1
|
Bukanova JV, Kondratenko RV, Solntseva EI. Interaction Between Allopregnanolone and Amiloride Binding Sites on the GABA A Receptor. Cell Biochem Biophys 2024:10.1007/s12013-024-01654-6. [PMID: 39730891 DOI: 10.1007/s12013-024-01654-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2024] [Indexed: 12/29/2024]
Abstract
Allopregnanolone (Allo) is a positive allosteric modulator of the GABAA receptor, and amiloride (Ami) is a competitive antagonist of the GABAA receptor. The purpose of this work was to study the combined effect of Allo and Ami on functional activity of GABAA receptor. The GABA-induced chloride current (IGABA) was measured in isolated Purkinje cells of rat cerebellum using the patch-clamp technique and a system of fast application. Our results indicate that Allo suppresses the inhibitory effect of Ami on IGABA, the IC50 value of Ami concentration-response curve was increased from 164 to 547 µM (P < 0.001) in the presence of Allo. Next, GABA concentration-response curves (EC50 = 5.8 µM) were constructed in the presence of Allo (EC50 = 1.2 µM), Ami (EC50 = 25.5 µM), and the combination of Allo+Ami (EC50 = 3.2 µM). Changes in EC50 values as a percentage relative to the control were calculated. The blocking effect of Ami is reduced in the presence of Allo (340% vs 150%, P < 0.01) and the potentiating effect of Allo does not change in the presence of Ami (78% vs 87%, P > 0.05). The results suggest that there is an allosteric relationship between the Allo and Ami binding sites on GABAA receptor that operates in one direction, from Allo sites to Ami site, but not vice versa.
Collapse
|
2
|
Fischer L, Paschke B, Gareis F, Schumacher M, Liere P, Hiergeist A, Gessner A, Rupprecht R, Neumann ID, Bosch OJ. The translocator protein 18 kDa (TSPO) ligand etifoxine in an animal model of anxiety: line- and sex-dependent effects on emotionality, stress reactivity, spine density, oxytocin receptors, steroids, and microbiome composition. Neuropharmacology 2024:110282. [PMID: 39725124 DOI: 10.1016/j.neuropharm.2024.110282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
The treatment of stress-related disorders such as anxiety and depression is still challenging. One potential therapeutical option are neurosteroids. Their synthesis is promoted by ligands of the mitochondrial translocator protein 18 kDa (TSPO). We tested the TSPO ligand etifoxine (ETX) in a rat model of hyper-anxiety and depression-like behavior, i.e., in female and male HAB (high anxiety-related behavior) rats, as well as in respective low anxiety (LAB) and non-selected control (NAB) rats for behavioral, molecular, cellular, and physiological parameters. Daily acute i.p. treatment with ETX or vehicle over 5 or 9 days revealed that ETX was most effective in female HAB rats; it reduced anxiety levels (5 days) and OXT-R binding brain site-specifically (5 and 9 days), and increased spine density (5 days). The behavioral ETX effect exclusively found in female HABs was accompanied by increased 3β5α-THDOC levels, without any effect in female LABs and NABs and on other neurosteroids. In males of all breeding lines, ETX changed a total of 10 out of 23 brain steroids. Passive stress-coping during 10-min forced swimming was not affected by 9-day treatment with ETX, the resulting stress-induced plasma corticosterone levels were higher in ETX-treated NAB rats of both sexes compared with their VEH-treated groups. The fecal bacterial composition was similar but beta diversity differed between HABs and LABs and from NABs independent of sex; ETX treatment had no effect. Therefore, we propose considering the aspect of sex in treatment strategies for anxiety disorders. This is particularly important to establish better treatment regimens for women.
Collapse
Affiliation(s)
- Lilith Fischer
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Bjarne Paschke
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Franziska Gareis
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Michael Schumacher
- U1195 Inserm and University Paris-Saclay, 80 rue du Général Leclerc, Le Kremlin-Bicêtre, 94276, France.
| | - Philippe Liere
- U1195 Inserm and University Paris-Saclay, 80 rue du Général Leclerc, Le Kremlin-Bicêtre, 94276, France.
| | - Andreas Hiergeist
- Institute of Clinical Microbiology and Hygiene, University Medical Center, 93053 Regensburg, Germany.
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Medical Center, 93053 Regensburg, Germany.
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany.
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| | - Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
3
|
Qian M, Xu Y, Shu HJ, Chen ZW, Wang L, Zorumski CF, Evers AS, Mennerick S, Covey DF. Synthesis and evaluation of photoaffinity labeling reagents for identifying binding sites of sulfated neurosteroids on NMDA and GABA A receptors. RSC Adv 2024; 14:36352-36369. [PMID: 39539530 PMCID: PMC11559520 DOI: 10.1039/d4ra07074g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
The endogenous neurosteroids dehydroepiandrosterone sulfate (DHEAS) and pregnenolone sulfate (PS) are allosteric modulators of γ-aminobutyric acid type A (GABAA) and N-methyl-d-aspartate (NMDA) type glutamate receptors. Analogues of these endogenous steroid sulfates can be either positive or negative allosteric modulators (PAMs or NAMs, respectively) of these receptors, but there is limited information about the steroid-protein binding interactions that mediate these effects and photoaffinity labeling reagents (PALs) of sulfated steroids have not been reported previously. The synthesis of a panel of ten sulfated steroid analogues containing a diazirine group, five of which also contain an alkyne group for click chemistry reactions, for use in photoaffinity labeling studies to identify binding sites for steroid sulfates that are either positive or negative allosteric modulators is reported. Electrophysiological measurements on cultured rat hippocampal neurons were made to determine the modes of allosteric modulation in comparison to those of PS on both receptors. PALs with the activity profile of PS (NMDA PAM, GABAA NAM) were identified. Unexpectedly, PALs with PAM activity at both receptors were also found. Photolabeling of both receptors by two of the PALs was performed to demonstrate their utility, and by inference those of the other PALs, for future studies to identify binding sites for endogenous steroid sulfates on both receptors.
Collapse
Affiliation(s)
- Mingxing Qian
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Yuanjian Xu
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Hong-Jin Shu
- Department of Psychiatry, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Zi-Wei Chen
- Department of Anesthesiology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Lei Wang
- Department of Anesthesiology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430022 China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education China
| | - Charles F Zorumski
- Department of Psychiatry, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Alex S Evers
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Anesthesiology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Steven Mennerick
- Department of Psychiatry, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Psychiatry, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Anesthesiology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| |
Collapse
|
4
|
Chintala SM, Tateiwa H, Qian M, Xu Y, Amtashar F, Chen ZW, Kirkpatrick CC, Bracamontes J, Germann AL, Akk G, Covey DF, Evers AS. Direct measurements of neurosteroid binding to specific sites on GABA A receptors. Br J Pharmacol 2024; 181:4229-4244. [PMID: 38978389 PMCID: PMC11585924 DOI: 10.1111/bph.16490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 05/12/2024] [Accepted: 05/30/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Neurosteroids are allosteric modulators of GABAA currents, acting through several functional binding sites although their affinity and specificity for each site are unknown. The goal of this study was to measure steady-state binding affinities of various neurosteroids for specific sites on the GABAA receptor. EXPERIMENTAL APPROACH Two methods were developed to measure neurosteroid binding affinity: (1) quenching of specific tryptophan residues in neurosteroid binding sites by the neurosteroid 17-methylketone group, and (2) FRET between MQ290 (an intrinsically fluorescent neurosteroid) and tryptophan residues in the binding sites. The assays were developed using ELIC-α1GABAAR, a chimeric receptor containing transmembrane domains of the α1-GABAA receptor. Tryptophan mutagenesis was used to identify specific interactions. KEY RESULTS Allopregnanolone (3α-OH neurosteroid) was shown to bind at intersubunit and intrasubunit sites with equal affinity, whereas epi-allopregnanolone (3β-OH neurosteroid) binds at the intrasubunit site. MQ290 formed a strong FRET pair with W246, acting as a site-specific probe for the intersubunit site. The affinity and site-specificity of several neurosteroid agonists and inverse agonists was measured using the MQ290 binding assay. The FRET assay distinguishes between competitive and allosteric inhibition of MQ290 binding and demonstrated an allosteric interaction between the two neurosteroid binding sites. CONCLUSIONS AND IMPLICATIONS The affinity and specificity of neurosteroid binding to two sites in the ELIC-α1GABAAR were directly measured and an allosteric interaction between the sites was revealed. Adaptation of the MQ290 FRET assay to a plate-reader format will enable screening for high affinity agonists and antagonists for neurosteroid binding sites.
Collapse
Affiliation(s)
| | - Hiroki Tateiwa
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kochi, Japan
| | - Mingxing Qian
- Department of Developmental Biology (Pharmacology), Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yuanjian Xu
- Department of Developmental Biology (Pharmacology), Washington University School of Medicine, St. Louis, Missouri, USA
| | - Fatima Amtashar
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zi-Wei Chen
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Taylor Family Institute for Innovative Psychiatric Research, St. Louis, Missouri, USA
| | | | - John Bracamontes
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Allison L. Germann
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gustav Akk
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Taylor Family Institute for Innovative Psychiatric Research, St. Louis, Missouri, USA
| | - Douglas F. Covey
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kochi, Japan
- Taylor Family Institute for Innovative Psychiatric Research, St. Louis, Missouri, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alex S. Evers
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kochi, Japan
- Taylor Family Institute for Innovative Psychiatric Research, St. Louis, Missouri, USA
| |
Collapse
|
5
|
Pierce SR, Germann AL, Covey DF, Evers AS, Steinbach JH, Akk G. Inhibitory Actions of Potentiating Neuroactive Steroids in the Human α1β3γ2L γ-Aminobutyric Acid Type A Receptor. Mol Pharmacol 2024; 106:264-277. [PMID: 39214710 PMCID: PMC11493365 DOI: 10.1124/molpharm.124.000960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
The γ-aminobutyric acid type A (GABAA) receptor is modulated by a number of neuroactive steroids. Sulfated steroids and 3β-hydroxy steroids inhibit, while 3α-hydroxy steroids typically potentiate the receptor. Here, we have investigated inhibition of the α1β3γ2L GABAA receptor by the endogenous neurosteroid 3α-hydroxy-5β-pregnan-20-one (3α5βP) and the synthetic neuroactive steroid 3α-hydroxy-5α-androstane-17β-carbonitrile (ACN). The receptors were expressed in Xenopus oocytes. All experiments were done using two-electrode voltage-clamp electrophysiology. In the presence of low concentrations of GABA, 3α5βP and ACN potentiate the GABAA receptor. To reveal inhibition, we conducted the experiments on receptors activated by the combination of a saturating concentration of GABA and propofol to fully activate the receptors and mask potentiation, or on mutant receptors in which potentiation is ablated. Under these conditions, both steroids inhibited the receptor with IC50s of ∼13 μM and maximal inhibitory effects of 70-90%. Receptor inhibition by 3α5βP was sensitive to substitution of the α1 transmembrane domain (TM) 2-2' residue, previously shown to ablate inhibition by pregnenolone sulfate. However, results of coapplication studies and the apparent lack of state dependence suggest that pregnenolone sulfate and 3α5βP inhibit the GABAA receptor independently and through distinct mechanisms. Mutations to the neurosteroid binding sites in the α1 and β3 subunits statistically significantly, albeit weakly and incompletely, reduced inhibition by 3α5βP and ACN. SIGNIFICANCE STATEMENT: The heteromeric GABAA receptor is inhibited by sulfated steroids and 3β-hydroxy steroids, while 3α-hydroxy steroids are considered to potentiate the receptor. We show here that 3α-hydroxy steroids have inhibitory effects on the α1β3γ2L receptor, which are observed in specific experimental settings and are expected to manifest under different physiological conditions.
Collapse
Affiliation(s)
- Spencer R Pierce
- Departments of Anesthesiology (S.R.P., A.L.G., D.F.C., A.S.E., J.H.S., G.A.), Developmental Biology (D.F.C., A.S.E.), and the Taylor Family Institute for Innovative Psychiatric Research (D.F.C., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St Louis, Missouri
| | - Allison L Germann
- Departments of Anesthesiology (S.R.P., A.L.G., D.F.C., A.S.E., J.H.S., G.A.), Developmental Biology (D.F.C., A.S.E.), and the Taylor Family Institute for Innovative Psychiatric Research (D.F.C., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St Louis, Missouri
| | - Douglas F Covey
- Departments of Anesthesiology (S.R.P., A.L.G., D.F.C., A.S.E., J.H.S., G.A.), Developmental Biology (D.F.C., A.S.E.), and the Taylor Family Institute for Innovative Psychiatric Research (D.F.C., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St Louis, Missouri
| | - Alex S Evers
- Departments of Anesthesiology (S.R.P., A.L.G., D.F.C., A.S.E., J.H.S., G.A.), Developmental Biology (D.F.C., A.S.E.), and the Taylor Family Institute for Innovative Psychiatric Research (D.F.C., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St Louis, Missouri
| | - Joe Henry Steinbach
- Departments of Anesthesiology (S.R.P., A.L.G., D.F.C., A.S.E., J.H.S., G.A.), Developmental Biology (D.F.C., A.S.E.), and the Taylor Family Institute for Innovative Psychiatric Research (D.F.C., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St Louis, Missouri
| | - Gustav Akk
- Departments of Anesthesiology (S.R.P., A.L.G., D.F.C., A.S.E., J.H.S., G.A.), Developmental Biology (D.F.C., A.S.E.), and the Taylor Family Institute for Innovative Psychiatric Research (D.F.C., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
6
|
Chen ZW, Chintala SM, Bracamontes J, Sugasawa Y, Pierce SR, Varga BR, Smith EH, Edge CJ, Franks NP, Cheng WWL, Akk G, Evers AS. Three classes of propofol binding sites on GABA A receptors. J Biol Chem 2024; 300:107778. [PMID: 39270821 PMCID: PMC11490885 DOI: 10.1016/j.jbc.2024.107778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Propofol is a widely used anesthetic and sedative that acts as a positive allosteric modulator of gamma-aminobutyric acid type A (GABAA) receptors. Several potential propofol binding sites that may mediate this effect have been identified using propofol-analogue photoaffinity labeling. Ortho-propofol diazirine (o-PD) labels β-H267, a pore-lining residue, whereas AziPm labels residues β-M286, β-M227, and α-I239 in the two membrane-facing interfaces [β(+)/α(-) and α(+)/β(-)] between α and β subunits. This study used photoaffinity labeling of α1β3 GABAA receptors to reconcile the apparently conflicting results obtained with AziPm and o-PD labeling, focusing on whether β3-H267 identifies specific propofol binding site(s). The results show that propofol, but not AziPm protects β3-H267 from labeling by o-PD, whereas both propofol and o-PD protect against AziPm labeling of β3-M286, β3-M227, and α1I239. These data indicate that there are three distinct classes of propofol binding sites, with AziPm binding to two of the classes and o-PD to all three. Analysis of binding stoichiometry using native mass spectrometry in β3 homomeric receptors, demonstrated a minimum of five AziPm labeled residues and three o-PD labeled residues per pentamer, suggesting that there are two distinct propofol binding sites per β-subunit. The native mass spectrometry data, coupled with photolabeling performed in the presence of zinc, indicate that the binding site(s) identified by o-PD are adjacent to, but not within the channel pore, since the pore at the 17' H267 residue can accommodate only one propofol molecule. These data validate the existence of three classes of specific propofol binding sites on α1β3 GABAA receptors.
Collapse
Affiliation(s)
- Zi-Wei Chen
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA; The Taylor Family Institute for Innovative Psychiatric Research Washington University School of Medicine, St Louis, Missouri, USA
| | | | - John Bracamontes
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Yusuke Sugasawa
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA; Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Spencer R Pierce
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Balazs R Varga
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Edward H Smith
- Department of Life Sciences, Imperial College, London, UK
| | | | - Nicholas P Franks
- Department of Life Sciences, Imperial College, London, UK; UK Dementia Research Institute, Imperial College, London, UK
| | - Wayland W L Cheng
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Gustav Akk
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA; The Taylor Family Institute for Innovative Psychiatric Research Washington University School of Medicine, St Louis, Missouri, USA
| | - Alex S Evers
- Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri, USA; Department of Developmental Biology, Washington University School of Medicine, St Louis, Missouri, USA; The Taylor Family Institute for Innovative Psychiatric Research Washington University School of Medicine, St Louis, Missouri, USA.
| |
Collapse
|
7
|
Riebel M, Brunner LM, Nothdurfter C, Wein S, Schwarzbach J, Liere P, Schumacher M, Rupprecht R. Neurosteroids and translocator protein 18 kDa (TSPO) ligands as novel treatment options in depression. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01843-7. [PMID: 38976049 DOI: 10.1007/s00406-024-01843-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024]
Abstract
Recently, the gamma-aminobutyric acid (GABA) system has come into focus for the treatment of anxiety, postpartum depression, and major depressive disorder. Endogenous 3α-reduced steroids such as allopregnanolone are potent positive allosteric modulators of GABAA receptors and have been known for decades. Current industry developments and first approvals by the U.S. food and drug administration (FDA) for the treatment of postpartum depression with exogenous analogues of these steroids represent a major step forward in the field. 3α-reduced steroids target both synaptic and extrasynaptic GABAA receptors, unlike benzodiazepines, which bind to synaptic receptors. The first FDA-approved 3α-reduced steroid for postpartum depression is brexanolone, an intravenous formulation of allopregnanolone. It has been shown to provide rapid relief of depressive symptoms. An orally available 3α-reduced steroid is zuranolone, which also received FDA approval in 2023 for the treatment of postpartum depression. Although a number of studies have been conducted, the efficacy data were not sufficient to achieve approval of zuranolone in major depressive disorder by the FDA in 2023. The most prominent side effects of these 3α-reduced steroids are somnolence, dizziness and headache. In addition to the issue of efficacy, it should be noted that current data limit the use of these compounds to two weeks. An alternative to exogenous 3α-reduced steroids may be the use of substances that induce endogenous neurosteroidogenesis, such as the translocator protein 18 kDa (TSPO) ligand etifoxine. TSPO has been extensively studied for its role in steroidogenesis, in addition to other functions such as anti-inflammatory and neuroregenerative properties. Currently, etifoxine is the only clinically available TSPO ligand in France for the treatment of anxiety disorders. Studies are underway to evaluate its antidepressant potential. Hopefully, neurosteroid research will lead to the development of fast-acting antidepressants.
Collapse
Affiliation(s)
- Marco Riebel
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany.
| | - Lisa-Marie Brunner
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| | - Caroline Nothdurfter
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| | - Simon Wein
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| | - Jens Schwarzbach
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| | - Philippe Liere
- U1195 Inserm and University Paris-Saclay, Le Kremlin-Bicêtre, Paris, 94276, France
| | - Michael Schumacher
- U1195 Inserm and University Paris-Saclay, Le Kremlin-Bicêtre, Paris, 94276, France
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| |
Collapse
|
8
|
Krogman WL, Woodard T, McKay RSF. Anesthetic Mechanisms: Synergistic Interactions With Lipid Rafts and Voltage-Gated Sodium Channels. Anesth Analg 2024; 139:92-106. [PMID: 37968836 DOI: 10.1213/ane.0000000000006738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Despite successfully utilizing anesthetics for over 150 years, the mechanism of action remains relatively unknown. Recent studies have shown promising results, but due to the complex interactions between anesthetics and their targets, there remains a clear need for further mechanistic research. We know that lipophilicity is directly connected to anesthetic potency since lipid solubility relates to anesthetic partition into the membrane. However, clinically relevant concentrations of anesthetics do not significantly affect lipid bilayers but continue to influence various molecular targets. Lipid rafts are derived from liquid-ordered phases of the plasma membrane that contain increased concentrations of cholesterol and sphingomyelin and act as staging platforms for membrane proteins, including ion channels. Although anesthetics do not perturb membranes at clinically relevant concentrations, they have recently been shown to target lipid rafts. In this review, we summarize current research on how different types of anesthetics-local, inhalational, and intravenous-bind and affect both lipid rafts and voltage-gated sodium channels, one of their major targets, and how those effects synergize to cause anesthesia and analgesia. Local anesthetics block voltage-gated sodium channel pores while also disrupting lipid packing in ordered membranes. Inhalational anesthetics bind to the channel pore and the voltage-sensing domain while causing an increase in the number, size, and diameter of lipid rafts. Intravenous anesthetics bind to the channel primarily at the voltage-sensing domain and the selectivity filter, while causing lipid raft perturbation. These changes in lipid nanodomain structure possibly give proteins access to substrates that have translocated as a result of these structural alterations, resulting in lipid-driven anesthesia. Overall, anesthetics can impact channel activity either through direct interaction with the channel, indirectly through the lipid raft, or both. Together, these result in decreased sodium ion flux into the cell, disrupting action potentials and producing anesthetic effects. However, more research is needed to elucidate the indirect mechanisms associated with channel disruption through the lipid raft, as not much is known about anionic lipid products and their influence over voltage-gated sodium channels. Anesthetics' effect on S-palmitoylation, a promising mechanism for direct and indirect influence over voltage-gated sodium channels, is another auspicious avenue of research. Understanding the mechanisms of different types of anesthetics will allow anesthesiologists greater flexibility and more specificity when treating patients.
Collapse
Affiliation(s)
- William L Krogman
- From the Department of Anesthesiology, University of Kansas School of Medicine - Wichita, Wichita, Kansas
| | | | | |
Collapse
|
9
|
Mortensen M, Bright DP, Fagotti J, Dorovykh V, Cerna B, Smart TG. Forty Years Searching for Neurosteroid Binding Sites on GABA A Receptors. Neuroscience 2024:S0306-4522(24)00257-4. [PMID: 38852898 DOI: 10.1016/j.neuroscience.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Brain inhibition is a vital process for controlling and sculpting the excitability of the central nervous system in healthy individuals. This level of control is provided over several timescales and involves the neurotransmitter GABA acting at inhibitory synapses to: rapidly inhibit neurons by activating the GABAA receptor; over a slower timescale, to tonically activate extrasynaptic GABAA receptors to provide a low level of background inhibition; and finally, to activate G-protein coupled GABAB receptors to control transmitter release by inhibiting presynaptic Ca2+ channels whilst providing postsynaptic inhibition via K+ channel activation. From this plethora of roles for GABA and its receptors, the GABAA receptor isoform is of major interest due to its dynamic functional plasticity, which in part, is due to being targeted by modulatory brain neurosteroids derived from sex and stress hormones. This family of neurosteroids can, depending on their structure, potentiate, activate and also inhibit the activity of GABAA receptors to affect brain inhibition. This review tracks the methods that have been deployed in probing GABAA receptors, and charts the sterling efforts made by several groups to locate the key neurosteroid binding sites that affect these important receptors. Increasing our knowledge of these binding sites will greatly facilitate our understanding of the physiological roles of neurosteroids and will help to advance their use as novel therapeutics to combat debilitating brain diseases.
Collapse
Affiliation(s)
- Martin Mortensen
- University College London, Dept Neuroscience, Physiology & Pharmacology, Gower Street, London WC1E 6BT, United Kingdom
| | - Damian P Bright
- University College London, Dept Neuroscience, Physiology & Pharmacology, Gower Street, London WC1E 6BT, United Kingdom
| | - Juliane Fagotti
- University College London, Dept Neuroscience, Physiology & Pharmacology, Gower Street, London WC1E 6BT, United Kingdom
| | - Valentina Dorovykh
- University College London, Dept Neuroscience, Physiology & Pharmacology, Gower Street, London WC1E 6BT, United Kingdom
| | - Barbora Cerna
- University College London, Dept Neuroscience, Physiology & Pharmacology, Gower Street, London WC1E 6BT, United Kingdom
| | - Trevor G Smart
- University College London, Dept Neuroscience, Physiology & Pharmacology, Gower Street, London WC1E 6BT, United Kingdom.
| |
Collapse
|
10
|
Sugasawa Y. Neurosteroid Binding and Actions on GABA A Receptors. JUNTENDO IJI ZASSHI = JUNTENDO MEDICAL JOURNAL 2024; 70:239-244. [PMID: 39429691 PMCID: PMC11487359 DOI: 10.14789/jmj.jmj24-0002-r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/22/2024] [Indexed: 10/22/2024]
Abstract
Neurosteroids positively modulate GABAA receptor (GABAAR) channel activity by binding to a transmembrane domain intersubunit site. Using photo-affinity labeling and an ELIC-α1GABAAR chimera, we investigated the impact of mutations within the intersubunit site on neurosteroid binding. These mutations reduce neither photolabeling within the intersubunit site nor competitive prevention of labeling by allopregnanolone. Instead, these mutations change the orientation of neurosteroid photolabeling. The data indicate that mutations at Gln242 or Trp246 that eliminate neurosteroid effects do not eliminate neurosteroid binding within the intersubunit site, but significantly alter the preferred orientation of the neurosteroid within the site. The interactions formed by Gln242 and Trp246 within this pocket play a vital role in determining the orientation of the neurosteroid. We also examined how site-specific binding to three identified neurosteroid-binding sites in the α1β3GABAAR contributes to neurosteroid allosteric modulation. We found that the potentiating neurosteroid, allopregnanolone, but not its inhibitory 3β-epimer epi-allopregnanolone, binds to the canonical β3(+)-α1(-) intersubunit site that mediates receptor activation by neurosteroids. In contrast, both allopregnanolone and epi-allopregnanolone bind to intrasubunit sites in the β3 subunit, promoting receptor desensitization and the α1 subunit promoting effects that vary between neurosteroids. Two neurosteroid analogues with diazirine moieties replacing the 3-hydroxyl bind to all three sites, but do not potentiate GABAAR currents. One is a desensitizing agent, whereas the other is devoid of allosteric activity. 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 GABAAR.
Collapse
|
11
|
Bukanova JV, Solntseva EI, Skrebitsky VG. Factors promoting the release of picrotoxin from the trap in the GABA(A) receptor pore. Neurochem Int 2024; 175:105703. [PMID: 38395151 DOI: 10.1016/j.neuint.2024.105703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 02/25/2024]
Abstract
Picrotoxin (PTX), a convulsant of plant origin, has been used in many studies as research tool. PTX is the open channel blocker of the GABAA receptor (GABAAR). Being in the pore, PTX initiates transfer of the channel to the closed state and thus it falls into the "trap". The consequence of this PTX trapping is so-called aftereffect, i.e. continuation of the blockade of the GABA-induced chloride current (IGABA) after removal of PTX from the external solution. The present work shows that the positive allosteric modulators (PAMs) of the GABAA receptor, allopregnanolone (Allo) and zolpidem (Zolp) as well as a high concentration of GABA shortened the PTX aftereffect. Experiments were carried out on isolated Purkinje neurons of the rat cerebellum using the whole-cell patch-clamp method. IGABA was induced by applications of 5 μM GABA (EC30) for 1 s with 30 s intervals. 50 μM PTX completely blocked IGABA, and recovery upon PTX washout occurred with a time constant (τrec) of 20.2 min. 1 μM Allo reduced the blocking effect of PTX by 30% and accelerated the recovery of IGABA by almost 10 times (τrec = 2.4 min). 0.5 μM Zolp did not change the IGABA block in the presence of PTX but accelerated the recovery of IGABA by more than 3 times (τrec = 5.6 min). Increasing the GABA concentration to 20 μM did not change the blocking effect of PTX, but accelerated recovery by 6 times (τrec = 3.3 min). The mechanism of the shortening of the PTX aftereffect is presumably the expansion of the GABAAR pore in the presence of PAMs and a high concentration of the agonist and, as a consequence, the escape of PTX from the "trap". The work describes new pharmacological properties of Allo and Zolp.
Collapse
Affiliation(s)
- Julia V Bukanova
- Brain Research Institute, Research Center of Neurology, Moscow, Russia
| | - Elena I Solntseva
- Brain Research Institute, Research Center of Neurology, Moscow, Russia.
| | | |
Collapse
|
12
|
Tateiwa H, Evers AS. Neurosteroids and their potential as a safer class of general anesthetics. J Anesth 2024; 38:261-274. [PMID: 38252143 PMCID: PMC10954990 DOI: 10.1007/s00540-023-03291-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 11/25/2023] [Indexed: 01/23/2024]
Abstract
Neurosteroids (NS) are a class of steroids that are synthesized within the central nervous system (CNS). Various NS can either enhance or inhibit CNS excitability and they play important biological roles in brain development, brain function and as mediators of mood. One class of NS, 3α-hydroxy-pregnane steroids such as allopregnanolone (AlloP) or pregnanolone (Preg), inhibits neuronal excitability; these endogenous NS and their analogues have been therapeutically applied as anti-depressants, anti-epileptics and general anesthetics. While NS have many favorable properties as anesthetics (e.g. rapid onset, rapid recovery, minimal cardiorespiratory depression, neuroprotection), they are not currently in clinical use, largely due to problems with formulation. Recent advances in understanding NS mechanisms of action and improved formulations have rekindled interest in development of NS as sedatives and anesthetics. In this review, the synthesis of NS, and their mechanism of action will be reviewed with specific emphasis on their binding sites and actions on γ-aminobutyric acid type A (GABAA) receptors. The potential advantages of NS analogues as sedative and anesthetic agents will be discussed.
Collapse
Affiliation(s)
- Hiroki Tateiwa
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kochi, Japan
| | - Alex S Evers
- Department of Anesthesiology, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO, 63110, USA.
| |
Collapse
|
13
|
Richardson RJ, Petrou S, Bryson A. Established and emerging GABA A receptor pharmacotherapy for epilepsy. Front Pharmacol 2024; 15:1341472. [PMID: 38449810 PMCID: PMC10915249 DOI: 10.3389/fphar.2024.1341472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024] Open
Abstract
Drugs that modulate the GABAA receptor are widely used in clinical practice for both the long-term management of epilepsy and emergency seizure control. In addition to older medications that have well-defined roles for the treatment of epilepsy, recent discoveries into the structure and function of the GABAA receptor have led to the development of newer compounds designed to maximise therapeutic benefit whilst minimising adverse effects, and whose position within the epilepsy pharmacologic armamentarium is still emerging. Drugs that modulate the GABAA receptor will remain a cornerstone of epilepsy management for the foreseeable future and, in this article, we provide an overview of the mechanisms and clinical efficacy of both established and emerging pharmacotherapies.
Collapse
Affiliation(s)
- Robert J. Richardson
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Austin Health, Heidelberg, VIC, Australia
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Praxis Precision Medicines, Boston, MA, United States
| | - Alexander Bryson
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Department of Neurology, Austin Health, Heidelberg, VIC, Australia
- Department of Neurology, Eastern Health, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Maguire JL, Mennerick S. Neurosteroids: mechanistic considerations and clinical prospects. Neuropsychopharmacology 2024; 49:73-82. [PMID: 37369775 PMCID: PMC10700537 DOI: 10.1038/s41386-023-01626-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/15/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2023]
Abstract
Like other classes of treatments described in this issue's section, neuroactive steroids have been studied for decades but have risen as a new class of rapid-acting, durable antidepressants with a distinct mechanism of action from previous antidepressant treatments and from other compounds covered in this issue. Neuroactive steroids are natural derivatives of progesterone but are proving effective as exogenous treatments. The best understood mechanism is that of positive allosteric modulation of GABAA receptors, where subunit selectivity may promote their profile of action. Mechanistically, there is some reason to think that neuroactive steroids may separate themselves from liabilities of other GABA modulators, although research is ongoing. It is also possible that intracellular targets, including inflammatory pathways, may be relevant to beneficial actions. Strengths and opportunities for further development include exploiting non-GABAergic targets, structural analogs, enzymatic production of natural steroids, precursor loading, and novel formulations. The molecular mechanisms of behavioral effects are not fully understood, but study of brain network states involved in emotional processing demonstrate a robust influence on affective states not evident with at least some other GABAergic drugs including benzodiazepines. Ongoing studies with neuroactive steroids will further elucidate the brain and behavioral effects of these compounds as well as likely underpinnings of disease.
Collapse
Affiliation(s)
- Jamie L Maguire
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Steven Mennerick
- Department of Psychiatry and Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis School of Medicine, 660 S. Euclid Ave., St. Louis, MO, 63110, USA.
| |
Collapse
|
15
|
Qian M, Xu Y, Covey DF. Synthetic routes to trifluoromethylphenyl diazirine photolabeling reagents containing an alkyne substituent (TPDYNE) for chemical biology applications. RSC Adv 2023; 13:36484-36492. [PMID: 38099258 PMCID: PMC10719903 DOI: 10.1039/d3ra07260f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023] Open
Abstract
The trifluoromethylphenyl diazirine (TPD) group is widely used in photoaffinity labeling studies. The TPDYNE group (TPD with an additional alkyne substituent on the phenyl ring) enables the use of click chemistry in conjunction with photoaffinity labeling and expands the utility of the TPD group. New methods for preparing previously known as well as new TPDYNE reagents are reported. Additional methods for preparation of a TPDYNE precursor from which the TPDYNE group can be generated once the precursor is attached to the molecule of interest are also described. Procedures for attaching the TPDYNE or TPDYNE precursor to carboxyl, amino, hydroxyl and alkyne groups are demonstrated using steroids as examples.
Collapse
Affiliation(s)
- Mingxing Qian
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Yuanjian Xu
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Anesthesiology, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Department of Psychiatry, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis 660 S. Euclid Ave. St. Louis MO 63110 USA
| |
Collapse
|
16
|
Lawande NV, Conklin EA, Christian‐Hinman CA. Sex and gonadectomy modify behavioral seizure susceptibility and mortality in a repeated low-dose kainic acid systemic injection paradigm in mice. Epilepsia Open 2023; 8:1512-1522. [PMID: 37715318 PMCID: PMC10690657 DOI: 10.1002/epi4.12828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023] Open
Abstract
OBJECTIVE Sex differences in epilepsy appear driven in part due to effects of gonadal steroids, with varying results in experimental models based on species, strain, and method of seizure induction. Furthermore, removing the main source of these steroids via gonadectomy may impact seizure characteristics differently in males and females. Repeated low-dose kainic acid (RLDKA) systemic injection paradigms were recently shown to reliably induce status epilepticus (SE) and hippocampal histopathology in C57BL/6J mice. Here, we investigated whether seizure susceptibility in a RLDKA injection protocol exhibits a sex difference and whether gonadectomy differentially influences response to this seizure induction paradigm in males and females. METHODS Adult C57BL/6J mice were left gonad-intact as controls or gonadectomized (females: ovariectomized, OVX; males: orchidectomized, ORX). At least 2 weeks later, KA was injected ip, every 30 minutes at 7.5 mg/kg or less until the animal reached SE, defined by at least 5 generalized seizures (GS, Racine stage 3 or higher). Parameters of susceptibility to GS induction, SE development, and mortality rates were quantified. RESULTS No differences in seizure susceptibility or mortality were observed between control males and control females. Gonadectomized mice exhibited increased susceptibility and reduced latency to both GS and SE in comparison to corresponding controls of the same sex, but the effects were stronger in males. In addition, ORX males, but not OVX females, exhibited strongly increased seizure-induced mortality. SIGNIFICANCE The RLDKA protocol is notable for its efficacy in inducing SE and seizure-induced histopathology in C57BL/6J mice, the background for many transgenic strains in current use in epilepsy research. The present results indicate that this protocol may be beneficial for investigating the effects of gonadal hormone replacement on seizure susceptibility, mortality, and seizure-induced histopathology, and that gonadectomy unmasks sex differences in susceptibility to seizures and mortality not observed in gonad-intact controls.
Collapse
Affiliation(s)
- Niraj V. Lawande
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | | | - Catherine A. Christian‐Hinman
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
17
|
Dharavath RN, Pina-Leblanc C, Tang VM, Sloan ME, Nikolova YS, Pangarov P, Ruocco AC, Shield K, Voineskos D, Blumberger DM, Boileau I, Bozinoff N, Gerretsen P, Vieira E, Melamed OC, Sibille E, Quilty LC, Prevot TD. GABAergic signaling in alcohol use disorder and withdrawal: pathological involvement and therapeutic potential. Front Neural Circuits 2023; 17:1218737. [PMID: 37929054 PMCID: PMC10623140 DOI: 10.3389/fncir.2023.1218737] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 09/04/2023] [Indexed: 11/07/2023] Open
Abstract
Alcohol is one of the most widely used substances. Alcohol use accounts for 5.1% of the global disease burden, contributes substantially to societal and economic costs, and leads to approximately 3 million global deaths yearly. Alcohol use disorder (AUD) includes various drinking behavior patterns that lead to short-term or long-lasting effects on health. Ethanol, the main psychoactive molecule acting in alcoholic beverages, directly impacts the GABAergic system, contributing to GABAergic dysregulations that vary depending on the intensity and duration of alcohol consumption. A small number of interventions have been developed that target the GABAergic system, but there are promising future therapeutic avenues to explore. This review provides an overview of the impact of alcohol on the GABAergic system, the current interventions available for AUD that target the GABAergic system, and the novel interventions being explored that in the future could be included among first-line therapies for the treatment of AUD.
Collapse
Affiliation(s)
| | - Celeste Pina-Leblanc
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Victor M. Tang
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Addiction Division, CAMH, Toronto, ON, Canada
- Division of Neurosciences and Clinical Translation, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Institute of Mental Health Policy Research, CAMH, Toronto, ON, Canada
| | - Matthew E. Sloan
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Addiction Division, CAMH, Toronto, ON, Canada
- Division of Neurosciences and Clinical Translation, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Psychological Clinical Science, University of Toronto Scarborough, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Institute of Mental Health Policy Research, CAMH, Toronto, ON, Canada
| | - Yuliya S. Nikolova
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Peter Pangarov
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
| | - Anthony C. Ruocco
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Temerty Centre for Therapeutic Brain Intervention, CAMH, Toronto, ON, Canada
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Kevin Shield
- Institute of Mental Health Policy Research, CAMH, Toronto, ON, Canada
| | - Daphne Voineskos
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Temerty Centre for Therapeutic Brain Intervention, CAMH, Toronto, ON, Canada
| | - Daniel M. Blumberger
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Temerty Centre for Therapeutic Brain Intervention, CAMH, Toronto, ON, Canada
| | - Isabelle Boileau
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Brain Health Imaging Centre, CAMH, Toronto, ON, Canada
| | - Nikki Bozinoff
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| | - Philip Gerretsen
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Brain Health Imaging Centre, CAMH, Toronto, ON, Canada
| | - Erica Vieira
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Osnat C. Melamed
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Lena C. Quilty
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Thomas D. Prevot
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Mortensen M, Xu Y, Shehata MA, Krall J, Ernst M, Frølund B, Smart TG. Pregnenolone sulfate analogues differentially modulate GABA A receptor closed/desensitised states. Br J Pharmacol 2023; 180:2482-2499. [PMID: 37194503 PMCID: PMC10952582 DOI: 10.1111/bph.16143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 02/07/2023] [Accepted: 05/05/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND AND PURPOSE GABAA receptors are regulated by numerous classes of allosteric modulators. However, regulation of receptor macroscopic desensitisation remains largely unexplored and may offer new therapeutic opportunities. Here, we report the emerging potential for modulating desensitisation with analogues of the endogenous inhibitory neurosteroid, pregnenolone sulfate. EXPERIMENTAL APPROACH New pregnenolone sulfate analogues were synthesised incorporating various heterocyclic substitutions located at the C-21 position on ring D. The pharmacological profiles of these compounds were assessed using electrophysiology and recombinant GABAA receptors together with mutagenesis, molecular dynamics simulations, structural modelling and kinetic simulations. KEY RESULTS All seven analogues retained a negative allosteric modulatory capability whilst exhibiting diverse potencies. Interestingly, we observed differential effects on GABA current decay by compounds incorporating either a six- (compound 5) or five-membered heterocyclic ring (compound 6) on C-21, which was independent of their potencies as inhibitors. We propose that differences in molecular charges, and the targeted binding of analogues to specific states of the GABAA receptor, are the most likely cause of the distinctive functional profiles. CONCLUSIONS AND IMPLICATIONS Our findings reveal that heterocyclic addition to inhibitory neurosteroids not only affected their potency and macroscopic efficacy but also affected innate receptor mechanisms that underlie desensitisation. Acute modulation of macroscopic desensitisation will determine the degree and duration of GABA inhibition, which are vital for the integration of neural circuit activity. Discovery of this form of modulation could present an opportunity for next-generation GABAA receptor drug design and development.
Collapse
Affiliation(s)
- Martin Mortensen
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| | - Yue Xu
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Mohamed A. Shehata
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Jacob Krall
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Present address:
Xellia Pharmaceuticals ApSCopenhagenDenmark
| | - Margot Ernst
- Department of Pathology of the Nervous System, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Bente Frølund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Trevor G. Smart
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
19
|
Sun C, Zhu H, Clark S, Gouaux E. Cryo-EM structures reveal native GABA A receptor assemblies and pharmacology. Nature 2023; 622:195-201. [PMID: 37730991 PMCID: PMC10550821 DOI: 10.1038/s41586-023-06556-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023]
Abstract
Type A γ-aminobutyric acid receptors (GABAARs) are the principal inhibitory receptors in the brain and the target of a wide range of clinical agents, including anaesthetics, sedatives, hypnotics and antidepressants1-3. However, our understanding of GABAAR pharmacology has been hindered by the vast number of pentameric assemblies that can be derived from 19 different subunits4 and the lack of structural knowledge of clinically relevant receptors. Here, we isolate native murine GABAAR assemblies containing the widely expressed α1 subunit and elucidate their structures in complex with drugs used to treat insomnia (zolpidem (ZOL) and flurazepam) and postpartum depression (the neurosteroid allopregnanolone (APG)). Using cryo-electron microscopy (cryo-EM) analysis and single-molecule photobleaching experiments, we uncover three major structural populations in the brain: the canonical α1β2γ2 receptor containing two α1 subunits, and two assemblies containing one α1 and either an α2 or α3 subunit, in which the single α1-containing receptors feature a more compact arrangement between the transmembrane and extracellular domains. Interestingly, APG is bound at the transmembrane α/β subunit interface, even when not added to the sample, revealing an important role for endogenous neurosteroids in modulating native GABAARs. Together with structurally engaged lipids, neurosteroids produce global conformational changes throughout the receptor that modify the ion channel pore and the binding sites for GABA and insomnia medications. Our data reveal the major α1-containing GABAAR assemblies, bound with endogenous neurosteroid, thus defining a structural landscape from which subtype-specific drugs can be developed.
Collapse
Affiliation(s)
- Chang Sun
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Hongtao Zhu
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Sarah Clark
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA
| | - Eric Gouaux
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA.
- Howard Hughes Medical Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
20
|
Rupprecht R, Pradhan AK, Kufner M, Brunner LM, Nothdurfter C, Wein S, Schwarzbach J, Puig X, Rupprecht C, Rammes G. Neurosteroids and translocator protein 18 kDa (TSPO) in depression: implications for synaptic plasticity, cognition, and treatment options. Eur Arch Psychiatry Clin Neurosci 2023; 273:1477-1487. [PMID: 36574032 DOI: 10.1007/s00406-022-01532-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/30/2022] [Indexed: 12/28/2022]
Abstract
There is need for novel fast acting treatment options in affective disorders. 3α-reduced neurosteroids such as allopregnanolone are powerful positive allosteric modulators of GABAA receptors and target also extrasynaptic receptors. Their synthesis is mediated by the translocator protein 18 kDa (TSPO). TSPO ligands not only promote endogenous neurosteroidogenesis, but also exert a broad spectrum of functions involving modulation of mitochondrial activity and acting as anti-inflammatory and neuroregenerative agents. Besides affective symptoms, in depression cognitive impairment can be frequently observed, which may be ameliorated through targeting of extrasynaptic GABAA receptors either via TSPO ligands or exogenously administered 3α-reduced neurosteroids. Interestingly, recent findings indicate an enhanced activation of the complement system, e.g., enhanced expression of C1q, both in depression and dementia. It is of note that benzodiazepines have been shown to reduce long-term potentiation and to cause cognitive decline. Intriguingly, TSPO may be crucial in mediating the effects of benzodiazepines on synaptic pruning. Here, we discuss how benzodiazepines and TSPO may interfere with synaptic pruning. Moreover, we highlight recent developments of TSPO ligands and 3α-reduced neurosteroids as therapeutic agents. Etifoxine is the only clinically available TSPO ligand so far and has been studied in anxiety disorders. Regarding 3α-reduced neurosteroids, brexanolone, an intravenous formulation of allopregnanolone, has been approved for the treatment of postpartum depression and zuranolone, an orally available 3α-reduced neurosteroid, is currently being studied in major depressive disorder and postpartum depression. As such, 3α-reduced neurosteroids and TSPO ligands may constitute promising treatment approaches for affective disorders.
Collapse
Affiliation(s)
- Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany.
| | - Arpit Kumar Pradhan
- Experimental Neuropharmacology, Department of Anesthesiology, Technical University Munich, Munich, Germany
| | - Marco Kufner
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| | - Lisa Marie Brunner
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| | - Caroline Nothdurfter
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| | - Simon Wein
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| | - Jens Schwarzbach
- Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053, Regensburg, Germany
| | - Xenia Puig
- Experimental Neuropharmacology, Department of Anesthesiology, Technical University Munich, Munich, Germany
| | - Christian Rupprecht
- Experimental Neuropharmacology, Department of Anesthesiology, Technical University Munich, Munich, Germany
| | - Gerhard Rammes
- Experimental Neuropharmacology, Department of Anesthesiology, Technical University Munich, Munich, Germany
| |
Collapse
|
21
|
Walton NL, Antonoudiou P, Maguire JL. Neurosteroid influence on affective tone. Neurosci Biobehav Rev 2023; 152:105327. [PMID: 37499891 PMCID: PMC10528596 DOI: 10.1016/j.neubiorev.2023.105327] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 07/07/2023] [Accepted: 07/23/2023] [Indexed: 07/29/2023]
Abstract
Affective disorders such as depression and anxiety are among the most prevalent psychiatric illnesses and causes of disability worldwide. The recent FDA-approval of a novel antidepressant treatment, ZULRESSO® (Brexanolone), a synthetic neurosteroid has fueled interest into the role of neurosteroids in the pathophysiology of depression as well as the mechanisms mediating the antidepressant effects of these compounds. The majority of studies examining the impact of neurosteroids on affective states have relied on the administration of exogenous neurosteroids; however, neurosteroids can also be synthesized endogenously from cholesterol or steroid hormone precursors. Despite the well-established influence of exogenous neurosteroids on affective states, we still lack an understanding of the role of endogenous neurosteroids in modulating affective tone. This review aims to summarize the current literature supporting the influence of neurosteroids on affective states in clinical and preclinical studies, as well as recent evidence suggesting that endogenous neurosteroids may set a baseline affective tone.
Collapse
Affiliation(s)
- Najah L Walton
- Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA; Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Pantelis Antonoudiou
- Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA; Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Jamie L Maguire
- Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA; Department of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
22
|
Legesse DH, Fan C, Teng J, Zhuang Y, Howard RJ, Noviello CM, Lindahl E, Hibbs RE. Structural insights into opposing actions of neurosteroids on GABA A receptors. Nat Commun 2023; 14:5091. [PMID: 37607940 PMCID: PMC10444788 DOI: 10.1038/s41467-023-40800-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023] Open
Abstract
γ-Aminobutyric acid type A (GABAA) receptors mediate fast inhibitory signaling in the brain and are targets of numerous drugs and endogenous neurosteroids. A subset of neurosteroids are GABAA receptor positive allosteric modulators; one of these, allopregnanolone, is the only drug approved specifically for treating postpartum depression. There is a consensus emerging from structural, physiological and photolabeling studies as to where positive modulators bind, but how they potentiate GABA activation remains unclear. Other neurosteroids are negative modulators of GABAA receptors, but their binding sites remain debated. Here we present structures of a synaptic GABAA receptor bound to allopregnanolone and two inhibitory sulfated neurosteroids. Allopregnanolone binds at the receptor-bilayer interface, in the consensus potentiator site. In contrast, inhibitory neurosteroids bind in the pore. MD simulations and electrophysiology support a mechanism by which allopregnanolone potentiates channel activity and suggest the dominant mechanism for sulfated neurosteroid inhibition is through pore block.
Collapse
Affiliation(s)
| | - Chen Fan
- Dept. of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Jinfeng Teng
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Yuxuan Zhuang
- Dept. of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Rebecca J Howard
- Dept. of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Colleen M Noviello
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Erik Lindahl
- Dept. of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden.
- Dept. of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden.
| | - Ryan E Hibbs
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
23
|
Lawande NV, Conklin EA, Christian-Hinman CA. Sex and gonadectomy modify behavioral seizure susceptibility and mortality in a repeated low-dose kainic acid systemic injection paradigm in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541824. [PMID: 37292790 PMCID: PMC10245840 DOI: 10.1101/2023.05.22.541824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Objective Sex differences in epilepsy appear driven in part due to effects of gonadal steroids, with varying results in experimental models based on species, strain, and method of seizure induction. Furthermore, removing a main source of these steroids via gonadectomy may impact seizure characteristics differently in males and females. Repeated low-dose kainic acid (RLDKA) systemic injection paradigms were recently shown to reliably induce status epilepticus (SE) and hippocampal histopathology in C57BL/6J mice. Here, we investigated whether seizure susceptibility in a RLDKA injection protocol exhibits a sex difference, and whether gonadectomy differentially influences response to this seizure induction paradigm in males and females. Methods Adult C57BL/6J mice were left gonad-intact as controls or gonadectomized (females: ovariectomized, OVX; males: orchidectomized, ORX). At least 2 weeks later, KA was injected i.p. every 30 minutes at 7.5 mg/kg or less until the animal reached SE, defined by at least 5 generalized seizures (GS, Racine stage 3 or higher). Parameters of susceptibility to GS induction, SE development, and mortality rates were quantified. Results No differences in seizure susceptibility or mortality were observed between control males and control females. ORX males exhibited increased susceptibility and reduced latency to both GS and SE, but OVX females exhibited increased susceptibility and reduced latency to SE only. However, ORX males, but not OVX females, exhibited strongly increased seizure-induced mortality. Significance The RLDKA protocol is notable for its efficacy in inducing SE and seizure-induced histopathology in C57BL/6J mice, the background for many transgenic strains in current use in epilepsy research. The present results indicate that this protocol may be beneficial for investigating the effects of gonadal hormone replacement on seizure susceptibility, mortality, and seizure-induced histopathology, and that gonadectomy unmasks sex differences in susceptibility to seizures and mortality not observed in gonad-intact controls.
Collapse
Affiliation(s)
- Niraj V. Lawande
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| | - Elisabeth A. Conklin
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| | - Catherine A. Christian-Hinman
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, 61801 USA
| |
Collapse
|
24
|
Walton NL, Antonoudiou P, Barros L, Dargan T, DiLeo A, Evans-Strong A, Gabby J, Howard S, Paracha R, Sánchez EJ, Weiss GL, Kong D, Maguire JL. Impaired Endogenous Neurosteroid Signaling Contributes to Behavioral Deficits Associated With Chronic Stress. Biol Psychiatry 2023; 94:249-261. [PMID: 36736870 PMCID: PMC10363189 DOI: 10.1016/j.biopsych.2023.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/21/2022] [Accepted: 01/14/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Chronic stress is a major risk factor for psychiatric illnesses, including depression. However, the pathophysiological mechanisms whereby stress leads to mood disorders remain unclear. Allopregnanolone acts as a positive allosteric modulator preferentially on δ subunit-containing GABAA (gamma-aminobutyric acid A) receptors. Accumulating clinical and preclinical evidence supports the antidepressant effects of exogenous administration of allopregnanolone analogs; yet, the role of endogenous allopregnanolone in the pathophysiology of depression remains unknown. METHODS We utilized a chronic unpredictable stress (CUS) mouse model, followed by behavioral and biochemical assays, to examine whether altered neurosteroid signaling contributes to behavioral outcomes following CUS. We subsequently performed in vivo CRISPR (clustered regularly interspaced short palindromic repeats) knockdown of rate-limiting enzymes involved in allopregnanolone synthesis, 5α-reductase type 1 and 2 (5α1/2), in addition to lentiviral overexpression of 5α1/2 in the basolateral amygdala (BLA) of mice that underwent CUS to assess the impact of 5α1/2 on behavioral outcomes. RESULTS The expression of δ subunit-containing GABAA receptors and endogenous levels of allopregnanolone were reduced in the BLA following CUS. Treatment with an exogenous allopregnanolone analog, SGE-516, was sufficient to increase allopregnanolone levels in the BLA following CUS. Knockdown of 5α1/2 in the BLA mimicked the behavioral outcomes associated with CUS. Conversely, overexpression of 5α1/2 in the BLA improved behavioral outcomes following CUS. CONCLUSIONS Our findings demonstrate that chronic stress impairs endogenous neurosteroid signaling in the BLA, which is sufficient to induce behavioral deficits. Further, these studies suggest that allopregnanolone-based treatments may directly target the underlying pathophysiology of mood disorders suggesting that targeting endogenous neurosteroidogenesis may offer a novel therapeutic strategy.
Collapse
Affiliation(s)
- Najah L Walton
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Pantelis Antonoudiou
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Lea Barros
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts; Building Diversity in Biomedical Sciences Program, Tufts University School of Medicine, Boston, Massachusetts; Department of Biology, Hamilton College, Clinton, New York
| | - Tauryn Dargan
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Alyssa DiLeo
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Aidan Evans-Strong
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Jenah Gabby
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts; Building Diversity in Biomedical Sciences Program, Tufts University School of Medicine, Boston, Massachusetts; Louis Stokes Alliance for Minority Participation, Tufts University, Medford, Massachusetts
| | - Samantha Howard
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts; Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Rumzah Paracha
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Edgardo J Sánchez
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts; Building Diversity in Biomedical Sciences Program, Tufts University School of Medicine, Boston, Massachusetts; Department of Chemistry, University of Puerto Rico, Cayey, Puerto Rico
| | - Grant L Weiss
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Dong Kong
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts; Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jamie L Maguire
- Department of Neuroscience, Program of Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
25
|
Covey DF, Evers AS, Izumi Y, Maguire JL, Mennerick SJ, Zorumski CF. Neurosteroid enantiomers as potentially novel neurotherapeutics. Neurosci Biobehav Rev 2023; 149:105191. [PMID: 37085023 PMCID: PMC10750765 DOI: 10.1016/j.neubiorev.2023.105191] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/23/2023]
Abstract
Endogenous neurosteroids and synthetic neuroactive steroids (NAS) are important targets for therapeutic development in neuropsychiatric disorders. These steroids modulate major signaling systems in the brain and intracellular processes including inflammation, cellular stress and autophagy. In this review, we describe studies performed using unnatural enantiomers of key neurosteroids, which are physiochemically identical to their natural counterparts except for rotation of polarized light. These studies led to insights in how NAS interact with receptors, ion channels and intracellular sites of action. Certain effects of NAS show high enantioselectivity, consistent with actions in chiral environments and likely direct interactions with signaling proteins. Other effects show no enantioselectivity and even reverse enantioselectivity. The spectrum of effects of NAS enantiomers raises the possibility that these agents, once considered only as tools for preclinical studies, have therapeutic potential that complements and in some cases may exceed their natural counterparts. Here we review studies of NAS enantiomers from the perspective of their potential development as novel neurotherapeutics.
Collapse
Affiliation(s)
- Douglas F Covey
- Departments of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA; Anesthesiology Washington University School of Medicine, St. Louis, MO, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Alex S Evers
- Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA; Anesthesiology Washington University School of Medicine, St. Louis, MO, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Yukitoshi Izumi
- Departments of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Jamie L Maguire
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Steven J Mennerick
- Departments of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F Zorumski
- Departments of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
26
|
Hantsoo L, Payne JL. Towards understanding the biology of premenstrual dysphoric disorder: From genes to GABA. Neurosci Biobehav Rev 2023; 149:105168. [PMID: 37059403 PMCID: PMC10176022 DOI: 10.1016/j.neubiorev.2023.105168] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/10/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Premenstrual dysphoric disorder (PMDD) is a severe mood disorder, with affective symptoms that rise and fall in concert with the hormonal fluctuations of the menstrual cycle. PMDD's pathophysiology is poorly understood. This review describes recent research on potential biological contributors to PMDD, with a focus on neuroactive steroids, genetics, neuroimaging and cellular studies. Studies suggest that a key contributor is abnormal central nervous system (CNS) response to fluctuations in neuroactive steroid hormones. Imaging studies are limited but support alterations in serotonergic and GABA transmission. Genetic studies suggest heritability, yet specific genetic contributors have not been characterized. Finally, recent cutting-edge cellular studies indicate an underlying vulnerability to the effect of sex hormones at a cellular level. Overall the findings across studies do not yet fit together into a complete description of the underlying biology of PMDD. It is possible that PMDD consists of biological subtypes, and future research may benefit from a subtyping approach.
Collapse
Affiliation(s)
- Liisa Hantsoo
- Department of Psychiatry & Behavioral Sciences, The Johns Hopkins University School of Medicine, 550 N. Broadway Street, Baltimore, MD 21205, USA.
| | - Jennifer L Payne
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, PO Box 800548, Charlottesville, VA 22908, USA
| |
Collapse
|
27
|
Pierce SR, Germann AL, Xu SQ, Menon SL, Ortells MO, Arias HR, Akk G. Mutational Analysis of Anesthetic Binding Sites and Their Effects on GABA A Receptor Activation and Modulation by Positive Allosteric Modulators of the α7 Nicotinic Receptor. Biomolecules 2023; 13:698. [PMID: 37189445 PMCID: PMC10135968 DOI: 10.3390/biom13040698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
The positive allosteric modulators (PAMs) of the α7 nicotinic receptor N-(5-Cl-2-hydroxyphenyl)-N'-[2-Cl-5-(trifluoromethyl)phenyl]-urea (NS-1738) and (E)-3-(furan-2-yl)-N-(p-tolyl)-acrylamide (PAM-2) potentiate the α1β2γ2L GABAA receptor through interactions with the classic anesthetic binding sites located at intersubunit interfaces in the transmembrane domain of the receptor. In the present study, we employed mutational analysis to investigate in detail the involvement and contributions made by the individual intersubunit interfaces to receptor modulation by NS-1738 and PAM-2. We show that mutations to each of the anesthetic-binding intersubunit interfaces (β+/α-, α+/β-, and γ+/β-), as well as the orphan α+/γ- interface, modify receptor potentiation by NS-1738 and PAM-2. Furthermore, mutations to any single interface can fully abolish potentiation by the α7-PAMs. The findings are discussed in the context of energetic additivity and interactions between the individual binding sites.
Collapse
Affiliation(s)
- Spencer R. Pierce
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Allison L. Germann
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sophia Q. Xu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Saumith L. Menon
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marcelo O. Ortells
- Facultad de Medicina, Universidad de Morón, CONICET, Morón 1708, Argentina
| | - Hugo R. Arias
- Department of Pharmacology and Physiology, Oklahoma State University College of Osteopathic Medicine, Tahlequah, OK 74464, USA
| | - Gustav Akk
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
28
|
Sivcev S, Kudova E, Zemkova H. Neurosteroids as positive and negative allosteric modulators of ligand-gated ion channels: P2X receptor perspective. Neuropharmacology 2023; 234:109542. [PMID: 37040816 DOI: 10.1016/j.neuropharm.2023.109542] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/06/2023] [Accepted: 04/07/2023] [Indexed: 04/13/2023]
Abstract
Neurosteroids are steroids synthesized de novo in the brain from cholesterol in an independent manner from peripheral steroid sources. The term "neuroactive steroid" includes all steroids independent of their origin, and newly synthesized analogs of neurosteroids that modify neuronal activities. In vivo application of neuroactive steroids induces potent anxiolytic, antidepressant, anticonvulsant, sedative, analgesic and amnesic effects, mainly through interaction with the γ-aminobutyric acid type-A receptor (GABAAR). However, neuroactive steroids also act as positive or negative allosteric regulators on several ligand-gated channels including N-methyl-d-aspartate receptors (NMDARs), nicotinic acetylcholine receptors (nAChRs) and ATP-gated purinergic P2X receptors. Seven different P2X subunits (P2X1-7) can assemble to form homotrimeric or heterotrimeric ion channels permeable for monovalent cations and calcium. Among them, P2X2, P2X4, and P2X7 are the most abundant within the brain and can be regulated by neurosteroids. Transmembrane domains are necessary for neurosteroid binding, however, no generic motif of amino acids can accurately predict the neurosteroid binding site for any of the ligand-gated ion channels including P2X. Here, we will review what is currently known about the modulation of rat and human P2X by neuroactive steroids and the possible structural determinants underlying neurosteroid-induced potentiation and inhibition of the P2X2 and P2X4 receptors.
Collapse
Affiliation(s)
- Sonja Sivcev
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Eva Kudova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Hana Zemkova
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
29
|
McKinstry-Wu AR, Wasilczuk AZ, Dailey WP, Eckenhoff RG, Kelz MB. In Vivo Photoadduction of Anesthetic Ligands in Mouse Brain Markedly Extends Sedation and Hypnosis. J Neurosci 2023; 43:2338-2348. [PMID: 36849414 PMCID: PMC10072292 DOI: 10.1523/jneurosci.1884-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/01/2023] Open
Abstract
Photoaffinity ligands are best known as tools used to identify the specific binding sites of drugs to their molecular targets. However, photoaffinity ligands have the potential to further define critical neuroanatomic targets of drug action. In the brains of WT male mice, we demonstrate the feasibility of using photoaffinity ligands in vivo to prolong anesthesia via targeted yet spatially restricted photoadduction of azi-m-propofol (aziPm), a photoreactive analog of the general anesthetic propofol. Systemic administration of aziPm with bilateral near-ultraviolet photoadduction in the rostral pons, at the border of the parabrachial nucleus and locus coeruleus, produced a 20-fold increase in the duration of sedative and hypnotic effects compared with control mice without UV illumination. Photoadduction that missed the parabrachial-coerulean complex also failed to extend the sedative or hypnotic actions of aziPm and was indistinguishable from nonadducted controls. Paralleling the prolonged behavioral and EEG consequences of on target in vivo photoadduction, we conducted electrophysiologic recordings in rostral pontine brain slices. Using neurons within the locus coeruleus to further highlight the cellular consequences of irreversible aziPm binding, we demonstrate transient slowing of spontaneous action potentials with a brief bath application of aziPm that becomes irreversible on photoadduction. Together, these findings suggest that photochemistry-based strategies are a viable new approach for probing CNS physiology and pathophysiology.SIGNIFICANCE STATEMENT Photoaffinity ligands are drugs capable of light-induced irreversible binding, which have unexploited potential to identify the neuroanatomic sites of drug action. We systemically administer a centrally acting anesthetic photoaffinity ligand in mice, conduct localized photoillumination within the brain to covalently adduct the drug at its in vivo sites of action, and successfully enrich irreversible drug binding within a restricted 250 µm radius. When photoadduction encompassed the pontine parabrachial-coerulean complex, anesthetic sedation and hypnosis was prolonged 20-fold, thus illustrating the power of in vivo photochemistry to help unravel neuronal mechanisms of drug action.
Collapse
Affiliation(s)
- Andrew R McKinstry-Wu
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Philadelphia 19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Philadelphia 19104
| | - Andrzej Z Wasilczuk
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Philadelphia 19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Philadelphia 19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Philadelphia 19104
| | - William P Dailey
- Department of Chemistry, University of Pennsylvania School of Arts and Sciences, Philadelphia, Pennsylvania 19104
| | - Roderic G Eckenhoff
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Philadelphia 19104
| | - Max B Kelz
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Philadelphia 19104
- Mahoney Institute for Neurosciences, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Philadelphia 19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Philadelphia 19104
| |
Collapse
|
30
|
Faleschini T, Syafni N, Schulte HL, Garifulina A, Hering S, Espindola LS, Hamburger M. A neolignan from Connarus tuberosus as an allosteric GABA A receptor modulator at the neurosteroid binding site. Biomed Pharmacother 2023; 161:114498. [PMID: 36906973 DOI: 10.1016/j.biopha.2023.114498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
In a screening of a small library of extracts from plants of the Amazonian and Cerrado biomes, a hexane extract of Connarus tuberosus roots was found to significantly potentiate the GABA induced fluorescence in a fluorescence (FLIPR) assay in CHO cells stably expressing the α1β2γ2 subtype of human GABAA receptors. With the aid of HPLC-based activity profiling the activity was linked to the neolignan connarin. In CHO cells the activity of connarin was not abolished by increasing concentrations of flumazenil, while the effect of diazepam was increased by increasing concentrations of connarin. The effect of connarin was abolished by pregnenolone sulfate (PREGS) in a concentration-dependent manner, and the effect of allopregnanolone was further increased by increasing concentrations of connarin. In a two-microelectrode voltage clamp assay with Xenopus laevis oocytes transiently expressing GABAA receptors composed of human α1β2γ2S and α1β2 subunits connarin potentiated the GABA-induced currents, with EC50 values of 1.2 ± 0.3 μM (α1β2γ2S) and 1.3 ± 0.4 μM (α1β2), and with a maximum enhancement of currents Emax of 1959 ± 70% (α1β2γ2S) and 185 ± 48% (α1β2). The activation induced by connarin was abolished by increasing concentrations of PREGS.
Collapse
Affiliation(s)
- Teresa Faleschini
- Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland
| | - Nova Syafni
- Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; Faculty of Pharmacy and Sumatran Biota Laboratory, Andalas University, 25163 Padang, West Sumatra, Indonesia
| | - Heidi Luise Schulte
- Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; Universidade de Brasília, Laboratório de Farmacognosia, Campus Universitário Darcy Ribeiro, 70910-900 Brasília, DF, Brazil
| | - Aleksandra Garifulina
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Steffen Hering
- Universidade de Brasília, Laboratório de Farmacognosia, Campus Universitário Darcy Ribeiro, 70910-900 Brasília, DF, Brazil
| | - Laila Salmen Espindola
- Universidade de Brasília, Laboratório de Farmacognosia, Campus Universitário Darcy Ribeiro, 70910-900 Brasília, DF, Brazil
| | - Matthias Hamburger
- Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
31
|
Tateiwa H, Chintala SM, Chen Z, Wang L, Amtashar F, Bracamontes J, Germann AL, Pierce SR, Covey DF, Akk G, Evers AS. The Mechanism of Enantioselective Neurosteroid Actions on GABA A Receptors. Biomolecules 2023; 13:341. [PMID: 36830708 PMCID: PMC9953308 DOI: 10.3390/biom13020341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The neurosteroid allopregnanolone (ALLO) and pregnanolone (PREG), are equally effective positive allosteric modulators (PAMs) of GABAA receptors. Interestingly, the PAM effects of ALLO are strongly enantioselective, whereas those of PREG are not. This study was aimed at determining the basis for this difference in enantioselectivity. The oocyte electrophysiology studies showed that ent-ALLO potentiates GABA-elicited currents in α1β3 GABAA receptors with lower potency and efficacy than ALLO, PREG or ent-PREG. The small PAM effect of ent-ALLO was prevented by the α1(Q242L) mutation in the intersubunit neurosteroid binding site between the β3 and α1 subunits. Consistent with this result, neurosteroid analogue photolabeling with mass spectrometric readout, showed that ent-ALLO binds weakly to the β3-α1 intersubunit binding site in comparison to ALLO, PREG and ent-PREG. Rigid body docking predicted that ent-ALLO binds in the intersubunit site with a preferred orientation 180° different than ALLO, PREG or ent-PREG, potentially explaining its weak binding and effect. Photolabeling studies did not identify differences between ALLO and ent-ALLO binding to the α1 or β3 intrasubunit binding sites that also mediate neurosteroid modulation of GABAA receptors. The results demonstrate that differential binding of ent-ALLO and ent-PREG to the β3-α1 intersubunit site accounts for the difference in enantioselectivity between ALLO and PREG.
Collapse
Affiliation(s)
- Hiroki Tateiwa
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kochi 7838505, Japan
| | | | - Ziwei Chen
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Taylor Institute for Innovative Psychiatric Research, St. Louis, MO 63110, USA
| | - Lei Wang
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430074, China
| | - Fatima Amtashar
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John Bracamontes
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Allison L. Germann
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Spencer R. Pierce
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Douglas F. Covey
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Taylor Institute for Innovative Psychiatric Research, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Developmental Biology (Pharmacology), Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gustav Akk
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Taylor Institute for Innovative Psychiatric Research, St. Louis, MO 63110, USA
| | - Alex S. Evers
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Taylor Institute for Innovative Psychiatric Research, St. Louis, MO 63110, USA
- Department of Developmental Biology (Pharmacology), Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
32
|
Bryson A, Reid C, Petrou S. Fundamental Neurochemistry Review: GABA A receptor neurotransmission and epilepsy: Principles, disease mechanisms and pharmacotherapy. J Neurochem 2023; 165:6-28. [PMID: 36681890 DOI: 10.1111/jnc.15769] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/12/2022] [Accepted: 01/04/2023] [Indexed: 01/23/2023]
Abstract
Epilepsy is a common neurological disorder associated with alterations of excitation-inhibition balance within brain neuronal networks. GABAA receptor neurotransmission is the most prevalent form of inhibitory neurotransmission and is strongly implicated in both the pathophysiology and treatment of epilepsy, serving as a primary target for antiseizure medications for over a century. It is now established that GABA exerts a multifaceted influence through an array of GABAA receptor subtypes that extends far beyond simply negating excitatory activity. As the role of GABAA neurotransmission within inhibitory circuits is elaborated, this will enable the development of precision therapies that correct the network dysfunction underlying epileptic pathology.
Collapse
Affiliation(s)
- Alexander Bryson
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.,Department of Neurology, Austin Health, Heidelberg, Victoria, Australia
| | - Christopher Reid
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Steven Petrou
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.,Praxis Precision Medicines, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
33
|
Gao Q, Sun W, Wang YR, Li ZF, Zhao F, Geng XW, Xu KY, Chen D, Liu K, Xing Y, Liu W, Wei S. Role of allopregnanolone-mediated γ-aminobutyric acid A receptor sensitivity in the pathogenesis of premenstrual dysphoric disorder: Toward precise targets for translational medicine and drug development. Front Psychiatry 2023; 14:1140796. [PMID: 36937732 PMCID: PMC10017536 DOI: 10.3389/fpsyt.2023.1140796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/09/2023] [Indexed: 03/06/2023] Open
Abstract
Premenstrual dysphoric disorder (PMDD) can be conceptualized as a disorder of suboptimal sensitivity to neuroactive steroid hormones. Its core symptoms (emotional instability, irritability, depression, and anxiety) are related to the increase of stress sensitivity due to the fluctuation of hormone level in luteal phase of the menstrual cycle. In this review, we describe the emotional regulatory effect of allopregnanolone (ALLO), and summarize the relationship between ALLO and γ-aminobutyric acid A (GABAA) receptor subunits based on rodent experiments and clinical observations. A rapid decrease in ALLO reduces the sensitivity of GABAA receptor, and reduces the chloride influx, hindered the inhibitory effect of GABAergic neurons on pyramidal neurons, and then increased the excitability of pyramidal neurons, resulting in PMDD-like behavior. Finally, we discuss in depth the treatment of PMDD with targeted GABAA receptors, hoping to find a precise target for drug development and subsequent clinical application. In conclusion, PMDD pathophysiology is rooted in GABAA receptor sensitivity changes caused by rapid changes in ALLO levels. Targeting GABAA receptors may alleviate the occurrence of PMDD.
Collapse
Affiliation(s)
- Qian Gao
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yue-Rui Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zi-Fa Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Feng Zhao
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xi-Wen Geng
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kai-Yong Xu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dan Chen
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kun Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Xing
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Liu
- Department of Encephalopathy, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Wei Liu,
| | - Sheng Wei
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- Chinese Medicine and Brain Science Core Facility, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Sheng Wei,
| |
Collapse
|
34
|
Petroff JT, Dietzen NM, Santiago-McRae E, Deng B, Washington MS, Chen LJ, Trent Moreland K, Deng Z, Rau M, Fitzpatrick JAJ, Yuan P, Joseph TT, Hénin J, Brannigan G, Cheng WWL. Open-channel structure of a pentameric ligand-gated ion channel reveals a mechanism of leaflet-specific phospholipid modulation. Nat Commun 2022; 13:7017. [PMID: 36385237 PMCID: PMC9668969 DOI: 10.1038/s41467-022-34813-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022] Open
Abstract
Pentameric ligand-gated ion channels (pLGICs) mediate synaptic transmission and are sensitive to their lipid environment. The mechanism of phospholipid modulation of any pLGIC is not well understood. We demonstrate that the model pLGIC, ELIC (Erwinia ligand-gated ion channel), is positively modulated by the anionic phospholipid, phosphatidylglycerol, from the outer leaflet of the membrane. To explore the mechanism of phosphatidylglycerol modulation, we determine a structure of ELIC in an open-channel conformation. The structure shows a bound phospholipid in an outer leaflet site, and structural changes in the phospholipid binding site unique to the open-channel. In combination with streamlined alchemical free energy perturbation calculations and functional measurements in asymmetric liposomes, the data support a mechanism by which an anionic phospholipid stabilizes the activated, open-channel state of a pLGIC by specific, state-dependent binding to this site.
Collapse
Affiliation(s)
- John T Petroff
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Noah M Dietzen
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ezry Santiago-McRae
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Brett Deng
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Maya S Washington
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Lawrence J Chen
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - K Trent Moreland
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Zengqin Deng
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael Rau
- Center for Cellular Imaging, Washington University School of Medicine, Saint Louis, MO, USA
| | - James A J Fitzpatrick
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for Cellular Imaging, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University School of Medicine, Saint Louis, MO, USA
| | - Peng Yuan
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Thomas T Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jérôme Hénin
- Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique, Université Paris Cité, CNRS UPR 9080, Paris, France
| | - Grace Brannigan
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
- Department of Physics, Rutgers University, Camden, NJ, USA
| | - Wayland W L Cheng
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
35
|
Bukanova JV, Kondratenko RV, Solntseva EI. Positive allosteric modulators of GABA A receptor restore chloride current from blockade by competitive antagonists in a ligand-dependent manner. J Steroid Biochem Mol Biol 2022; 224:106158. [PMID: 35931327 DOI: 10.1016/j.jsbmb.2022.106158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/26/2022] [Accepted: 07/30/2022] [Indexed: 10/31/2022]
Abstract
γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter. GABA receptor type A (GABAAR) possesses binding sites for a large group of pharmacological agents which are supposed to interact allosterically with each other. The aim of this work was to study the interaction between the positive allosteric modulators (PAMs) and the competitive antagonists of GABAARs. The GABA-induced chloride current (IGABA) was measured in isolated Purkinje cells of rat cerebellum using the patch-clamp technique. PAMs, neurosteroid allopregnanolone (Allo) and zolpidem (Zolp), a drug that positively modulates the GABAAR through interaction with the benzodiazepine (BDZ) site, doubled the IGABA amplitude in the control solution. Competitive antagonist of GABAARs, bicuculline (Bic, 5 µM) blocked the IGABA by 90%. The addition of 1 μM Allo or 0.5 µM Zolp to the Bic solution caused an unblocking effect, so that the IGABA amplitude increased 10 and 4 times from control value, correspondingly. This unblocking effect developed slowly, as evidenced by a threefold increase in the current rise time. Competitive antagonist of GABAARs, gabazine (GBZ, 0.5 µM) blocked the IGABA by 87%. The addition of 1 μM Allo to the GBZ solution caused an unblocking effect, so that the IGABA amplitude increased 7-fold. However, the addition of 0.5 µM Zolp to the GBZ solution did not cause an unblocking effect. So, Allo appeared to have a stronger unblocking potential than Zolp, and Bic binding site showed a higher sensitivity to the action of unblocking PAMs than GBZ binding site. The results indicate for the first time the existence of an allosteric relationship between the sites binding PAMs and the competitive antagonists of GABAAR.
Collapse
|
36
|
Tongta S, Daendee S, Kalandakanond-Thongsong S. Effects of estrogen receptor β or G protein-coupled receptor 30 activation on anxiety-like behaviors in relation to GABAergic transmission in stress-ovariectomized rats. Neurosci Lett 2022; 789:136885. [PMID: 36152742 DOI: 10.1016/j.neulet.2022.136885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 12/24/2022]
Abstract
For mental disorders such as anxiety and depression, stress and stressful events are considered as precipitating causes that may be enhanced by estrogen variability. This condition is proven by the higher vulnerability of women than men. Despite the complexity of underlying mechanisms, the gamma-aminobutyric acid (GABA) system piques interest as its receptor contains multiple psychoactive modulatory sites including neurosteroids. Moreover, according to clinical and experimental reports, GABA-associated genes can be altered by stress and hormonal status. Therefore, this study investigated the effects of estrogen receptor β (ERβ) or G protein-coupled receptor 30 (GPR30) activation on anxiety/depression-like behaviors and the alterations in the GABA-associated gene of ovariectomized rats under chronic mild stress (CMS). Mild stressors were focused on because they represent a realistic simulation of daily life stress. In this study, ovariectomized rats were treated with vehicle, estradiol (E2), diarylpropionitrile (DPN; ERβ agonist) or G1 (GPR30 agonist) and exposed to 4-week CMS. The results showed that E2, DPN, and G1 treatments reduced anxiety-like behaviors without affecting depression-like behaviors. Concurrently, the GABA level and most GABA- and neurosteroid-associated mRNAs were altered by E2. Similar mRNA profiles were observed in DPN- and E2-administrations but not in G1 treatment. Collectively, these data suggest that estrogen exerts an anxiolytic-like action through either ERβ and/or GPR30 activation, and the modulatory effects of estrogen on GABAergic system are likely to be modulated through ERβ. The findings of this study therefore further provide insights into the roles of estrogen and daily mild stressors in GABA-related activity and behavioral responses, especially anxiety.
Collapse
Affiliation(s)
- Sushawadee Tongta
- Department of Veterinary Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Suwaporn Daendee
- College of Medicine and Public Health, Ubon Ratchathani University, Ubon Ratchathani, Thailand
| | | |
Collapse
|
37
|
Shin HR, Citron YR, Wang L, Tribouillard L, Goul CS, Stipp R, Sugasawa Y, Jain A, Samson N, Lim CY, Davis OB, Castaneda-Carpio D, Qian M, Nomura DK, Perera RM, Park E, Covey DF, Laplante M, Evers AS, Zoncu R. Lysosomal GPCR-like protein LYCHOS signals cholesterol sufficiency to mTORC1. Science 2022; 377:1290-1298. [PMID: 36007018 PMCID: PMC10023259 DOI: 10.1126/science.abg6621] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Lysosomes coordinate cellular metabolism and growth upon sensing of essential nutrients, including cholesterol. Through bioinformatic analysis of lysosomal proteomes, we identified lysosomal cholesterol signaling (LYCHOS, previously annotated as G protein-coupled receptor 155), a multidomain transmembrane protein that enables cholesterol-dependent activation of the master growth regulator, the protein kinase mechanistic target of rapamycin complex 1 (mTORC1). Cholesterol bound to the amino-terminal permease-like region of LYCHOS, and mutating this site impaired mTORC1 activation. At high cholesterol concentrations, LYCHOS bound to the GATOR1 complex, a guanosine triphosphatase (GTPase)-activating protein for the Rag GTPases, through a conserved cytoplasm-facing loop. By sequestering GATOR1, LYCHOS promotes cholesterol- and Rag-dependent recruitment of mTORC1 to lysosomes. Thus, LYCHOS functions in a lysosomal pathway for cholesterol sensing and couples cholesterol concentrations to mTORC1-dependent anabolic signaling.
Collapse
Affiliation(s)
- Hijai R. Shin
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Y. Rose Citron
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lei Wang
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Laura Tribouillard
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, G1R 3S3, Canada
| | - Claire S. Goul
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Robin Stipp
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yusuke Sugasawa
- Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Aakriti Jain
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nolwenn Samson
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, G1R 3S3, Canada
| | - Chun-Yan Lim
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Oliver B. Davis
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - David Castaneda-Carpio
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mingxing Qian
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Daniel K. Nomura
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Rushika M. Perera
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eunyong Park
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Douglas F. Covey
- Department of Developmental Biology and Biochemistry, Washington University School of Medicine, St Louis, MO 63110, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Mathieu Laplante
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, G1R 3S3, Canada
| | - Alex S. Evers
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
- Department of Developmental Biology and Biochemistry, Washington University School of Medicine, St Louis, MO 63110, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
38
|
Solntseva EI, Bukanova JV, Skrebitsky VG, Kudova E. Pregnane neurosteroids exert opposite effects on GABA and glycine-induced chloride current in isolated rat neurons. Hippocampus 2022; 32:552-563. [PMID: 35703084 DOI: 10.1002/hipo.23449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/11/2022]
Abstract
The ability of endogenous neurosteroids (NSs) with pregnane skeleton modified at positions C-3 and C-5 to modulate the functional activity of inhibitory glycine receptors (GlyR) and ionotropic ɣ-aminobutyric acid receptors (GABAA R) was estimated. The glycine and GABA-induced chloride current (IGly and IGABA ) were measured in isolated pyramidal neurons of the rat hippocampus and in isolated rat cerebellar Purkinje cells, respectively. Our experiments demonstrated that pregnane NSs affected IGABA and IGly in a different manner. At low concentrations (up to 5 μM), tested pregnane NSs increased or did not change the peak amplitude of the IGABA , but reduced the IGly by decreasing the peak amplitude and/or accelerating desensitization. Namely, allopregnanolone (ALLO), epipregnanolone (EPI), pregnanolone (PA), pregnanolone sulfate (PAS) and 5β-dihydroprogesterone (5β-DHP) enhanced the IGABA in Purkinje cells. Dose-response curves plotted in the concentration range from 1 nM to 100 μM were smooth for EPI and 5β-DHP, but bell-shaped for ALLO, PA and PAS. The peak amplitude of the IGly was reduced by PA, PAS, and 5α- and 5β-DHP. In contrast, ALLO, ISO and EPI did not modulate it. Dose-response curves for the inhibition of the IGly peak amplitude were smooth for all active compounds. All NSs accelerated desensitization of the IGly . The dose-response relationship for this effect was smooth for ALLO, PA, PAS and 5β-DHP, but it was U-shaped for EPI, 5α-DHP and ISO. These results, together with our previous results on NSs with androstane skeleton, offer comprehensive overview for understanding the mechanisms of effects of NSs on IGly and IGABA .
Collapse
Affiliation(s)
- Elena I Solntseva
- Functional Synaptology Laboratory, Brain Research Department, Research Center of Neurology, Moscow, Russia
| | - Julia V Bukanova
- Functional Synaptology Laboratory, Brain Research Department, Research Center of Neurology, Moscow, Russia
| | - Vladimir G Skrebitsky
- Functional Synaptology Laboratory, Brain Research Department, Research Center of Neurology, Moscow, Russia
| | - Eva Kudova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
39
|
Rupprecht R, Wetzel CH, Dorostkar M, Herms J, Albert NL, Schwarzbach J, Schumacher M, Neumann ID. Translocator protein (18kDa) TSPO: a new diagnostic or therapeutic target for stress-related disorders? Mol Psychiatry 2022; 27:2918-2926. [PMID: 35444254 DOI: 10.1038/s41380-022-01561-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 12/11/2022]
Abstract
Efficient treatment of stress-related disorders, such as depression, is still a major challenge. The onset of antidepressant drug action is generally quite slow, while the anxiolytic action of benzodiazepines is considerably faster. However, their long-term use is impaired by tolerance development, abuse liability and cognitive impairment. Benzodiazepines act as positive allosteric modulators of ɣ-aminobutyric acid type A (GABAA) receptors. 3α-reduced neurosteroids such as allopregnanolone also are positive allosteric GABAA receptor modulators, however, through a site different from that targeted by benzodiazepines. Recently, the administration of neurosteroids such as brexanolone or zuranolone has been shown to rapidly ameliorate symptoms in post-partum depression or major depressive disorder. An attractive alternative to the administration of exogenous neurosteroids is promoting endogenous neurosteroidogenesis via the translocator protein 18k Da (TSPO). TSPO is a transmembrane protein located primarily in mitochondria, which mediates numerous biological functions, e.g., steroidogenesis and mitochondrial bioenergetics. TSPO ligands have been used in positron emission tomography (PET) studies as putative markers of microglia activation and neuroinflammation in stress-related disorders. Moreover, TSPO ligands have been shown to modulate neuroplasticity and to elicit antidepressant and anxiolytic therapeutic effects in animals and humans. As such, TSPO may open new avenues for understanding the pathophysiology of stress-related disorders and for the development of novel treatment options.
Collapse
Affiliation(s)
- Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany.
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Mario Dorostkar
- Center for Neuropathology and Prion Research, Ludwig-Maximilian-University Munich, 81377, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig-Maximilian-University Munich, 81377, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig-Maximilian-University Munich, 81377, Munich, Germany
| | - Jens Schwarzbach
- Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Michael Schumacher
- Research Unit 1195, INSERM and University Paris-Saclay, 94276, Le Kremlin-Bicêtre, France
| | - Inga D Neumann
- Department of Neurobiology and Animal Physiology, University Regensburg, 93040, Regensburg, Germany
| |
Collapse
|
40
|
Das R, Ragagnin G, Sjöstedt J, Johansson M, Haage D, Druzin M, Johansson S, Bäckström T. Medroxyprogesterone acetate positively modulates specific GABA A-receptor subtypes - affecting memory and cognition. Psychoneuroendocrinology 2022; 141:105754. [PMID: 35395561 DOI: 10.1016/j.psyneuen.2022.105754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/14/2022] [Accepted: 03/28/2022] [Indexed: 11/28/2022]
Abstract
Medroxyprogesterone acetate (MPA) is a progestin widely used in humans as hormone replacement therapy and at other indications. Many progestin metabolites, as the progesterone metabolite allopregnanolone, have GABAA-receptor modulatory effects and are known to affect memory, learning, appetite, and mood. In women, 4 years chronic treatment with MPA doubles the frequency of dementia and in rats, MPA causes cognitive impairment related to the GABAergic system. Activation of the membrane bound GABAA receptor results in a chloride ion flux that can be studied by whole-cell patch-clamp electrophysiological recordings. The purpose of this study was to clarify the modulatory effects of MPA and specific MPA metabolites, with structures like known GABAA-receptor modulators, on different GABAA-receptor subtypes. An additional aim was to verify the results as steroid effects on GABA response in single cells taken from rat hypothalamus. HEK-293 cell-lines permanently expressing the recombinant human GABAA-receptor subtype α1β2γ2L or α5β3γ2L or α2β3γ2S were created. The MPA metabolites 3α5α-MPA,3β5α-MPA and 3β5β-MPA were synthesised and purified for electrophysiological patch-clamp measurements with a Dynaflow system. The effects of MPA and tetrahydrodeoxycorticosterone were also studied. None of the studied MPA metabolites affected the responses mediated by α1β2γ2L or α5β3γ2L GABAA receptors. Contrary, MPA clearly acted both as a positive modulator and as a direct activator of the α5β3γ2L and α2β3γ2S GABAA receptors. However, in concentrations up to 10 μM, MPA was inactive at the α1β2γ2L GABAA receptor. In the patch-clamp recordings from dissociated cells of the preoptic area in rats, MPA increased the amplitude of responses to GABA. In addition, MPA alone without added GABA, evoked a current response. In conclusion, MPA acts as a positive modulator of specific GABAA receptor subtypes expressed in HEK cells and at native GABA receptors in single cells from the hypothalamic preoptic area.
Collapse
Affiliation(s)
- Roshni Das
- Department of Integrative medical biology, Umeå University, SE-901 87 Umeå, Sweden; Umecrine AB, Norrlands University Hospital Umeå, Building 6 M 4th floor, Sweden
| | - Gianna Ragagnin
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Jessica Sjöstedt
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Maja Johansson
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden; Umecrine AB, Norrlands University Hospital Umeå, Building 6 M 4th floor, Sweden
| | - David Haage
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden; Department of Nursing Sciences, Mid Sweden University, Sundsvall, Sweden; Umecrine AB, Norrlands University Hospital Umeå, Building 6 M 4th floor, Sweden
| | - Michael Druzin
- Department of Integrative medical biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Staffan Johansson
- Department of Integrative medical biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Torbjörn Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical sciences, Umeå University, SE-901 85 Umeå, Sweden; Umecrine AB, Norrlands University Hospital Umeå, Building 6 M 4th floor, Sweden.
| |
Collapse
|
41
|
Syafni N, Faleschini MT, Garifulina A, Danton O, Gupta MP, Hering S, Hamburger M. Clerodane Diterpenes from Casearia corymbosa as Allosteric GABA A Receptor Modulators. JOURNAL OF NATURAL PRODUCTS 2022; 85:1201-1210. [PMID: 35475609 PMCID: PMC9150179 DOI: 10.1021/acs.jnatprod.1c00840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Indexed: 06/14/2023]
Abstract
An EtOAc extract of Casearia corymbosa leaves led to an allosteric potentiation of the GABA signal in a fluorometric imaging plate reader (FLIPR) assay on Chinese hamster ovary (CHO) cells stably expressing GABAA receptors with an α1β2γ2 subunit composition. The activity was tracked by HPLC-based activity profiling, and four known (2, 3, 4, and 8) and five new clerodane-type diterpenoids (1, 5-7, and 9) were isolated. Compounds 1-8 were obtained from the active time window. The absolute configuration of all compounds was established by ECD. Compounds 3, 7, and 8 exhibited EC50 values of 0.5, 4.6, and 1.4 μM, respectively. To explore possible binding sites at the receptor, the most abundant diterpenoid 8 was tested in combination with diazepam, etazolate, and allopregnanolone. An additive potentiation of the GABA signal was observed with these compounds, while the effect of 8 was not inhibited by flumazenil, a negative allosteric modulator at the benzodiazepine binding site. Finally, the activity was validated in voltage clamp studies on Xenopus laevis oocytes transiently expressing GABAA receptors of the α1β2γ2S and α1β2 subtypes. Compound 8 potentiated GABA-induced currents with both receptor subunit compositions [EC50 (α1β2γ2S) = 43.6 μM; Emax = 809% and EC50 (α1β2) = 57.6 μM; Emax = 534%]. The positive modulation of GABA-induced currents was not inhibited by flumazenil, thereby confirming an allosteric modulation independent of the benzodiazepine binding site.
Collapse
Affiliation(s)
- Nova Syafni
- Pharmaceutical
Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
- Faculty
of Pharmacy and Sumatran Biota Laboratory, Andalas University, Kampus Limau Manis, Padang, West Sumatra 25175, Indonesia
| | - Maria Teresa Faleschini
- Pharmaceutical
Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Aleksandra Garifulina
- Division
of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Pharmaziezentrum, Althanstrasse 14, 1090 Vienna, Austria
| | - Ombeline Danton
- Pharmaceutical
Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Mahabir P. Gupta
- Center
for Pharmacognostic Research on Panamanian Flora, Faculty of Pharmacy, University of Panama, Panama City 0801, Panama
| | - Steffen Hering
- Division
of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Pharmaziezentrum, Althanstrasse 14, 1090 Vienna, Austria
| | - Matthias Hamburger
- Pharmaceutical
Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| |
Collapse
|
42
|
Koniuszewski F, Vogel FD, Bampali K, Fabjan J, Seidel T, Scholze P, Schmiedhofer PB, Langer T, Ernst M. Molecular Mingling: Multimodal Predictions of Ligand Promiscuity in Pentameric Ligand-Gated Ion Channels. Front Mol Biosci 2022; 9:860246. [PMID: 35615739 PMCID: PMC9124788 DOI: 10.3389/fmolb.2022.860246] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/28/2022] [Indexed: 01/23/2023] Open
Abstract
Background: Human pentameric ligand-gated ion channels (pLGICs) comprise nicotinic acetylcholine receptors (nAChRs), 5-hydroxytryptamine type 3 receptors (5-HT3Rs), zinc-activated channels (ZAC), γ-aminobutyric acid type A receptors (GABAARs) and glycine receptors (GlyRs). They are recognized therapeutic targets of some of the most prescribed drugs like general anesthetics, anxiolytics, smoking cessation aids, antiemetics and many more. Currently, approximately 100 experimental structures of pLGICs with ligands bound exist in the protein data bank (PDB). These atomic-level 3D structures enable the generation of a comprehensive binding site inventory for the superfamily and the in silico prediction of binding site properties. Methods: A panel of high throughput in silico methods including pharmacophore screening, conformation analysis and descriptor calculation was applied to a selection of allosteric binding sites for which in vitro screens are lacking. Variant abundance near binding site forming regions and computational docking complement the approach. Results: The structural data reflects known and novel binding sites, some of which may be unique to individual receptors, while others are broadly conserved. The membrane spanning domain, comprising four highly conserved segments, contains ligand interaction sites for which in vitro assays suitable for high throughput screenings are critically lacking. This is also the case for structurally more variable novel sites in the extracellular domain. Our computational results suggest that the phytocannabinoid Δ9-tetrahydrocannabinol (Δ9-THC) can utilize multiple pockets which are likely to exist on most superfamily members. Conclusion: With this study, we explore the potential for polypharmacology among pLGICs. Our data suggest that ligands can display two forms of promiscuity to an extent greater than what has been realized: 1) Ligands can interact with homologous sites in many members of the superfamily, which bears toxicological relevance. 2) Multiple pockets in distinct localizations of individual receptor subtypes share common ligands, which counteracts efforts to develop selective agents. Moreover, conformational states need to be considered for in silico drug screening, as certain binding sites display considerable flexibility. In total, this work contributes to a better understanding of polypharmacology across pLGICs and provides a basis for improved structure guided in silico drug development and drug derisking.
Collapse
Affiliation(s)
- Filip Koniuszewski
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Florian D. Vogel
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Konstantina Bampali
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Jure Fabjan
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Thomas Seidel
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Petra Scholze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Philip B. Schmiedhofer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Thierry Langer
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Margot Ernst
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| |
Collapse
|
43
|
Wang L, Covey DF, Akk G, Evers AS. Neurosteroid Modulation of GABA A Receptor Function by Independent Action at Multiple Specific Binding Sites. Curr Neuropharmacol 2022; 20:886-890. [PMID: 34856904 PMCID: PMC9881108 DOI: 10.2174/1570159x19666211202150041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/28/2021] [Accepted: 11/28/2021] [Indexed: 11/22/2022] Open
Abstract
Neurosteroids are endogenous modulators of GABAA receptors that mediate anxiety, pain, mood and arousal. The 3-hydroxyl epimers, allopregnanolone (3α-OH) and epiallopregnanolone (3β-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 3β-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.
Collapse
Affiliation(s)
- Lei Wang
- Department of Anesthesiology (LW, DFC, GA, ASE),,Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Douglas F. Covey
- Department of Anesthesiology (LW, DFC, GA, ASE),,Department of Developmental Biology (Pharmacology);,Department of Psychiatry (DFC) and
| | - Gustav Akk
- Department of Anesthesiology (LW, DFC, GA, ASE),,The Taylor Institute for Innovative Psychiatric Research (DFC, GA, ASE), Washington University School of Medicine, St. Louis, MO63110; U.S.A
| | - Alex S. Evers
- Department of Anesthesiology (LW, DFC, GA, ASE),,Department of Developmental Biology (Pharmacology);,The Taylor Institute for Innovative Psychiatric Research (DFC, GA, ASE), Washington University School of Medicine, St. Louis, MO63110; U.S.A,Address correspondence to this author at the Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Ave, Box # 8054, St. Louis, MO 63110, USA; Tel: +1 314-362-8557; E-mail:
| |
Collapse
|
44
|
Arias HR, Borghese CM, Germann AL, Pierce SR, Bonardi A, Nocentini A, Gratteri P, Thodati TM, Lim NJ, Adron Harris R, Akk G. (+)-Catharanthine potentiates the GABA A receptor by binding to a transmembrane site at the β(+)/α(-) interface near the TM2-TM3 loop. Biochem Pharmacol 2022; 199:114993. [PMID: 35304861 PMCID: PMC9178925 DOI: 10.1016/j.bcp.2022.114993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/17/2022]
Abstract
(+)-Catharanthine, a coronaridine congener, potentiates the γ-aminobutyric acid type A receptor (GABAAR) and induces sedation through a non-benzodiazepine mechanism, but the specific site of action and intrinsic mechanism have not beendefined. Here, we describe GABAAR subtype selectivity and location of the putative binding site for (+)-catharanthine using electrophysiological, site-directed mutagenesis, functional competition, and molecular docking experiments. Electrophysiological and in silico experiments showed that (+)-catharanthine potentiates the responses to low, subsaturating GABA at β2/3-containing GABAARs 2.4-3.5 times more efficaciously than at β1-containing GABAARs. The activity of (+)-catharanthine is reduced by the β2(N265S) mutation that decreases GABAAR potentiation by loreclezole, but not by the β3(M286C) or α1(Q241L) mutations that reduce receptor potentiation by R(+)-etomidate or neurosteroids, respectively. Competitive functional experiments indicated that the binding site for (+)-catharanthine overlaps that for loreclezole, but not those for R(+)-etomidate or potentiating neurosteroids. Molecular docking experiments suggested that (+)-catharanthine binds at the β(+)/α(-) intersubunit interface near the TM2-TM3 loop, where it forms H-bonds with β2-D282 (TM3), β2-K279 (TM2-TM3 loop), and β2-N265 and β2-R269 (TM2). Site-directed mutagenesis experiments supported the in silico results, demonstrating that the K279A and D282A substitutions, that lead to a loss of H-bonding ability of the mutated residue, and the N265S mutation, impair the gating efficacy of (+)-catharanthine. We infer that (+)-catharanthine potentiates the GABAAR through several H-bond interactions with a binding site located in the β(+)/α(-) interface in the transmembrane domain, near the TM2-TM3 loop, where it overlaps with loreclezole binding site.
Collapse
Affiliation(s)
- Hugo R Arias
- Department of Pharmacology and Physiology, Oklahoma State University College of Osteopathic Medicine, Tahlequah, OK, USA.
| | - Cecilia M Borghese
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, USA.
| | - Allison L Germann
- Department of Anesthesiology, the Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA.
| | - Spencer R Pierce
- Department of Anesthesiology, the Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA.
| | - Alessandro Bonardi
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences, Laboratory of Molecular Modeling Cheminformatics & QSAR, University of Florence, Florence, Italy.
| | - Alessio Nocentini
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences, Laboratory of Molecular Modeling Cheminformatics & QSAR, University of Florence, Florence, Italy.
| | - Paola Gratteri
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmaceutical and Nutraceutical Sciences, Laboratory of Molecular Modeling Cheminformatics & QSAR, University of Florence, Florence, Italy.
| | - Thanvi M Thodati
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, USA.
| | - Natalie J Lim
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, USA.
| | - R Adron Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, USA.
| | - Gustav Akk
- Department of Anesthesiology, the Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
45
|
Cheng WWL, Arcario MJ, Petroff JT. Druggable Lipid Binding Sites in Pentameric Ligand-Gated Ion Channels and Transient Receptor Potential Channels. Front Physiol 2022; 12:798102. [PMID: 35069257 PMCID: PMC8777383 DOI: 10.3389/fphys.2021.798102] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
Lipids modulate the function of many ion channels, possibly through direct lipid-protein interactions. The recent outpouring of ion channel structures by cryo-EM has revealed many lipid binding sites. Whether these sites mediate lipid modulation of ion channel function is not firmly established in most cases. However, it is intriguing that many of these lipid binding sites are also known sites for other allosteric modulators or drugs, supporting the notion that lipids act as endogenous allosteric modulators through these sites. Here, we review such lipid-drug binding sites, focusing on pentameric ligand-gated ion channels and transient receptor potential channels. Notable examples include sites for phospholipids and sterols that are shared by anesthetics and vanilloids. We discuss some implications of lipid binding at these sites including the possibility that lipids can alter drug potency or that understanding protein-lipid interactions can guide drug design. Structures are only the first step toward understanding the mechanism of lipid modulation at these sites. Looking forward, we identify knowledge gaps in the field and approaches to address them. These include defining the effects of lipids on channel function in reconstituted systems using asymmetric membranes and measuring lipid binding affinities at specific sites using native mass spectrometry, fluorescence binding assays, and computational approaches.
Collapse
Affiliation(s)
- Wayland W L Cheng
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
| | - Mark J Arcario
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
| | - John T Petroff
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
46
|
Andersen ME, Guerrero T. Assessing Modes of Action, Measures of Tissue Dose and Human Relevance of Rodent Toxicity Endpoints with Octamethylcyclotetrasiloxane (D4). Toxicol Lett 2022; 357:57-72. [PMID: 34995712 DOI: 10.1016/j.toxlet.2021.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/07/2021] [Accepted: 12/30/2021] [Indexed: 10/19/2022]
Abstract
Octamethylcyclotetrasiloxane (D4), a highly lipophilic, volatile compound with low water solubility, is metabolized to lower molecular weight, linear silanols. Toxicity has been documented in several tissues in animals following mixed vapor/aerosol exposures by inhalation at near saturating vapor concentrations or with gavage dosing in vegetable oil vehicles. These results, together with more mechanism-based studies and detailed pharmacokinetic information, were used to assess likely modes of action (MOAs) and the tissue dose measures of D4 and metabolites that would serve as key events leading to these biological responses. This MOA analysis indicates that pulmonary effects arise from direct epithelial contact with mixed vapor/aerosol atmospheres of D4; liver hypertrophy and hepatocyte proliferation arise from adaptive, rodent-specific actions of D4 with nuclear receptor signaling pathways; and, nephropathy results from silanol metabolites binding with alpha-2μ globulin (a rat specific protein). At this time, the MOAs of other liver effects - pigment accumulation and bile duct hyperplasia (BDH) preferentially observed in Sprague-Dawley (SD) rats- are not known. Hypothalamic actions of D4 delaying the rat mid-cycle gonadotrophin releasing hormone (GnRH) surge that result in reproductive effects and subsequent vaginal/uterine/ovarian tissue responses, including small increases in incidence of benign endometrial adenomas, are associated with prolongation of endogenous estrogen exposures due to delays in ovulation. Human reproduction is not controlled by a mid-cycle GnRH surge. Since the rodent-specific reproductive and the vaginal/uterine/ovarian tissue responses are not relevant for risk assessments in human populations, D4 should neither be classified as a CMR (i.e., carcinogenic, mutagenic, or toxic for reproduction) substance nor be regarded as an endocrine disruptor. Bile duct hyperplasia (BDH) and pigment accumulation in liver seen in SD rats are endpoints that could serve to define a Benchmark Dose or No-Observed-Effect-Level (NOEL) for D4.
Collapse
Affiliation(s)
- Melvin E Andersen
- Andersen ToxConsulting LLC, 424 Granite Lake Ct., Denver, NC 28037, United States.
| | - Tracy Guerrero
- American Chemistry Council Director, Silicones, Environmental, Health, and Safety Center, 700 2nd Street, NE, Washington, DC, 20002, United States.
| |
Collapse
|
47
|
Littlejohn EL, Boychuk CR. Protein Kinase C-Dependent Effects of Neurosteroids on Synaptic GABA A Receptor Inhibition Require the δ-Subunit. Front Physiol 2021; 12:742838. [PMID: 34759836 PMCID: PMC8573421 DOI: 10.3389/fphys.2021.742838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/25/2021] [Indexed: 11/13/2022] Open
Abstract
The dorsal motor nucleus of the vagus (DMV) contains preganglionic motor neurons important for interpreting sensory input from the periphery, integrating that information, and coding the appropriate parasympathetic (vagal) output to target organs. Despite the critical role of hormonal regulation of vagal motor output, few studies examine the role of neurosteroids in the regulation of the DMV. Of the few examinations, no studies have investigated the potential impact of allopregnanolone (Allo), a neuroactive progesterone-derivative, in the regulation of neurotransmission on the DMV. Since DMV neuronal function is tightly regulated by GABAA receptor activity and Allo is an endogenous GABAA receptor ligand, the present study used in vitro whole cell patch clamp to investigate whether Allo alters GABAergic neurotransmission to DMV neurons. Although Allo did not influence GABAergic neurotransmission during initial application (5-20 min), a TTX-insensitive prolongment of decay time and increase in frequency of GABAergic currents was established after Allo was removed from the bath for at least 30 min (LtAllo). Inhibition of protein kinase C (PKC) abolished these effects, suggesting that PKC is largely required to mediate Allo-induced inhibition of the DMV. Using mice that lack the δ-subunit of the GABAA receptor, we further confirmed that PKC-dependent activity of LtAllo required this subunit. Allo also potentiated GABAA receptor activity after a repeated application of δ-subunit agonist, suggesting that the presence of Allo encodes stronger δ-subunit-mediated inhibition over time. Using current clamp recording, we demonstrated that LtAllo-induced inhibition is sufficient to decrease action potential firing and excitability within DMV neurons. We conclude that the effects of LtAllo on GABAergic inhibition are dependent on δ-subunit and PKC activation. Taken together, DMV neurons can undergo long lasting Allo-dependent GABAA receptor plasticity.
Collapse
Affiliation(s)
| | - Carie R. Boychuk
- Department of Cellular and Integrative Physiology, Long College of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
48
|
Malyshev AV, Sukhanova IA, Zlobin AS, Gedzun VR, Pavshintsev VV, Vasileva EV, Zalevsky AO, Doronin II, Mitkin NA, Golovin AV, Lovat ML, Kovalev GI, Zolotarev YA, Kuchumov AR, Babkin GA, Luscher B. In silico Screening and Behavioral Validation of a Novel Peptide, LCGA-17, With Anxiolytic-Like Properties. Front Neurosci 2021; 15:705590. [PMID: 34421525 PMCID: PMC8372404 DOI: 10.3389/fnins.2021.705590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/13/2021] [Indexed: 12/01/2022] Open
Abstract
The aim of the study was to develop better anxiolytics and antidepressants. We focused on GABAA receptors and the α2δ auxiliary subunit of V-gated Ca2+ channels as putative targets because they are established as mediators of efficacious anxiolytics, antidepressants, and anticonvulsants. We further focused on short peptides as candidate ligands because of their high safety and tolerability profiles. We employed a structural bioinformatics approach to develop novel tetrapeptides with predicted affinity to GABAA receptors and α2δ. In silico docking studies of one of these peptides, LCGA-17, showed a high binding score for both GABAA receptors and α2δ, combined with anxiolytic-like properties in a Danio rerio behavioral screen. LCGA-17 showed anxiolytic-like effects in the novel tank test, the light–dark box, and the social preference test, with efficacy comparable to fluvoxamine and diazepam. In binding assays using rat brain membranes, [3H]-LCGA-17 was competed more effectively by gabapentinoid ligands of α2δ than ligands of GABAA receptors, suggesting that α2δ represents a likely target for LCGA-17. [3H]-LCGA-17 binding to brain lysates was unaffected by competition with ligands for GABAB, glutamate, dopamine, serotonin, and other receptors, suggesting specific interaction with α2δ. Dose-finding studies in mice using acute administration of LCGA-17 (i.p.) demonstrated anxiolytic-like effects in the open field test, elevated plus maze, and marble burying tests, as well as antidepressant-like properties in the forced swim test. The anxiolytic effects were effectively blocked by bicuculline. Therefore, LCGA-17 is a novel candidate anxiolytic and antidepressant that may act through α2δ, with possible synergism by GABAA receptors.
Collapse
Affiliation(s)
| | | | - Alexander S Zlobin
- Lactocore, Inc., Plymouth, MI, United States.,Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia.,Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Sirius University of Science and Technology, Sochi, Russia
| | - Vasilina R Gedzun
- Lactocore, Inc., Plymouth, MI, United States.,Department of Human and Animal Physiology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Ekaterina V Vasileva
- Federal State Budgetary Institution, Research Zakusov Institute of Pharmacology, Moscow, Russia
| | - Arthur O Zalevsky
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | | | - Andrey V Golovin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia.,Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Sirius University of Science and Technology, Sochi, Russia
| | - Maxim L Lovat
- Department of Human and Animal Physiology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Georgy I Kovalev
- Federal State Budgetary Institution, Research Zakusov Institute of Pharmacology, Moscow, Russia
| | - Yurii A Zolotarev
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Moscow, Russia
| | | | | | - Bernhard Luscher
- Department of Biology, The Pennsylvania State University, University Park, PA, United States.,Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
49
|
Krishnan K, Qian M, Feltes M, Chen ZW, Gale S, Wang L, Sugasawa Y, Reichert DE, Schaffer JE, Ory DS, Evers AS, Covey DF. Validation of Trifluoromethylphenyl Diazirine Cholesterol Analogues As Cholesterol Mimetics and Photolabeling Reagents. ACS Chem Biol 2021; 16:1493-1507. [PMID: 34355883 DOI: 10.1021/acschembio.1c00364] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aliphatic diazirine analogues of cholesterol have been used previously to elaborate the cholesterol proteome and identify cholesterol binding sites on proteins. Cholesterol analogues containing the trifluoromethylphenyl diazirine (TPD) group have not been reported. Both classes of diazirines have been prepared for neurosteroid photolabeling studies and their combined use provided information that was not obtainable with either diazirine class alone. Hence, we prepared cholesterol TPD analogues and used them along with previously reported aliphatic diazirine analogues as photoaffinity labeling reagents to obtain additional information on the cholesterol binding sites of the pentameric Gloeobacter ligand-gated ion channel (GLIC). We first validated the TPD analogues as cholesterol substitutes and compared their actions with those of previously reported aliphatic diazirines in cell culture assays. All the probes bound to the same cholesterol binding site on GLIC but with differences in photolabeling efficiencies and residues identified. Photolabeling of mammalian (HEK) cell membranes demonstrated differences in the pattern of proteins labeled by the two classes of probes. Collectively, these date indicate that cholesterol photoaffinity labeling reagents containing an aliphatic diazirine or TPD group provide complementary information and will both be useful tools in future studies of cholesterol biology.
Collapse
|
50
|
Luo Y, Kusay AS, Jiang T, Chebib M, Balle T. Delta-containing GABA A receptors in pain management: Promising targets for novel analgesics. Neuropharmacology 2021; 195:108675. [PMID: 34153311 DOI: 10.1016/j.neuropharm.2021.108675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/01/2021] [Accepted: 06/11/2021] [Indexed: 12/26/2022]
Abstract
Communication between nerve cells depends on the balance between excitatory and inhibitory circuits. GABA, the major inhibitory neurotransmitter, regulates this balance and insufficient GABAergic activity is associated with numerous neuropathological disorders including pain. Of the various GABAA receptor subtypes, the δ-containing receptors are particularly interesting drug targets in management of chronic pain. These receptors are pentameric ligand-gated ion channels composed of α, β and δ subunits and can be activated by ambient levels of GABA to generate tonic conductance. However, only a few ligands preferentially targeting δ-containing GABAA receptors have so far been identified, limiting both pharmacological understanding and drug-discovery efforts, and more importantly, understanding of how they affect pain pathways. Here, we systemically review and discuss the known drugs and ligands with analgesic potential targeting δ-containing GABAA receptors and further integrate the biochemical nature of the receptors with clinical perspectives in pain that might generate interest among researchers and clinical physicians to encourage analgesic discovery efforts leading to more efficient therapies.
Collapse
Affiliation(s)
- Yujia Luo
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia; Brain and Mind Centre, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Ali Saad Kusay
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia; Brain and Mind Centre, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Tian Jiang
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia; Brain and Mind Centre, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Mary Chebib
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia; Brain and Mind Centre, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Thomas Balle
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia; Brain and Mind Centre, The University of Sydney, Camperdown, NSW, 2050, Australia.
| |
Collapse
|