Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters










Publication year range
1.
ACS Chem Neurosci ; 14(10): 1870-1883, 2023 05 17.
Article in English | MEDLINE | ID: mdl-37126803

ABSTRACT

Multiple molecular targets have been identified to mediate membrane-delimited and nongenomic effects of natural and synthetic steroids, but the influence of steroid metabolism on neuroactive steroid signaling is not well understood. To begin to address this question, we set out to identify major metabolites of a neuroprotective synthetic steroid 20-oxo-5ß-pregnan-3α-yl l-glutamyl 1-ester (pregnanolone glutamate, PAG) and characterize their effects on GABAA and NMDA receptors (GABARs, NMDARs) and their influence on zebrafish behavior. Gas chromatography-mass spectrometry was used to assess concentrations of PAG and its metabolites in the hippocampal tissue of juvenile rats following intraperitoneal PAG injection. PAG is metabolized in the peripheral organs and nervous tissue to 20-oxo-17α-hydroxy-5ß-pregnan-3α-yl l-glutamyl 1-ester (17-hydroxypregnanolone glutamate, 17-OH-PAG), 3α-hydroxy-5ß-pregnan-20-one (pregnanolone, PA), and 3α,17α-dihydroxy-5ß-pregnan-20-one (17-hydroxypregnanolone, 17-OH-PA). Patch-clamp electrophysiology experiments in cultured hippocampal neurons demonstrate that PA and 17-OH-PA are potent positive modulators of GABARs, while PAG and 17-OH-PA have a moderate inhibitory effect at NMDARs. PAG, 17-OH-PA, and PA diminished the locomotor activity of zebrafish larvae in a dose-dependent manner. Our results show that PAG and its metabolites are potent modulators of neurotransmitter receptors with behavioral consequences and indicate that neurosteroid-based ligands may have therapeutic potential.


Subject(s)
Pregnanolone , Receptors, N-Methyl-D-Aspartate , Rats , Animals , Pregnanolone/pharmacology , Pregnanolone/chemistry , Zebrafish , Glutamic Acid , Esters , gamma-Aminobutyric Acid , Receptors, GABA-A
2.
Curr Neuropharmacol ; 20(5): 886-890, 2022.
Article in English | MEDLINE | ID: mdl-34856904

ABSTRACT

Neurosteroids are endogenous modulators of GABAA receptors that mediate anxiety, pain, mood and arousal. The 3-hydroxyl epimers, allopregnanolone (3α-OH) and epiallopregnanolone (3ß-OH) are both prevalent in the mammalian brain and produce opposite effects on GABAA receptor function, acting as positive and negative allosteric modulators, respectively. This Perspective provides a model to explain the actions of 3α-OH and 3ß-OH neurosteroids. The model is based on evidence that the neurosteroid epimers bind to an overlapping subset of specific sites on GABAA receptors, with their net functional effect on channel gating being the sum of their independent effects at each site.


Subject(s)
Neurosteroids , Animals , Binding Sites , Humans , Mammals/metabolism , Pregnanolone/chemistry , Pregnanolone/metabolism , Receptors, GABA-A/chemistry , gamma-Aminobutyric Acid
3.
Mol Pharmacol ; 100(1): 19-31, 2021 07.
Article in English | MEDLINE | ID: mdl-33958479

ABSTRACT

Prior work employing functional analysis, photolabeling, and X-ray crystallography have identified three distinct binding sites for potentiating steroids in the heteromeric GABAA receptor. The sites are located in the membrane-spanning domains of the receptor at the ß-α subunit interface (site I) and within the α (site II) and ß subunits (site III). Here, we have investigated the effects of mutations to these sites on potentiation of the rat α1ß2γ2L GABAA receptor by the endogenous neurosteroid allopregnanolone (3α5αP). The mutations were introduced alone or in combination to probe the additivity of effects. We show that the effects of amino acid substitutions in sites I and II are energetically additive, indicating independence of the actions of the two steroid binding sites. In site III, none of the mutations tested reduced potentiation by 3α5αP, nor did a mutation in site III modify the effects of mutations in sites I or II. We infer that the binding sites for 3α5αP act independently. The independence of steroid action at each site is supported by photolabeling data showing that mutations in either site I or site II selectively change steroid orientation in the mutated site without affecting labeling at the unmutated site. The findings are discussed in the context of linking energetic additivity to empirical changes in receptor function and ligand binding. SIGNIFICANCE STATEMENT: Prior work has identified three distinct binding sites for potentiating steroids in the heteromeric γ-aminobutyric acid type A receptor. This study shows that the sites act independently and additively in the presence of the steroid allopregnanolone and provide estimates of energetic contributions made by steroid binding to each site.


Subject(s)
Amino Acid Substitution , Pregnanolone/pharmacology , Receptors, GABA-A/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Pregnanolone/chemistry , Rats , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism
4.
Cells ; 10(3)2021 03 21.
Article in English | MEDLINE | ID: mdl-33801063

ABSTRACT

Microglial cells are key players in neural pathogenesis and microglial function regulation appears to be pivotal in controlling neuroinflammatory/neurological diseases. Here, we investigated the effects and mechanism of action of neurosteroid allopregnanolone (ALLO) on murine microglial BV-2 cells and primary microglia in order to determine ALLO-induced immunomodulatory potential and to provide new insights for the development of both natural and safe neuroprotective strategies targeting microglia. Indeed, ALLO-treatment is increasingly suggested as beneficial in various models of neurological disorders but the underlying mechanisms have not been elucidated. Therefore, the microglial cells were cultured with various serum concentrations to mimic the blood-brain-barrier rupture and to induce their activation. Proliferation, viability, RT-qPCR, phagocytosis, and morphology analyzes, as well as migration with time-lapse imaging and quantitative morphodynamic methods, were combined to investigate ALLO actions on microglia. BV-2 cells express subunits of GABA-A receptor that mediates ALLO activity. ALLO (10µM) induced microglial cell process extension and decreased migratory capacity. Interestingly, ALLO modulated the phagocytic activity of BV-2 cells and primary microglia. Our results, which show a direct effect of ALLO on microglial morphology and phagocytic function, suggest that the natural neurosteroid-based approach may contribute to developing effective strategies against neurological disorders that are evoked by microglia-related abnormalities.


Subject(s)
Cell Shape , Microglia/cytology , Microglia/metabolism , Neuroprotection , Neurosteroids/metabolism , Phagocytosis , Pregnanolone/metabolism , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Shape/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation/drug effects , Humans , Mice, Inbred C57BL , Microglia/drug effects , Models, Biological , Neuroprotection/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Phagocytosis/drug effects , Pregnanolone/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA/genetics , Receptors, GABA/metabolism , Serum
5.
Elife ; 92020 09 21.
Article in English | MEDLINE | ID: mdl-32955433

ABSTRACT

This study examines how site-specific binding to three identified neurosteroid-binding sites in the α1ß3 GABAA receptor (GABAAR) contributes to neurosteroid allosteric modulation. We found that the potentiating neurosteroid, allopregnanolone, but not its inhibitory 3ß-epimer epi-allopregnanolone, binds to the canonical ß3(+)-α1(-) intersubunit site that mediates receptor activation by neurosteroids. In contrast, both allopregnanolone and epi-allopregnanolone bind to intrasubunit sites in the ß3 subunit, promoting receptor desensitization and the α1 subunit promoting effects that vary between neurosteroids. Two neurosteroid analogues with diazirine moieties replacing the 3-hydroxyl (KK148 and KK150) bind to all three sites, but do not potentiate GABAAR currents. KK148 is a desensitizing agent, whereas KK150 is devoid of allosteric activity. These compounds provide potential chemical scaffolds for neurosteroid antagonists. Collectively, these data show that differential occupancy and efficacy at three discrete neurosteroid-binding sites determine whether a neurosteroid has potentiating, inhibitory, or competitive antagonist activity on GABAARs.


Subject(s)
Neurosteroids , Receptors, GABA-A , Animals , Binding Sites , Cells, Cultured , Electrophysiological Phenomena/drug effects , Molecular Docking Simulation , Neurosteroids/antagonists & inhibitors , Neurosteroids/chemistry , Neurosteroids/metabolism , Neurosteroids/pharmacology , Oocytes/metabolism , Pregnanolone/chemistry , Pregnanolone/metabolism , Pregnanolone/pharmacology , Protein Binding , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Xenopus laevis
6.
Mol Pharmacol ; 98(4): 280-291, 2020 10.
Article in English | MEDLINE | ID: mdl-32675382

ABSTRACT

The ρ1 GABAA receptor is prominently expressed in the retina and is present at lower levels in several brain regions and other tissues. Although the ρ1 receptor is insensitive to many anesthetic drugs that modulate the heteromeric GABAA receptor, it maintains a rich and multifaceted steroid pharmacology. The receptor is negatively modulated by 5ß-reduced steroids, sulfated or carboxylated steroids, and ß-estradiol, whereas many 5α-reduced steroids potentiate the receptor. In this study, we analyzed modulation of the human ρ1 GABAA receptor by several neurosteroids, individually and in combination, in the framework of the coagonist concerted transition model. Experiments involving coapplication of two or more steroids revealed that the receptor contains at least three classes of distinct, nonoverlapping sites for steroids, one each for the inhibitory steroids pregnanolone (3α5ßP), 3α5ßP sulfate, and ß-estradiol. The site for 3α5ßP can accommodate the potentiating steroid 5αTHDOC. The findings are discussed with respect to receptor modulation by combinations of endogenous neurosteroids. SIGNIFICANCE STATEMENT: The study describes modulation of the ρ1 GABAA receptor by neurosteroids. The coagonist concerted transition model was used to determine overlap of binding sites for several inhibitory and potentiating steroids.


Subject(s)
Desoxycorticosterone/analogs & derivatives , Neurosteroids/pharmacology , Pregnanolone/pharmacology , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Xenopus laevis/genetics , Animals , Animals, Genetically Modified , Binding Sites , Desoxycorticosterone/chemistry , Desoxycorticosterone/pharmacology , Drug Synergism , Drug Therapy, Combination , Humans , Models, Molecular , Molecular Structure , Neurosteroids/chemistry , Pregnanolone/chemistry , Receptors, GABA-A/genetics
7.
Sci Rep ; 9(1): 15997, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31690811

ABSTRACT

Activation of GABAA receptors consisting of α4, ß2 (or ß3), and δ subunits is a major contributor to tonic inhibition in several brain regions. The goal of this study was to analyze the function of the α4ß2δ receptor in the presence of GABA and other endogenous and clinical activators and modulators under steady-state conditions. We show that the receptor has a high constitutive open probability (~0.1), but is only weakly activated by GABA that has a maximal peak open probability (POpen,peak) of 0.4, taurine (maximal POpen,peak = 0.4), or the endogenous steroid allopregnanolone (maximal POpen,peak = 0.2). The intravenous anesthetic propofol is a full agonist (maximal POpen,peak = 0.99). Analysis of currents using a cyclic three-state Resting-Active-Desensitized model indicates that the maximal steady-state open probability of the α4ß2δ receptor is ~0.45. Steady-state open probability in the presence of combinations of GABA, taurine, propofol, allopregnanolone and/or the inhibitory steroid pregnenolone sulfate closely matched predicted open probability calculated assuming energetic additivity. The results suggest that the receptor is active in the presence of physiological concentrations of GABA and taurine, but, surprisingly, that receptor activity is only weakly potentiated by propofol.


Subject(s)
Receptors, GABA-A/chemistry , Animals , GABA-A Receptor Agonists/chemistry , GABA-A Receptor Agonists/metabolism , Humans , Kinetics , Pregnanolone/chemistry , Pregnanolone/metabolism , Propofol/chemistry , Propofol/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Xenopus laevis
8.
J Biol Chem ; 294(19): 7892-7903, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30923128

ABSTRACT

Many neuroactive steroids potently and allosterically modulate pentameric ligand-gated ion channels, including GABAA receptors (GABAAR) and nicotinic acetylcholine receptors (nAChRs). Allopregnanolone and its synthetic analog alphaxalone are GABAAR-positive allosteric modulators (PAMs), whereas alphaxalone and most neuroactive steroids are nAChR inhibitors. In this report, we used 11ß-(p-azidotetrafluorobenzoyloxy)allopregnanolone (F4N3Bzoxy-AP), a general anesthetic and photoreactive allopregnanolone analog that is a potent GABAAR PAM, to characterize steroid-binding sites in the Torpedo α2ßγδ nAChR in its native membrane environment. We found that F4N3Bzoxy-AP (IC50 = 31 µm) is 7-fold more potent than alphaxalone in inhibiting binding of the channel blocker [3H]tenocyclidine to nAChRs in the desensitized state. At 300 µm, neither steroid inhibited binding of [3H]tetracaine, a closed-state selective channel blocker, or of [3H]acetylcholine. Photolabeling identified three distinct [3H]F4N3Bzoxy-AP-binding sites in the nAChR transmembrane domain: 1) in the ion channel, identified by photolabeling in the M2 helices of ßVal-261 and δVal-269 (position M2-13'); 2) at the interface between the αM1 and αM4 helices, identified by photolabeling in αM1 (αCys-222/αLeu-223); and 3) at the lipid-protein interface involving γTrp-453 (M4), a residue photolabeled by small lipophilic probes and promegestone, a steroid nAChR antagonist. Photolabeling in the ion channel and αM1 was higher in the nAChR-desensitized state than in the resting state and inhibitable by promegestone. These results directly indicate a steroid-binding site in the nAChR ion channel and identify additional steroid-binding sites also occupied by other lipophilic nAChR antagonists.


Subject(s)
Fish Proteins/chemistry , Molecular Docking Simulation , Pregnanolone , Receptors, Nicotinic/chemistry , Steroids/chemistry , Animals , Binding Sites , Fish Proteins/metabolism , Pregnanolone/analogs & derivatives , Pregnanolone/chemistry , Receptors, Nicotinic/metabolism , Steroids/metabolism , Tetracaine/chemistry , Torpedo
9.
Channels (Austin) ; 13(1): 48-61, 2019 12.
Article in English | MEDLINE | ID: mdl-30672394

ABSTRACT

Voltage-activated calcium channels play an important role in excitability of sensory nociceptive neurons in acute and chronic pain models. We have previously shown that low-voltage-activated calcium channels, or T-type channels (T-channels), increase excitability of sensory neurons after surgical incision in rats. We have also found that endogenous 5ß-reduced neuroactive steroid epipregnanolone [(3ß,5ß)-3-hydroxypregnan-20-one] blocked isolated T-currents in dorsal root ganglion (DRG) cells in vitro, and reduced nociceptive behavior in vivo, after local intraplantar application into the foot pads of heathy rats and mice. Here, we investigated if epipregnanolone exerts an antinociceptive effect after intrathecal (i.t.) application in healthy rats, as well as an antihyperalgesic effect in a postsurgical pain model. We also studied if this endogenous neurosteroid blocks currents originating from high voltage-activated (HVA) calcium channels in rat sensory neurons. In in vivo studies, we found that epipregnanolone alleviated thermal and mechanical nociception in healthy rats after i.t. administration without affecting their sensory-motor abilities. Furthermore, epipregnanolone effectively reduced mechanical hyperalgesia after i.t application in rats after surgery. In subsequent in vitro studies, we found that epipregnanolone blocked isolated HVA currents in nociceptive sensory neurons with an IC50 of 3.3 µM in a G-protein-dependent fashion. We conclude that neurosteroids that have combined inhibitory effects on T-type and HVA calcium currents may be suitable for development of novel pain therapies during the perioperative period.


Subject(s)
Anesthetics/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Pregnanolone/pharmacology , Spinal Cord/drug effects , Surgical Wound/drug therapy , Anesthetics/administration & dosage , Anesthetics/chemistry , Animals , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/chemistry , Electrophysiology , Female , Injections, Spinal , Models, Animal , Molecular Conformation , Patch-Clamp Techniques , Pregnanolone/administration & dosage , Pregnanolone/chemistry , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism
10.
Neurochem Int ; 118: 145-151, 2018 09.
Article in English | MEDLINE | ID: mdl-29886074

ABSTRACT

The ability of pregnanolone glutamate (PA-Glu), pregnanolone hemisuccinate (PA-hSuc) and pregnanolone hemipimelate (PA-hPim), neuroactive steroids with a negative modulatory effect on excitatory N-methyl-d-aspartate receptors, to influence the functional activity of inhibitory γ-aminobutyric acid and glycine receptors was estimated. The GABA- and glycine-induced chloride currents (IGABA and IGly) were measured in isolated pyramidal neurons of the rat hippocampus using the patch-clamp technique. Compound PA-Glu was found to potentiate IGABA and to inhibit IGly, while PA-hSuc and PA-hPim inhibited both IGABA and IGly. Moreover, PA-Glu, PA-hSuc, and PA-hPim had a greater effect on desensitization than on the peak amplitude of IGly. At a high concentration of glycine (500 µM), the effect of neurosteroids on the peak amplitude of IGly disappeared, and the acceleration of desensitization remained. The conversion of PA-Glu into androstane glutamate (AND-Glu), an analogue that lacks the C-17 acetyl moiety, completely eliminated the effects on these receptors. Our results indicate that the C-17 acetyl moiety is crucial for the action on IGABA and IGly. Our results indicate that the pregnanolone derivatives, in contrast to the androstane analogues, modulate IGABA and IGly at low micromolar concentrations and this family of neurosteroids can be useful for future structure-activity relationship studies of the steroid modulation of other receptor types.


Subject(s)
Hippocampus/physiology , Neurons/physiology , Pregnanolone/pharmacology , Receptors, GABA-A/physiology , Receptors, Glycine/physiology , Animals , Dose-Response Relationship, Drug , Hippocampus/drug effects , Neurons/drug effects , Neurotransmitter Agents/chemistry , Neurotransmitter Agents/pharmacology , Organ Culture Techniques , Pregnanolone/chemistry , Pyramidal Cells , Rats , Rats, Wistar
11.
J Steroid Biochem Mol Biol ; 182: 72-80, 2018 09.
Article in English | MEDLINE | ID: mdl-29705269

ABSTRACT

Neurosteroids are the principal endogenous modulators of the γ-Aminobutyric acid receptors (GABAARs), pentameric membrane-bound proteins that can be assembled from at least 19 subunits. In the most abundant GABAAR arrangement (α1ß2γ2), neurosteroids can potentiate the GABA action as well as produce a direct activation of the channel. The recent crystal structures of neurosteroids bound to α homopentameric GABAAR reveal binding to five equivalent sites. However, these results have been obtained using receptors that are not physiologically relevant, suggesting a need to investigate neurosteroid binding to heteropentameric receptors that exist in the central nervous system. In a previous work, we predicted the neurosteroid binding site by applying molecular modeling methods on the ß3 homopentamer. Here we construct a homology model of the transmembrane domain of the heteropentameric α1ß2γ2 receptor and then, by combining docking and molecular dynamics simulations, we analyzed neurosteroid binding. Results show that the five neurosteroid cavities are conserved in the α1ß2γ2 receptor and all of them are able to bind neurosteroids. Two different binding modes were detected depending on the identity of the residue at position 241 in the transmembrane helix 1. These theoretical findings provide microscopic insights into neurosteroid binding at the heteropentameric GABAAR. The existence of two classes of sites may be associated with how neurosteroids modulate GABAAR. Our finding would represent the essential first step to reach a comprehensive understanding of how these endogenous molecules regulate the central nervous system.


Subject(s)
Pregnanolone/chemistry , Pregnanolone/metabolism , Protein Conformation , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Amino Acid Sequence , Binding Sites , Humans , Models, Molecular , Molecular Dynamics Simulation , Protein Domains , Receptors, GABA-A/classification , Sequence Homology
12.
J Pharmacol Exp Ther ; 365(3): 583-601, 2018 06.
Article in English | MEDLINE | ID: mdl-29602830

ABSTRACT

Neurosteroids are powerful modulators of γ-aminobutyric acid (GABA)-A receptors. Ganaxolone (3α-hydroxy-3ß-methyl-5α-pregnan-20-one, GX) and synthetic analogs of the neurosteroid allopregnanolone (AP) are designed to treat epilepsy and related conditions. However, their precise mechanism of action in native neurons remains unclear. Here, we sought to determine the mode of action of GX and its analogs at GABA-A receptors in native hippocampal neurons by analyzing extrasynaptic receptor-mediated tonic currents and synaptic receptor-mediated phasic currents. Concentration-response profiles of GX were determined in two cell types: δ-containing dentate gyrus granule cells (DGGCs) and γ2-containing CA1 pyramidal cells (CA1PCs). GX produced significantly greater potentiation of the GABA-A receptor-activated chloride currents in DGGCs (500%) than CA1PCs (200%). In the absence of GABA, GX evoked 2-fold greater inward currents in DGGCs than CA1PCs, which were 2-fold greater than AP within DGGCs. In hippocampus slices, GX potentiated and directly activated tonic currents in DGGCs. These responses were significantly diminished in DGGCs from δ-subunit knockout (δKO) mice, confirming GX's selectivity for δGABA-A receptors. Like AP, GX potentiation of tonic currents was prevented by protein kinase C inhibition. Furthermore, GX's protection against hippocampus-kindled seizures was significantly diminished in δKO mice. GX analogs exhibited greater potency and efficacy than GX on δGABA-A receptor-mediated tonic inhibition. In summary, these results provide strong evidence that GX and its analogs are preferential allosteric modulators and direct activators of extrasynaptic δGABA-A receptors regulating network inhibition and seizures in the dentate gyrus. Therefore, these findings provide a mechanistic rationale for the clinical use of synthetic neurosteroids in epilepsy and seizure disorders.


Subject(s)
Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , GABA-A Receptor Antagonists/chemistry , GABA-A Receptor Antagonists/pharmacology , Pregnanolone/analogs & derivatives , Receptors, GABA-A/metabolism , Allosteric Regulation/drug effects , Animals , Dentate Gyrus/cytology , GABA-A Receptor Antagonists/therapeutic use , Ion Channel Gating/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Pregnanolone/chemistry , Pregnanolone/pharmacology , Pregnanolone/therapeutic use , Protein Kinase C/antagonists & inhibitors , Seizures/drug therapy , Synapses/drug effects , Synapses/metabolism
13.
Regen Med ; 13(2): 141-157, 2018 03.
Article in English | MEDLINE | ID: mdl-29160149

ABSTRACT

AIM: This in vitro and in vivo study reports on silk fibroin (SF) scaffold, functionalized for in situ delivery of GABA and/or allopregnanolone (ALLO), as biomaterial for potential application in tissue engineering and nerve regeneration. MATERIALS & METHODS: We evaluated the feasibility to design 2D scaffolds (films) made of regenerated Bombyx mori SF, functionalized with GABA and/or ALLO to enhance in vitro biological functions, health, survival and growth of Schwann cells and sensitive neurons of the dorsal root ganglia. RESULTS & CONCLUSION: Our 2D-SF film showed an efficient loading and controllable release of drugs promoting nerve regeneration. SF functionalized film may be helpful for the development of bioengineered conduits and, in principle, have great potential for long-gap nerve injury repair.


Subject(s)
Fibroins/physiology , Neurons/cytology , Schwann Cells/cytology , Silk , Tissue Engineering/methods , Animals , Biocompatible Materials , Bombyx , Fibroins/chemistry , Materials Testing , Nerve Regeneration , Pregnanolone/chemistry , Pregnanolone/physiology , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/physiology
14.
Steroids ; 128: 1-5, 2017 12.
Article in English | MEDLINE | ID: mdl-29024671

ABSTRACT

Treatment of steroid sapogenins with H2O2 in CF3COOH for 15min followed by reflux in CH3OH/H2O afforded good yields of pregnan-3ß,16ß,20-triol 3-monoacetates. When the hydrolysis step was carried out with KOH in refluxing methanol excellent yields pregnantriols were obtained. The resulting compounds were characterized by their melting points and NMR spectral data. An X-ray diffraction analysis of compound 3a confirmed the proposed structure and provided detailed information about the bond lengths, bond angles and conformation.


Subject(s)
Pregnanolone/chemistry , Sapogenins/chemistry , Steroids/chemistry , Crystallography, X-Ray , Hydrogen Peroxide/chemistry , Hydrolysis , Magnetic Resonance Spectroscopy , Molecular Conformation , Molecular Structure , Sapogenins/chemical synthesis , Stereoisomerism , Steroids/chemical synthesis , Transition Temperature
15.
J Med Chem ; 60(18): 7810-7819, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28753313

ABSTRACT

Certain classes of neuroactive steroids (NASs) are positive allosteric modulators (PAM) of synaptic and extrasynaptic GABAA receptors. Herein, we report new SAR insights in a series of 5ß-nor-19-pregnan-20-one analogues bearing substituted pyrazoles and triazoles at C-21, culminating in the discovery of 3α-hydroxy-3ß-methyl-21-(4-cyano-1H-pyrazol-1'-yl)-19-nor-5ß-pregnan-20-one (SAGE-217, 3), a potent GABAA receptor modulator at both synaptic and extrasynaptic receptor subtypes, with excellent oral DMPK properties. Compound 3 has completed a phase 1 single ascending dose (SAD) and multiple ascending dose (MAD) clinical trial and is currently being studied in parallel phase 2 clinical trials for the treatment of postpartum depression (PPD), major depressive disorder (MDD), and essential tremor (ET).


Subject(s)
Allosteric Regulation/drug effects , GABA-A Receptor Agonists/chemistry , GABA-A Receptor Agonists/pharmacology , Pregnanolone/analogs & derivatives , Receptors, GABA-A/metabolism , Animals , Depression, Postpartum/drug therapy , Depressive Disorder, Major/drug therapy , Female , GABA-A Receptor Agonists/pharmacokinetics , Mice , Pregnanolone/chemistry , Pregnanolone/pharmacokinetics , Pregnanolone/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Rats
16.
BMC Complement Altern Med ; 17(1): 54, 2017 Jan 18.
Article in English | MEDLINE | ID: mdl-28100216

ABSTRACT

BACKGROUND: The present research was carried out to investigate pharmacological properties of Buxus papillosa C.K. Schneid. (Buxaceae). METHODS: Buxus papillosa extracts of leaves (BpL), stem (BpS), roots (BpR) and BpL fractions: hexane (BpL-H), aqueous (BpL-A) also plant constituent, cyclomicrobuxine effect were studied in jejunum, atria, aorta and tracheal preparations from rabbit and guine-peg. RESULTS: Ca++ antagonistic effect of BpS, BpR, BpL-H, BpL-A and cyclomicrobuxine were conclusively suggested, when spontaneous contractions of rabbit jejunal preparation was relaxed along with subsequent relaxation of potassium chloride (80 mM) induced contractions. Ca++ antagonistic effect was further confirmed, when a prominent right shift like that of verapamil was observed in Ca++ concentration-response curves, drawn in a tissue pretreated with BpL (0.3-1.0 mg/mL). In rabbit tracheal tissues BpL, BpS, BpR, BpL-H and BpL-A produced a prominent relaxation in contractions induced by potassium chloride (80 mM) and carbachol (1 µm). When tested in rabbit aortic rings, BpL, BpS, BpR, BpL-H and BpL-A showed concentration-dependent (0.1-3.0 mg/mL) vasorelaxant effect against phenylephrine (1 µM) and high K+-induced contractions. In isolated guinea-pig right atria, BpL, BpS, BpR, BpL-H and BpL-A suppressed atrial force of spontaneous contractions, with BpL-A being most potent. CONCLUSIONS: Our results reveal that Buxus papillosa possesses gut, airways and cardiovascular inhibitory actions.


Subject(s)
Bronchodilator Agents/pharmacology , Buxus/chemistry , Myocardial Contraction/drug effects , Parasympatholytics/pharmacology , Plant Extracts/pharmacology , Vasodilator Agents/pharmacology , Animals , Aorta/drug effects , Guinea Pigs , Jejunum/drug effects , Molecular Structure , Plant Leaves/chemistry , Pregnanolone/analogs & derivatives , Pregnanolone/chemistry , Pregnanolone/isolation & purification , Pregnanolone/pharmacology , Rabbits , Trachea/drug effects
17.
Steroids ; 118: 25-31, 2017 02.
Article in English | MEDLINE | ID: mdl-27913096

ABSTRACT

The concentrations of allopregnanolone (Allopreg), pregnenolone (Preg) and androsterone (ADT) are very low in the circulation, especially in postmenopausal women, resulting in a considerable challenge for their accurate measurements in serum or plasma. In this report, a sensitive and reliable LC-MS/MS assay method has been developed using a simple sample preparation and the 1-Amino-4-methylpiperazine (AMP) derivatization procedure. A 5pg/ml (0.1pg on column) of low limit of quantitation has been achieved for Allopreg, Preg and ADT, with a sensitivity comparable to data obtained with the commercial reagent. The major benefit of this reagent is to limit the matrix effect since the excess amount of reagent can be removed during the reaction. Multiple reaction monitoring (MRM) from the derivatization of AMP not only increases the detection of these compounds but also provides a good resolution for Allopreg, Preg and ADT from interferences, especially for Allopreg from its isomers. Within the calibration range of 5pg/ml to 2000pg/ml, a good linearity was obtained with R>0.99 where the weighing factor is 1/X. Bias and coefficients of variance are within 15% for all QC levels. The matrix effect has been evaluated, well meeting the acceptance criteria according to the FDA guidelines. With this method, the concentrations of Allopreg, Preg and ADT in postmenopausal serum are in the range of 6.4-53.6pg/ml, 16.2-68.0pg/ml and 23.9-114.0pg/ml, respectively, while the ranges in premenopausal serum are 8.2-701.5pg/ml, 31.2-135.2pg/ml and 47.8-310.0pg/ml, respectively.


Subject(s)
Androsterone/blood , Chromatography, Liquid/methods , Piperazines/chemistry , Postmenopause/blood , Pregnanolone/blood , Pregnenolone/blood , Premenopause/blood , Tandem Mass Spectrometry/methods , Androsterone/chemistry , Biological Assay/methods , Female , Humans , Molecular Structure , Pregnanolone/chemistry , Pregnenolone/chemistry , Reproducibility of Results
18.
J Neurosci ; 36(7): 2161-75, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26888927

ABSTRACT

Postsynaptic N-methyl-d-aspartate receptors (NMDARs) phasically activated by presynaptically released glutamate are critical for synaptic transmission and plasticity. However, under pathological conditions, excessive activation of NMDARs by tonically increased ambient glutamate contributes to excitotoxicity associated with various acute and chronic neurological disorders. Here, using heterologously expressed GluN1/GluN2A and GluN1/GluN2B receptors and rat autaptic hippocampal microisland cultures, we show that pregnanolone sulfate inhibits NMDAR currents induced by a prolonged glutamate application with a higher potency than the NMDAR component of EPSCs. For synthetic pregnanolone derivatives substituted with a carboxylic acid moiety at the end of an aliphatic chain of varying length and attached to the steroid skeleton at C3, the difference in potency between tonic and phasic inhibition increased with the length of the residue. The steroid with the longest substituent, pregnanolone hemipimelate, had no effect on phasically activated receptors while inhibiting tonically activated receptors. In behavioral tests, pregnanolone hemipimelate showed neuroprotective activity without psychomimetic symptoms. These results provide insight into the influence of steroids on neuronal function and stress their potential use in the development of novel therapeutics with neuroprotective action. SIGNIFICANCE STATEMENT: Synaptic activation of N-methyl-d-aspartate receptors (NMDARs) plays a key role in synaptic plasticity, but excessive tonic NMDAR activation mediates excitotoxicity associated with many neurological disorders. Therefore, there is much interest in pharmacological agents capable of selectively blocking tonically activated NMDARs while leaving synaptically activated NMDARs intact. Here, we show that an endogenous neurosteroid pregnanolone sulfate is more potent at inhibiting tonically than synaptically activated NMDARs. Further, we report that a novel synthetic analog of pregnanolone sulfate, pregnanolone hemipimelate, inhibits tonic NMDAR currents without inhibiting the NMDAR component of the EPSC and shows neuroprotective activity in vivo without inducing psychomimetic side effects. These results suggest steroids may have a clinical advantage over other known classes of NMDAR inhibitors.


Subject(s)
Pregnanes/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Avoidance Learning/drug effects , Excitatory Postsynaptic Potentials/drug effects , HEK293 Cells , Hippocampus/metabolism , Humans , Male , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques , Pregnanes/chemistry , Pregnanolone/chemistry , Pregnanolone/pharmacology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/genetics , Structure-Activity Relationship , Synaptic Transmission/drug effects
19.
J Neurosci Res ; 94(6): 568-78, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26308557

ABSTRACT

Alterations in the ratio of excitatory to inhibitory transmission are emerging as a common component of many nervous system disorders, including autism spectrum disorders (ASDs). Tonic γ-aminobutyric acidergic (GABAergic) transmission provided by peri- and extrasynaptic GABA type A (GABAA ) receptors powerfully controls neuronal excitability and plasticity and, therefore, provides a rational therapeutic target for normalizing hyperexcitable networks across a variety of disorders, including ASDs. Our previous studies revealed tonic GABAergic deficits in principal excitatory neurons in the basolateral amygdala (BLA) in the Fmr1(-/y) knockout (KO) mouse model fragile X syndrome. To correct amygdala deficits in tonic GABAergic neurotransmission in Fmr1(-/y) KO mice, we developed a novel positive allosteric modulator of GABAA receptors, SGE-872, based on endogenously active neurosteroids. This study shows that SGE-872 is nearly as potent and twice as efficacious for positively modulating GABAA receptors as its parent molecule, allopregnanolone. Furthermore, at submicromolar concentrations (≤1 µM), SGE-872 is selective for tonic, extrasynaptic α4ß3δ-containing GABAA receptors over typical synaptic α1ß2γ2 receptors. We further find that SGE-872 strikingly rescues the tonic GABAergic transmission deficit in principal excitatory neurons in the Fmr1(-/y) KO BLA, a structure heavily implicated in the neuropathology of ASDs. Therefore, the potent and selective action of SGE-872 on tonic GABAA receptors containing α4 subunits may represent a novel and highly useful therapeutic avenue for ASDs and related disorders involving hyperexcitability of neuronal networks.


Subject(s)
Amygdala/drug effects , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/pathology , GABA Modulators/pharmacology , Membrane Potentials/drug effects , gamma-Aminobutyric Acid/metabolism , Amygdala/metabolism , Amygdala/pathology , Animals , Animals, Newborn , CHO Cells , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , GABA Agents/pharmacology , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/pharmacology , In Vitro Techniques , Membrane Potentials/genetics , Mice , Mice, Knockout , Patch-Clamp Techniques , Pregnanolone/analogs & derivatives , Pregnanolone/chemistry , Pregnanolone/pharmacology , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Transfection , gamma-Aminobutyric Acid/pharmacology
20.
Sci Rep ; 5: 10935, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26086919

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) mediate synaptic plasticity, and their dysfunction is implicated in multiple brain disorders. NMDARs can be allosterically modulated by numerous compounds, including endogenous neurosteroid pregnanolone sulfate. Here, we identify the molecular basis of the use-dependent and voltage-independent inhibitory effect of neurosteroids on NMDAR responses. The site of action is located at the extracellular vestibule of the receptor's ion channel pore and is accessible after receptor activation. Mutations in the extracellular vestibule in the SYTANLAAF motif disrupt the inhibitory effect of negatively charged steroids. In contrast, positively charged steroids inhibit mutated NMDAR responses in a voltage-dependent manner. These results, in combination with molecular modeling, characterize structure details of the open configuration of the NMDAR channel. Our results provide a unique opportunity for the development of new therapeutic neurosteroid-based ligands to treat diseases associated with dysfunction of the glutamate system.


Subject(s)
Mutation , Pregnanolone , Receptors, N-Methyl-D-Aspartate , Vestibule, Labyrinth , Amino Acid Motifs , Humans , Pregnanolone/chemistry , Pregnanolone/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Vestibule, Labyrinth/chemistry , Vestibule, Labyrinth/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...