Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.551
Filter
1.
Eur J Endocrinol ; 191(3): 288-299, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39219353

ABSTRACT

BACKGROUND: Adrenal-origin and peripheral tissue-transformed 11-oxygenated androgens are recognized as significant androgens. However, our current understanding of the synthesis of 11-oxygenated androgens, including the organs and cell types involved, remains limited. METHODS: We performed comprehensive analyses on an extensive dataset of normal human tissues, which included bulk RNA data from 30 tissues, single-cell RNA sequencing (scRNA) data from 16 tissues and proteomics data from 29 tissues, to characterize the expression profiles of enzyme-encoding genes. To validate the findings, immunohistochemical and liquid chromatography-tandem mass spectrometry (LC-MS/MS) techniques were employed. RESULTS: Our investigation revealed that the gene expression levels of the enzymes HSD11B2 and AKR1C3 were notably elevated in the kidney and intestines. Intriguingly, within these organs, we observed an increasing trend in enzyme expression with age in women, while a decreasing trend was apparent in men. scRNA analysis revealed that HSD11B2 was predominantly expressed in collecting duct principal cells in the kidney, while AKR1C3 was primarily expressed in the proximal tubules. Intriguingly, nearly all epithelial cells in the intestine expressed these key enzymes. Further analysis using LC-MS/MS revealed that the kidney exhibited the highest levels of 11-ketoandrostenedione (11KA4) and 11-ketotestosterone (11KT) among the seven tissues examined, and substantial synthesis of 11KA4 and 11KT was also observed in the intestine. Finally, we developed the TransMap website (http://gxmujyzmolab.cn:16245/TransMap/) to provide comprehensive visualization of all currently available transcriptome data. CONCLUSION: This study offers an overarching perspective on tracing the synthesis of 11-oxygenated androgens in peripheral tissues, thereby providing valuable insights into the potential role of these androgens in humans.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3 , Androgens , Tandem Mass Spectrometry , Humans , Chromatography, Liquid , Male , Aldo-Keto Reductase Family 1 Member C3/metabolism , Aldo-Keto Reductase Family 1 Member C3/genetics , Female , Androgens/biosynthesis , Androgens/metabolism , Kidney/metabolism , Kidney/enzymology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Adult , Middle Aged , Gene Expression , Liquid Chromatography-Mass Spectrometry
3.
J Steroid Biochem Mol Biol ; 243: 106561, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38866189

ABSTRACT

The role of mitochondria in steroidogenesis is well established. However, the specific effects of mitochondrial dysfunction on androgen synthesis are not fully understood. In this study, we investigate the effects of various mitochondrial and metabolic inhibitors in H295R adrenal cells and perform a comprehensive analysis of steroid and metabolite profiling. We report that mitochondrial complex I inhibition by rotenone shifts cells toward anaerobic metabolism with a concomitant hyperandrogenic phenotype characterized by rapid stimulation of dehydroepiandrosterone (DHEA, 2 h) and slower accumulation of androstenedione and testosterone (24 h). Screening of metabolic inhibitors confirmed DHEA stimulation, which included mitochondrial complex III and mitochondrial pyruvate carrier inhibition. Metabolomic studies revealed truncated tricarboxylic acid cycle with an inverse correlation between citric acid and DHEA production as a common metabolic marker of hyperandrogenic inhibitors. The current study sheds light on a direct interplay between energy metabolism and androgen biosynthesis that could be further explored to identify novel molecular targets for efficient treatment of androgen excess disorders.


Subject(s)
Androgens , Dehydroepiandrosterone , Mitochondria , Humans , Mitochondria/metabolism , Androgens/metabolism , Androgens/biosynthesis , Dehydroepiandrosterone/metabolism , Testosterone/metabolism , Androstenedione/metabolism , Rotenone/pharmacology , Adrenal Glands/metabolism , Energy Metabolism , Cell Line , Cell Line, Tumor , Electron Transport Complex I/metabolism
4.
Eur J Endocrinol ; 187(1): 1-14, 2022 May 12.
Article in English | MEDLINE | ID: mdl-35521709

ABSTRACT

Context: A sex discordance in COVID exists, with males disproportionately affected. Although sex steroids may play a role in this discordance, no definitive genetic data exist to support androgen-mediated immune suppression neither for viral susceptibility nor for adrenally produced androgens. Objective: The common adrenal-permissive missense-encoding variant HSD3B1(1245C) that enables androgen synthesis from adrenal precursors and that has been linked to suppression of inflammation in severe asthma was investigated in COVID susceptibility and outcomes reported in the UK Biobank. Methods: The UK Biobank is a long-term study with detailed medical information and health outcomes for over 500 000 genotyped individuals. We obtained COVID test results, inpatient hospital records, and death records and tested for associations between COVID susceptibility or outcomes and HSD3B1(1245A/C) genotype. Primary analyses were performed on the UK Biobank Caucasian cohort. The outcomes were identification as a COVID case among all subjects, COVID positivity among COVID-tested subjects, and mortality among subjects identified as COVID cases. Results: Adrenal-permissive HSD3B1(1245C) genotype was associated with identification as a COVID case (odds ratio (OR): 1.11 per C allele, 95% CI: 1.04-1.18, P = 0.0013) and COVID-test positivity (OR: 1.09, 95% CI: 1.02-1.17, P = 0.011) in older (≥70 years of age) women. In women identified as COVID cases, there was a positive linear relationship between age and 1245C allele frequency (P < 0.0001). No associations were found between genotype and mortality or between genotype and circulating sex hormone levels. Conclusion: Our study suggests that a common androgen synthesis variant regulates immune susceptibility to COVID infection in women, with increasingly strong effects as women age.


Subject(s)
Androgens , COVID-19 , Aged , Alleles , Androgens/biosynthesis , Biological Specimen Banks , COVID-19/epidemiology , COVID-19/genetics , Female , Humans , Male , Multienzyme Complexes/genetics , Progesterone Reductase , Steroid Isomerases , United Kingdom/epidemiology
5.
Gynecol Endocrinol ; 38(1): 2-9, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34787028

ABSTRACT

Polycystic ovary syndrome (PCOS) is a disease whose diagnosis is based on the detection of hyperandrogenism (HA) and ovulatory dysfunction. Women with PCOS frequently develop insulin resistance (IR), which generates a metabolic condition that involves a decrease in the action of insulin at the cellular level and is linked to compensatory hyperinsulinemia (HI). In PCOS, the ovary remains sensitive to the action of insulin. Additionally, it has been observed that the main effect of insulin in the ovary is the stimulation of androgen synthesis, resulting in HA, one of the fundamental characteristics of the PCOS. In this sense, the excess of androgens favors the development of IR, thus perpetuating the cycle of IR-HI-HA, and therefore PCOS. Moreover, mitochondrial dysfunction is present in PCOS patients and is a common feature in both IR and HA. This review places electron transfer as a key element in HA and IR development, with emphasis on the relationship between androgen biosynthesis and mitochondrial function. Indeed, metformin has been involved in repair mitochondrial dysfunction, decrease of oxidative stress, reduction of androgens levels and the enhancing of insulin sensitivity. Therefore, we propose that treatment with metformin could decrease HI and consequently HA, restoring, at least in part, the metabolic and hormonal disorders of PCOS.


Subject(s)
Feedback, Physiological/physiology , Hyperandrogenism/physiopathology , Insulin Resistance/physiology , Polycystic Ovary Syndrome/physiopathology , Androgens/biosynthesis , Electron Transport/physiology , Female , Humans , Hyperandrogenism/drug therapy , Hyperinsulinism/drug therapy , Insulin/physiology , Metformin/therapeutic use , Mitochondria/drug effects , Mitochondria/physiology , Ovary/metabolism
6.
Rev. chil. obstet. ginecol. (En línea) ; Rev. chil. obstet. ginecol;86(6): 516-520, dic. 2021. tab
Article in Spanish | LILACS | ID: biblio-1388692

ABSTRACT

INTRODUCCIÓN Y OBJETIVO: El rol de la testosterona exógena en la función sexual femenina ha sido estudiado durante muchos años, con resultados contradictorios. En el último tiempo se ha promovido el uso de pellets de testosterona como una solución para mejorar la libido femenina, la cognición, la fuerza muscular y los sistemas cardiovascular y óseo, e incluso evitar el envejecimiento. Por ello, revisamos las publicaciones para tratar de responder si esto es una moda o el tratamiento más innovador del último tiempo. MÉTODO: Se analizaron las bases de datos PubMed/Medline, Trip Database, Cochrane, SciELO, Scopus, UpToDate, Ovid, ProQuest, Science Direct y ResearchGate. RESULTADOS: De acuerdo con la evidencia, la mejor testosterona disponible es la transdérmica y debe ser usada solo en el trastorno del deseo sexual hipoactivo (TDSH). Los trabajos que evalúan los pellets de testosterona tienen sesgos metodológicos importantes. Si bien son útiles para mejorar la función sexual femenina, producen concentraciones plasmáticas suprafisiológicas de testosterona, por lo que no se puede establecer su seguridad a largo plazo. Tampoco hay datos suficientes que avalen su uso para mejorar el rendimiento cognitivo y el bienestar general, en el tratamiento de enfermedades cardiovasculares o en la prevención de enfermedad ósea. CONCLUSIONES: La testosterona solo se recomienda en el tratamiento del TDSH por vía transdérmica. No recomendamos el uso de pellets de testosterona para el tratamiento de la disfunción sexual ni como hormona antienvejecimiento, ya que no hay estudios consistentes sobre su seguridad, eficacia y efectos adversos a largo plazo.


INTRODUCTION AND OBJECTIVE: The role of exogenous testosterone in female sexual function has been studied for many years with contradictory results. In recent times, the use of testosterone pellets has been promoted as a solution to improve female libido, cognition, muscle strength, cardiovascular system, bone and even prevent aging. Therefore, we will review the publications in order to answer whether this is a fad or the most innovative treatment of recent times. METHOD: The databases PubMed/Medline, Trip Database, Cochrane, SciELO, Scopus, UpToDate, Ovid, ProQuest, Science Direct and ResearchGate were analyzed. RESULTS: So far, the evidence best testosterone available is transdermal testosterone and that it should be used only in hypoactive sexual desire disorder (HSDD). Papers evaluating testosterone pellets have significant methodological biases. While they are useful in improving female sexual function, they produce supra-physiological plasma levels of testosterone, so their long-term safety cannot be established. There is also insufficient data to support their use in improving cognitive performance and general well-being, treatment of cardiovascular disease or prevention of bone disease. CONCLUSIONS: Testosterone is only recommended for the tratment of HSDD via the transdermal route. We do not recommended the use of testosterone pellets for the treatment of sexual dysfunction or as an anti aging hormone, as there are no consistent studies on its safety, efficacy, and long-term adverse effects.


Subject(s)
Humans , Female , Testosterone/administration & dosage , Sexual Dysfunctions, Psychological/drug therapy , Drug Implants , Androgens/biosynthesis
7.
Biochemistry ; 60(43): 3262-3271, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34662099

ABSTRACT

Steroid metabolism in humans originates from cholesterol and involves several enzyme reactions including dehydrogenation, hydroxylation, and carbon-carbon bond cleavage that occur at regio- and stereo-specific points in the four-membered ring structure. Cytochrome P450s occur at critical junctions that control the production of the male sex hormones (androgens), the female hormones (estrogens) as well as the mineralocorticoids and glucocorticoids. An important branch point in human androgen production is catalyzed by cytochrome P450 CYP17A1 and involves an initial Compound I-mediated hydroxylation at the 17-position of either progesterone (PROG) or pregnenolone (PREG) to form 17-hydroxy derivatives, 17OH-PROG and 17OH-PREG, with approximately similar efficiencies. Subsequent processing of the 17-hydroxy substrates involves a C17-C20 bond scission (lyase) activity that is heavily favored for 17OH-PREG in humans. The mechanism for this lyase reaction has been debated for several decades, some workers favoring a Compound I-mediated process, with others arguing that a ferric peroxo- is the active oxidant. Mutations in CYP17A1 can have profound clinical manifestations. For example, the replacement of the glutamic acid side with a glycine chain at position 305 in the CYP17A1 structure causes a clinically relevant steroidopathy; E305G CYP17A1 displays a dramatic decrease in the production of dehydroepiandrosterone from pregnenolone but surprisingly increases the activity of the enzyme toward the formation of androstenedione from progesterone. To better understand the functional consequences of this mutation, we self-assembled wild-type and the E305G mutant of CYP17A1 into nanodiscs and examined the detailed catalytic mechanism. We measured substrate binding, spin state conversion, and solvent isotope effects in the hydroxylation and lyase pathways for these substrates. Given that, following electron transfer, the ferric peroxo- species is the common intermediate for both mechanisms, we used resonance Raman spectroscopy to monitor the positioning of important hydrogen-bonding interactions of the 17-OH group with the heme-bound peroxide. We discovered that the E305G mutation changes the orientation of the lyase substrate in the active site, which alters a critical hydrogen bonding of the 17-alcohol to the iron-bound peroxide. The observed switch in substrate specificity of the enzyme is consistent with this result if the hydrogen bonding to the proximal peroxo oxygen is necessary for a proposed nucleophilic peroxoanion-mediated mechanism for CYP17A1 in carbon-carbon bond scission.


Subject(s)
Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/ultrastructure , Steroids/metabolism , Androgens/biosynthesis , Androgens/metabolism , Androstenedione/metabolism , Catalytic Domain , Dehydroepiandrosterone/metabolism , Humans , Hydrogen Bonding , Hydroxylation , Mutation , Polymorphism, Single Nucleotide/genetics , Pregnenolone/metabolism , Progesterone/metabolism , Spectrum Analysis, Raman/methods , Steroid 17-alpha-Hydroxylase/metabolism , Steroids/biosynthesis , Substrate Specificity , Translocation, Genetic
8.
Science ; 374(6564): 216-224, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34618582

ABSTRACT

The microbiota comprises the microorganisms that live in close contact with the host, with mutual benefit for both counterparts. The contribution of the gut microbiota to the emergence of castration-resistant prostate cancer (CRPC) has not yet been addressed. We found that androgen deprivation in mice and humans promotes the expansion of defined commensal microbiota that contributes to the onset of castration resistance in mice. Specifically, the intestinal microbial community in mice and patients with CRPC was enriched for species capable of converting androgen precursors into active androgens. Ablation of the gut microbiota by antibiotic therapy delayed the emergence of castration resistance even in immunodeficient mice. Fecal microbiota transplantation (FMT) from CRPC mice and patients rendered mice harboring prostate cancer resistant to castration. In contrast, tumor growth was controlled by FMT from hormone-sensitive prostate cancer patients and Prevotella stercorea administration. These results reveal that the commensal gut microbiota contributes to endocrine resistance in CRPC by providing an alternative source of androgens.


Subject(s)
Androgens/biosynthesis , Bacteria/metabolism , Gastrointestinal Microbiome/physiology , Host Microbial Interactions , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/microbiology , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/genetics , Cell Line, Tumor , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasms, Experimental , Prevotella/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Symbiosis , Xenograft Model Antitumor Assays
9.
Sci Rep ; 11(1): 19855, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34615913

ABSTRACT

The eyestalk of crustaceans, such as Macrobrachium nipponense, contains many neurosecretory hormones affecting the process of reproduction, molting, metabolism of glucose, and other functions. In this study, important metabolic pathways and candidate genes involved in male sexual development were selected from M. nipponense. The methodology involved performing long-read and next generation transcriptome sequencing of genes from the androgenic gland after eyestalk ablation. qPCR analysis revealed that the mRNA expression of Mn-IAG was significantly increased after ablation of both the single-side (SS) and double-side (DS) eyestalk, compared with the control group (CG). The long-read transcriptome generated 49,840 non-redundant transcripts. A total of 1319, 2092 and 4351 differentially expressed genes (DEGs) were identified between CG versus SS, SS versus DS and CG versus DS, respectively. These data indicated that ablation of the double-sided eyestalk played stronger regulatory roles than the single-side ablation on male sexual development in M. nipponense. This was consistent with the qPCR analysis. Cell Cycle, Cellular Senescence, Oxidative Phosphorylation, Glycolysis/Gluconeogenesis and Steroid Hormone Biosynthesis were the primary enriched metabolic pathways in all three comparisons, and the important genes from these metabolic pathways were also selected. qPCR permitted secondary confirmation of ten DEGs identified through RNA-seq. RNAi-mediated silencing analyses of Hydroxysteroid dehydrogenase like 1 (HSDL1) revealed that HSDL1 has a positive regulatory effect on testes development. This study provides valuable insight into male sexual development in M. nipponense, including metabolic pathways and genes, paving the way for advanced studies on male sexual development in this species and in other crustaceans.


Subject(s)
Androgens/biosynthesis , Gonads/metabolism , Palaemonidae/genetics , Palaemonidae/metabolism , Animals , Biomarkers , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , RNA Interference , Sex Differentiation/genetics
10.
J Biol Chem ; 297(2): 100969, 2021 08.
Article in English | MEDLINE | ID: mdl-34273352

ABSTRACT

Cytochrome P450 (P450) 17A1 catalyzes the 17α-hydroxylation of progesterone and pregnenolone as well as the subsequent lyase cleavage of both products to generate androgens. However, the selective inhibition of the lyase reactions, particularly with 17α-hydroxy pregnenolone, remains a challenge for the treatment of prostate cancer. Here, we considered the mechanisms of inhibition of drugs that have been developed to inhibit P450 17A1, including ketoconazole, seviteronel, orteronel, and abiraterone, the only approved inhibitor used for prostate cancer therapy, as well as clotrimazole, known to inhibit P450 17A1. All five compounds bound to P450 17A1 in a multistep process, as observed spectrally, over a period of 10 to 30 s. However, no lags were observed for the onset of inhibition in rapid-quench experiments with any of these five compounds. Furthermore, the addition of substrate to inhibitor-P450 17A1 complexes led to an immediate formation of product, without a lag that could be attributed to conformational changes. Although abiraterone has been previously described as showing slow-onset inhibition (t1/2 = 30 min), we observed rapid and strong inhibition. These results are in contrast to inhibitors of P450 3A4, an enzyme with a larger active site in which complete inhibition is not observed with ketoconazole and clotrimazole until the changes are completed. Overall, our results indicate that both P450 17A1 reactions-17α-hydroxylation and lyase activity-are inhibited by the initial binding of any of these inhibitors, even though subsequent conformational changes occur.


Subject(s)
Androgens/biosynthesis , Antineoplastic Agents, Hormonal/pharmacology , Catalytic Domain , Pregnenolone/metabolism , Progesterone/metabolism , Prostatic Neoplasms/drug therapy , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Androstenes/pharmacology , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A/metabolism , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Ketoconazole/pharmacology , Kinetics , Male , Naphthalenes/pharmacology , Prostatic Neoplasms/enzymology , Steroid 17-alpha-Hydroxylase/metabolism
11.
Toxicology ; 456: 152779, 2021 05 30.
Article in English | MEDLINE | ID: mdl-33862173

ABSTRACT

Dibutyltin (DBT) is an organotine widely applied in stabilizing plastics and de-worm poultry agents. But the effects of DBT on immature Leydig cells remain elusive. Thus, the present study aims to investigate whether in vitro exposure to DBT affects immature Leydig cell function of androgen production and delineate the underlying mechanisms. 35 days old rat immature Leydig cells were isolated and exposed to DBT at different concentrations (0, 0.1, 0.5, and 1 µM). It was found that 0.5 and 1 µM DBT lowered androgen production from immature Leydig cells under basal conditions. DBT at 1 µM lowered androgen production from immature Leydig cells under the stimulations from luteinizing hormone or 8-Br-cAMP. DBT at 1 µM lowered 22R-hydroxycholesterol and pregnenolone-mediated androgen production from immature Leydig cells. DBT at 0.1, 0.5, and 1 µM down-regulated the mRNA expression levels of Lhcgr, Star, Cyp11a1, Hsd3b1, and Nr5a1. Further investigation found that DBT at 1 µM directly inhibited CYP11A1 and 3ß-HSD1 enzyme activities. In conclusion, this study told us that in vitro exposure to DBT inhibited androgen biosynthesis in immature Leydig cells by selectively interfering with the expressions and enzyme activities of CYP11A1 and 3ß-HSD1.


Subject(s)
Androgen Antagonists/toxicity , Androgens/biosynthesis , Leydig Cells/drug effects , Leydig Cells/metabolism , Organotin Compounds/toxicity , Age Factors , Animals , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/antagonists & inhibitors , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley
12.
Int J Mol Sci ; 22(6)2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33808818

ABSTRACT

Dichlorodiphenyltrichloroethane (DDT) is the most widespread, persistent pollutant and endocrine disruptor on the planet. Although DDT has been found to block androgen receptors, the effects of its low-dose exposure in different periods of ontogeny on the male reproductive system remain unclear. We evaluate sex steroid hormone production in the pubertal period and after maturation in male Wistar rats exposed to low doses of o,p'-DDT, either during prenatal and postnatal development or postnatal development alone. Prenatally and postnatally exposed rats exhibit lower testosterone production and increased estradiol and estriol serum levels after maturation, associated with the delayed growth of gonads. Postnatally exposed rats demonstrate accelerated growth of gonads and higher testosterone production in the pubertal period. In contrast to the previous group, they do not present raised estradiol production. All of the exposed animals exhibit a reduced conversion of progesterone to 17OH-progesterone after sexual maturation, which indicates putative attenuation of sex steroid production. Thus, the study reveals age-dependent outcomes of low-dose exposure to DDT. Prenatal onset of exposure results in the later onset of androgen production and the enhanced conversion of androgens to estrogens after puberty, while postnatal exposure induces the earlier onset of androgen secretion.


Subject(s)
Androgens/biosynthesis , DDT/pharmacology , Endocrine Disruptors/pharmacology , Environmental Exposure/adverse effects , Estrogens/biosynthesis , Animals , DDT/administration & dosage , Endocrine Disruptors/administration & dosage , Female , Genitalia, Male/drug effects , Genitalia, Male/metabolism , Gonadal Steroid Hormones/biosynthesis , Gonads/drug effects , Gonads/metabolism , Male , Rats
13.
Endocrinology ; 162(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-33784378

ABSTRACT

Two specialized functions of cholesterol during fetal development include serving as a precursor to androgen synthesis and supporting hedgehog (HH) signaling activity. Androgens are produced by the testes to facilitate masculinization of the fetus. Recent evidence shows that intricate interactions between the HH and androgen signaling pathways are required for optimal male sex differentiation and defects of either can cause birth anomalies indicative of 46,XY male variations of sex development (VSD). Further, perturbations in cholesterol synthesis can cause developmental defects, including VSD, that phenocopy those caused by disrupted androgen or HH signaling, highlighting the functional role of cholesterol in promoting male sex differentiation. In this review, we focus on the role of cholesterol in systemic androgen and local HH signaling events during fetal masculinization and their collective contributions to pediatric VSD.


Subject(s)
Androgens/biosynthesis , Cholesterol/physiology , Hedgehog Proteins/metabolism , Sex Differentiation/physiology , Signal Transduction/physiology , Animals , Cholesterol/biosynthesis , Disorders of Sex Development , Fetal Development/physiology , Fetus/metabolism , Humans , Leydig Cells/physiology , Male , Testis/embryology , Testis/metabolism
14.
J Biol Chem ; 296: 100571, 2021.
Article in English | MEDLINE | ID: mdl-33753170

ABSTRACT

It has been recognized for >50 years that cytochrome b5 (b5) stimulates some cytochrome P450 (P450)-catalyzed oxidations, but the basis of this function is still not understood well. The strongest stimulation of catalytic activity by b5 is in the P450 17A1 lyase reaction, an essential step in androgen synthesis from 21-carbon (C21) steroids, making this an excellent model system to interrogate b5 function. One of the issues in studying b5-P450 interactions has been the limited solution assay methods. We constructed a fluorescently labeled variant of human b5 that can be used in titrations. The labeled b5 bound to WT P450 17A1 with a Kd of 2.5 nM and rapid kinetics, on the order of 1 s-1. Only weak binding was observed with the clinical P450 17A1 variants E305G, R347H, and R358Q; these mutants are deficient in lyase activity, which has been hypothesized to be due to attenuated b5 binding. Kd values were not affected by the presence of P450 17A1 substrates. A peptide containing the P450 17A1 Arg-347/Arg-358 region attenuated Alexa 488-T70C-b5 fluorescence at higher concentrations. The addition of NADPH-P450 reductase (POR) to an Alexa 488-T70C-b5:P450 17A1 complex resulted in a concentration-dependent partial restoration of b5 fluorescence, indicative of a ternary P450:b5:POR complex, which was also supported by gel filtration experiments. Overall, these results are interpreted in the context of a dynamic and tight P450 17A1:b5 complex that also binds POR to form a catalytically competent ternary complex, and variants that disrupt this interaction have low catalytic activity.


Subject(s)
Androgens/biosynthesis , Cytochromes b5/metabolism , Lyases/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , Humans , Kinetics , Mutation , Protein Binding , Steroid 17-alpha-Hydroxylase/genetics
15.
Reprod Fertil Dev ; 33(6): 392-400, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33685580

ABSTRACT

Spix's cavy is a potentially good experimental model for research on reproductive biology and sexual development. The aim of the present study was to evaluate the ontogeny of the steroidogenic enzymes involved in testicular androgen synthesis during prenatal development. Testes were investigated on Days 25, 30, 40 and >50 of gestation. Immunohistochemistry and immunoblotting were used to establish the site and relative amount of androgenic enzymes, including 5α-reductase, cytosolic 17ß-hydroxysteroid dehydrogenase (17ß-HSDI) and mitochondrial microsomal 3ß-hydroxysteroid dehydrogenase (3ß-HSDII), throughout prenatal development. The testicular parenchyma began to organise on Day 25 of gestation, with the development of recognisable testicular cords. The mesonephros was established after Day 25 of gestation and the ducts differentiated to form the epididymis, as testicular cords were beginning to proliferate and the interstitium to organise by Day 30 of gestation, continuing thereafter. The androgen-synthesising enzymes 5α-reductase, 17ß-HSDI and 3ß-HSDII were evident in Leydig cells as they differentiated at all subsequent gestational ages studied. In addition, immunoblotting showed an increase in immunoreactivity for the enzymes at Days 30 and 40 of gestation (P<0.05) and a decrease at Day 50 of gestation (P<0.05). It is concluded that the increase in androgenic enzymes in Leydig cells coincides with the functional differentiation of the testes, and with the stabilisation and differentiation of mesonephric ducts forming the epididymis.


Subject(s)
Androgens/biosynthesis , Guinea Pigs/embryology , Testis/embryology , Testis/metabolism , 17-Hydroxysteroid Dehydrogenases/analysis , Animals , Cholestenone 5 alpha-Reductase/analysis , Female , Gestational Age , Immunohistochemistry/veterinary , Leydig Cells/enzymology , Male , Pregnancy , Progesterone Reductase/analysis
16.
Oncogene ; 40(7): 1205-1216, 2021 02.
Article in English | MEDLINE | ID: mdl-33420371

ABSTRACT

Targeting the androgen receptor (AR) signaling axis has been, over decades, the mainstay of prostate cancer therapy. More potent inhibitors of androgen synthesis and antiandrogens have emerged and have been successfully implemented in clinical practice. That said, the stronger inhibition of the AR signaling axis has led in recent years to an increase of prostate cancers that de-differentiate into AR-negative disease. Unfortunately, this process is intimately linked with a poor prognosis. Here, we review the molecular mechanisms that enable cancer cells to switch from an AR-positive to an AR-negative disease and efforts to prevent/revert this process and thereby maintain/restore AR-dependence.


Subject(s)
Androgens/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Androgens/biosynthesis , Humans , Male , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Signal Transduction/genetics
17.
Biochem Biophys Res Commun ; 540: 83-89, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33450484

ABSTRACT

Intracrine androgen synthesis plays a critical role in the development of castration-resistant prostate cancer (CRPC). Aldo-keto reductase family 1 member C3 (AKR1C3) is a vital enzyme in the intracrine androgen synthesis pathway. In this study, mesoporous silica nanoparticles (MSNs) were employed to deliver small interfering RNA targeting AKR1C3 (siAKR1C3) to downregulate AKR1C3 expression in CPRC cells. The optimal weight ratio of MSNs/siAKR1C3 was determined by a gel retardation assay. Prostate cancer cells such as VCaP cells, which intracrinally express AKR1C3, and LNCaP-AKR1C3 cells stably transfected with AKR1C3 were used to investigate the antitumour effect of MSNs-siAKR1C3. Fluorescence detection and Western blot analyses were applied to confirm the entrance of MSNs-siAKR1C3 into the cells. A SRB (Sulforhodamine B) assay was employed to assess the cell viability, and a radioimmunoassay was used to measure the androgen concentration. Moreover, real-time PCR (RT-PCR), Western blot analysis and ELISA were used to determine the transcription and expression of prostate-specific antigen (PSA), AKR1C3 and androgen receptor (AR). Meanwhile, a reporter gene assay was performed to determine the AR activity. Additionally, a castrated nude mouse xenograft tumour model was produced to verify the inhibitory effect of MSNs-siAKR1C3 in vivo. The results showed that the optimal weight ratio of MSNs/siAKR1C3 was 140:1, and the complex could effectively enter cells, downregulate AKR1C3 expression, reduce the androgen concentration, inhibit AR activation, and inhibit CRPC development both in vitro and in vivo. These results indicate that decreasing intracrine androgen synthesis and inactivating AR signals by MSNs-siAKR1C3 may be a potential effective method for CRPC treatment.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/genetics , Androgens/biosynthesis , Nanoparticles/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/therapy , RNA, Small Interfering/therapeutic use , Silicon Dioxide/therapeutic use , Aldo-Keto Reductase Family 1 Member C3/deficiency , Aldo-Keto Reductase Family 1 Member C3/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA, Small Interfering/genetics , Receptors, Androgen/genetics , Testosterone/biosynthesis , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
18.
Prostate ; 81(6): 309-317, 2021 05.
Article in English | MEDLINE | ID: mdl-33503318

ABSTRACT

BACKGROUND: Castration resistant prostate cancer progression is associated with an acquired intratumoral androgen synthesis. Signaling pathways that can upregulate androgen production in prostate tumor microenvironment are not entirely known. In this study, we investigate the potential effect of a secreted signaling protein named semaphorin 3C (SEMA3C) on steroidogenic activities of prostatic stromal cells. METHODS: We treated human primary prostate stromal cells (PrSC) with 1uM recombinant SEMA3C protein and androgen precursor named dehydroepiandrosterone (DHEA) 1.7uM. Also, to test SEMA3C's effect on the conversion of DHEA to androgens, we exposed PrSCs to the conditioned media derived from LNCaP cells that were transduced with a lentiviral vector harboring full length SEMA3C gene or empty vector (CM-LNSEMA3C or CM-LNVector ). Then, liquid chromatography-mass spectrometry was performed on steroids isolated from PrSCs media. The messnger RNA expression of steroidogenic enzymes in PrSCs was quantified by quantitative polymerase chain reaction. RESULTS: Recombinant SEMA3C had no effect on steroidogenic activities in PrSCs. However, key steroidogenic enzymes expression and androgen synthesis were upregulated in PrSCs treated with CM-LNSEMA3C , compared to those treated with CM-LNVector . These results suggest that steroidogenic activities in PrSCs were upregulated in response to a signaling factor in CM-LNSEMA3C , other than SEMA3C. We hypothesized that SEMA3C overexpression in LNCaP cells affected androgen synthesis in PrSCs through sonic hedgehog (Shh) pathway activation in PrSCs. We verified this effect by blocking Shh signaling with smoothened antagonist. CONCLUSION: Based on known ability of Shh signaling pathway to activate steroidogenesis in stromal cells, we suggest that SEMA3C overexpression in LNCaP cells can upregulate Shh which in turn is able to stimulate steroidogenic activities in prostatic stromal cells.


Subject(s)
Androgens/biosynthesis , Hedgehog Proteins/metabolism , Prostate/metabolism , Semaphorins/metabolism , Stromal Cells/metabolism , Dehydroepiandrosterone/metabolism , Humans , Male , Paracrine Communication , Prostate/cytology , Semaphorins/genetics , Up-Regulation
19.
In Vitro Cell Dev Biol Anim ; 56(9): 799-807, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33000385

ABSTRACT

Granulosa cells (GCs) and theca cells (TCs) are the main components of follicles, and the interactions between GCs and TCs play a significant role in steroidogenesis, follicular growth, and atresia. However, the effects of GCs in the form of conditioned medium on steroidogenesis in buffalo TCs remain unclear. In the present study, the impacts of GC-conditioned medium (GCCM) on androgen synthesis in buffalo TCs were examined. The results showed that GCCM collected at 48 h promoted both the expression levels of androgen synthesis-related genes (CYP11A1, CYP17A1, 3ß-HSD, and Star) and the secretion levels of testosterone in TCs. The treatment time of 48 h in GCCM improved both the expression levels of androgen synthesis-related genes (CYP11A1, CYP17A1, 3ß-HSD, and Star) and the secretion levels of testosterone in TCs. Furthermore, GCCM that was collected at 48 h and applied to TCs for 48 h (48 h and 48 h) promoted the sensitivity of buffalo TCs to LH. This study indicated that GCCM (48 h and 48 h) enhanced the steroidogenic competence of TCs mainly through facilitating the responsiveness of TCs to LH in buffalo. This study provides a basis for further exploration of interactions between GCs and TCs for steroidogenesis in the ovary.


Subject(s)
Culture Media, Conditioned/pharmacology , Granulosa Cells/chemistry , Steroids/metabolism , Theca Cells/metabolism , Androgens/biosynthesis , Animals , Buffaloes , Cell Separation , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Luteinizing Hormone/metabolism , Theca Cells/drug effects , Time Factors
20.
Toxicology ; 444: 152577, 2020 11.
Article in English | MEDLINE | ID: mdl-32898603

ABSTRACT

Trimethyltin (TMT) is widely used in industry and agriculture. The present study aims to clarify the effects of in vitro TMT exposure on androgen biosynthesis and metabolism in immature Leydig cells (ILCs), and to unveil the underlying mechanism. It was found that 1-10µM TMT decreased ILC androgen productions under basal conditions. TMT at 10µM decreased luteinizing hormone (LH) or 8-Br-cAMP (8BR)-stimulated androgen productions from ILCs. TMT at 10µM decreased 22R-hydroxycholesterol (22R) and androstenedione (D4)-mediated androgen productions from ILCs. TMT at 0.1-10µM down-regulated the mRNA or protein expression levels of STAR, CYP11A1, 17ß-HSD3, or NR5A1. TMT at 10µM directly inhibited the enzyme activities of CYP11A1 and 17ß-HSD3. In conclusion, the present study demonstrated that in vitro TMT exposure decreased ILC function of androgen production, via exerting negative effects on the mRNA/protein expression levels, or enzyme activities of STAR, CYP11A1, 17ß-HSD3, or NR5A1.


Subject(s)
Androgens/biosynthesis , Leydig Cells/drug effects , Trimethyltin Compounds/toxicity , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Leydig Cells/metabolism , Male , Phosphoproteins/genetics , Phosphoproteins/metabolism , Rats, Sprague-Dawley , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL