Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 307
1.
Mini Rev Med Chem ; 23(8): 941-952, 2023.
Article En | MEDLINE | ID: mdl-36121077

Dehydroepiandrosterone (DHEA) is the most abundant steroid hormone in primates, which is predominantly synthesized in the adrenal cortex. A characteristic curve of growth and decline of its synthesis during life was observed, together with the corresponding formation of its sulphate ester (DHEAS). High levels of plasma circulating DHEA are suggested as a marker of human longevity, and various pathophysiological conditions lead to a decreased DHEA level, including adrenal insufficiency, severe systemic diseases, acute stress, and anorexia. More recent studies have established the importance of DHEA in the central nervous system (CNS). A specific intranuclear receptor for DHEA has not yet been identified; however, highly specific membrane receptors have been detected in endothelial cells, the heart, kidney, liver, and the brain. Research shows that DHEA and DHEAS, as well as their metabolites, have a wide range of effects on numerous organs and organ systems, which places them in the group of potential pharmacological agents useful in various clinical entities. Their action as neurosteroids is especially interesting due to potential neuroprotective, pro-cognitive, anxiolytic, and antidepressant effects. Evidence from clinical studies supports the use of DHEA in hypoadrenal individuals and in treating depression and associated cognitive disorders. However, there is also an increasing trend of recreational DHEA misuse in healthy people, as it is classified as a dietary supplement in some countries. This article aims to provide a critical review regarding the biological and pharmacological effects of DHEA, its mechanism of action, and potential therapeutic use, especially in CNS disorders.


Dehydroepiandrosterone , Endothelial Cells , Animals , Humans , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Endothelial Cells/metabolism , Dehydroepiandrosterone Sulfate/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Brain/metabolism , Steroids
2.
Mol Pharmacol ; 101(2): 68-77, 2022 02.
Article En | MEDLINE | ID: mdl-34853153

The GABAA receptor is inhibited by the endogenous sulfated steroids pregnenolone sulfate (PS) and dehydroepiandrosterone sulfate (DHEAS). It has been proposed in previous work that these steroids act by enhancing desensitization of the receptor. Here, we have investigated the modulatory effects of the steroids on the human α1ß3γ2L GABAA receptor. Using electrophysiology and quantitative model-based data analysis, we show that exposure to the steroid promotes occupancy of a nonconducting state that retains high affinity to the transmitter but whose properties differ from those of the classic, transmitter-induced desensitized state. From the analysis of the inhibitory actions of two combined steroids, we infer that PS and DHEAS act through shared or overlapping binding sites. SIGNIFICANCE STATEMENT: Previous work has proposed that sulfated neurosteroids inhibit the GABAA receptor by enhancing the rate of entry into the desensitized state. This study shows that the inhibitory steroids pregnenolone sulfate and dehydroepiandrosterone sulfate act through a common interaction site by stabilizing a distinct nonconducting state.


Dehydroepiandrosterone Sulfate/pharmacology , GABA Antagonists/pharmacology , Pregnenolone/pharmacology , Receptors, GABA-A/metabolism , Animals , Dehydroepiandrosterone Sulfate/chemistry , Dose-Response Relationship, Drug , Female , GABA Antagonists/chemistry , Humans , Neurosteroids/chemistry , Neurosteroids/pharmacology , Pregnenolone/chemistry , Protein Stability , Receptors, GABA-A/chemistry , Xenopus laevis
3.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article En | MEDLINE | ID: mdl-34948452

Sex hormone steroidal drugs were reported to have modulating actions on the ion channel TRPM3. Pregnenolone sulphate (PS) presents the most potent known endogenous chemical agonist of TRPM3 and affects several gating modes of the channel. These includes a synergistic action of PS and high temperatures on channel opening and the PS-induced opening of a noncanonical pore in the presence of other TRPM3 modulators. Moreover, human TRPM3 variants associated with neurodevelopmental disease exhibit an increased sensitivity for PS. However, other steroidal sex hormones were reported to influence TRPM3 functions with activating or inhibiting capacity. Here, we aimed to answer how DHEAS, estradiol, progesterone and testosterone act on the various modes of TRPM3 function in the wild-type channel and two-channel variants associated with human disease. By means of calcium imaging and whole-cell patch clamp experiments, we revealed that all four drugs are weak TRPM3 agonists that share a common steroidal interaction site. Furthermore, they exhibit increased activity on TRPM3 at physiological temperatures and in channels that carry disease-associated mutations. Finally, all steroids are able to open the noncanonical pore in wild-type and DHEAS also in mutant TRPM3. Collectively, our data provide new valuable insights in TRPM3 gating, structure-function relationships and ligand sensitivity.


Dehydroepiandrosterone Sulfate/pharmacology , Estradiol/pharmacology , Progesterone/pharmacology , TRPM Cation Channels/metabolism , Testosterone/pharmacology , Binding Sites , Calcium/metabolism , Dehydroepiandrosterone Sulfate/chemistry , Estradiol/chemistry , HEK293 Cells , Humans , Molecular Structure , Mutation , Progesterone/chemistry , Structure-Activity Relationship , TRPM Cation Channels/agonists , TRPM Cation Channels/chemistry , TRPM Cation Channels/genetics , Temperature , Testosterone/chemistry , Up-Regulation
4.
Neurotox Res ; 38(2): 447-460, 2020 Aug.
Article En | MEDLINE | ID: mdl-32410195

Several pathophysiological processes involve Hypoxia conditions, where the nervous system is affected as well. We postulate that the GABAergic system is especially sensitive. Furthermore, drugs improving the resistance to hypoxia have been investigated, such as the neurosteroid dehydroepiandrosterone sulfate (DHEAS) which has shown beneficial effects in hypoxic processes in mammals; however, at the cellular level, its exact mechanism of action has yet to be fully elucidated. Here, we used a chemical hypoxia model through sodium sulfite (SS) exposure in Caenorhabditis elegans (C. elegans), a nematode whose response to hypoxia involves pathways and cellular processes conserved in mammals, and that allows study the direct effect of DHEAS without its conversion to sex hormones. This work aimed to determine the effect of DHEAS on damage to the GABAergic system associated with SS exposure in C. elegans. Worms were subjected to nose touch response (Not Assay) and observed in epifluorescence microscopy. DHEAS decreased the shrinkage response of Not Assay and the level of damage in GABAergic neurons on SS-exposed worms. Also, the enhanced nuclear localization of DAF-16 and consequently the overexpression of chaperone HSP-16.2 by hypoxia were significantly reduced in SS + DHEAS exposed worms. As well, DHEAS increased the survival rate of worms exposed to hydrogen peroxide. These results suggest that hypoxia-caused damage over the GABAergic system was prevented at least partially by DHEAS, probably through non-genomic mechanisms that involve its antioxidant properties related to its chemical structure.


Antioxidants/pharmacology , Caenorhabditis elegans Proteins/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Forkhead Transcription Factors/drug effects , GABAergic Neurons/drug effects , Heat-Shock Proteins/drug effects , Hypoxia/metabolism , Sulfites/toxicity , Animals , Behavior, Animal/drug effects , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Forkhead Transcription Factors/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Heat-Shock Proteins/metabolism , Hydrogen Peroxide/toxicity , Hypoxia/pathology , Microscopy, Fluorescence , Oxidants/toxicity , Signal Transduction , Survival Rate
5.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118600, 2020 02.
Article En | MEDLINE | ID: mdl-31760088

It is generally assumed that circulating dehydroepiandrosterone sulfate (DHEAS) can be desulfated and further metabolized to estrogen, which is of concern for all patients with estrogen-responsive breast cancer. We addressed this issue by comparing the effects of DHEAS, its desulfated form DHEA, and 17ß-estradiol on human metastatic, estrogen-responsive MCF-7 breast cancer cells. Physiological concentrations of DHEAS promoted phosphorylation of Erk1/2, whereas DHEA and 17ß-estradiol failed to stimulate Erk1/2 phosphorylation, indicating that the sulfated steroid acts as an autonomous hormone. Exposure of MCF-7 cells to 17ß-estradiol stimulated cell proliferation and the expression of pro-metastatic and pro-invasive elements such as claudin-1, matrix metalloproteinase 9 (MMP9), and the CC chemokine ligand 2 (CCL2). In contrast, treatment with DHEAS did not stimulate these responses but prevented all of the actions of 17ß-estradiol, and as a consequence cell migration and invasion were completely inhibited. The results of this study not only challenge the assumption that DHEAS poses a danger as an endogenous source of estrogen, they rather favor the idea that keeping DHEAS levels within a physiological range might be supportive in treating estrogen-responsive breast cancer.


Cell Movement/drug effects , Cell Proliferation/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Estradiol/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokine CCL2/metabolism , Claudin-1/metabolism , Female , Humans , MCF-7 Cells , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects
6.
Biol Aujourdhui ; 213(3-4): 131-140, 2019.
Article Fr | MEDLINE | ID: mdl-31829933

Discovered in the eighties by Pr Baulieu and colleagues, neurosteroids are a class of neuroactive brain-born steroids, which comprises the steroid hormones, their biosynthesis precursors and their metabolites. They can act through genomic as well as non-genomic pathways. Genomic pathways, only triggered by the neurosteroid hormones, are, in the brain, the same as those largely described in the periphery: the binding of these steroid hormones to nuclear receptors leads to transcription regulations. On the other hand, their precursors and metabolites, such as pregnenolone (PREG), dehydroepiandrosterone (DHEA), their respective sulfate esters, pregnenolone sulfate (PREG-S) and DHEA sulfate (DHEA-S) and allopregnanolone (ALLOP), are defined as neurosteroids, but no corresponding nuclear receptors have been identified so far. In fact, they trigger non-genomic pathways which consist in (i) inhibitory ionotropic receptors, (ii) excitatory ionotropic receptors and (iii) the microtubular system. Hence, inhibitory neurosteroids, whose mostly studied representative is ALLOP, positively modulate, or directly activate, the ionotropic GABA-A receptors. In contrast, excitatory neurosteroids, represented by PREG-S, DHEA-S and DHEA, inhibit the GABA-A receptors, and activate, directly or indirectly, through the sigma-1 receptors, the NMDA glutamate receptors. Neurosteroids of the third group, the microtubular neurosteroids, are able to bind microtubule associated proteins, in particular MAP2, to promote microtubule assembly, neurite outgrowth and in fine structural neuroplasticity. So far, PREG, DHEA and progesterone are the three identified microtubular neurosteroids. The pharmacological properties of neurosteroids have led to specific investigations for assessing their therapeutic potentialities in psychiatric diseases, using validated animal models. In some cases, clinical trials were also performed. These studies showed that ALLOP, the main inhibitory neurosteroid, displayed clear-cut anxiolytic-like and antidepressant-like efficacy in animals. It has been subsequently developed as Brexanolone and tested with success in phase III of clinical trials for the treatment of post-partum depression. Although showing pro-cognitive properties in animals, the sulfated neurosteroids, PREG-S and DHEA-S, were, in contrast, never tested in clinical trials, probably due to their poor stability and proconvulsivant side effects. Their respective non-sulfated forms, PREG and DHEA, showed antidepressant and antipsychotic efficacies in clinical trials, but these drugs never reached the phase III of clinical development because their therapeutic uses would have led to an overproduction of active metabolites responsible for intolerable side effects. The alternative strategy which has been selected consists of the development of non-metabolizable synthetic derivatives of these natural steroids, which keep the same neuroactive properties as their parent molecules, but are devoid of any hormonal side effects. An example of such innovative drugs is MAP4343, a synthetic derivative of PREG, which exhibits potent antidepressant-like efficacy in validated animal models. It is currently tested in depressed patients.


TITLE: Potentialités thérapeutiques des neurostéroïdes en psychiatrie. ABSTRACT: Les neurostéroïdes constituent une famille de molécules synthétisées par le cerveau, représentée par les hormones stéroïdes elles-mêmes, mais également par certains de leurs précurseurs et métabolites. Ils ont des propriétés neuroactives en stimulant des voies de signalisation non génomiques, spécifiques des neurones. Trois types de neurostéroïdes ont été identifiés selon les voies qu'ils activent, à savoir (i) les neurostéroïdes inhibiteurs, (ii) les neurostéroïdes excitateurs et (iii) les neurostéroïdes microtubulaires. Les neurostéroïdes inhibiteurs activent les récepteurs ionotropiques GABA-A, tandis que les neurostéroïdes excitateurs inhibent les courants GABAergiques et stimulent la neurotransmission glutamatergique (soit directement en activant les récepteurs NMDA, soit indirectement via la stimulation des récepteurs sigma-1). Enfin, les neurostéroïdes microtubulaires sont capables de se lier aux protéines associées aux microtubules, comme MAP2, pour favoriser la croissance des microtubules, et in fine la plasticité neuronale. En regard de leurs actions pharmacologiques, certains neurostéroïdes ont fait l'objet d'études cliniques pour le traitement de maladies psychiatriques. C'est le cas de l'alloprégnanolone, le principal neurostéroïde inhibiteur, qui a montré une efficacité dans le traitement de la dépression du post-partum et de l'anxiété. Contrairement à leurs dérivés sulfatés qui n'ont jamais été testés en clinique, la DHEA (déhydroépiandrostérone) et la prégnénolone ont montré des effets antidépresseurs et antipsychotiques. Cependant, la surproduction éventuelle d'hormones provoquée par leur métabolisation a conduit à développer des dérivés de synthèse non métabolisables. C'est le cas du composé MAP4343, un dérivé de la prégnénolone, qui a montré des effets de type antidépresseur dans différents modèles animaux. Il fait actuellement l'objet d'un développement clinique pour le traitement de la dépression.


Mental Disorders/drug therapy , Neurosteroids/therapeutic use , Psychiatry/trends , Animals , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone Sulfate/pharmacology , Depression/drug therapy , Drugs, Investigational/therapeutic use , Humans , Neuronal Plasticity/drug effects , Pregnanolone/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Therapies, Investigational/methods , Therapies, Investigational/trends
7.
Eur J Pharmacol ; 852: 198-206, 2019 Jun 05.
Article En | MEDLINE | ID: mdl-30935894

Stress-induced altered visceral sensation and impaired gut barrier play an important role in the pathophysiology of irritable bowel syndrome (IBS). These responses were demonstrated to be peripheral corticotropin-releasing factor (CRF) dependent and also mediated via proinflammatory cytokine in animal IBS model. Dehydroepiandrosterone sulfate (DHEA-S) is known to have anti-inflammatory properties by suppressing proinflammatory cytokine release. We hypothesized that DHEA-S improves stress-induced visceral changes and is beneficial for IBS treatment. We explored the effects of DHEA-S on lipopolysaccharide (LPS)- or repeated water avoidance stress (WAS)-induced visceral allodynia and increased colonic permeability (rat IBS models). The threshold of visceromotor response, i.e. abdominal muscle contractions induced by colonic balloon distention was electrophysiologically measured. Colonic permeability was estimated in vivo by quantifying the absorbed Evans blue in colonic tissue. DHEA-S abolished visceral allodynia and colonic hyperpermeability induced by LPS in a dose-dependent manner. It also blocked repeated WAS- or peripheral injection of CRF-induced visceral changes. These effects by DHEA-S in LPS model were reversed by bicuculline, a γ-aminobutyric acid (GABA)A receptor antagonist, NG-nitro-L-arginine methyl ester, a nitric oxide (NO) synthesis inhibitor, naloxone, an opioid receptor antagonist, or sulpiride, a dopamine D2 receptor antagonist. However, domperidone, a peripheral dopamine D2 receptor antagonist did not modify the effects. Peripheral injection of astressin2-B, a selective CRF receptor subtype 2 (CRF2) antagonist also reversed these effects. In conclusion, DHEA-S blocked stress-induced visceral changes via GABAA, NO, opioid, central dopamine D2 and peripheral CRF2 signaling. DHEA-S may be useful for IBS treating.


Colon/drug effects , Colon/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Irritable Bowel Syndrome/complications , Visceral Pain/complications , Visceral Pain/drug therapy , Animals , Cytokines/metabolism , Dehydroepiandrosterone Sulfate/therapeutic use , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/psychology , Male , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Stress, Psychological/complications
8.
Br J Anaesth ; 123(2): e215-e225, 2019 Aug.
Article En | MEDLINE | ID: mdl-31030988

BACKGROUND: The neurosteroid dehydroepiandrosterone sulphate (DHEAS) activates the sigma-1 receptor, inhibits gamma-aminobutyric acid A (GABAA) and glycine receptors, and induces hyperalgesic effects. Although its effects have been studied in various tissues of the nervous system, its synaptic mechanisms in nociceptive pathways remain to be elucidated. METHODS: The threshold of mechanical hypersensitivity and spontaneous pain behaviour was assessed using the von Frey test in adult male Wistar rats after intrathecal administration of DHEAS. We also investigated the effects of DHEAS on synaptic transmission in the spinal dorsal horn using slice patch-clamp electrophysiology. RESULTS: Intrathecally administered DHEAS elicited dose-dependent mechanical hyperalgesia and spontaneous pain behaviours (withdrawal threshold: saline; 51.0 [20.1] g, 3 µg DHEAS; 14.0 [7.8] g, P<0.01, 10 µg DHEAS; 6.9 [5.2] g, 15 min after administration, P<0.001). DHEAS at 100 µM increased the frequency of miniature postsynaptic currents in the rat dorsal spinal horn; this increase was extracellular Ca2+-dependent but not sigma-1 and N-methyl-d-aspartate receptor-dependent. DHEAS suppressed the frequency of miniature inhibitory postsynaptic currents in a GABAA receptor- and sigma-1 receptor-dependent manner. CONCLUSIONS: These results suggest that DHEAS participates in the pathophysiology of nociceptive synaptic transmission in the spinal cord by potentiation of glutamate release and inhibition of the GABAA receptor.


Dehydroepiandrosterone Sulfate/pharmacology , Pain/physiopathology , Spinal Cord Dorsal Horn/physiopathology , Animals , Disease Models, Animal , Male , Patch-Clamp Techniques , Rats , Rats, Wistar , Synaptic Transmission/physiology
9.
Mol Neurobiol ; 56(1): 736-747, 2019 Jan.
Article En | MEDLINE | ID: mdl-29796990

Dehydroepiandrosterone sulfate (DHEAS), one of the most important neuroactive steroids, is produced in the adrenals and the brain. DHEAS is believed to play a critical role in modulating different forms of cellular control, including processes associated with human neural systems. Its production rate and level in serum, adrenals, and brain gradually decrease with advancing age. The decline of DHEAS level was associated with age-related neuronal dysfunction and degeneration, most probably because the steroids protect the central nervous system (CNS) neurons against neurotoxic challenges. Moreover, increasing studies show that matrix metalloproteinases (MMPs), MMP-9 especially, are upregulated by proinflammatory mediators in the CNS disorders. The increased MMP-9 as an inflammatory biomarker of several CNS disorders that may participate in the CNS inflammation and neurodegeneration. Herein, we investigate the effects of DHEAS on brain inflammation by the model we have defined of bradykinin (BK)-induced MMP-9 expression in rat brain astrocyte (RBA) and its mechanism. The results showed that DHEAS significantly reduce MMP-9 induced by BK. Pretreatment with DHEAS can inhibit BK-stimulated phosphorylation of c-Src and PYK2. Moreover, DHEAS attenuated BK-stimulated NADPH oxidase (Nox)-derived reactive oxygen species (ROS) production, suggesting that DHEAS has an antioxidative effect. We further demonstrated that DHEAS blocked activation of ERK1/2, Akt, and c-Fos/AP-1 by BK. Finally, DHEAS decreased MMP-9-related events including RBA migration and neuronal apoptosis. The results will provide new insights into the anti-inflammatory action of DHEAS, supporting that DHEAS may have a neuroprotective effect in the improvement of the CNS disorders by reducing neuroinflammation.


Astrocytes/enzymology , Bradykinin/adverse effects , Dehydroepiandrosterone Sulfate/pharmacology , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Astrocytes/drug effects , Brain/enzymology , Cell Line , Cell Movement/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 2/metabolism , Models, Biological , NADPH Oxidases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reactive Oxygen Species/metabolism , Transcription Factor AP-1/metabolism , Up-Regulation/drug effects , src-Family Kinases/metabolism
10.
J Cell Physiol ; 234(8): 12676-12684, 2019 08.
Article En | MEDLINE | ID: mdl-30536399

OBJECTIVES: Systemic lupus erythematosus| (SLE) is an autoimmune disease characterized by hyperactive B cells that produce various autoantibodies. Sex hormones have been documented to influence the development of SLE, in which women with SLE have low plasma level of dehydroepiandrosterone sulfate (DHEAS). A strong conclusion about the effect of DHEAS on apoptosis in SLE patients has not been provided. The aim of this study was to assess apoptotic effects of DHEAS on peripheral blood lymphocytes (PBLs) from SLE patients. METHODS: Twenty SLE patients and 20 age- and sex-matched healthy controls were included into this study. Concentration of DHEAS was measured using enzyme-linked immunosorbent assay in serum from all participants. Freshly isolated PBLs from each individual were treated with 7.5-µmol of DHEAS for 24 hr in cell culture medium to assess the effect of DHEAS on apoptosis using fluorescein isothiocyante-conjugated annexin V and propidium iodide. The messenger RNA (mRNA) expression level of apoptosis-related genes (Fas, Fas-L, Bcl-2, and Bax) in PBLs was measured using real-time PCR before and after treating with DHEAS. RESULTS: Level of DHEAS was low in SLE patients compared with healthy controls (p < 0.05). After treating with DHEAS, the percentage of apoptotic cells in SLE patients was decreased in comparison with healthy controls. DHEAS treatment increased the mRNA expression level of Bcl-2 in PBLs from SLE patients. CONCLUSIONS: DHEAS reduced the apoptosis rate in PBLs from SLE patients and may decrease the load of autoantigens. Therefore, DHEAS might be considered as a therapeutic tool in SLE patients.


Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Lupus Erythematosus, Systemic/metabolism , Adult , Female , Humans , Male , RNA, Messenger/metabolism
11.
Vitam Horm ; 108: 413-441, 2018.
Article En | MEDLINE | ID: mdl-30029737

Steroid hormones are important regulators of brain development, physiological function, and behavior. Among them, dehydroepiandrosterone (DHEA) and dehydroepiandrosterone-sulfate (DHEAS) also do modulate emotional processing and may have mood enhancement effects. This chapter reviews the studies that bear relation to DHEA and DHEAS [DHEA(S)] and brain emotional processing and behavior. A brief introduction to the mechanisms of action and variations of DHEA(S) levels throughout life has also been forward in this chapter. Higher DHEA(S) levels may reduce activity in brain regions involved in the generation of negative emotions and modulate activity in regions involved in regulatory processes. At the electrophysiological level, higher DHEA-to-cortisol and DHEAS-to-DHEA ratios were related to shorter P300 latencies and shorter P300 amplitudes during the processing of negative stimuli, suggesting less interference of negative stimuli with the task and less processing of the negative information, which in turn may suggest a protective mechanism against negative information overload. Present knowledge indicates that DHEA(S) may play a role in cortical development and plasticity, protecting against negative affect and depression, and at the same time enhancing attention and overall working memory, possibly at the cost of a reduction in emotional processing, emotional memory, and social understanding.


Brain/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Dehydroepiandrosterone/pharmacology , Emotions/drug effects , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone Sulfate/metabolism , Emotions/physiology , Humans
13.
J Steroid Biochem Mol Biol ; 182: 50-61, 2018 09.
Article En | MEDLINE | ID: mdl-29684479

Although dehydroepiandrosterone sulfate (DHEAS) constitutes the most abundant steroid in humans, in-depth investigations of its effects are rather scarce. We address here DHEAS effects on the estrogen receptor-positive metastatic human breast cancer cell line MCF-7. We focus on DHEAS-mediated signaling that might influence expression of claudin-1 and matrix metalloproteinase-9 (MMP-9), both known to be critical factors for migration and invasiveness of various cancers, including breast cancer cells. Physiological concentrations of DHEAS trigger persistent phosphorylation of Erk1/2 in MCF-7 cells. Exposure of these cells for 24 h to 1 µM DHEAS also leads to a significant reduction of claudin-1 expression that cannot be prevented by high concentrations of the steroid sulfatase inhibitor STX64, indicating that desulfation and further conversion of DHEAS to some other steroid hormone is not required for this action. In addition, exposure of MCF-7 cells to the same concentration of DHEAS completely abolishes MMP-9 expression and considerably impairs cell migratory behavior. Abrogation of Gnα11 expression by siRNA prevents the stimulatory effect of DHEAS on Erk1/2 phosphorylation, consistent with a G-protein-coupled receptor being involved in the DHEAS-induced signaling. Nevertheless, Gnα11 also has direct effects that do not depend on DHEAS; thus, when Gnα11 expression is suppressed, expression of claudin-1 and MMP-9 as well as cell migration are significantly reduced. This is the first report demonstrating direct involvement of DHEAS and Gnα11 in the regulation of claudin-1 and MMP-9 expression and migration of MCF-7 cells.


Breast Neoplasms/pathology , Cell Movement/drug effects , Claudin-1/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , GTP-Binding Protein alpha Subunits/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Matrix Metalloproteinase 9/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Claudin-1/genetics , Female , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/genetics , Humans , MCF-7 Cells , Matrix Metalloproteinase 9/genetics , Phosphorylation , RNA, Small Interfering/genetics , Signal Transduction
14.
Cancer Sci ; 108(10): 2011-2021, 2017 Oct.
Article En | MEDLINE | ID: mdl-28771887

Long-chain acyl-coenzyme A (CoA) synthetase 3 (ACSL3) is an androgen-responsive gene involved in the generation of fatty acyl-CoA esters. ACSL3 is expressed in both androgen-sensitive and castration-resistant prostate cancer (CRPC). However, its role in prostate cancer remains elusive. We overexpressed ACSL3 in androgen-dependent LNCaP cells and examined the downstream effectors of ACSL3. Furthermore, we examined the role of ACSL3 in the androgen metabolism of prostate cancer. ACSL3 overexpression led to upregulation of several genes such as aldo-keto reductase 1C3 (AKR1C3) involved in steroidogenesis, which utilizes adrenal androgen dehydroepiandrosterone sulfate (DHEAS) as substrate, and downregulated androgen-inactivating enzyme UDP-glucuronosyltransferase 2 (UGT2B). Exposure to DHEAS significantly increased testosterone levels and cell proliferative response in ACSL3-overexpressing cells when compared to that in control cells. A public database showed that ACSL3 level was higher in CRPC than in hormone-sensitive prostate cancer. CRPC cells showed an increased expression of ACSL3 and an expression pattern of AKR1C3 and UGT2B similar to ACSL3-overexpressing cells. DHEAS stimulation significantly promoted the proliferation of CRPC cells when compared to that of LNCaP cells. These findings suggest that ACSL3 contributes to the growth of CRPC through intratumoral steroidogenesis (i.e. promoting androgen synthesis from DHEAS and preventing the catabolism of active androgens).


Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Prostatic Neoplasms, Castration-Resistant/metabolism , Testosterone/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Aldo-Keto Reductase Family 1 Member C3 , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucuronosyltransferase/metabolism , Humans , Hydroxyprostaglandin Dehydrogenases/metabolism , Lipogenesis , Male , Prostatic Neoplasms, Castration-Resistant/genetics
15.
Biochim Biophys Acta Mol Cell Res ; 1864(8): 1382-1392, 2017 Aug.
Article En | MEDLINE | ID: mdl-28495656

Tight junctions (TJ) between brain endothelial cells are essential for formation and maintenance of the blood-brain barrier (BBB). Although loss of BBB integrity is associated with several neuropathological disorders, treatments that augment or stabilise the BBB are scarce. Here we show that physiological concentrations of dehydroepiandrosterone sulfate (DHEAS) stimulate the expression of the TJ proteins zonula occludens-1 (ZO-1) and claudin-3 in the brain-derived endothelial cell line bEnd.3 and promote TJ formation between neighbouring cells, demonstrated by augmented transendothelial resistance across cell monolayers. Silencing androgen receptor expression by siRNA does not prevent DHEAS-induced stimulation of ZO-1 expression, indicating that conversion of DHEAS into testosterone is not required for its actions. Suppression of Gnα11 expression by siRNA prevents DHEAS actions, pointing towards a G-protein-coupled receptor as being a mediator of the DHEAS effects. These results are consistent with the idea that DHEAS, acting as a hormone in its own right, supports the integrity of the BBB. The current findings might help in developing new strategies for the prevention or treatment of neurological disorders associated with BBB defects.


Blood-Brain Barrier/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Endothelial Cells/drug effects , Tight Junctions/drug effects , Zonula Occludens-1 Protein/genetics , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/ultrastructure , Brain/blood supply , Brain/drug effects , Cell Line , Claudin-3/genetics , Claudin-3/metabolism , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Gene Expression Regulation , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction , Tight Junctions/metabolism , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein/agonists , Zonula Occludens-1 Protein/metabolism
16.
Drug Des Devel Ther ; 10: 1201-15, 2016.
Article En | MEDLINE | ID: mdl-27051273

BACKGROUND: Neurosteroid dehydroepiandrosterone sulfate (DHEAS) has been associated with important brain functions, including neuronal survival, memory, and behavior, showing therapeutic potential in various neuropsychiatric and cognitive disorders. However, the antagonistic effects of DHEAS on γ-amino-butyric acidA receptors and its facilitatory action on glutamatergic neurotransmission might lead to enhanced brain excitability and seizures and thus limit DHEAS therapeutic applications. The aim of this study was to investigate possible age and sex differences in the neuronal excitability of the mice following acute and chronic DHEAS administration. METHODS: DHEAS was administered intraperitoneally in male and female adult and old mice either acutely or repeatedly once daily for 4 weeks in a 10 mg/kg dose. To investigate the potential proconvulsant properties of DHEAS, we studied the effects of acute and chronic DHEAS treatment on picrotoxin-, pentylentetrazole-, and N-methyl-D-aspartate-induced seizures in mice. The effects of acute and chronic DHEAS administration on the locomotor activity, motor coordination, and body weight of the mice were also studied. We also investigated the effects of DHEAS treatment on [(3)H]flunitrazepam binding to the mouse brain membranes. RESULTS: DHEAS did not modify the locomotor activity, motor coordination, body weight, and brain [(3)H]flunitrazepam binding of male and female mice. The results failed to demonstrate significant effects of single- and long-term DHEAS treatment on the convulsive susceptibility in both adult and aged mice of both sexes. However, small but significant changes regarding sex differences in the susceptibility to seizures were observed following DHEAS administration to mice. CONCLUSION: Although our findings suggest that DHEAS treatment might be safe for various potential therapeutic applications in adult as well as in old age, they also support subtle interaction of DHEAS with male and female hormonal status, which may underline observed sex differences in the relationship between DHEAS and various health outcomes.


Brain/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Motor Activity/drug effects , Neurons/drug effects , Age Factors , Animals , Brain/metabolism , Dehydroepiandrosterone Sulfate/administration & dosage , Drug Administration Schedule , Female , Flunitrazepam/metabolism , Male , Mice , Mice, Inbred CBA , Neurons/metabolism , Seizures/etiology , Sex Factors
17.
Psychoneuroendocrinology ; 69: 116-22, 2016 07.
Article En | MEDLINE | ID: mdl-27104812

Cardiovascular stress reactivity is a predictor of atherosclerosis and cardiac events. Dehydroepiandrosterone (DHEA) protects against cardiovascular diseases, but results among previous studies have been inconsistent. We investigated the association between dehydroepiandrosterone-sulfate (DHEA-s) and cardiovascular stress reactivity in Japanese women and men. Among 979 healthy Japanese subjects (641 women and 338 men), serum levels of DHEA-s, systolic blood pressure (SBP) and diastolic blood pressure (DBP), heart rate, heart rate variability, and peripheral blood flow were measured under rest and two types of task. Mean differences in measured variables during tasks and a post-task period were calculated as changes in stress reactivity. Variables of stress reactivity were adjusted for multiple potential confounding factors. In women, DHEA-s levels showed positive associations with changes in SBP and DBP (standardized beta=0.12, p=0.020; 0.17, 0.002, respectively). Stratification by menopausal status and other lifestyle factors (e.g., smoking status, alcohol consumption) were conducted. Significant positive associations remained in pre-menopausal (standardized beta=0.13, p=0.037; 0.18, 0.005), non-smoking (0.12, 0.010; 0.18, <0.001), and non-drinking women (0.14, 0.021; 0.21, 0.001), and women without a medical history (0.15, 0.020; 0.20, 0.001). In men, there was no significant association between DHEA-s levels and changes in stress reactivity. DHEA-s levels were positively associated with high blood-pressure reactivity to stress in women, and being menopausal, smoking, and alcohol consumption modified this association.


Dehydroepiandrosterone Sulfate/pharmacology , Adult , Age Factors , Blood Flow Velocity , Blood Pressure/physiology , Cardiovascular Diseases , Cardiovascular Physiological Phenomena , Dehydroepiandrosterone/blood , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone Sulfate/blood , Dehydroepiandrosterone Sulfate/metabolism , Female , Heart Rate/physiology , Humans , Hypertension , Japan , Life Style , Male , Menopause , Middle Aged , Sex Factors , Smoking , Stress, Physiological/physiology
18.
Steroids ; 105: 50-8, 2016 Jan.
Article En | MEDLINE | ID: mdl-26666359

Dehydroepiandrosterone sulfate (DHEAS) and estrone sulfate (E1S) are two of the most abundant steroids in the human circulation. The enzyme steroid sulfatase (STS) cleaves the sulfate group of DHEAS and E1S leading to biosynthesis of endogenous hormones such as testosterone and estrone. In the current study we aimed at determining the effect of E1S and DHEAS on estrogen receptor (ER) and androgen receptor (AR) transactivation. Using luciferase reporter gene assays, the ER and AR transactivities of E1S and DHEAS were determined by direct cell exposure; as well as upon extraction from human serum using a method to extract perfluorinated alkyl acids (PFAAs). By direct cell exposure, both E1S and DHEAS transactivated the ER and the AR in dose-dependent manners. The DHEAS-induced AR transactivity could be abolished by the STS inhibitor STX64. Immunoassay analysis confirmed the presence of E1S and DHEAS in the serum PFAA extracts with mean recoveries below 2.5%. For the PFAA extracts of human male and female serum, only the AR was significantly transactivated. The AR transactivity of the sulfated steroids in the extracts was abolished by STX64 to obtain the net PFAA induced xenohormone transactivity, but further cleanup might be needed at high concentrations of E1S.


Dehydroepiandrosterone Sulfate/pharmacology , Estrogens/genetics , Estrone/analogs & derivatives , Receptors, Estrogen/genetics , Transcriptional Activation/drug effects , Animals , CHO Cells , Cell Death/drug effects , Cricetinae , Cricetulus , Dehydroepiandrosterone Sulfate/blood , Dehydroepiandrosterone Sulfate/chemistry , Estrogens/metabolism , Estrone/blood , Estrone/chemistry , Estrone/pharmacology , Female , Fluorocarbons/chemistry , Humans , Male , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Serum/metabolism , Transcriptional Activation/genetics
19.
Endocrinology ; 156(7): 2704-12, 2015 Jul.
Article En | MEDLINE | ID: mdl-25919187

Estriol biosynthesis in human placenta requires the uptake of a fetal liver-derived estriol precursor, 16α-hydroxydehydroepiandrosterone sulfate (16α-OH DHEAS), by placental syncytiotrophoblasts at their basal plasma membrane (BM), which faces the fetal circulation. The aim of this work is to identify the transporter(s) mediating 16α-OH DHEAS uptake at the fetal side of syncytiotrophoblasts by using human placental BM-enriched vesicles and to examine the contribution of the putative transporter to estriol synthesis at the cellular level, using choriocarcinoma JEG-3 cells. Organic anion transporter (OAT)-4 and organic anion transporting polypeptide 2B1 proteins were enriched in human placental BM vesicles compared with crude membrane fraction. Uptake of [(3)H]16α-OH DHEAS by BM vesicles was partially inhibited in the absence of sodium but was significantly increased in the absence of chloride and after preloading glutarate. Uptake of [(3)H]16α-OH DHEAS by BM vesicles was significantly inhibited by OAT4 substrates such as dehydroepiandrosterone sulfate, estrone-3-sulfate, and bromosulfophthalein but not by cyclosporin A, tetraethylammonium, p-aminohippuric acid, or cimetidine. These characteristics of vesicular [(3)H]16α-OH DHEAS uptake are in good agreement with those of human OAT4-transfected COS-7 cells as well as forskolin-differentiated JEG-3 cells. Estriol secretion from differentiated JEG-3 cells was detected when the cells were incubated with 16α-OH DHEAS for 8 hours but was inhibited in the presence of 50 µM bromosulfophthalein. Our results indicate that OAT4 at the BM of human placental syncytiotrophoblasts plays a predominant role in the uptake of 16α-OH DHEAS for placental estriol synthesis.


Dehydroepiandrosterone/analogs & derivatives , Organic Anion Transporters, Sodium-Independent/metabolism , Trophoblasts/metabolism , Adult , Animals , COS Cells , Cell Line, Tumor , Cell Membrane/metabolism , Chlorocebus aethiops , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Estriol/biosynthesis , Estriol/metabolism , Estrone/analogs & derivatives , Estrone/pharmacology , Female , Fetus , HEK293 Cells , Humans , Male , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Placenta/drug effects , Placenta/metabolism , Pregnancy , Radioisotopes , Sulfobromophthalein/pharmacology , Transport Vesicles/metabolism , Tritium , Trophoblasts/drug effects
20.
J Pharm Sci ; 104(9): 3128-35, 2015 Sep.
Article En | MEDLINE | ID: mdl-25820021

Mechanisms regulating fetal transfer of olmesartan, an angiotensin-II receptor type 1 antagonist, are important as potential determinants of life-threatening adverse fetal effects. The purpose of this study was to examine the olmesartan transport mechanism through the basal plasma membrane (BM) of human syncytiotrophoblasts forming the placental barrier. Uptake of olmesartan by human placental BM vesicles was potently inhibited by dehydroepiandrosterone sulfate (DHEAS), estrone 3-sulfate, and bromosulfophthalein, which are all typical substrates of organic anion transporter (OAT) 4 localized at the BM of syncytiotrophoblasts, and was increased in the absence of chloride. In tetracycline-inducible OAT4-expressing cells, [(3) H]olmesartan uptake was increased by tetracycline treatment. Olmesartan uptake via OAT4 was concentration dependent with a Km of 20 µM, and was increased in the absence of chloride. [(3) H]Olmesartan efflux via OAT4 was also observed and was trans-stimulated by extracellular chloride and DHEAS. Thus, OAT4 mediates bidirectional transport of olmesartan and appears to regulate fetal transfer of olmesartan at the BM of syncytiotrophoblasts. Efflux transport of olmesartan via OAT4 from syncytiotrophoblasts to the fetal circulation might be facilitated in the presence of an inwardly directed physiological chloride gradient and extracellular DHEAS.


Cell Membrane/metabolism , Imidazoles/metabolism , Organic Anion Transporters/metabolism , Placenta/metabolism , Tetrazoles/metabolism , Angiotensin II Type 1 Receptor Blockers/metabolism , Biological Transport/drug effects , Biological Transport/physiology , Cell Membrane/drug effects , Chlorides/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Estrone/analogs & derivatives , Estrone/pharmacology , Female , Humans , Placenta/drug effects , Pregnancy , Sulfobromophthalein/pharmacology , Trophoblasts/drug effects , Trophoblasts/metabolism
...