1
|
Bukanova JV, Kondratenko RV, Solntseva EI. Interaction Between Allopregnanolone and Amiloride Binding Sites on the GABA A Receptor. Cell Biochem Biophys 2025; 83:2453-2459. [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] [MESH Headings] [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
|
Zorumski CF, Covey DF, Izumi Y, Evers AS, Maguire JL, Mennerick SJ. New directions in neurosteroid therapeutics in neuropsychiatry. Neurosci Biobehav Rev 2025; 172:106119. [PMID: 40127877 DOI: 10.1016/j.neubiorev.2025.106119] [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: 12/02/2024] [Revised: 02/22/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
In recent years three neuroactive steroids (NAS), brexanolone (allopregnanolone, AlloP), ganaxolone and zuranolone, have been approved for the treatment of neuropsychiatric illnesses including postpartum depression and seizures in a neurodevelopmental syndrome. The approved agents are pregnane steroids and strong positive allosteric modulators (PAMs) of gamma-aminobutyric acid type A receptors (GABAARs). Broad effects on GABAARs play important roles in therapeutic benefits. However, these NAS also have actions on non-GABAR targets that could be important for clinical outcomes. Thus, understanding the broader effects of NAS is potentially important for expanding the therapeutic landscape of these important modulators. The approved NAS as well as other structurally distinct NAS and oxysterols have effects on non-GABAAR receptors and ion channels, along with intracellular actions that could have therapeutic importance, including modulation of cellular stress mechanisms, neuroinflammation, mitochondrial function and autophagy, among others. In this review, we explore GABAergic and other cellular effects of pregnane steroids including novel molecules that have potential therapeutic importance. This work discusses the complex chemical nature of NAS and what is being learned at cellular, molecular, synaptic and brain network levels about key sites of action including GABAARs and other targets.
Collapse
Affiliation(s)
- Charles F Zorumski
- Departments of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA.
| | - 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; 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; 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; 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
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
Kumar A, Qian M, Xu Y, Benz A, Covey DF, Zorumski CF, Mennerick S. Multifaceted Actions of Neurosteroids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634297. [PMID: 39896603 PMCID: PMC11785204 DOI: 10.1101/2025.01.22.634297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Background and purpose Neurosteroids modulate neuronal function and are promising therapeutic agents for neuropsychiatric disorders. Neurosteroid analogues are approved for treating postpartum depression and are of interest in other disorders. GABA-A receptors are well characterized targets of natural neurosteroids, but other biological pathways are likely relevant to therapeutic mechanisms and/or to off-target effects. We performed hypothesis-generating in silico analyses and broad in vitro biological screens to assess the range of actions of neurosteroids analogues of varying structural attributes. Key Results We employed in silico molecular similarity analysis and network pharmacology to elucidate likely targets. This analysis confirmed likely targets beyond GABA-A receptors. We then functionally screened 19 distinct neurosteroid structures across 78 targets representing interconnected signaling pathways, complemented with a limited screen of kinase activation. Results revealed unanticipated modulation of targets by neurosteroids with some structural selectivity. Many compounds-initiated androgen receptor translocation with little or no enantioselectivity. Modulation of multiple G-protein receptors was also unexpected. Conclusions and implications Neurosteroids are ascendant treatments in neuropsychiatry, but their full spectrum of actions remains unclear. This virtual and biological screening discovery approach opens new vistas for exploring mechanism of neurosteroids analogues. The multifaceted approach provides an unbiased, holistic exploration of the potential effects of neurosteroids across various molecular targets and provides a platform for future validation studies to aid drug discovery.
Collapse
|
4
|
Michałowski MA, Kłopotowski K, Wiera G, Czyżewska MM, Mozrzymas JW. Molecular mechanisms of the GABA type A receptor function. Q Rev Biophys 2025; 58:e3. [PMID: 39806800 DOI: 10.1017/s0033583524000179] [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] [Indexed: 01/16/2025]
Abstract
The GABA type A receptor (GABAAR) belongs to the family of pentameric ligand-gated ion channels and plays a key role in inhibition in adult mammalian brains. Dysfunction of this macromolecule may lead to epilepsy, anxiety disorders, autism, depression, and schizophrenia. GABAAR is also a target for multiple physiologically and clinically relevant modulators, such as benzodiazepines (BDZs), general anesthetics, and neurosteroids. The first GABAAR structure appeared in 2014, but the past years have brought a particularly abundant surge in structural data for these receptors with various ligands and modulators. Although the open conformation remains elusive, this novel information has pushed the structure-function studies to an unprecedented level. Electrophysiology, mutagenesis, photolabeling, and in silico simulations, guided by novel structural information, shed new light on the molecular mechanisms of receptor functioning. The main goal of this review is to present the current knowledge of GABAAR functional and structural properties. The review begins with an outline of the functional and structural studies of GABAAR, accompanied by some methodological considerations, especially biophysical methods, enabling the reader to follow how major breakthroughs in characterizing GABAAR features have been achieved. The main section provides a comprehensive analysis of the functional significance of specific structural elements in GABAARs. We additionally summarize the current knowledge on the binding sites for major GABAAR modulators, referring to the molecular underpinnings of their action. The final chapter of the review moves beyond examining GABAAR as an isolated macromolecule and describes the interactions of the receptor with other proteins in a broader context of inhibitory plasticity. In the final section, we propose a general conclusion that agonist binding to the orthosteric binding sites appears to rely on local interactions, whereas conformational transitions of bound macromolecule (gating) and allosteric modulation seem to reflect more global phenomena involving vast portions of the macromolecule.
Collapse
Affiliation(s)
- Michał A Michałowski
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Karol Kłopotowski
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Grzegorz Wiera
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Marta M Czyżewska
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Jerzy W Mozrzymas
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
5
|
Akkerman V, Reinholdt P, Schnoor-Madsen R, Lauritsen L, Bader J, Qian M, Xu Y, Akk G, Scheidt HA, Müller P, Covey DF, Evers AS, Kongsted J, Wüstner D. Stereospecific Properties and Intracellular Transport of Novel Intrinsically Fluorescent Neurosteroids. ACS Chem Neurosci 2024; 15:4322-4336. [PMID: 39574303 PMCID: PMC11892034 DOI: 10.1021/acschemneuro.4c00571] [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] [Indexed: 03/12/2025] Open
Abstract
Allopregnanolone (AlloP) is an example of neuroactive steroids (NAS), which is a potent allosteric activator of the γ-aminobutyric acid A (GABAA) receptor. The mechanisms underlying the biological activity of AlloP and other NAS are only partially understood. Here, we present intrinsically fluorescent analogs of AlloP (MQ-323) and its 3β-epimer, epi-allopregnanolone (E-AlloP) (YX-11), and show, by a combination of spectroscopic and computational studies, that these analogs mimic the membrane properties of AlloP and E-AlloP very well. We found stereospecific differences in the orientation and dynamics of the NAS as well as in their impact on membrane permeability. However, all NAS are unable to condense the lipid bilayer, in stark contrast to cholesterol. Using Förster resonance energy transfer (FRET) and electrophysiological measurements, we show that MQ-323 but not YX-11 binds at the intersubunit site of the ELICα1GABAA receptor and potentiates GABA-induced receptor currents. In aqueous solvents, YX-11 forms aggregates at much lower concentrations than MQ-323, and loading both analogs onto cyclodextrin allows for their uptake by human astrocytes, where they become enriched in lipid droplets (LDs), as shown by quantitative fluorescence microscopy. Trafficking of the NAS analogs is stereospecific, as uptake and lipid droplet targeting is more pronounced for YX-11 compared to MQ-323. In summary, we present novel minimally modified analogs of AlloP and E-AlloP, which enable us to reveal stereospecific membrane properties, allosteric receptor activation, and intracellular transport of these neurosteroids. Our fluorescence design strategy will be very useful for the analysis of other NAS in the future.
Collapse
Affiliation(s)
- Vibeke Akkerman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Peter Reinholdt
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Rasmus Schnoor-Madsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Line Lauritsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Jad Bader
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Minxing Qian
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Yuanjiang Xu
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Gustav Akk
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Holger A Scheidt
- Institute for Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| | - Peter Müller
- Department of Biology, Humboldt University Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| | - Douglas F Covey
- Department of Developmental Biology and Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Alex S Evers
- Department of Developmental Biology and Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Jacob Kongsted
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
Gonda X, Tarazi FI, Dome P. The emergence of antidepressant drugs targeting GABA A receptors: A concise review. Biochem Pharmacol 2024; 228:116481. [PMID: 39147329 DOI: 10.1016/j.bcp.2024.116481] [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: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
Depression is among the most common psychiatric illnesses, which imposes a major socioeconomic burden on patients, caregivers, and the public health system. Treatment with classical antidepressants (e.g. tricyclic antidepressants and selective serotonine reuptake inhibitors), which primarily affect monoaminergic systems has several limitations, such as delayed onset of action and moderate efficacy in a relatively large proportion of depressed patients. Furthermore, depression is highly heterogeneus, and its different subtypes, including post-partum depression, involve distinct neurobiology, warranting a differential approach to pharmacotherapy. Given these shortcomings, the need for novel antidepressants that are superior in efficacy and faster in onset of action is fully justified. The development and market introduction of rapid-acting antidepressants has accelerated in recent years. Some of these new antidepressants act through the GABAergic system. In this review, we discuss the discovery, efficacy, and limitations of treatment with classic antidepressants. We provide a detailed discussion of GABAergic neurotransmission, with a special focus on GABAA receptors, and possible explanations for the mood-enhancing effects of GABAergic medications (in particular neurosteroids acting at GABAA receptors), and, ultimately, we present the most promising molecules belonging to this family which are currently used in clinical practice or are in late phases of clinical development.
Collapse
Affiliation(s)
- Xenia Gonda
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary.
| | - Frank I Tarazi
- Department of Psychiatry and Neurology, Harvard Medical School and McLean Hospital, Boston, MA, USA
| | - Peter Dome
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Nyiro Gyula National Institute of Psychiatry and Addictology, Budapest, Hungary
| |
Collapse
|
10
|
Borghese CM, Goldschen-Ohm MP. State-dependent energetics of GABA A receptor modulators. Biophys J 2024; 123:1903-1906. [PMID: 38303510 PMCID: PMC11309981 DOI: 10.1016/j.bpj.2024.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024] Open
Affiliation(s)
- Cecilia M Borghese
- University of Texas at Austin, Department of Neuroscience, Austin, Texas
| | | |
Collapse
|
11
|
Pierce SR, Xu SQ, Germann AL, Steinbach JH, Akk G. Potentiation of the GABA AR reveals variable energetic contributions by etiocholanolone and propofol. Biophys J 2024; 123:1954-1967. [PMID: 37752702 PMCID: PMC11442032 DOI: 10.1016/j.bpj.2023.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 09/28/2023] Open
Abstract
The properties of a potentiator are typically evaluated by measuring its ability to enhance the magnitude of the control response. Analysis of the ability of drugs to potentiate responses from receptor channels takes place in the context of particular models to extract parameters for functional effects. In the often-used coagonist model, the agonist generating control activity and the potentiator enhancing the control activity make additive energetic contributions to stabilize the active state of the receptor. The energetic contributions are fixed and, once known, enable calculation of predicted receptor behavior at any concentration combination of agonist and potentiator. Here, we have examined the applicability of the coagonist model by measuring the relationship between the magnitude of receptor potentiation and the level of background activity. Ternary αβγ GABAA receptors were activated by GABA or the allosteric agonist propofol, or by a gain-of-function mutation, and etiocholanolone- or propofol-mediated potentiation of peak responses was measured. We show that the free energy change contributed by the modulators etiocholanolone or propofol is reduced at higher levels of control activity, thereby being in disagreement with basic principles of the coagonist model. Possible mechanisms underlying this discrepancy are discussed.
Collapse
Affiliation(s)
- Spencer R Pierce
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Sophia Q Xu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Allison L Germann
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Joe Henry Steinbach
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri
| | - Gustav Akk
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
Chen MJ, Chou CH, Hsiao TH, Wu TY, Li CY, Chen YL, Chao KH, Lee TH, Gicana RG, Shih CJ, Brandon-Mong GJ, Lai YL, Li PT, Tseng YL, Wang PH, Chiang YR. Clostridium innocuum, an opportunistic gut pathogen, inactivates host gut progesterone and arrests ovarian follicular development. Gut Microbes 2024; 16:2424911. [PMID: 39508647 PMCID: PMC11545266 DOI: 10.1080/19490976.2024.2424911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/04/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
\Levels of progesterone, an endogenous female hormone, increase after ovulation; progesterone is crucial in the luteal phase to maintain successful pregnancy and prevent early miscarriage. Both endogenous and exogenous progesterone are recycled between the liver and gut; thus, the gut microbiota regulate host progesterone levels by inhibiting enterohepatic progesterone circulation. Our data indicated Clostridium innocuum as a major species involved in gut progesterone metabolism in women with infertility. C. innocuum converts progesterone into the neurosteroid epipregnanolone (with negligible progestogenic activity). We purified and characterized the corresponding enzyme, namely NADPH-dependent 5β-dihydroprogesterone reductase, which is highly oxygen sensitive and whose corresponding genes are prevalent in C. innocuum. Moreover, C. innocuum-administered female C57BL/6 mice (aged 7 weeks) exhibited decreased plasma progesterone levels (~35%). Clostridium-specific antibiotics (metronidazole) restored low plasma progesterone levels in these mice. Furthermore, prolonged C. innocuum administration (12 weeks) arrested ovarian follicular development in female mice. Cytological and histological analyses indicated that C. innocuum may cause luteal phase insufficiency and affect menstrual regularity. Our findings suggest C. innocuum as a causal factor of progesterone resistance in women taking progesterone.
Collapse
Affiliation(s)
- Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
- Livia Shan-Yu Wan Chair Professor of Obstetrics and Gynecology, National Taiwan University, Taipei, Taiwan
| | - Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsun-Hsien Hsiao
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Tien-Yu Wu
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Chi-Ying Li
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Lung Chen
- Department of Microbiology, Soochow University, Taipei, Taiwan
- Department of Agricultural Chemistry, National Taiwan University, Taipei, Taiwan
| | - Kuang-Han Chao
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzong-Huei Lee
- Institute of Fisheries Science, National Taiwan University, Taipei, Taiwan
| | | | - Chao-Jen Shih
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | | | - Yi-Li Lai
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Po-Ting Li
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Lin Tseng
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Hsiang Wang
- Graduate Institute of Environmental Engineering, National Central University, Taoyuan, Taiwan
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Yin-Ru Chiang
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
- Department of Agricultural Chemistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
17
|
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
|
18
|
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: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
19
|
Solntseva EI, Bukanova JV, Kondratenko R, Kudova E. Corticosteroids as Selective and Effective Modulators of Glycine Receptors. ACS Chem Neurosci 2023; 14:3132-3142. [PMID: 37584305 PMCID: PMC10485894 DOI: 10.1021/acschemneuro.3c00287] [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: 05/02/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023] Open
Abstract
The mechanism of the negative impact of corticosteroids on the induction and progress of mental illness remains unclear. In this work, we studied the effects of corticosteroids on the activity of neuronal glycine receptors (GlyR) and GABA-A receptors (GABAAR) by measuring the chloride current induced by the application of GABA (2 or 5 μM) to isolated cerebellar Purkinje cells (IGABA) and by the application of glycine (100 μM) to pyramidal neurons of the rat hippocampus (IGly). It was found that corticosterone, 5α-dihydrodeoxycorticosterone, allotetrahydrocorticosterone, cortisol, and 17α,21-dihydroxypregnenolone were able to accelerate the desensitization of the IGly at physiological concentrations (IC50 values varying from 0.39 to 0.72 μM). Next, cortisone, 11-deoxycortisol, 11-deoxycorticosterone, 5β-dihydrodeoxycorticosterone, and tetrahydrocorticosterone accelerated the desensitization of IGly with IC50 values varying from 10.3 to 15.2 μM. Allotetrahydrocorticosterone and tetrahydrocorticosterone potentiated the IGABA albeit with high EC50 values (18-23 μM). The rest of the steroids had no effect on IGABA in the range of concentrations of 1-100 μM. Finally, our study has suggested a structural relationship of the 3β-hydroxyl group/3-oxo group with the selective modulatory activity on GlyRs in contrast to the 3α-hydroxyl group that is pivotal for GABAARs. In summary, our results suggest that increased GlyR desensitization by corticosteroids may contribute to brain dysfunction under chronic stress and identify corticosteroids for further development as selective modulators of GlyRs.
Collapse
Affiliation(s)
- Elena I. Solntseva
- Functional
Synaptology Laboratory, Brain Research Institute,
Research Center of Neurology, Moscow 125367, Russia
| | - Julia V. Bukanova
- Functional
Synaptology Laboratory, Brain Research Institute,
Research Center of Neurology, Moscow 125367, Russia
| | - Rodion Kondratenko
- Functional
Synaptology Laboratory, Brain Research Institute,
Research Center of Neurology, Moscow 125367, Russia
| | - Eva Kudova
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 166 10, Czech Republic
| |
Collapse
|
20
|
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
|
21
|
Sikes-Keilp C, Rubinow DR. GABA-ergic Modulators: New Therapeutic Approaches to Premenstrual Dysphoric Disorder. CNS Drugs 2023; 37:679-693. [PMID: 37542704 DOI: 10.1007/s40263-023-01030-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/20/2023] [Indexed: 08/07/2023]
Abstract
Premenstrual dysphoric disorder (PMDD) is characterized by the predictable onset of mood and physical symptoms secondary to gonadal steroid fluctuation during the luteal phase of the menstrual cycle. Although menstrual-related affective dysfunction is responsible for considerable functional impairment and reduction in quality of life worldwide, currently approved treatments for PMDD are suboptimal in their effectiveness. Research over the past two decades has suggested that the interaction between allopregnanolone, a neurosteroid derivative of progesterone, and the gamma-aminobutyric acid (GABA) system represents an important relationship underlying symptom genesis in reproductive-related mood disorders, including PMDD. The objective of this narrative review is to discuss the plausible link between changes in GABAergic transmission secondary to the fluctuation of allopregnanolone during the luteal phase and mood impairment in susceptible individuals. As part of this discussion, we explore promising findings from early clinical trials of several compounds that stabilize allopregnanolone signaling during the luteal phase, including dutasteride, a 5-alpha reductase inhibitor; isoallopregnanolone, a GABA-A modulating steroid antagonist; and ulipristal acetate, a selective progesterone receptor modulator. We then reflect on the implications of these therapeutic advances, including how they may promote our knowledge of affective regulation more generally. We conclude that these and other studies of PMDD may yield critical insight into the etiopathogenesis of affective disorders, considering that (1) symptoms in PMDD have a predictable onset and offset, allowing for examination of affective state kinetics, and (2) GABAergic interventions in PMDD can be used to better understand the relationship between mood states, network regulation, and the balance between excitatory and inhibitory signaling in the brain.
Collapse
Affiliation(s)
- Christopher Sikes-Keilp
- Department of Psychiatry, University of North Carolina Hospitals, 101 Manning Drive, Chapel Hill, NC, 27514, USA.
| | - David R Rubinow
- Department of Psychiatry, University of North Carolina Hospitals, 101 Manning Drive, Chapel Hill, NC, 27514, USA
| |
Collapse
|
22
|
Altê GA, Rodrigues ALS. Exploring the Molecular Targets for the Antidepressant and Antisuicidal Effects of Ketamine Enantiomers by Using Network Pharmacology and Molecular Docking. Pharmaceuticals (Basel) 2023; 16:1013. [PMID: 37513925 PMCID: PMC10383558 DOI: 10.3390/ph16071013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Ketamine, a racemic mixture of esketamine (S-ketamine) and arketamine (R-ketamine), has received particular attention for its rapid antidepressant and antisuicidal effects. NMDA receptor inhibition has been indicated as one of the main mechanisms of action of the racemic mixture, but other pharmacological targets have also been proposed. This study aimed to explore the possible multiple targets of ketamine enantiomers related to their antidepressant and antisuicidal effects. To this end, targets were predicted using Swiss Target Prediction software for each ketamine enantiomer. Targets related to depression and suicide were collected by the Gene Cards database. The intersections of targets were analyzed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Network pharmacology analysis was performed using Gene Mania and Cytoscape software. Molecular docking was used to predict the main targets of the network. The results indicated that esketamine and arketamine share some biological targets, particularly NMDA receptor and phosphodiesterases 3A, 7A, and 5A but have specific molecular targets. While esketamine is predicted to interact with the GABAergic system, arketamine may interact with macrophage migration inhibitory factor (MIF). Both ketamine enantiomers activate neuroplasticity-related signaling pathways and show addiction potential. Our results identified novel, poorly explored molecular targets that may be related to the beneficial effects of esketamine and arketamine against depression and suicide.
Collapse
Affiliation(s)
- Glorister A Altê
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis 88037-000, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis 88037-000, SC, Brazil
| |
Collapse
|
23
|
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: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
24
|
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
|
25
|
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: 4] [Impact Index Per Article: 2.0] [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
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
Recent Insight into Lipid Binding and Lipid Modulation of Pentameric Ligand-Gated Ion Channels. Biomolecules 2022; 12:biom12060814. [PMID: 35740939 PMCID: PMC9221113 DOI: 10.3390/biom12060814] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023] Open
Abstract
Pentameric ligand-gated ion channels (pLGICs) play a leading role in synaptic communication, are implicated in a variety of neurological processes, and are important targets for the treatment of neurological and neuromuscular disorders. Endogenous lipids and lipophilic compounds are potent modulators of pLGIC function and may help shape synaptic communication. Increasing structural and biophysical data reveal sites for lipid binding to pLGICs. Here, we update our evolving understanding of pLGIC–lipid interactions highlighting newly identified modes of lipid binding along with the mechanistic understanding derived from the new structural data.
Collapse
|
30
|
Menzikov SA, Zaichenko DM, Moskovtsev AA, Morozov SG, Kubatiev AA. Physiological Role of ATPase for GABA A Receptor Resensitization. Int J Mol Sci 2022; 23:ijms23105320. [PMID: 35628132 PMCID: PMC9141714 DOI: 10.3390/ijms23105320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 02/01/2023] Open
Abstract
γ-Aminobutyric acid type A receptors (GABAARs) mediate primarily inhibitory synaptic transmission in the central nervous system. Following fast-paced activation, which provides the selective flow of mainly chloride (Cl−) and less bicarbonate (HCO3−) ions via the pore, these receptors undergo desensitization that is paradoxically prevented by the process of their recovery, referred to as resensitization. To clarify the mechanism of resensitization, we used the cortical synaptoneurosomes from the rat brain and HEK 293FT cells. Here, we describe the effect of γ-phosphate analogues (γPAs) that mimic various states of ATP hydrolysis on GABAAR-mediated Cl− and HCO3− fluxes in response to the first and repeated application of the agonist. We found that depending on the presence of bicarbonate, opened and desensitized states of the wild or chimeric GABAARs had different sensitivities to γPAs. This study presents the evidence that recovery of neuronal Cl− and HCO3− concentrations after desensitization is accompanied by a change in the intracellular ATP concentration via ATPase performance. The transition between the desensitization and resensitization states was linked to changes in both conformation and phosphorylation. In addition, the chimeric β3 isoform did not exhibit the desensitization of the GABAAR-mediated Cl− influx but only the resensitization. These observations lend a new physiological significance to the β3 subunit in the manifestation of GABAAR resensitization.
Collapse
Affiliation(s)
- Sergey A. Menzikov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, 8, Baltiyskaya St., 125315 Moscow, Russia; (D.M.Z.); (A.A.M.); (S.G.M.); (A.A.K.)
- Correspondence: ; Tel.: +7-(499)-151-1756; Fax: +7-(495)-601-2366
| | - Danila M. Zaichenko
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, 8, Baltiyskaya St., 125315 Moscow, Russia; (D.M.Z.); (A.A.M.); (S.G.M.); (A.A.K.)
| | - Aleksey A. Moskovtsev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, 8, Baltiyskaya St., 125315 Moscow, Russia; (D.M.Z.); (A.A.M.); (S.G.M.); (A.A.K.)
- Russian Medical Academy of Postdoctoral Education, Federal State Budgetary Educational Institution of Further Professional Education of the Ministry of Healthcare of the Russian Federation, 2/1, Barrykadnaya St., 125993 Moscow, Russia
| | - Sergey G. Morozov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, 8, Baltiyskaya St., 125315 Moscow, Russia; (D.M.Z.); (A.A.M.); (S.G.M.); (A.A.K.)
| | - Aslan A. Kubatiev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, 8, Baltiyskaya St., 125315 Moscow, Russia; (D.M.Z.); (A.A.M.); (S.G.M.); (A.A.K.)
- Russian Medical Academy of Postdoctoral Education, Federal State Budgetary Educational Institution of Further Professional Education of the Ministry of Healthcare of the Russian Federation, 2/1, Barrykadnaya St., 125993 Moscow, Russia
| |
Collapse
|
31
|
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
|
32
|
Sikstus S, Benkherouf AY, Soini SL, Uusi-Oukari M. The Influence of AA29504 on GABA A Receptor Ligand Binding Properties and Its Implications on Subtype Selectivity. Neurochem Res 2022; 47:667-678. [PMID: 34727270 PMCID: PMC8847198 DOI: 10.1007/s11064-021-03475-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/03/2021] [Accepted: 10/27/2021] [Indexed: 10/26/2022]
Abstract
The unique pharmacological properties of δ-containing γ-aminobutyric acid type A receptors (δ-GABAARs) make them an attractive target for selective and persistent modulation of neuronal excitability. However, the availability of selective modulators targeting δ-GABAARs remains limited. AA29504 ([2-amino-4-(2,4,6-trimethylbenzylamino)-phenyl]-carbamic acid ethyl ester), an analog of K+ channel opener retigabine, acts as an agonist and a positive allosteric modulator (Ago-PAM) of δ-GABAARs. Based on electrophysiological studies using recombinant receptors, AA29504 was found to be a more potent and effective agonist in δ-GABAARs than in γ2-GABAARs. In comparison, AA29504 positively modulated the activity of recombinant δ-GABAARs more effectively than γ2-GABAARs, with no significant differences in potency. The impact of AA29504's efficacy- and potency-associated GABAAR subtype selectivity on radioligand binding properties remain unexplored. Using [3H]4'-ethynyl-4-n-propylbicycloorthobenzoate ([3H]EBOB) binding assay, we found no difference in the modulatory potency of AA29504 on GABA- and THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol)-induced responses between native forebrain GABAARs of wild type and δ knock-out mice. In recombinant receptors expressed in HEK293 cells, AA29504 showed higher efficacy on δ- than γ2-GABAARs in the GABA-independent displacement of [3H]EBOB binding. Interestingly, AA29504 showed a concentration-dependent stimulation of [3H]muscimol binding to γ2-GABAARs, which was absent in δ-GABAARs. This was explained by AA29504 shifting the low-affinity γ2-GABAAR towards a higher affinity desensitized state, thereby rising new sites capable of binding GABAAR agonists with low nanomolar affinity. Hence, the potential of AA29504 to act as a desensitization-modifying allosteric modulator of γ2-GABAARs deserves further investigation for its promising influence on shaping efficacy, duration and plasticity of GABAAR synaptic responses.
Collapse
Affiliation(s)
- Sylvia Sikstus
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland
| | - Ali Y Benkherouf
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland
| | - Sanna L Soini
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland
| | - Mikko Uusi-Oukari
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014, Turku, Finland.
| |
Collapse
|
33
|
Mechanisms of inhibition and activation of extrasynaptic αβ GABA A receptors. Nature 2022; 602:529-533. [PMID: 35140402 PMCID: PMC8850191 DOI: 10.1038/s41586-022-04402-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 12/22/2021] [Indexed: 12/21/2022]
Abstract
Type A GABA (γ-aminobutyric acid) receptors represent a diverse population in the mammalian brain, forming pentamers from combinations of α-, β-, γ-, δ-, ε-, ρ-, θ- and π-subunits1. αβ, α4βδ, α6βδ and α5βγ receptors favour extrasynaptic localization, and mediate an essential persistent (tonic) inhibitory conductance in many regions of the mammalian brain1,2. Mutations of these receptors in humans are linked to epilepsy and insomnia3,4. Altered extrasynaptic receptor function is implicated in insomnia, stroke and Angelman and Fragile X syndromes1,5, and drugs targeting these receptors are used to treat postpartum depression6. Tonic GABAergic responses are moderated to avoid excessive suppression of neuronal communication, and can exhibit high sensitivity to Zn2+ blockade, in contrast to synapse-preferring α1βγ, α2βγ and α3βγ receptor responses5,7–12. Here, to resolve these distinctive features, we determined structures of the predominantly extrasynaptic αβ GABAA receptor class. An inhibited state bound by both the lethal paralysing agent α-cobratoxin13 and Zn2+ was used in comparisons with GABA–Zn2+ and GABA-bound structures. Zn2+ nullifies the GABA response by non-competitively plugging the extracellular end of the pore to block chloride conductance. In the absence of Zn2+, the GABA signalling response initially follows the canonical route until it reaches the pore. In contrast to synaptic GABAA receptors, expansion of the midway pore activation gate is limited and it remains closed, reflecting the intrinsic low efficacy that characterizes the extrasynaptic receptor. Overall, this study explains distinct traits adopted by αβ receptors that adapt them to a role in tonic signalling. Cryo-electron microscopy structures are used to identify mechanisms underlying distinct features of extrasynaptic type A γ-aminobutyric acid receptors.
Collapse
|
34
|
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
|
35
|
Dietzen NM, Arcario MJ, Chen LJ, Petroff JT, Moreland KT, Krishnan K, Brannigan G, Covey DF, Cheng WW. Polyunsaturated fatty acids inhibit a pentameric ligand-gated ion channel through one of two binding sites. eLife 2022; 11:74306. [PMID: 34982031 PMCID: PMC8786314 DOI: 10.7554/elife.74306] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/31/2021] [Indexed: 01/01/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) inhibit pentameric ligand-gated ion channels (pLGICs) but the mechanism of inhibition is not well understood. The PUFA, docosahexaenoic acid (DHA), inhibits agonist responses of the pLGIC, ELIC, more effectively than palmitic acid, similar to the effects observed in the GABAA receptor and nicotinic acetylcholine receptor. Using photo-affinity labeling and coarse-grained molecular dynamics simulations, we identified two fatty acid binding sites in the outer transmembrane domain (TMD) of ELIC. Fatty acid binding to the photolabeled sites is selective for DHA over palmitic acid, and specific for an agonist-bound state. Hexadecyl-methanethiosulfonate modification of one of the two fatty acid binding sites in the outer TMD recapitulates the inhibitory effect of PUFAs in ELIC. The results demonstrate that DHA selectively binds to multiple sites in the outer TMD of ELIC, but that state-dependent binding to a single intrasubunit site mediates DHA inhibition of ELIC.
Collapse
Affiliation(s)
- Noah M Dietzen
- Department of Anesthesiology, Washington University in St. Louis, St Louis, United States
| | - Mark J Arcario
- Department of Anesthesiology, Washington University in St. Louis, St Louis, United States
| | - Lawrence J Chen
- Department of Anesthesiology, Washington University in St. Louis, St Louis, United States
| | - John T Petroff
- Department of Anesthesiology, Washington University in St. Louis, St Louis, United States
| | - K Trent Moreland
- Department of Anesthesiology, Washington University in St. Louis, St Louis, United States
| | - Kathiresan Krishnan
- Department of Developmental Biology, Washington University in St. Louis, St Louis, United States
| | - Grace Brannigan
- Center for the Computational and Integrative Biology, Rutgers University, Camden, United States.,Department of Physics, Rutgers University, Camden, United States
| | - Douglas F Covey
- Department of Anesthesiology, Washington University in St. Louis, St Louis, United States.,Department of Developmental Biology, Washington University in St. Louis, St Louis, United States.,Department of Psychiatry, Washington University in St. Louis, St. Louis, United States.,Taylor Institute for Innovative Psychiatric Research, Washington University in St. Louis, St. Louis, United States
| | - Wayland Wl Cheng
- Department of Anesthesiology, Washington University in St. Louis, St Louis, United States
| |
Collapse
|
36
|
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
|
37
|
Mokrov G, Pantileev A, Yarkova M, Gudasheva T, Seredenin S. Design, synthesis, and pharmacological activity of new pyrrolo[1,2-a]pyrazine translocator protein (tspo) ligands. Med Chem 2021; 18:497-508. [PMID: 34365957 DOI: 10.2174/1573406417666210806095051] [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: 09/30/2020] [Revised: 04/22/2021] [Accepted: 05/04/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Translocator protein 18 kDa (TSPO) is a promising target for the creation of effective and safe neuropsychotropic drugs. The ligands of TSPO exhibit anxiolytic, antidepressant, neuroprotective and other activities without the side effects of benzodiazepines. METHODS New TSPO ligands in the series of N,1-diphenylpyrrolo[1,2-a]pyrazine-3-carboxamides derivatives were designed using calculated pharmacophore model and molecular docking analysis. The synthesis of new compounds was carried out by two schemes using [3+3]-cycloaddition reaction of 2-azidoacrylic acid derivatives with pyrrolphenylketone as a key stage. The anxiolytic activity of new substances has been established using open field test with flash. RESULTS Several synthesized N,1-diphenylpyrrolo[1,2-a]pyrazine-3-carboxamides derivatives significantly increased the total motor activity of Balb/c mice compared to the control. The structure-activity relationship was investigated. The most effective compound was found to be GML-11 (N-benzyl-N,1-diphenylpyrrolo[1,2-a]pyrazine-3-carboxamide), which had anxiolytic action in the dose range from 0.001 to 0.100 mg/kg (Balb/c, i.p.). This compound is two orders of magnitude higher in dose activity than all other pyrrolo[1,2-a]pyrazine TSPO ligands. CONCLUSION Molecular modelling methods allowed us to create new TSPO ligands in the series of N,1-diphenylpyrrolo[1,2-a]pyrazine-3-carboxamides with high anxiolytic activity.
Collapse
Affiliation(s)
- Grigory Mokrov
- FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya 8, Moscow, 125315. Russian Federation
| | - Andry Pantileev
- FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya 8, Moscow, 125315. Russian Federation
| | - Milada Yarkova
- FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya 8, Moscow, 125315. Russian Federation
| | - Tatiana Gudasheva
- FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya 8, Moscow, 125315. Russian Federation
| | - Sergei Seredenin
- FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya 8, Moscow, 125315. Russian Federation
| |
Collapse
|
38
|
Germann AL, Pierce SR, Tateiwa H, Sugasawa Y, Reichert DE, Evers AS, Steinbach JH, Akk G. Intrasubunit and Intersubunit Steroid Binding Sites Independently and Additively Mediate α1 β2 γ2L GABA A Receptor Potentiation by the Endogenous Neurosteroid Allopregnanolone. Mol Pharmacol 2021; 100:19-31. [PMID: 33958479 PMCID: PMC8256884 DOI: 10.1124/molpharm.121.000268] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/22/2021] [Indexed: 12/18/2022] Open
Abstract
Prior work employing functional analysis, photolabeling, and X-ray crystallography have identified three distinct binding sites for potentiating steroids in the heteromeric GABAA receptor. The sites are located in the membrane-spanning domains of the receptor at the β-α subunit interface (site I) and within the α (site II) and β subunits (site III). Here, we have investigated the effects of mutations to these sites on potentiation of the rat α1β2γ2L GABAA receptor by the endogenous neurosteroid allopregnanolone (3α5αP). The mutations were introduced alone or in combination to probe the additivity of effects. We show that the effects of amino acid substitutions in sites I and II are energetically additive, indicating independence of the actions of the two steroid binding sites. In site III, none of the mutations tested reduced potentiation by 3α5αP, nor did a mutation in site III modify the effects of mutations in sites I or II. We infer that the binding sites for 3α5αP act independently. The independence of steroid action at each site is supported by photolabeling data showing that mutations in either site I or site II selectively change steroid orientation in the mutated site without affecting labeling at the unmutated site. The findings are discussed in the context of linking energetic additivity to empirical changes in receptor function and ligand binding. SIGNIFICANCE STATEMENT: Prior work has identified three distinct binding sites for potentiating steroids in the heteromeric γ-aminobutyric acid type A receptor. This study shows that the sites act independently and additively in the presence of the steroid allopregnanolone and provide estimates of energetic contributions made by steroid binding to each site.
Collapse
Affiliation(s)
- Allison L Germann
- Departments of Anesthesiology (A.L.G., S.R.P., H.T., A.S.E., J.H.S., G.A.) and Radiology (D.E.R.), and the Taylor Family Institute for Innovative Psychiatric Research (D.E.R., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St. Louis, Missouri; and Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo, Japan (Y.S.)
| | - Spencer R Pierce
- Departments of Anesthesiology (A.L.G., S.R.P., H.T., A.S.E., J.H.S., G.A.) and Radiology (D.E.R.), and the Taylor Family Institute for Innovative Psychiatric Research (D.E.R., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St. Louis, Missouri; and Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo, Japan (Y.S.)
| | - Hiroki Tateiwa
- Departments of Anesthesiology (A.L.G., S.R.P., H.T., A.S.E., J.H.S., G.A.) and Radiology (D.E.R.), and the Taylor Family Institute for Innovative Psychiatric Research (D.E.R., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St. Louis, Missouri; and Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo, Japan (Y.S.)
| | - Yusuke Sugasawa
- Departments of Anesthesiology (A.L.G., S.R.P., H.T., A.S.E., J.H.S., G.A.) and Radiology (D.E.R.), and the Taylor Family Institute for Innovative Psychiatric Research (D.E.R., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St. Louis, Missouri; and Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo, Japan (Y.S.)
| | - David E Reichert
- Departments of Anesthesiology (A.L.G., S.R.P., H.T., A.S.E., J.H.S., G.A.) and Radiology (D.E.R.), and the Taylor Family Institute for Innovative Psychiatric Research (D.E.R., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St. Louis, Missouri; and Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo, Japan (Y.S.)
| | - Alex S Evers
- Departments of Anesthesiology (A.L.G., S.R.P., H.T., A.S.E., J.H.S., G.A.) and Radiology (D.E.R.), and the Taylor Family Institute for Innovative Psychiatric Research (D.E.R., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St. Louis, Missouri; and Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo, Japan (Y.S.)
| | - Joe Henry Steinbach
- Departments of Anesthesiology (A.L.G., S.R.P., H.T., A.S.E., J.H.S., G.A.) and Radiology (D.E.R.), and the Taylor Family Institute for Innovative Psychiatric Research (D.E.R., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St. Louis, Missouri; and Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo, Japan (Y.S.)
| | - Gustav Akk
- Departments of Anesthesiology (A.L.G., S.R.P., H.T., A.S.E., J.H.S., G.A.) and Radiology (D.E.R.), and the Taylor Family Institute for Innovative Psychiatric Research (D.E.R., A.S.E., J.H.S., G.A.), Washington University School of Medicine, St. Louis, Missouri; and Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, Tokyo, Japan (Y.S.)
| |
Collapse
|
39
|
Epipregnanolone as a Positive Modulator of GABA A Receptor in Rat Cerebellar and Hippocampus Neurons. Biomolecules 2021; 11:biom11060791. [PMID: 34074021 PMCID: PMC8225200 DOI: 10.3390/biom11060791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 11/24/2022] Open
Abstract
Epipregnanolone (3β-hydroxy-5β-pregnan-20-one, Epi) is an endogenous steroid with important physiological effects and high affinity for GABAA receptors. The effect of Epi on GABA-induced chloride current (IGABA) in native neurons has hardly been studied. In this work, we studied the influence of Epi on the IGABA in the Purkinje cells of rat cerebellum and pyramidal neurons of rat hippocampus with the patch clamp technique. We showed that Epi is a positive modulator of the IGABA with EC50 of 5.7 µM in Purkinje cells and 9.3 µM in hippocampal neurons. Epi-induced potentiation of the IGABA was more potent at low vs. high GABA concentrations. Isopregnanolone (3β-hydroxy-5α-pregnan-20-one, Iso) counteracted Epi, reducing its potentiating effect by 2–2.3 times. Flumazenil, a nonsteroidal GABAA receptor antagonist, does not affect the Epi-induced potentiation. Comparison of the potentiating effects of Epi and allopregnanolone (3α-hydroxy-5α-pregnan-20-one, ALLO) showed that ALLO is, at least, a four times more potent positive modulator than Epi. The combined application of ALLO and Epi showed that the effects of these two steroids are not additive. We conclude that Epi has a dual effect on the IGABA increasing the current in the control solution and decreasing the stimulatory effect of ALLO.
Collapse
|
40
|
Niu C, Leavitt LS, Lin Z, Paguigan ND, Sun L, Zhang J, Torres JP, Raghuraman S, Chase K, Cadeddu R, Karthikeyan M, Bortolato M, Reilly CA, Hughen RW, Light AR, Olivera BM, Schmidt EW. Neuroactive Type-A γ-Aminobutyric Acid Receptor Allosteric Modulator Steroids from the Hypobranchial Gland of Marine Mollusk, Conus geographus. J Med Chem 2021; 64:7033-7043. [PMID: 33949869 DOI: 10.1021/acs.jmedchem.1c00562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In a program to identify pain treatments with low addiction potential, we isolated five steroids, conosteroids A-E (1-5), from the hypobranchial gland of the mollusk Conus geographus. Compounds 1-5 were active in a mouse dorsal root ganglion (DRG) assay that suggested that they might be analgesic. A synthetic analogue 6 was used for a detailed pharmacological study. Compound 6 significantly increased the pain threshold in mice in the hot-plate test at 2 and 50 mg/kg. Compound 6 at 500 nM antagonizes type-A γ-aminobutyric acid receptors (GABAARs). In a patch-clamp experiment, out of the six subunit combinations tested, 6 exhibited subtype selectivity, most strongly antagonizing α1β1γ2 and α4β3γ2 receptors (IC50 1.5 and 1.0 μM, respectively). Although the structures of 1-6 differ from those of known neuroactive steroids, they are cell-type-selective modulators of GABAARs, expanding the known chemical space of neuroactive steroids.
Collapse
Affiliation(s)
- Changshan Niu
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Lee S Leavitt
- School of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Zhenjian Lin
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Noemi D Paguigan
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Lili Sun
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jie Zhang
- Department of Anesthesiology, School of Medicine, University of Utah, Salt Lake City, Utah 84112, United States
| | - Joshua P Torres
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Shrinivasan Raghuraman
- School of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Kevin Chase
- School of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Roberto Cadeddu
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Manju Karthikeyan
- School of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Christopher A Reilly
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Ronald W Hughen
- Department of Anesthesiology, School of Medicine, University of Utah, Salt Lake City, Utah 84112, United States
| | - Alan R Light
- Department of Anesthesiology, School of Medicine, University of Utah, Salt Lake City, Utah 84112, United States
| | - Baldomero M Olivera
- School of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Eric W Schmidt
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
41
|
Castellano D, Shepard RD, Lu W. Looking for Novelty in an "Old" Receptor: Recent Advances Toward Our Understanding of GABA ARs and Their Implications in Receptor Pharmacology. Front Neurosci 2021; 14:616298. [PMID: 33519367 PMCID: PMC7841293 DOI: 10.3389/fnins.2020.616298] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
Diverse populations of GABAA receptors (GABAARs) throughout the brain mediate fast inhibitory transmission and are modulated by various endogenous ligands and therapeutic drugs. Deficits in GABAAR signaling underlie the pathophysiology behind neurological and neuropsychiatric disorders such as epilepsy, anxiety, and depression. Pharmacological intervention for these disorders relies on several drug classes that target GABAARs, such as benzodiazepines and more recently neurosteroids. It has been widely demonstrated that subunit composition and receptor stoichiometry impact the biophysical and pharmacological properties of GABAARs. However, current GABAAR-targeting drugs have limited subunit selectivity and produce their therapeutic effects concomitantly with undesired side effects. Therefore, there is still a need to develop more selective GABAAR pharmaceuticals, as well as evaluate the potential for developing next-generation drugs that can target accessory proteins associated with native GABAARs. In this review, we briefly discuss the effects of benzodiazepines and neurosteroids on GABAARs, their use as therapeutics, and some of the pitfalls associated with their adverse side effects. We also discuss recent advances toward understanding the structure, function, and pharmacology of GABAARs with a focus on benzodiazepines and neurosteroids, as well as newly identified transmembrane proteins that modulate GABAARs.
Collapse
Affiliation(s)
- David Castellano
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Ryan David Shepard
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Wei Lu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|