrenal in men and within the ovary and adrenal in females. This was confirmed in vitro with inhibition of the lyase activity by both genistein and daidzein. In addition, 3ßHSD2 was inhibited perhaps accounting for decreased A4 levels observed in females. The decreased DHEAS and A4 levels together with the inhibition of the 17,20 lyase activity of CYP17A1, may impact production of androgens in clinical conditions associated with androgen excess. ISRCTN number: ISRCTN55827330 ISRCTN number: ISRCTN 90604927.

9.
J Steroid Biochem Mol Biol ; 189: 116-126, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30825506

RESUMO

The 11ß-hydroxysteroid dehydrogenase (11ßHSD) types 1 and 2 are primarily associated with glucocorticoid inactivation and reactivation. Several adrenal C11-oxy C19 and C11-oxy C21 steroids, which have been identified in prostate cancer, 21-hydroxylase deficiency and polycystic ovary syndrome, are substrates for these isozymes. This study describes the kinetic parameters of 11ßHSD1 and 11ßHSD2 towards the C11-keto and C11-hydroxy derivatives of the C19 and C21 steroids. The apparent Km and Vmax values indicate the more prominent 11ßHSD2 activity towards 11ß-hydroxy androstenedione, 11ß-hydroxytestosterone and 11ß-hydroxyprogesterone in contrast to the 11ßHSD1 reduction of the C11-keto steroids, as was demonstrated in the LNCaP cell model in the production of 11-ketotestosterone and 11-ketodihydrotestosterone. Data highlighted the role of 11ßHSD2 and cytochrome P450 17A1 in the contribution of C11-oxy C21 steroids to the C11-oxy C19 steroid pool in the C11-oxy backdoor pathway. In addition, 11ßHSD2 activity, catalysing 11-ketotestosterone biosynthesis, was shown to be key in the production of prostate specific antigen and in the progression of prostate cancer to castration resistant prostate cancer. The study at hand thus provides evidence that 11ßHSD isozymes play key roles in pathophysiological states, more so than was previously put forward.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Androstenos/metabolismo , Progesterona/análogos & derivados , Testosterona/análogos & derivados , Vias Biossintéticas , Linhagem Celular Tumoral , Células HEK293 , Humanos , Masculino , Progesterona/metabolismo , Neoplasias da Próstata/metabolismo , Isoformas de Proteínas/metabolismo , Especificidade por Substrato , Testosterona/metabolismo
10.
Steroids ; 138: 1-5, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29883615

RESUMO

The purpose of this study was to identify the C11-oxy C19 and C11-oxy C21 steroids in male and female neonate plasma. At birth, the most abundant C11-oxy steroids detected in neonatal plasma were 11ß-hydroxyandrostenedione, ∼13 nM, and 11-ketoprogesterone, ∼23 nM. C11-oxy C19 steroids were higher than C19 steroids in neonatal plasma, 22.2 nM vs 5.4 nM. The inclusion of C11-oxy C19 and C21 steroid reference ranges in routine steroid analyses will assist the characterization of disorders associated with impaired steroidogenic enzyme expression and the identification of potential biomarkers.


Assuntos
Androstenóis/sangue , Progesterona/análogos & derivados , Esteroides/sangue , Espectrometria de Massas em Tandem/métodos , Testosterona/análogos & derivados , Feminino , Humanos , Recém-Nascido , Masculino , Progesterona/sangue , Testosterona/sangue
11.
Mol Cell Endocrinol ; 461: 265-276, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-28939401

RESUMO

Although the adrenal C19 steroids, androstenedione and testosterone, contribute to prostate cancer (PCa) progression the full complement of adrenal androgens, including the C11-oxy C19 steroids, 11ß-hydroxyandrostenedione (11OHA4) and 11ß-hydroxytestosterone (11OHT) and their androgenic metabolites, 11keto-testosterone (11KT) and 11keto-dihydrotestosterone (11KDHT) have, to date, not been considered. This study investigated the contribution of 11OHA4 and 11OHT to the pool of active androgens in the prostate. Steroid profiles were determined in LNCaP, C4-2B and VCaP cell models, in PCa tissue, and in plasma focussing on the inactivation, reactivation and glucuronidation of 11OHA4, 11OHT and their downstream products using ultra-performance convergence chromatography tandem mass spectrometry (UPC2-MS/MS). The C11-oxy C19 steroids were the predominant steroids with the production of 11KT and 11KDHT in prostate cell models identifying 11ß-hydroxysteroid dehydrogenase type 2 activity. Active:inactive steroid ratios indicated efficient inactivation of dihydrotestosterone (DHT) and 11KDHT by 3α-hydroxysteroid dehydrogenases, while the reactivation of DHT by retinol-like dehydrogenases was greater than the reactivation of 11KDHT. In PCa tissue, inactive C11-oxy C19 steroids ranged from 27 to 30 ng/g, whereas inactive C19 steroids were below 1 ng/g. Steroid glucuronidation was impeded: in VCaP cells, the C11-oxy C19 steroids were unconjugated and the C19 steroids fully conjugated; in C4-2B cells, all steroids were unconjugated, except for DHT of which 50% was conjugated; in LNCaP cells only androsterone, 11KT and 11ß-hydroxyandrosterone were unconjugated. In PCa patients' plasma 11KDHT was present only in the unconjugated form, with 11KT also predominantly unconjugated (90-95%). Even though plasma and tissue sample numbers were limited, this study serves to demonstrate the abundance of C11-oxy C19 steroids, with notable differences in their metabolism, dictated by steroidogenic enzymes and hampered conjugation, affecting active androgen levels. Larger cohorts are required to analyse profiles in modulated metabolic pathways, in order to shed light on treatment outcomes. The C11-oxy C19 steroids are involved in PCa, with impeded glucuronidation in PCa ascribing a dominant role to these steroids in disease progression.


Assuntos
Glândulas Suprarrenais/metabolismo , Androgênios/metabolismo , Androstenodiona/análogos & derivados , Glucuronosiltransferase/metabolismo , Metaboloma , Neoplasias da Próstata/metabolismo , Esteroides/metabolismo , Androgênios/sangue , Androstenodiona/metabolismo , Linhagem Celular Tumoral , Humanos , Masculino , Modelos Biológicos , Neoplasias da Próstata/sangue
12.
J Steroid Biochem Mol Biol ; 178: 203-212, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29277707

RESUMO

Increased circulating 11ß-hydroxyprogesterone (11OHP4), biosynthesised in the human adrenal, is associated with 21-hydroxylase deficiency in congenital adrenal hyperplasia. 17α-hydroxyprogesterone levels are also increased, with the steroid's metabolism to dihydrotestosterone in the backdoor pathway contributing to hyperandrogenic clinical conditions. In this study we investigated the in vitro biosynthesis and downstream metabolism of 11OHP4. Both cytochrome P450 11ß-hydroxylase and aldosterone synthase catalyse the biosynthesis of 11OHP4 from progesterone (P4) which is converted to 11-ketoprogesterone (11KP4) by 11ß-hydroxysteroid dehydrogenase type 2, while type 1 readily catalysed the reverse reaction. We showed in HEK-293 cells that these C11-oxy C21 steroids were metabolised by steroidogenic enzymes in the backdoor pathway-5α-reductase (SRD5A) and 3α-hydroxysteroid type 3 (AKR1C2) converted 11OHP4 to 5α-pregnan-11ß-ol,3,20-dione and 5α-pregnan-3α,11ß-diol-20-one, while 11KP4 was converted to 5α-pregnan-3,11,20-trione and 5α-pregnan-3α-ol-11,20-dione (alfaxalone), respectively. Cytochrome P450 17α-hydroxylase/17,20-lyase catalysed the hydroxylase and lyase reaction to produce the C11-oxy C19 steroids demonstrated in the conversion of alfaxalone to 11-oxy steroids demonstrated in the conversion of alfaxalone to 11ketoandrosterone. In LNCaP cells, a prostate cancer cell model endogenously expressing the relevant enzymes, 11OHP4 and 11KP4 were metabolised to the potent androgen, 11-ketodihydrotestosterone (11KDHT), thus suggesting the C11-oxy C21 steroids contribute to the pool of validating the in vitro biosynthesis of C11-oxy C19 steroids from C11-oxy C21 steroids. The in vitro reduction of 11KP4 at C3 and C5 by AKR1C2 and SRD5A has confirmed the metabolic route of the urinary metabolite, 3α,20α-dihydroxy-5ß-pregnan-11-one. Although our assays have demonstrated the conversion of 11OHP4 and 11KP4 by steroidogenic enzymes in the backdoor pathway yielding 11KDHT, thus suggesting the C11-oxy C21 steroids contribute to the pool of potent androgens, the in vivo confirmation of this metabolic route remains challenging.


Assuntos
Hidroxiprogesteronas/metabolismo , Progesterona/análogos & derivados , Neoplasias da Próstata/metabolismo , Testosterona/análogos & derivados , Células HEK293 , Humanos , Hidroxiesteroide Desidrogenases/metabolismo , Técnicas In Vitro , Masculino , Progesterona/metabolismo , Neoplasias da Próstata/patologia , Esteroide 17-alfa-Hidroxilase/metabolismo , Testosterona/metabolismo , Células Tumorais Cultivadas
13.
J Steroid Biochem Mol Biol ; 174: 86-95, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28774496

RESUMO

21-Hydroxylase deficiency presents with increased levels of cytochrome P450 21-hydroxylase substrates, progesterone and 17α-hydroxyprogesterone, which have been implicated in the production of androgens via the backdoor pathway. This study shows the biosynthesis of C11-oxy C21 steroids, 21-deoxycortisol and 21-deoxycortisone, and their metabolism by steroidogenic enzymes in the backdoor pathway yielding novel steroid metabolites: 5α-pregnan-11ß,17α-diol-3,20-dione; 5α-pregnan-17α-ol-3,11,20-trione; 5α-pregnan-3α,11ß,17α-triol-20-one and 5α-pregnan-3α,17α-diol-11,20-dione. The metabolism of 21-deoxycortisol was validated in LNCaP cells expressing the relevant steroidogenic enzymes showing for the first time that the steroid, produced at high levels in 21OHD, is metabolised via the C11-oxy derivatives of 5α-pregnan-17α-ol-3,20-dione and 5α-pregnan-3α,17α-diol-20-one to substrates for the lyase activity of CYP17A1, leading to the production of C11-oxy C19 steroids. 21-Deoxycortisol thus contributes to the pool of potent androgens in 21OHD, with novel steroid metabolites also presenting possible biomarkers in disease identification.


Assuntos
Oxirredutases/metabolismo , Esteroides/biossíntese , Esteroides/metabolismo , Glândulas Suprarrenais/metabolismo , Linhagem Celular Tumoral , Escherichia coli/genética , Células HEK293 , Humanos , Oxirredutases/genética
14.
J Steroid Biochem Mol Biol ; 166: 54-67, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27345701

RESUMO

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.


Assuntos
Glândulas Suprarrenais/metabolismo , Androstenodiona/análogos & derivados , Neoplasias da Próstata/metabolismo , Testosterona/análogos & derivados , Idoso , Androgênios/metabolismo , Androstenodiona/metabolismo , Linhagem Celular Tumoral , Di-Hidrotestosterona/metabolismo , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Ácido Glucurônico/química , Humanos , Hidroxitestosteronas/metabolismo , Masculino , Esteroides/química , Esteroides/metabolismo , Espectrometria de Massas em Tandem , Testosterona/metabolismo
15.
Mol Cell Endocrinol ; 441: 86-98, 2017 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-27664517

RESUMO

16α-hydroxyprogesterone (16OHP4) is not well characterised in terms of metabolism and receptor interaction. We therefore investigated its metabolism by adrenal CYP11B and peripheral steroidogenic enzymes, SRD5A and AKR1C2. UHPLC-MS/MS analyses identified novel steroids: the biosynthesis of 4-pregnen-11ß,16α-diol-3,20-dione catalysed by CYP11B2; the 5α-reduction of the latter and 16OHP4 catalysed by SRD5A yielding 5α-pregnan-11ß,16α-diol-3,20-diovne and 5α-pregnan-16α-ol-3,20-dione (16OH-DHP4); and 16OH-DHP4 converted by AKR1C2 to 5α-pregnan-3α,16α-diol-20-one. Receptor studies showed 16OHP4, 16OH-DHP4, progesterone and dihydroprogesterone (DHP4) were weak partial AR agonists; 16OHP4, 16OH-DHP4 and DHP4 exhibited weak partial agonist activity towards PR-B with DHP4 also exhibiting partial agonist activity towards PR-A. Data showed that while the 5α-reduction of P4 decreased PR activation significantly, 16OHP4 and 16OH-DHP4 exhibited comparable receptor activation. Although the clinical relevance of 16OHP4 remains unclear the elevated 16OHP4 levels characteristic of 21OHD, CAH, PCOS, prostate cancer, testicular feminization syndrome and cryptorchidism likely contribute towards these clinical conditions, inducing receptor-activated target genes.


Assuntos
Hidroxiprogesteronas/metabolismo , Receptores Androgênicos/metabolismo , Receptores de Progesterona/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/metabolismo , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP11B2/metabolismo , Células HEK293 , Humanos , Hidroxilação , Hidroxiesteroide Desidrogenases/metabolismo , Proteínas de Membrana/metabolismo , Peso Molecular , Esteroide 11-beta-Hidroxilase/metabolismo , Espectrometria de Massas em Tandem
16.
Oncotarget ; 6(31): 30730-44, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26436695

RESUMO

Glucocorticoids are among the most effective anti-inflammatory drugs, and are widely used for cancer therapy. Unfortunately, chronic treatment with glucocorticoids results in multiple side effects. Thus, there was an intensive search for selective glucocorticoid receptor (GR) activators (SEGRA), which retain therapeutic potential of glucocorticoids, but with fewer adverse effects. GR regulates gene expression by transactivation (TA), by binding as homodimer to gene promoters, or transrepression (TR), via diverse mechanisms including negative interaction between monomeric GR and other transcription factors. It is well accepted that metabolic and atrophogenic effects of glucocorticoids are mediated by GR TA. Here we summarized the results of extensive international collaboration that led to discovery and characterization of Compound A (CpdA), a unique SEGRA with a proven "dissociating" GR ligand profile, preventing GR dimerization and shifting GR activity towards TR both in vitro and in vivo. We outlined here the unusual story of compound's discovery, and presented a comprehensive overview of CpdA ligand properties, its anti-inflammatory effects in numerous animal models of inflammation and autoimmune diseases, as well as its anti-cancer effects. Finally, we presented mechanistic analysis of CpdA and glucocorticoid effects in skin, muscle, bone, and regulation of glucose and fat metabolism to explain decreased CpdA side effects compared to glucocorticoids. Overall, the results obtained by our and other laboratories underline translational potential of CpdA and its derivatives for treatment of inflammation, autoimmune diseases and cancer.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Extratos Vegetais/uso terapêutico , Receptores de Glucocorticoides/agonistas , Salsola/química , Animais , Anti-Inflamatórios/efeitos adversos , Anti-Inflamatórios/isolamento & purificação , Antineoplásicos Fitogênicos/efeitos adversos , Antineoplásicos Fitogênicos/isolamento & purificação , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Fitoterapia , Extratos Vegetais/efeitos adversos , Extratos Vegetais/isolamento & purificação , Plantas Medicinais , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Steroid Biochem Mol Biol ; 153: 80-92, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25869556

RESUMO

The comprehensive evaluation of the adrenal steroidogenic pathway, given its complexity, requires methodology beyond the standard techniques currently employed. Advances in LC-MS/MS, coupled with in vitro cell models that produce all the steroid metabolites of the mineralo-, glucocorticoid and androgen arms, present a powerful approach for the comprehensive evaluation of adrenal steroidogenesis in response to compounds of interest including bioactives, drug treatments and endocrine disrupting compounds. UHPLC-MS/MS analysis of steroid panels in forskolin, angiotensin II and K(+) stimulated H295R cells provides a snapshot of their effect on intermediates and end products of adrenal steroidogenesis. The impact of full steroid panel evaluations by LC- and GC-MS/MS extends to clinical profiling with the characterization of normal pediatric steroid reference ranges in sexual development and of disease-specific profiles improving diagnosis and sub classification. Comprehensive analyses of steroid profiles may potentially improve patient outcomes together with the application of treatments specifically suited to clinical subgroups. LC-MS/MS and GC-MS/MS applications in the analyses of comprehensive steroid panels are demonstrated in clinical conditions such as congenital adrenal hyperplasia in newborns requiring accurate diagnoses and in predicting metabolic risk in polycystic ovary syndrome patients. Most notable perhaps is the impact of LC-MS/MS evaluations on our understanding of the basic biochemistry of steroidogenesis with the detection of the long forgotten adrenal steroid, 11ß-hydroxyandrostenedione, at significant levels. The characterization of its metabolism to androgen receptor ligands in the LNCaP prostate cancel cell model, specifically within the context of recurring prostate cancer, lends new perspectives to old dogmas. We demonstrate that UHPLC-MS/MS has enabled the analyses of novel metabolites of the enzymes, SRD5A, 11ßHSD and 17ßHSD, in LNCaP cells. Undoubtedly, the continuous advances in the analytical methodologies used for steroid profiling and quantification will give impetus to the unraveling of the remaining enigmas, old and new, of both hormone biosynthesis and metabolism.


Assuntos
Transdução de Sinais , Esteroides/análise , Esteroides/metabolismo , Espectrometria de Massas em Tandem/métodos , 11-beta-Hidroxiesteroide Desidrogenases/metabolismo , Glândulas Suprarrenais/metabolismo , Animais , Cromatografia Gasosa/métodos , Cromatografia Líquida de Alta Pressão/métodos , Feminino , Humanos , Masculino , Síndrome do Ovário Policístico/metabolismo , Neoplasias da Próstata/metabolismo
18.
Mol Cell Endocrinol ; 408: 114-23, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25542845

RESUMO

11ß-Hydroxyandrostenedione (11OHA4), a major C19 steroid produced by the adrenal, was first reported in the 1950s. Initially the subject of numerous studies, interest dwindled due to the apparent lack of physiological function and, by the end of the century, 11OHA4 was no longer considered as an adrenal C19 steroid. Our recent studies, however, showed that 11OHA4 is the precursor to novel active androgens which include 11-ketodihydrotestosterone (11KDHT) which has been implicated in prostate cancer, thereby renewing interest in 11OHA4. In this paper we review the biosynthesis and downstream metabolism of 11OHA4. We discuss the extra-adrenal biosynthesis of 11OHA4 in humans and in other species, highlighting the well-documented role of 11OHA4 in the testes of male fish in which the steroid functions as an active androgen. Finally, we discuss the physiological relevance of 11OHA4 metabolism in castration resistant prostate cancer and outline future prospects.


Assuntos
11-beta-Hidroxiesteroide Desidrogenases/metabolismo , 17-Hidroxiesteroide Desidrogenases/metabolismo , Androstenodiona/análogos & derivados , Proteínas de Membrana/metabolismo , Androgênios/biossíntese , Androgênios/química , Androgênios/metabolismo , Androstenodiona/biossíntese , Androstenodiona/química , Androstenodiona/metabolismo , Animais , Humanos , Especificidade por Substrato
19.
Mol Nutr Food Res ; 58(3): 537-49, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24022885

RESUMO

SCOPE: To determine the effect of Rooibos (Aspalathus linearis) on glucocorticoid biosynthesis and inactivation in vivo and in vitro. METHODS AND RESULTS: Ultra-performance liquid chromatography/tandem mass spectrometry (UPLC-MS/MS) analyses of in vivo studies showed that human Rooibos consumption increased cortisone plasma levels in males (p = 0.0465) and reduced cortisol:cortisone ratios in males and females (p = 0.0486) at risk for cardiovascular disease. In rats, corticosterone (CORT) (p = 0.0275) and deoxycorticosterone (p = 0.0298) levels as well as the CORT:testosterone ratio (p = 0.0009) decreased following Rooibos consumption. The inactivation of cortisol was investigated in vitro by expressing 11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1) and type 2 (11ßHSD2) in CHO-K1 cells. Rooibos inhibited 11ßHSD1, which resulted in a significant reduction in the cortisol:cortisone ratio (p < 0.01). No significant effect was detected on 11ßHSD2. In vitro studies in adrenal H295R cells showed that Rooibos and rutin, one of the more stable flavonoid compounds present in Rooibos, significantly reduced the levels of cortisol and CORT in cells stimulated with forskolin to mimic a stress response. CONCLUSION: In vivo studies demonstrate that Rooibos significantly decreased glucocorticoid levels in rats and steroid metabolite ratios linked to metabolic disorders--cortisol:cortisone in humans and CORT:testosterone in rats. Results obtained at cellular level elucidate possible mechanisms by which these effects were achieved.


Assuntos
Aspalathus/química , Glucocorticoides/metabolismo , Esteroides/metabolismo , 11-beta-Hidroxiesteroide Desidrogenases/genética , 11-beta-Hidroxiesteroide Desidrogenases/metabolismo , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Animais , Células CHO/efeitos dos fármacos , Doenças Cardiovasculares/dietoterapia , Doenças Cardiovasculares/prevenção & controle , Corticosterona/sangue , Cortisona/sangue , Cricetulus , Suplementos Nutricionais , Feminino , Glucocorticoides/sangue , Hidrocortisona/sangue , Masculino , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/análise , Extratos Vegetais/química , Ratos Wistar , Rutina/farmacologia , Esteroides/sangue
20.
Molecules ; 18(11): 13228-44, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-24165582

RESUMO

The biological significance of 11ß-hydroxyandrostenedione (11OHA4) has eluded researchers for the past six decades. It is now known that 11OHA4 is biosynthesized in the androgen arm of the adrenal steroidogenesis pathway and subsequently metabolized by steroidogenic enzymes in vitro, serving as precursor to recognized and novel androgenic steroids. These in vitro findings extend beyond the adrenal, suggesting that 11OHA4 could be metabolized in steroid-responsive peripheral tissues, as is the case for androgen precursor metabolites of adrenal origin. The significance thereof becomes apparent when considering that the metabolism of 11OHA4 in LNCaP androgen dependent prostate cancer cells yields androgenic steroid metabolites. It is thus possible that 11OHA4 may be metabolized to yield ligands for steroid receptors in not only the prostate but also in other steroid-responsive tissues. Future investigations of 11OHA4 may therefore characterize it as a vital steroid with far-reaching physiological consequences. An overview of the research on 11OHA4 since its identification in 1953 will be presented, with specific focus on the most recent works that have advanced our understanding of its biological role, thereby underscoring its relevance in health and disease.


Assuntos
Androstenodiona/análogos & derivados , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/metabolismo , Androstenodiona/biossíntese , Androstenodiona/metabolismo , Di-Hidrotestosterona/metabolismo , Humanos , Hidroxiesteroide Desidrogenases/metabolismo , Masculino , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/metabolismo , Esteroide 11-beta-Hidroxilase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA