Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Pharmacol ; 101(2): 68-77, 2022 02.
Article in English | MEDLINE | ID: mdl-34853153

ABSTRACT

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.


Subject(s)
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
2.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34948452

ABSTRACT

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.


Subject(s)
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
3.
J Cell Physiol ; 234(8): 12676-12684, 2019 08.
Article in English | MEDLINE | ID: mdl-30536399

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Lupus Erythematosus, Systemic/metabolism , Adult , Female , Humans , Male , RNA, Messenger/metabolism
4.
Br J Anaesth ; 123(2): e215-e225, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31030988

ABSTRACT

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.


Subject(s)
Dehydroepiandrosterone Sulfate/pharmacology , Pain/physiopathology , Spinal Cord Dorsal Horn/physiopathology , Animals , Disease Models, Animal , Male , Patch-Clamp Techniques , Rats , Rats, Wistar , Synaptic Transmission/physiology
5.
Biochim Biophys Acta Mol Cell Res ; 1864(8): 1382-1392, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28495656

ABSTRACT

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.


Subject(s)
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
6.
Cancer Sci ; 108(10): 2011-2021, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28771887

ABSTRACT

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).


Subject(s)
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
7.
Horm Behav ; 70: 64-72, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25747464

ABSTRACT

One of the mechanisms of cocaine's actions in the central nervous system is its antidepressant action. This effect might be responsible for increased usage of the drug by individuals with mood disorders. Higher endogenous levels of the excitatory neurosteroid dehydroepiandrosterone sulfate (DHEAS) were reported to correlate with successful abstinence from cocaine use in addicts, but a clinical trial showed that supplementation with a high dose of DHEA increased cocaine usage instead. Such ambiguous effects of DHEA(S) could potentially be linked to its influence on the antidepressant effect of cocaine. In this study we tested DHEAS and its metabolite, androsterone, for interactions with cocaine in animal model of depression (forced swim test) and examined the effects of both steroids and cocaine on serotoninergic neurotransmission. All substances were also tested for influence on locomotor activity. A cocaine dose of 5mg/kg, which had no significant effect on locomotor activity, was chosen for the forced swim test. Neither DHEAS nor androsterone showed any antidepressant action in this test, while cocaine manifested a clear antidepressant effect. Androsterone slightly reduced the antidepressant influence of cocaine while DHEAS markedly, dose-dependently enhanced it. Such an effect might be caused by the influence of DHEAS on serotonin neurotransmission, as this steroid decreased serotonin concentration and turnover in the striatum. When DHEAS and cocaine were administered together, the levels of serotonin in the striatum and hippocampus remained unchanged. This phenomenon may explain the additive antidepressant action of DHEAS and cocaine and why co-administration of DHEAS and cocaine increases drug use.


Subject(s)
Androsterone/pharmacology , Antidepressive Agents/pharmacology , Cocaine/pharmacology , Dehydroepiandrosterone Sulfate/pharmacology , Serotonin/physiology , Synaptic Transmission/drug effects , Animals , Brain Chemistry/drug effects , Dose-Response Relationship, Drug , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Swimming/psychology
8.
Biochim Biophys Acta ; 1833(12): 3064-3075, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23988737

ABSTRACT

Dehydroepiandrosterone sulfate (DHEAS) is a circulating steroid produced in the adrenal cortex, brain, and gonads. Whereas a series of investigations attest to neuroprotective effects of the steroid in the brain, surprisingly little is known about the physiological effects of DHEAS on cells of the reproductive system. Here we demonstrate that DHEAS acting on the spermatogenic cell line GC-2 induces a time- and concentration-dependent phosphorylation of c-Src and Erk1/2 and activates the transcription factors activating transforming factor-1 (ATF-1) and cyclic AMP-responsive element binding protein (CREB). These actions are consistent with the non-classical signaling pathway of testosterone and suggest that DHEAS is a pro-androgen that is converted into testosterone in order to exert its biological activity. The fact, however, that steroid sulfatase mRNA was not detected in the GC-2 cells and the clear demonstration of DHEAS-induced activation of Erk1/2, ATF-1 and CREB after silencing the androgen receptor by small interfering RNA (siRNA) clearly contradict this assumption and make it appear unlikely that DHEAS has to be converted in the cytosol into a different steroid in order to activate the kinases and transcription factors mentioned. Instead, it is likely that the DHEAS-induced signaling is mediated through the interaction of the steroid with a membrane-bound G-protein-coupled receptor, since silencing of Guanine nucleotide-binding protein subunit alpha-11 (Gnα11) leads to the abolition of the DHEAS-induced stimulation of Erk1/2, ATF-1, and CREB. The investigation presented here shows a hormone-like activity of DHEAS on a spermatogenic cell line. Since DHEAS is produced in male and female reproductive organs, these findings could help to define new roles for DHEAS in the physiology of reproduction.


Subject(s)
Activating Transcription Factor 1/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptors, G-Protein-Coupled/metabolism , Spermatozoa/metabolism , Animals , Blotting, Western , Cell Line , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique , Gene Silencing/drug effects , Male , Mice , Phosphorylation/drug effects , RNA, Small Interfering/metabolism , Receptors, Androgen/metabolism , Spermatozoa/drug effects , Spermatozoa/enzymology , Steryl-Sulfatase/metabolism , src-Family Kinases/metabolism
9.
FASEB J ; 27(2): 725-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23150523

ABSTRACT

Neurosteroids are cholesterol-derived molecules synthesized within the brain, which exert trophic and protective actions. Infection by human and feline immunodeficiency viruses (HIV and FIV, respectively) causes neuroinflammation and neurodegeneration, leading to neurological deficits. Secretion of neuroinflammatory host and viral factors by glia and infiltrating leukocytes mediates the principal neuropathogenic mechanisms during lentivirus infections, although the effect of neurosteroids on these processes is unknown. We investigated the interactions between neurosteroid-mediated effects and lentivirus infection outcomes. Analyses of HIV-infected (HIV(+)) and uninfected human brains disclosed a reduction in neurosteroid synthesis enzyme expression. Human neurons exposed to supernatants from HIV(+) macrophages exhibited suppressed enzyme expression without reduced cellular viability. HIV(+) human macrophages treated with sulfated dehydroepiandrosterone (DHEA-S) showed suppression of inflammatory gene (IL-1ß, IL-6, TNF-α) expression. FIV-infected (FIV(+)) animals treated daily with 15 mg/kg body weight. DHEA-S treatment reduced inflammatory gene transcripts (IL-1ß, TNF-α, CD3ε, GFAP) in brain compared to vehicle-(ß-cyclodextrin)-treated FIV(+) animals similar to levels found in vehicle-treated FIV(-) animals. DHEA-S treatment also increased CD4(+) T-cell levels and prevented neurobehavioral deficits and neuronal loss among FIV(+) animals, compared to vehicle-treated FIV(+) animals. Reduced neuronal neurosteroid synthesis was evident in lentivirus infections, but treatment with DHEA-S limited neuroinflammation and prevented neurobehavioral deficits. Neurosteroid-derived therapies could be effective in the treatment of virus- or inflammation-mediated neurodegeneration.


Subject(s)
AIDS Dementia Complex/immunology , AIDS Dementia Complex/virology , Brain/immunology , Brain/virology , Dehydroepiandrosterone Sulfate/immunology , Immunity, Innate , AIDS Dementia Complex/metabolism , Animals , Behavior, Animal , Brain/drug effects , Brain/metabolism , Cats , Dehydroepiandrosterone Sulfate/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Feline Acquired Immunodeficiency Syndrome/immunology , Feline Acquired Immunodeficiency Syndrome/psychology , Feline Acquired Immunodeficiency Syndrome/virology , Female , HIV-1/immunology , HIV-1/pathogenicity , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunodeficiency Virus, Feline/immunology , Immunodeficiency Virus, Feline/pathogenicity , Pregnancy , Virulence/drug effects , Virulence/immunology , Virus Replication
10.
Reprod Biomed Online ; 28(2): 191-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24365022

ABSTRACT

Baseline dehydroepiandrostendione sulphate (DHEAS) has been demonstrated to discriminate between young, expected poor responders with favourable clinical pregnancy prospects after IVF treatment and their counterparts with significantly lower pregnancy chances. This study investigated DHEAS ability to predict live birth before starting the first gonadotrophin-releasing hormone (GnRH) antagonist ovarian stimulation for IVF/intracytoplasmic sperm injection in young women (⩽37years) with low serum AMH (<6.5pmol/l). Medical records of 90patients were analysed. DHEAS was predictive for live birth (AUC-ROC 0.69, 95% CI 0.59-0.79). Its predictive accuracy for live birth was similar to that of the number of oocytes retrieved. The cut-off value for DHEAS of 5.4µmol/l offered the best discriminative performance between patients who achieved live birth and those who did not. The live birth rate per initiated cycle in women with DHEAS concentration >5.4µmol/l was 5-fold higher compared with women with DHEAS ⩽5.4µmol/l (38.9% versus 7.4%, P<0.001) despite similar oocyte yield in both groups. In conclusion, the association between baseline DHEAS and probability of live birth after the GnRH antagonist IVF cycle in young women with low AMH was demonstrated. This association could not be explained by the effect of DHEAS on the oocyte yield. This study demonstrates an association of baseline dehydroepinadrostendione sulphate (DHEAS) concentration with implantation rate and live birth rate after the first gonadotrophin-releasing hormone antagonist IVF cycle in young women who are expected to respond poorly to ovarian stimulation. Compared with the number of oocytes retrieved, DHEAS showed a similar ability to distinguish women who will achieve a live birth from those who will not. However, as a parameter available to clinicians and patients before commencing an ovarian stimulation for IVF, DHEAS could be used in predicting probability of live birth at the initial consultation. Women whose DHEAS concentrations were above the derived cut-off value 5.4µmol/l had 5-fold more favourable prognosis for live birth than their counterparts with DHEAS concentrations below the cut-off value. The findings could not be explained by the difference in the oocyte yield and/or the number of embryos transferred but rather by the possible association between the endogenous DHEAS concentration and the oocyte competence to produce a viable embryo.


Subject(s)
Anti-Mullerian Hormone/deficiency , Dehydroepiandrosterone Sulfate/pharmacology , Fertilization in Vitro/methods , Infertility, Female/drug therapy , Infertility, Female/therapy , Adult , Area Under Curve , Croatia , Dehydroepiandrosterone Sulfate/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Humans , Live Birth , Pregnancy , ROC Curve
11.
Biochim Biophys Acta ; 1822(10): 1638-42, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22771498

ABSTRACT

Airway diseases such as asthma, emphysema, and chronic bronchitis are, in part, characterized by reversible airflow obstruction and inflammation. In severe disease, marked decreases in lung function are associated with airway smooth muscle proliferation and airway neutrophilia. Inhaled glucocorticoids attenuate increased airflow obstruction and airway inflammation that occur, in part, due to increased smooth muscle migration and proliferation, as well as the airway neutrophilia. Glucocorticoids, however, have adverse side effects and, in some patients, are ineffective despite high doses. Recent research has explored the effects of non-traditional steroids on attenuation of inflammation associated with airway diseases. These non-traditional steroids have improved side effect profiles in comparison to glucocorticoid therapy. Our studies assessed effects of dehydroepiandrosterone-3-sulfate (DHEA-S) on migration of both human peripheral blood neutrophils (PMN) and human airway smooth muscle cells (HASM). DHEA-S dose-dependently inhibited chemotaxis of PMN and HASM while having no effect on the phosphorylation levels of Akt, ERK1/2, p38 MAPK or PKC, canonical positive regulators of cell migration. These studies demonstrate direct effects of DHEA-S on cell migration, thereby suggesting that DHEA-S may attenuate airway inflammation and cell migration.


Subject(s)
Cell Movement/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Neutrophils/drug effects , Cells, Cultured , Glucocorticoids/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Phosphorylation/drug effects , Platelet-Derived Growth Factor/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Respiratory System/drug effects , Respiratory System/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Biochem Biophys Res Commun ; 440(4): 756-61, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24120945

ABSTRACT

Derived from adrenal cortical, dehydroepiandrosterone-sulfate (DHEAS) is a precursor to androgens and estrogens, with various bioactivities. Although it has the property of anti-diabetes, the long-term effect of DHEAS on insulin secretion in beta-cells is still unclear. In this study, the effect of DHEAS on the insulin secretion activity in MIN6 cell lines in vitro was assessed. Insulin biosynthesis and secretion were stimulated by DHEAS for 24h. DHEAS inhibited the AMPK activation and upregulated the expression of ACC-1. These findings indicate that DHEAS may exert prominent stimulatory effects on insulin secretion partly via AMPK inhibition and ACC-1 upregulation.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Dehydroepiandrosterone Sulfate/pharmacology , Insulin-Secreting Cells/drug effects , Insulin/metabolism , AMP-Activated Protein Kinases/metabolism , Acetyltransferases/biosynthesis , Acetyltransferases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Line , Hypoglycemic Agents/pharmacology , Insulin/biosynthesis , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Phosphorylation/drug effects , RNA, Messenger/biosynthesis , Ribonucleotides/pharmacology
13.
Eur Respir J ; 42(5): 1309-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23143540

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an ageing-related lung disorder characterised by expansion of the myofibroblast population and aberrant lung remodelling. Dehydroepiandrosterone (DHEA), a steroid pro-hormone, decreases with age but an exaggerated decline has been associated with chronic degenerative diseases. We quantified the plasma levels of DHEA and its sulfated form (DHEA-S) in 137 IPF patients and 58 controls and examined the effects of DHEA on human lung fibroblasts. Plasma DHEA/DHEA-S was significantly decreased in male IPF patients (median (range) DHEA: 4.4 (0.2-29.2) versus 6.7 (2.1-15.2) ng · mL(-1), p<0.01; DHEA-S: 47 (15.0-211) versus 85.2 (37.6-247.0) µg · dL(-1), p<0.001), while in females only DHEA-S was significantly decreased (32.6 (15.0-303.0) versus 68.3 (16.4-171) µg · dL(-1), p<0.001). DHEA caused a decrease in fibroblast proliferation and an approximately two-fold increase in fibroblast apoptosis, probably through the intrinsic pathway with activation of caspase-9. This effect was accompanied by upregulation of several pro-apoptotic proteins (Bax and cyclin-dependent kinase-inhibitor CDNK1A) and downregulation of anti-apoptotic proteins, such as cellular inhibitor of apoptosis (c-IAP)1 and c-IAP2. DHEA also caused a significant decrease of transforming growth factor-ß1-induced collagen production and fibroblast to myofibroblast differentiation, and inhibited platelet-derived growth factor-induced fibroblast migration. These findings demonstrate a disproportionate decrease of DHEA/DHEA-S in IPF patients and indicate that this molecule has multiple antifibrotic properties.


Subject(s)
Dehydroepiandrosterone/blood , Dehydroepiandrosterone/pharmacology , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Aged , Apoptosis , Bronchoalveolar Lavage , Case-Control Studies , Caspase 9/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Collagen/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Female , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Male , Middle Aged , Platelet-Derived Growth Factor/metabolism , Transforming Growth Factor beta1/metabolism
14.
Bull Exp Biol Med ; 156(1): 35-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24319722

ABSTRACT

Dehydroepiandrosterone sulfate (30 mg/kg) produced a hypotensive effect by preventing stress-induced surge of hypertensive hormone aldosterone in rats after manifold repeated, but not single stress exposure. Both effects were realized via µ-opioid receptors. Thus, µ-opioid mechanism of blockage of aldosterone surge can underlay the hypotensive effects of dehydroepiandrosterone sulfate at least under conditions of manifold repeated exposures.


Subject(s)
Aldosterone/blood , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Receptors, Opioid, mu/metabolism , Animals , Male , Rats , Rats, Wistar , Stress, Psychological/blood , Stress, Psychological/physiopathology
15.
Mini Rev Med Chem ; 23(8): 941-952, 2023.
Article in English | MEDLINE | ID: mdl-36121077

ABSTRACT

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.


Subject(s)
Dehydroepiandrosterone , Endothelial Cells , Animals , Humans , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Endothelial Cells/metabolism , Dehydroepiandrosterone Sulfate/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Brain/metabolism , Steroids
16.
Hippocampus ; 22(4): 707-22, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21484933

ABSTRACT

The neurosteroid dehydroepiandrosterone-sulfate (DHEAS) is a positive modulator of synaptic transmission in mammalian brains; however, the underlying molecular mechanisms are not fully understood. This report describes the acute effects of DHEAS on the synaptic transmission in the hippocampal dentate gyrus of rat brain slices. The application of DHEAS for 10 min augmented the optically recorded EPSP (op-EPSP) in a dose dependent manner. The effect became visible at 1 nM and saturated at 100 nM. We focused on the effect of DHEAS at 100 nM, where the op-EPSP amplitude was increased by 30%, and gradually decreased to the basal level in 30 min after wash out of the drug (short-term potentiation by DHEAS; STP(DHEAS)). DHEAS did not alter the presynaptic properties including the presynaptic fiber volley (PSFV) and paired pulse facilitation (PPF), thus indicating that the acute DHEAS effect is of postsynaptic origin. The involvement of putative DHEAS targets, GABA(A), NMDA, and σ1 receptors in STP(DHEAS) was also investigated; however, antagonists to these receptors only partially inhibited the acute effect of DHEAS. By contrast, STP(DHEAS) was totally inhibited by either the metabotropic glutamate receptor 5 (mGluR5) antagonist MPEP (10 µM) or the ryanodine receptor (RyR) inhibitors (ryanodine and ruthenium red), but not by the mGluR1 antagonist LY367385 and the IP3R antagonist 2-APB, suggesting that STP(DHEAS) is mediated by an mGluR5-RyR cascade in postsynaptic neurons. Consistent with this finding, the selective agonist for mGluR5 CHPG nearly perfectly mimicked the DHEAS effect. This is the first demonstration of mGluR involvement in the DHEAS action in regard to hippocampal synaptic transmission.


Subject(s)
Dehydroepiandrosterone Sulfate/pharmacology , Hippocampus/drug effects , Hippocampus/physiology , Receptors, Metabotropic Glutamate/agonists , Synaptic Transmission/drug effects , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Dehydroepiandrosterone Sulfate/administration & dosage , Dentate Gyrus/drug effects , Dentate Gyrus/physiology , Electric Stimulation , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glycine/analogs & derivatives , Glycine/pharmacology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Phenylacetates/pharmacology , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Synaptic Transmission/physiology
17.
J Pharmacol Sci ; 120(1): 54-8, 2012.
Article in English | MEDLINE | ID: mdl-22878600

ABSTRACT

Neurosteroids are known as allosteric modulators of the ligand-gated ion channel superfamily. Voltage-gated sodium channels (Na(v)) play an important role in mediating excitotoxic damages. Here we report the effects of neurosteroids on the function of Na(v), using voltage-clamp techniques in Xenopus oocytes expressed with the Na(v)1.2 α subunit. Pregnenolone sulphate, but not pregnenolone, inhibited sodium currents (I(Na)) at 3 - 100 µmol/L. The suppression of I(Na) by pregnenolone sulphate was due to increased inactivation with little change in activation. These findings suggest that pregnenolone sulphate, a metabolite of pregnenolone, suppresses the function of Na(v) via increased inactivation, which may contribute to the neuroprotection.


Subject(s)
NAV1.2 Voltage-Gated Sodium Channel/physiology , Pregnenolone/pharmacology , Sodium Channel Blockers/pharmacology , Animals , Cells, Cultured , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone Sulfate/pharmacology , Female , Oocytes/drug effects , Oocytes/physiology , Pregnenolone/metabolism , Xenopus laevis
18.
Pharmacology ; 89(3-4): 163-71, 2012.
Article in English | MEDLINE | ID: mdl-22433179

ABSTRACT

AIM: To investigate whether long-term exposure to the neurosteroid dehydroepiandrosterone sulfate (DHEAS) induces adaptive changes of GABA(A) receptors related to the development of tolerance and dependence. METHODS: We compared the parameters of [(3)H]DHEAS binding and the effects of DHEAS on [(3)H]flunitrazepam binding in the membranes of HEK 293 cells, nontransfected or stably transfected with recombinant α(1)ß(2)γ(2S) GABA(A) receptors. In HEK 293 cells expressing α(1)ß(2)γ(2S) GABA(A) receptors, we investigated the effects of long-term DHEAS treatment on the [(3)H]flunitrazepam and [(3)H]t-butylbicycloorthobenzoate ([(3)H]TBOB) binding and on their modulation with GABA. RESULTS: DHEAS behaves as an allosteric antagonist of the recombinant α(1)ß(2)γ(2S) GABA(A) receptors expressed in HEK 293 cells. Exposure of cells to 100 µmol/l DHEAS for 48 h did not change the number or affinity of benzodiazepine and convulsive binding sites. Long-term DHEAS treatment failed to affect functional allosteric interactions between GABA(A) receptor binding sites, as evidenced by an unchanged ability of GABA to stimulate or to inhibit [(3)H]flunitrazepam and [(3)H]TBOB binding, respectively. CONCLUSION: The findings that prolonged DHEAS treatment does not produce changes in GABA(A) receptor expression and functional coupling, assumed to underlie the development of tolerance and dependence, might have importance in the long-term therapy necessary for the observed beneficial effects of DHEAS.


Subject(s)
Dehydroepiandrosterone Sulfate/pharmacology , Receptors, GABA-A/metabolism , Recombinant Proteins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Cell Membrane/metabolism , Flunitrazepam/metabolism , HEK293 Cells , Humans
19.
Fiziol Cheloveka ; 38(5): 97-101, 2012.
Article in Russian | MEDLINE | ID: mdl-23101246

ABSTRACT

Effects of dehydroepiandrosteron sulfate on oxidative stress- and ethanol-induced morphological changes and level of apoptotic enzymes (Bcl-2 and caspase-3) of lymphocytes in healthy persons have been studied. This study showed that hydroperoxide induces apoptotic changes in lymphocytes. Ethanol did not change the basal concentration of apoptotic enzyme. Dehydroepiandrosteron sulfate prevented the augmentation of apoptosis in hydroperoxide-treated lymphocytes.


Subject(s)
Apoptosis/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Lymphocytes/metabolism , Oxidative Stress/drug effects , Adult , Caspase 3/metabolism , Ethanol/pharmacology , Female , Humans , Lymphocytes/cytology , Male , Proto-Oncogene Proteins c-bcl-2/metabolism , Solvents/pharmacology
20.
Horm Metab Res ; 43(10): 702-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21932174

ABSTRACT

Dehydroepiandrosterone (DHEA) and its sulfonated form dehydroepiandrosterone sulfate (DHEAS) are the main circulating steroid hormones and many epidemiological studies show an inverse relationship between DHEA/DHEAS levels and muscle loss for which the primary cause is the accelerated protein breakdown. The aim of this work was to determine whether DHEA/DHEAS supplementation in differentiating C2C12 skeletal muscle cells might influence the expression of the atrophy-related ubiquitin ligase, MuRF-1, and thereby impact key molecules of the differentiation program. DHEA is the prohormone crucial for sex steroid synthesis, and DHEAS is thought to be its reservoir. However, our preliminary experiments showed that DHEAS, but not DHEA, is able to influence MuRF-1 expression. Therefore, we treated differentiating C2C12 cells with various concentrations of DHEAS and analyzed the expression of MuRF-1, Hsp70, myosin heavy chain (MHC), myogenin, and the activity of creatine kinase. We observed that DHEAS at physiological concentrations downregulates MuRF-1 expression and affects muscle differentiation, as shown by the increased levels of MHC, which is a sarcomeric protein that undergoes MuRF-1-dependent degradation, and also by an increase in creatine kinase activity and myogenin expression, which are two other well-known markers of differentiation. Moreover, we found that DHEAS might have a protective effect on differentiating cells as suggested by the augmented levels of Hsp70, a member of heat shock proteins family that, besides its cytoprotective action, seems to have a regulatory role on key atrophy genes such as MuRF-1. In conclusion, our data shed light on the role of DHEAS at physiologic concentrations in maintaining muscle mass.


Subject(s)
Cell Differentiation/drug effects , Dehydroepiandrosterone Sulfate/pharmacology , Muscle, Skeletal/cytology , Animals , Cell Line , Creatine Kinase/metabolism , Gene Expression Regulation/drug effects , HSP70 Heat-Shock Proteins/metabolism , Mice , Muscle Cells/cytology , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle Proteins/drug effects , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myogenin/metabolism , Myosin Heavy Chains/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/drug effects , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL