Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
Drug Metab Dispos ; 46(4): 470-482, 2018 04.
Article in English | MEDLINE | ID: mdl-29436390

ABSTRACT

Galeterone and abiraterone acetate are antiandrogens developed for the treatment of metastatic castration-resistant prostate cancer. In the present study, we investigated the effect of these drugs on dehydroepiandrosterone (DHEA) sulfonation catalyzed by human liver and intestinal cytosols and human recombinant sulfotransferase enzymes (SULT2A1, SULT2B1b, and SULT2E1) and compared their effects to those of other antiandrogens (cyproterone acetate, spironolactone, and danazol). Each of these chemicals (10 µM) inhibited DHEA sulfonation catalyzed by human liver and intestinal cytosols. Enzyme kinetic analysis showed that galeterone and abiraterone acetate inhibited human liver cytosolic DHEA sulfonation with apparent Ki values at submicromolar concentrations, whereas cyproterone acetate, spironolactone, and danazol inhibited it with apparent Ki values at low micromolar concentrations. The temporal pattern of abiraterone formation and abiraterone acetate depletion suggested that the metabolite abiraterone, not the parent drug abiraterone acetate, was responsible for the inhibition of DHEA sulfonation in incubations containing human liver cytosol and abiraterone acetate. Consistent with this proposal, similar apparent Ki values were obtained, regardless of whether abiraterone or abiraterone acetate was added to the enzymatic incubation. Abiraterone was more effective than abiraterone acetate in inhibiting DHEA sulfonation when catalyzed by human recombinant SULT2A1 or SULT2B1b. In conclusion, galeterone and abiraterone are novel inhibitors of DHEA sulfonation, as determined in enzymatic incubations containing human tissue cytosol (liver or intestinal) or human recombinant SULT enzyme (SULT2A1, SULT2B1b, or SULT1E1). Our findings on galeterone and abiraterone may have implications in drug-drug interactions and biosynthesis of steroid hormones.


Subject(s)
Androstadienes/pharmacology , Androstenes/pharmacology , Benzimidazoles/pharmacology , Cytosol/metabolism , Dehydroepiandrosterone/antagonists & inhibitors , Liver/metabolism , Sulfotransferases/metabolism , Adolescent , Adult , Aged , Female , Humans , Kinetics , Male , Middle Aged , Young Adult
2.
Nutr Cancer ; 68(6): 978-87, 2016.
Article in English | MEDLINE | ID: mdl-27352233

ABSTRACT

Alkylresorcinols (ARs, 5-n-alkylresorcinols) are amphiphilic phenolic lipids in whole grain rye and wheat, with a long odd-numbered carbon chain. A preventive effect of whole grain diet on sex hormone-dependent cancers has been recognized, but the active component(s) or mechanisms are not known. We have investigated the effects of the ARs C15:0, C19:0, and C21:0, individually and in combination, on steroid hormone production by using the human adrenocortical cell line H295R. Decreased synthesis of dehydroepiandrosterone (DHEA), testosterone, and estradiol was demonstrated at low concentrations of C15:0 and C19:0. There were no indications of additive effects on steroid secretion from the combined treatment with equimolar concentrations of the three ARs. Gene expressions of CYP21A2, HSD3B2, and CYP19A1 were downregulated and CYP11A1 was upregulated by the ARs. The results on gene expression could not explain the effects on steroidogenesis, which may be due to direct effects on enzyme activities, such as inhibition of CYP17A1. Our results demonstrate suppressed synthesis of testosterone and estradiol by ARs suggesting a novel mechanism for ARs in the chemoprevention of prostate and breast cancer.


Subject(s)
Adrenal Cortex/metabolism , Anticarcinogenic Agents/metabolism , Dehydroepiandrosterone/antagonists & inhibitors , Estrogen Antagonists/metabolism , Gene Expression Regulation, Enzymologic , Resorcinols/metabolism , Testosterone/antagonists & inhibitors , Adrenal Cortex/enzymology , Alkylation , Anticarcinogenic Agents/chemistry , Aromatase/chemistry , Aromatase/genetics , Aromatase/metabolism , Cell Line, Tumor , Cholesterol Side-Chain Cleavage Enzyme/chemistry , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cytochrome P-450 Enzyme Inhibitors/chemistry , Cytochrome P-450 Enzyme Inhibitors/metabolism , Dehydroepiandrosterone/biosynthesis , Dietary Supplements , Estradiol/biosynthesis , Estrogen Antagonists/chemistry , Fatty Acids/chemistry , Fatty Acids/metabolism , Female , Humans , Male , Progesterone Reductase/antagonists & inhibitors , Progesterone Reductase/genetics , Progesterone Reductase/metabolism , Resorcinols/chemistry , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/metabolism , Steroid 21-Hydroxylase/antagonists & inhibitors , Steroid 21-Hydroxylase/genetics , Steroid 21-Hydroxylase/metabolism , Testosterone/biosynthesis
3.
J Neurochem ; 117(5): 879-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21434925

ABSTRACT

Dehydroepiandrosterone (DHEA) is one of the most abundant neurosteroids synthesized de novo in the CNS. We here found that sigma-1 receptor stimulation by DHEA improves cognitive function through phosphorylation of synaptic proteins in olfactory bulbectomized (OBX) mouse hippocampus. We have previously reported that calcium/calmodulin-dependent protein kinase II (CaMKII), protein kinase C (PKC) and extracellular signal-regulated kinase (ERK) were impaired in OBX mouse hippocampus. OBX mice were administered once a day for 7-8 days with DHEA (30 or 60 mg/kg p.o.) 10 days after operation. The spatial, cognitive and conditioned fear memories in OBX mice were significantly improved as assessed by Y-maze, novel object recognition and passive avoidance task, respectively. DHEA also improved impaired hippocampal long-term potentiation in OBX mice. Notably, DHEA treatment restored PKCα (Ser-657) autophosphorylation and NR1 (Ser-896) and myristoylated alanine-rich protein kinase C substrate (Ser-152/156) phosphorylation to the control levels in the hippocampal CA1 region. Likewise, DHEA treatment improved CaMKIIα (Thr-286) autophosphorylation and GluR1 (Ser-831) phosphorylation to the control levels in the CA1 region. Furthermore, DHEA treatment improved ERK and cAMP-responsive element-binding protein (Ser-133) phosphorylation to the control levels. Finally, NE-100, sigma-1 receptor antagonist, significantly inhibited the DHEA-induced improvement of memory-related behaviors and CaMKII, PKC and ERK phosphorylation in CA1 region. Taken together, sigma-1 receptor stimulation by DHEA ameliorates OBX-induced impairment in memory-related behaviors and long-term potentiation in the hippocampal CA1 region through activation of CaMKII, PKC and ERK.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cognition Disorders/drug therapy , Dehydroepiandrosterone/therapeutic use , Extracellular Signal-Regulated MAP Kinases/metabolism , Olfactory Bulb/physiology , Protein Kinase C/metabolism , Receptors, sigma/agonists , Animals , Anisoles/pharmacology , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Blotting, Western , CA1 Region, Hippocampal/metabolism , Dehydroepiandrosterone/antagonists & inhibitors , Electrophysiological Phenomena , Enzyme Activation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Male , Maze Learning , Memory Disorders/drug therapy , Memory Disorders/psychology , Mice , Phosphorylation , Propylamines/pharmacology , Psychomotor Performance/drug effects , Recognition, Psychology/drug effects , Sigma-1 Receptor
4.
J Pharmacol Exp Ther ; 338(2): 598-604, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21558439

ABSTRACT

Steroidogenic acute regulatory protein facilitates the translocation of cholesterol to the inner mitochondrial membrane, thereby initiating steroidogenesis. At the inner mitochondrial membrane, cytochrome P450 side-chain cleavage enzyme converts cholesterol to pregnenolone, an oxidative process requiring electrons from NADPH. Pregnenolone then serves as the substrate for the formation of progesterone or dehydroepiandrosterone by downstream enzymes. Studies have shown that cigarette smoke (CS) influences steroid hormone levels. To better understand the underlying mechanisms, we used a mouse model to study the effects of chronic CS exposure on steroidogenesis. Through radioimmunoassay and metabolic conversion assays, we found that CS reduced progesterone and dehydroepiandrosterone without affecting cytochrome P450 side-chain cleavage enzyme or 3ß-hydroxysteroid dehydrogenase 2 expression. However, CS did reduce expression of cytochrome c oxidase IV (COX IV), a component of the mitochondrial complex that serves as the last enzyme in the electron transport chain. Small interfering RNA-mediated COX IV knockdown indeed decreased progesterone synthesis in steroidogenic cells. In summary, COX IV likely plays a role in steroidogenesis, and passive smoking may negatively affect steroidogenesis by disrupting the electron transport chain.


Subject(s)
Dehydroepiandrosterone/biosynthesis , Electron Transport Complex IV/antagonists & inhibitors , Gene Expression Regulation, Enzymologic , Pregnenolone/biosynthesis , Progesterone/antagonists & inhibitors , Smoking/metabolism , Animals , COS Cells , Chlorocebus aethiops , Dehydroepiandrosterone/antagonists & inhibitors , Down-Regulation/genetics , Electron Transport Complex IV/biosynthesis , Female , Gene Knockdown Techniques/methods , Mice , Mice, Inbred C57BL , Pregnenolone/antagonists & inhibitors , Progesterone/biosynthesis , Random Allocation , Smoking/adverse effects , Steroids
5.
Cancer Res ; 52(5): 1372-6, 1992 Mar 01.
Article in English | MEDLINE | ID: mdl-1531325

ABSTRACT

We have previously demonstrated that the chemopreventive agent dehydroepiandrosterone (DHEA) inhibits the isoprenylation of cellular proteins by depletion of endogenous mevalonate. We now report that treatment of HT-29 SF human colonic adenocarcinoma cells with DHEA at concentrations ranging from 12.5 to 200 microM for up to 72 h inhibited growth and arrested cells in the G1 phase of the cell cycle in a time- and dose-dependent manner. Exposure to 25 or 50 microM DHEA also transiently delayed cells in G2M phase after 48 h. Addition of mevalonic acid partially overcame both the growth and cell cycle effects of 25 microM DHEA in the initial 48 h. During prolonged exposure (72 h), the addition of mevalonic acid as well as cholesterol was required to reconstitute cell cycle progression. This suggests that the depletion of endogenous mevalonate and other isoprenoids is involved in DHEA-mediated growth inhibition and cell cycle arrest.


Subject(s)
Adenocarcinoma/pathology , Cell Cycle/drug effects , Colonic Neoplasms/pathology , Dehydroepiandrosterone/pharmacology , Adenocarcinoma/metabolism , Cholesterol/pharmacology , Colonic Neoplasms/metabolism , Dehydroepiandrosterone/antagonists & inhibitors , Deoxyribonucleosides/pharmacology , Humans , Mevalonic Acid/pharmacology , Ribonucleosides/pharmacology , Squalene/pharmacology , Time Factors , Tumor Cells, Cultured
6.
Endocrinology ; 146(12): 5176-87, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16150906

ABSTRACT

A decline in dehydroepiandrosterone (DHEA) and GH levels with aging may be associated with frailty and morbidity. Little is known about the direct effects of DHEA on somatotropes. We recently reported that 17beta-estradiol (E2), a DHEA metabolite, stimulates the expression of GH in vitro in young female rats. To test the hypothesis that DHEA restores function in aging somatotropes, dispersed anterior pituitary (AP) cells from middle-aged (12-14 months) or young (3-4 months) female rats were cultured in vitro with or without DHEA or E2 and fixed for immunolabeling or in situ hybridization. E2 increased the percentage of AP cells with GH protein or mRNA in the aged rats to young levels. DHEA increased the percentages of somatotropes (detected by GH protein or mRNA) from 14-16 +/- 2% to 29-31 +/- 3% (P < or = 0.05) and of GH mRNA (detected by quantitative RT-PCR) only in aging rats. To test DHEA's in vivo effects, 18-month-old female rats were injected with DHEA or vehicle for 2.5 d, followed by a bolus of GHRH 1 h before death. DHEA treatment increased serum GH 1.8-fold (7 +/- 0.5 to 12 +/- 1.3 ng/ml; P = 0.02, by RIA) along with a similar increase (P = 0.02) in GH immunolabel. GHRH target cells also increased from 11 +/- 1% to 19 +/- 2% (P = 0.03). Neither GH nor GHRH receptor mRNAs levels were changed. To test the mechanisms behind DHEA's actions, AP cells from aging rats were treated with DHEA with or without inhibitors of DHEA metabolism. Trilostane, aminogluthemide, or ICI 182,780 completely blocked the stimulatory effects of DHEA, suggesting that DHEA metabolites may stimulate aging somatotropes via estrogen receptors.


Subject(s)
Aging/metabolism , Dehydroepiandrosterone/pharmacology , Growth Hormone/metabolism , Receptors, Estrogen/metabolism , Aminoglutethimide/pharmacology , Animals , Aromatase Inhibitors/pharmacology , Cells, Cultured , Dehydroepiandrosterone/antagonists & inhibitors , Dehydroepiandrosterone/metabolism , Dihydrotestosterone/analogs & derivatives , Dihydrotestosterone/pharmacology , Enzyme Inhibitors/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Gene Expression/drug effects , Growth Hormone/antagonists & inhibitors , Growth Hormone/genetics , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Estrogen/genetics , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism
7.
J Neuroimmunol ; 159(1-2): 41-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15652401

ABSTRACT

The cytochrome P4507B1 (P4507B1) is responsible for the 7alpha-hydroxylation of dehydroepiandrosterone (DHEA) and other 3beta-hydroxysteroids in the brain and other organs. The cDNA of human P4507B1 was used for DNA immunization of mice. The best responding mouse led to the production of monoclonal antibodies (mAbs). The clone D16-37 produced an IgM specific for P4507B1 with no cross-reaction with other human P450s. This antibody permitted the immunohistochemical detection of P4507B1 in slices of human hippocampus. P4507B1 was expressed in neurons only. This new tool will be used for the extensive examination of the P4507B1 presence and determination of its levels in slices of human normal and diseased brain and in other human tissues.


Subject(s)
Antibodies, Monoclonal/analysis , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/immunology , DNA, Complementary/administration & dosage , Steroid Hydroxylases/analysis , Steroid Hydroxylases/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/metabolism , Antibody Specificity , Binding Sites, Antibody , Catalysis , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P450 Family 7 , DNA, Complementary/immunology , Dehydroepiandrosterone/antagonists & inhibitors , Dehydroepiandrosterone/metabolism , Hippocampus/enzymology , Hippocampus/immunology , Humans , Immunoglobulin M/metabolism , Immunohistochemistry , Injections, Intramuscular , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
8.
Neurosci Lett ; 377(2): 130-5, 2005 Mar 29.
Article in English | MEDLINE | ID: mdl-15740851

ABSTRACT

Dehydroepiandrosterone (DHEA) is a neurosteroid synthesized de novo in the brain, in addition to the periphery, modulating some membrane, ion-gated channel neurotransmitter receptors. P450-17alpha-hydroxylase activity converting pregnenolone to DHEA, has not yet been identified in the brain of rodents. Studies in brain-derived primary cultures and cell lines, suggest a possible alternative pathway for DHEA synthesis involving oxygenated hydroxyperoxides. We investigated DHEA synthesis in the brains of castrated male mice before and after treatment with N-acetylcysteine amide (AD4) (a newly developed brain penetrating antioxidant). We found a significant increase in brain DHEA level 24 h after castration, which was totally blocked by AD4. This blockade of castration-induced increased brain DHEA synthesis, supports the assumption that this synthesis may also be affected by free radicals. This is the first in vivo study indicating the possible existence of an in-brain oxidative stress-related pathway leading to brain DHEA production.


Subject(s)
Acetylcysteine/analogs & derivatives , Brain/metabolism , Dehydroepiandrosterone/biosynthesis , Free Radicals/metabolism , Acetylcysteine/pharmacology , Animals , Brain/drug effects , Dehydroepiandrosterone/antagonists & inhibitors , Male , Mice , Mice, Inbred ICR , Orchiectomy
9.
Endocrinology ; 135(1): 67-75, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8013393

ABSTRACT

Metabolism of dehydroepiandrosterone sulfate (DHEAS) to dehydroepiandrosterone (DHEA) occurs within specific anatomical compartments in vivo through the actions of the enzyme DHEAS sulfatase. This enzymatic activity facilitates the conversion of hydrophilic DHEAS to the hydrophobic species DHEA, which can then be further metabolized to other steroid hormones. High levels of DHEAS sulfatase reside in tissues where the biological activity of DHEA or its downstream metabolites regulate cellular function. Therefore, control over the activity of DHEAS sulfatase may represent an important regulatory process for the production of DHEA and its metabolites. Homogeneous populations of macrophages from normal mice were found to effectively convert DHEAS to DHEA in vitro. DHEAS sulfatase activity could be markedly depressed after exposure of these cells to a variety of nonspecific macrophage activators [i.e. zymosan, polyinosine/cytosine, heat-killed bacteria, or bacterial lipopolysaccharide (LPS)]. Inhibition of DHEAS metabolism was found to require protein synthesis, because temporary abrogation of protein synthesis with cycloheximide eliminated the ability of LPS to depress the conversion of DHEAS to DHEA. Additionally, exposure of LPS-nonresponsive macrophages to supernatants derived from LPS-treated BALB/c macrophages inhibited their ability to convert DHEAS to DHEA. Potent inhibition of sulfatase activity could be achieved by directly exposing murine macrophages to interferon-alpha (IFN alpha), IFN beta, or tumor necrosis factor-alpha, but not interleukin-1, interleukin-6, granulocyte-macrophage colony-stimulating factor, transforming growth factor-beta, platelet-derived growth factor, or the T-cell product IFN gamma. Our results indicate that macrophage metabolism of DHEAS to DHEA is down-regulated after cellular activation. Furthermore, inhibition of DHEAS sulfatase activity appears to be mediated through the actions of the inflammatory cytokines tumor necrosis factor-alpha and IFN alpha/beta.


Subject(s)
Cytokines/physiology , Dehydroepiandrosterone/analogs & derivatives , Inflammation/metabolism , Macrophages/metabolism , Animals , Bone Marrow/metabolism , Bone Marrow Cells , Cells, Cultured , Dehydroepiandrosterone/antagonists & inhibitors , Dehydroepiandrosterone/biosynthesis , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone Sulfate , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
10.
J Clin Endocrinol Metab ; 80(11): 3201-8, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7593427

ABSTRACT

We have recently demonstrated that estrogen reduced the responsivity of the baboon fetal adrenal gland to ACTH with respect to the formation of dehydroepiandrosterone (DHA) and therefore have proposed that a regulatory system exists for feedback control of estrogen on fetal adrenal androgen production. Because the maternal adrenal also provides DHA and DHA sulfate (DHAS) for placental estrogen synthesis, we determined whether an estrogen-androgen feedback system is operative in the maternal-placental unit. Serum DHA/DHAS and cortisol concentrations were determined by RIA in maternal blood samples obtained at 1- to 2-day intervals from intact baboons untreated (n = 4) or treated sc with estradiol benzoate (n = 3, beginning with 1 mg/day and increasing by 1 mg each day) on days 150 to 184 (term); from animals in which fetal adrenal DHA and DHAS were eliminated by fetectomy on day 100 (n = 4); and from fetectomized baboons treated with estradiol on day 130 to term (n = 3). Maternal serum DHA and DHAS levels increased (P < 0.001) in controls between day 80 and term to means +/- SE of 46.8 +/- 2.4 nmol/L and 0.507 +/- 0.048 mumol/L, respectively, on days 150-184. Estrogen increased serum estradiol concentrations by 78% to 14.50 +/- 0.84 nmol/L and decreased (P < 0.001) DHA and DHAS to 17.2 +/- 1.3 nmol/L and 0.246 +/- 0.015 mumol/L, respectively, on days 150 to 184. After fetectomy, serum estradiol decreased to a level that was 5% of controls, and maternal DHA increased (P < 0.01) to 75.2 +/- 4.8 nmol/L. Estrogen treatment after fetectomy increased mean maternal serum estradiol concentration to 12.15 +/- 0.37 nmol/L and reduced (P < 0.01) DHA and DHAS to 11.9 +/- 0.7 nmol/L and 0.102 +/- 0.005 mumol/L. In contrast, serum cortisol levels were not altered in baboons by estrogen treatment. The estrogen-induced decrease in maternal DHA/DHAS levels reflected a decline in adrenal production; the MCRs (liters/day) of DHA and DHAS in three nonpregnant baboons were similar before (414 +/- 119 and 29.3 +/- 5.6, respectively) and after (359 +/- 66 and 30.4 +/- 3.4, respectively) estradiol treatment, which decreased (P < 0.05) serum DHA and DHAS levels by more than 90%. On the basis of these results and our previous observations in fetal baboons, we propose that a negative feedback system exists in utero whereby placental product estrogen regulates maternal and fetal adrenal C19-steroid production to maintain a physiologically normal balance of estrogen biosynthesis during primate pregnancy.


Subject(s)
Adrenal Glands/metabolism , Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/antagonists & inhibitors , Estrogens/physiology , Papio/physiology , Pregnancy, Animal/metabolism , Animals , Dehydroepiandrosterone/blood , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone Sulfate , Female , Hydrocortisone/blood , Metabolic Clearance Rate , Pregnancy
11.
Ageing Res Rev ; 3(2): 171-87, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15177053

ABSTRACT

Dehydroepiandrosterone (DHEA) is an abundantly produced adrenal steroid whose biological role has never been clarified. DHEA is a potent uncompetitive inhibitor of mammalian glucose-6-phosphate dehydrogenase (G6PDH) and as a consequence lowers NADPH levels and reduces NADPH-dependent oxygen-free radical production. Overproduction of oxygen-free radicals, or oxidative stress, upregulates inflammation and cellular proliferation and is believed to play a critical role in the development of cancer, atherosclerosis, and Alzheimer's disease, as well as the basic aging process. Both in vitro and in vivo experimental studies strongly indicate that DHEA and related steroids inhibit inflammation and associated epithelial hyperplasia, carcinogenesis, and atherosclerosis, at least in part, through the inhibition of G6PDH and oxygen-free radical formation. Recent epidemiological findings in Sardinian males bearing the Mediterranean variant of G6PDH deficiency are consistent with the hypothesis that reduced G6PDH activity has a beneficial effect on age-related disease development and longevity. Clinical trials with DHEA are encumbered by the high oral doses required as well as the conversion of DHEA into active androgens. The use of less androgenic congeners as well as non-oral formulations may facilitate testing of this class of compounds.


Subject(s)
Dehydroepiandrosterone/physiology , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Glucosephosphate Dehydrogenase/metabolism , Longevity/physiology , Animals , Dehydroepiandrosterone/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Longevity/drug effects
12.
Neuroscience ; 83(2): 413-28, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9460750

ABSTRACT

The sigma1 (sigma 1) receptor agonists exert potent anti-amnesic effects, as they apparently block the learning impairments either induced by the muscarinic receptor antagonist scopolamine, the N-methyl-D-aspartate receptor antagonist dizocilpine or inherently due to the age-related deficits in senescence-accelerated mice. We recently described the amnesia induced by the beta-amyloid-related peptide beta 25-35, administered centrally in an aggregated form, in mice. The deficits were sensitive to cholinomimetics or to N-methyl-D-aspartate/glycine modulatory site agonists. Herein, we examined the effects of sigma 1 receptor ligands on the beta 25-35 peptide-induced amnesia. The effects of neuro(active) steroids, which interact in vitro and in vivo with sigma 1 receptors were examined in parallel. Mnesic capacity was evaluated seven days after administration of aggregated beta 25-35 peptide (3 nmol), using spontaneous alternation in the Y-maze for spatial short-term memory, or after 14 days, using the step-down type passive avoidance test for long-term memory. The sigma 1 receptor agonists (+)-pentazocine, PRE-084, or SA4503 attenuated, in a dose-dependent and bell-shaped manner, the beta 25-35 peptide-induced deficits on both tests. These effects were antagonized by haloperidol or BMY-14802, confirming the sigma 1 receptor pharmacology. Pregnenolone, dehydroepiandrosterone, and their sulphate esters, but not progesterone, also dose-dependently attenuated the beta 25-35 peptide-induced deficits. Progesterone blocked the beneficial effects of each other neurosteroid, behaving as an antagonist. Furthermore, haloperidol blocked the effects induced by neurosteroids, whereas progesterone antagonized the effects of the non-steroidal sigma 1 receptor agonists, showing a clear crossed pharmacology of different drug classes. These results demonstrate that: (i) the anti-amnesic effect of sigma 1 receptor agonists may be of therapeutic relevance in pathological states affecting the cholinergic and/or glutamatergic systems, such as in pathological aging; (ii) neurosteroids play an important role in learning processes and may collectively constitute a therapeutic target; (iii) the interaction between sigma 1 systems and neurosteroids appears indeed of behavioural relevance.


Subject(s)
Amnesia/psychology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/pharmacology , Dehydroepiandrosterone/pharmacology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Pregnenolone/pharmacology , Receptors, sigma/agonists , Amnesia/chemically induced , Analgesics, Opioid/pharmacology , Animals , Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Dehydroepiandrosterone/antagonists & inhibitors , Haloperidol/pharmacology , Injections, Intraventricular , Male , Mice , Morpholines/pharmacology , Pentazocine/pharmacology , Piperazines/pharmacology , Pregnenolone/antagonists & inhibitors , Progesterone/pharmacology
13.
Obstet Gynecol ; 73(5 Pt 2): 901-5, 1989 May.
Article in English | MEDLINE | ID: mdl-2523030

ABSTRACT

A 21-year-old white woman presented with virilization, hirsutism, and acne of 1.5 years' duration. Endocrine testing demonstrated complete suppression of serum testosterone, from 5.3 to 0.6 ng/mL, and serum androstenedione, from 4.7 to 1.7 ng/mL, after oral administration of 50 micrograms of mestranol and 1 mg of norethindrone for 21 days. No suppression of either steroid was produced by dexamethasone, whereas serum dehydroepiandrosterone sulfate was suppressed from 5.2 to 1.9 micrograms/mL. A left salpingo-oophorectomy was performed for a 3 x 4-cm Sertoli-Leydig cell tumor of intermediate differentiation. Intraoperative studies demonstrated that the tumor secreted testosterone, androstenedione, 17 alpha-hydroxyprogesterone, and estradiol, but not dehydroepiandrosterone sulfate. These findings support the thesis that hormonal manipulation tests cannot differentiate between adrenal and ovarian virilizing tumors. Nor does the oral contraceptive suppression of testosterone secretion exclude an ovarian malignancy. The patient remains free of recurrence after 7 years.


Subject(s)
Contraceptives, Oral/pharmacology , Fertility , Leydig Cell Tumor , Ovarian Neoplasms , Testosterone/antagonists & inhibitors , Adult , Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/antagonists & inhibitors , Dehydroepiandrosterone/blood , Dehydroepiandrosterone Sulfate , Female , Follow-Up Studies , Gonadal Steroid Hormones/blood , Hirsutism/etiology , Humans , Leydig Cell Tumor/blood , Leydig Cell Tumor/complications , Leydig Cell Tumor/diagnosis , Leydig Cell Tumor/surgery , Luteinizing Hormone/blood , Ovarian Neoplasms/blood , Ovarian Neoplasms/complications , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/surgery , Ovariectomy , Testosterone/blood
14.
Eur J Pharmacol ; 228(1): 15-21, 1992 May 01.
Article in English | MEDLINE | ID: mdl-1397064

ABSTRACT

The inhibition of steroid and phenol sulfotransferase activities in human liver cytosol by a wide range of commonly used drugs was studied. Dehydroepiandrosterone (DHEA) and estrone sulfotransferase activities were strongly inhibited by a number of compounds, with IC50 values ranging between 440 pM and 147 microM. For DHEA sulfotransferase, clomiphene, testosterone, danazol and spironolactone were the best inhibitors, with IC50 values less than 5 microM, whereas for estrone sulfotransferase cyclizine, ibuprofen, chlorpheniramine and dimenhydrinate resulted in the strongest inhibition, again with IC50 values of less than 5 microM. The xenobiotic substrate 1-naphthol was refractory to substantial inhibition, with the exception of clomiphene. The majority of the drugs which inhibited steroid ST activities strongly were either synthetic steroids, antisteroidals or were tertiary amine drugs such as tricyclic antidepressants and antihistamines, many of which exhibit adverse side effects manifesting particularly as sexual dysfunction and disruption of hormone action in clinical use. The importance of these findings for our understanding of the molecular basis of adverse drug reactions is discussed.


Subject(s)
Dehydroepiandrosterone/antagonists & inhibitors , Liver/drug effects , Sulfotransferases , Sulfurtransferases/antagonists & inhibitors , Cytosol/drug effects , Cytosol/enzymology , Drug-Related Side Effects and Adverse Reactions , Humans , In Vitro Techniques , Liver/enzymology
15.
Eur J Pharmacol ; 390(1-2): 145-55, 2000 Feb 25.
Article in English | MEDLINE | ID: mdl-10708718

ABSTRACT

The neuroactive steroids pregnenolone (3beta-hydroxy-5-pregnen-20-one) and dehydroepiandrosterone (DHEA, 3alpha-hydroxy-5-androstene-17-one) are negative allosteric modulators of the GABA(A) receptors and positive modulators of acetylcholine, NMDA and sigma(1) receptors. Pregnenolone was recently shown to potentiate the neuronal damage induced by excessive glutamate in cell culture models, whereas dehydroepiandrosterone was reported to present some neuroprotective activity. The in vivo relevance of these effects was investigated in mice submitted to an hypoxic insult, the repeated exposure to carbon monoxide (CO) gas, a model that leads to neurodegeneration in the CA(1) hippocampal area and learning deficits. Recording spontaneous alternation behaviour in the Y-maze assessed short-term memory and long-term memory was examined using a passive avoidance task. After exposure to CO, mice showed a progressive deterioration of their learning ability, reaching significance after 3 days and being maximal after 7 days. Pregnenolone administered before CO significantly facilitated the hypoxia-related deficits, which could be measured 1 day after CO and appeared maximal after 3 days. Dizocilpine blocked the deficits in vehicle- and pregnenolone-treated CO-exposed animals, showing that pregnenolone selectively facilitated the NMDA receptor-dependent excitotoxicity. Dehydroepiandrosterone blocked the appearance of the CO-induced deficits, even after 7 days. Interestingly, the sigma(1) receptor antagonist N, N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine (NE-100) failed to affect the dehydroepiandrosterone-induced protection, showing the lack of involvement of sigma(1) receptors. Cresyl violet-stained sections of the mouse hippocampal formation showed that the neurodegeneration observed in the CA(1) area after exposure to CO was augmented by pregnenolone and blocked by dehydroepiandrosterone. These results show that pregnenolone and dehydroepiandrosterone, although being similarly involved in modulating the excitatory/inhibitory balance in the brain, do not equally affect the extent of excitotoxic insults.


Subject(s)
Behavior, Animal/drug effects , Carbon Monoxide Poisoning/psychology , Dehydroepiandrosterone/pharmacology , Pregnenolone/pharmacology , Animals , Anisoles/pharmacology , Antipsychotic Agents/pharmacology , Carbon Monoxide Poisoning/pathology , Dehydroepiandrosterone/antagonists & inhibitors , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/pathology , Learning/drug effects , Male , Memory/drug effects , Memory, Short-Term/drug effects , Mice , Nerve Degeneration/pathology , Nerve Degeneration/prevention & control , Pregnenolone/antagonists & inhibitors , Propylamines/pharmacology
16.
Toxicol Lett ; 91(2): 129-36, 1997 Apr 28.
Article in English | MEDLINE | ID: mdl-9175849

ABSTRACT

Dehydroepiandrosterone (DHEA), an adrenal steroid, causes lipid peroxidation in rat liver microsomes and mitochondria and induces hepatocarcinogenesis. It was investigated whether alpha-tocopherol, a naturally occurring free radical chain terminator, could decrease lipid peroxidation. When DHEA-free diet supplemented with increasing concentrations of alpha-tocopherol (25, 50, 100, 200, 400 and 1000 mg/kg diet) was fed to rats for 7 days, a marked lipid peroxidation (measured as thiobarbituric acid reactive substances formation) was observed at concentrations 25 and 50 mg/kg in liver microsomes and mitochondria isolated from these animals. Lipid peroxidation was significantly reduced at concentrations > or = 100 mg/kg. When DHEA (500 mg/kg diet) was fed to rats simultaneously with increasing concentrations of alpha-tocopherol, strong lipid peroxidation was observed at alpha-tocopherol concentrations < or = 200 mg/kg diet. However, microsomes and mitochondria isolated from livers of rats fed alpha-tocopherol at doses of 400 and 1000 mg/kg diet produced only negligible amounts of thiobarbituric acid reactive substances. The data show that high concentrations of alpha-tocopherol in the diet decrease DHEA-induced microsomal and mitochondrial lipid peroxidation. Our results support the concept that alpha-tocopherol can protect against DHEA-induced lipid peroxidation and consequently against steroid-induced liver cell damage and, perhaps, also tumour development.


Subject(s)
Dehydroepiandrosterone/toxicity , Lipid Peroxidation/drug effects , Microsomes, Liver/drug effects , Mitochondria, Liver/drug effects , Vitamin E/pharmacology , Animals , Dehydroepiandrosterone/antagonists & inhibitors , Food, Fortified , Male , Rats , Rats, Sprague-Dawley , Vitamin E/administration & dosage
17.
Arch Pharm Res ; 36(10): 1270-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23821476

ABSTRACT

Experimental studies indicate that some steroid derivatives have inotropic activity; nevertheless, there is scarce information about the effects of the dehydroisoandrosterone and its derivatives at cardiovascular level. In addition, to date the cellular site and mechanism of action of dehydroisoandrosterone at cardiovascular level is very confusing. In order, to clarify those phenomena in this study, a dehydroisoandrosterone derivative was synthesized with the objective of to evaluate its activity on perfusion pressure and coronary resistance and compare this phenomenon with the effect exerted by dehydroisoandrosterone. The Langendorff technique was used to measure changes on perfusion pressure and coronary resistance in an isolated rat heart model in absence or presence of dehydroisoandrosterone and its derivative. Additionally, to characterize the molecular mechanism involved in the inotropic activity induced by dehydroisoandrosterone derivative was evaluated by measuring left ventricular pressure in absence or presence of following compounds; flutamide, prazosin, metoprolol and nifedipine. The results showed that dehydroisoandrosterone derivative significantly increased the perfusion pressure and coronary resistance in comparison with the control conditions and dehydroisoandrosterone. Additionally, other data indicate that dehydroisoandrosterone derivative increase left ventricular pressure in a dose-dependent manner [1 × 10(-9)-1 × 10(-4) mmol]; nevertheless, this phenomenon was significantly inhibited by nifedipine at a dose of 1 × 10(-6) mmol. In conclusion, these data suggest that dehydroisoandrosterone derivative induces positive inotropic activity through of activation the L-type calcium channel.


Subject(s)
Cardiotonic Agents/pharmacology , Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/pharmacology , Androgen Antagonists/pharmacology , Animals , Antihypertensive Agents/pharmacology , Calcium Channel Blockers/pharmacology , Cardiotonic Agents/antagonists & inhibitors , Dehydroepiandrosterone/antagonists & inhibitors , Dose-Response Relationship, Drug , Flutamide/pharmacology , In Vitro Techniques , Male , Metoprolol/pharmacology , Models, Animal , Nifedipine/pharmacology , Prazosin/pharmacology , Rats , Ventricular Function, Left/drug effects
18.
J Steroid Biochem Mol Biol ; 129(3-5): 115-28, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22249003

ABSTRACT

Surgical or pharmacologic methods to control gonadal androgen biosynthesis are effective approaches in the treatment of a variety of non-neoplastic and neoplastic diseases. For example, androgen ablation and its consequent reduction in circulating levels of testosterone is an effective therapy for advanced prostate cancers. Unfortunately, the therapeutic effectiveness of this approach is often temporary because of disease progression to the 'castration resistant' (CRPC) state, a situation for which there are limited treatment options. One mechanism thought to be responsible for the development of CRPC is extra-gonadal androgen synthesis and the resulting impact of these residual extra-gonadal androgens on prostate tumor cell proliferation. An important enzyme responsible for the synthesis of extra-gonadal androgens is CYP17A1 which possesses both 17,20-lyase and 17-hydroxylase catalytic activities with the 17,20-lyase activity being key in the androgen biosynthetic process. Orteronel (TAK-700), a novel, selective, and potent inhibitor of 17,20-lyase is under development as a drug to inhibit androgen synthesis. In this study, we quantified the inhibitory activity and specificity of orteronel for testicular and adrenal androgen production by evaluating its effects on CYP17A1 enzymatic activity, steroid production in monkey adrenal cells and human adrenal tumor cells, and serum levels of dehydroepiandrosterone (DHEA), cortisol, and testosterone after oral dosing in castrated and intact male cynomolgus monkeys. We report that orteronel potently suppresses androgen production in monkey adrenal cells but only weakly suppresses corticosterone and aldosterone production; the IC(50) value of orteronel for cortisol was ~3-fold higher than that for DHEA. After single oral dosing, serum levels of DHEA, cortisol, and testosterone were rapidly suppressed in intact cynomolgus monkeys. In castrated monkeys treated twice daily with orteronel, suppression of DHEA and testosterone persisted throughout the treatment period. In both in vivo models and in agreement with our in vitro data, suppression of serum cortisol levels following oral dosing was less than that seen for DHEA. In terms of human CYP17A1 and human adrenal tumor cells, orteronel inhibited 17,20-lyase activity 5.4 times more potently than 17-hydroxylase activity in cell-free enzyme assays and DHEA production 27 times more potently than cortisol production in human adrenal tumor cells, suggesting greater specificity of inhibition between 17,20-lyase and 17-hydroxylase activities in humans vs monkeys. In summary, orteronel potently inhibited the 17,20-lyase activity of monkey and human CYP17A1 and reduced serum androgen levels in vivo in monkeys. These findings suggest that orteronel may be an effective therapeutic option for diseases where androgen suppression is critical, such as androgen sensitive and CRPC.


Subject(s)
Adrenal Glands/cytology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Naphthalenes/pharmacology , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroids/blood , Steroids/metabolism , Adrenal Glands/drug effects , Androgen Antagonists/pharmacology , Androgens/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Dehydroepiandrosterone/antagonists & inhibitors , Dehydroepiandrosterone/blood , Humans , Hydrocortisone/antagonists & inhibitors , Hydrocortisone/blood , Imidazoles/blood , Ketoconazole/pharmacology , Macaca fascicularis , Male , Naphthalenes/blood , Orchiectomy , Steroid 17-alpha-Hydroxylase/metabolism , Testosterone/antagonists & inhibitors , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL