1
|
Rong W, Qian X, Yin Y, Gu Y, Su W, Li J, Xu Y, Zhu H, Li J, Zhu Q. N-Demethylsinomenine Relieves Neuropathic Pain in Male Mice Mainly via Regulating α2-Subtype GABA A Receptors. CNS Neurosci Ther 2025; 31:e70197. [PMID: 39749638 PMCID: PMC11696256 DOI: 10.1111/cns.70197] [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: 08/30/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 01/04/2025] Open
Abstract
AIMS N-Demethylsinomenine (NDSM) demonstrates good analgesic efficacy in preclinical pain models. However, how NDSM exerts analgesic actions remains unknown. METHODS We examined the analgesic effects of NDSM using both pain-evoked and pain-suppressed behavioral assays in two persistent pain models. Then western blot assay and immunofluorescence staining were used to investigate the effects of NDSM on the expression of the GABAA receptor α2 subunit (GABRA2) and inflammatory factors in the spinal cord and brain tissues of male spared nerve injury (SNI) mice. Finally, the individual subtypes of GABAARs (α1, α2, α3, and α5) were respectively silenced by viral-mediated knockdown to explore the involvement of subtypes of GABAARs in the effects of NDSM on the pain-like behaviors in male SNI mice. RESULTS NDSM demonstrated significant analgesic effects against chronic pain both in pain-evoked and pain-suppressed behavioral assays. NDSM treatment significantly reversed the SNI induced down-regulation of GABRA2 and up-regulation of TNF-α and IL-1β. The analgesic effects of NDSM were completely blocked by silencing GABRA2 or partially blocked by silencing GABRA3. CONCLUSION This study provided the first evidence that the analgesic effects of NDSM are mediated primarily by GABRA2 and partially by GABRA3, and the inhibition of neuroinflammation also contributes to the analgesic effects of NDSM.
Collapse
Affiliation(s)
- Weiwei Rong
- School of PharmacyNantong UniversityNantongJiangsuChina
- Provincial Key Laboratory of Inflammation and Molecular Drug TargetNantongJiangsuChina
| | - Xunjia Qian
- School of PharmacyNantong UniversityNantongJiangsuChina
| | - Yujian Yin
- School of PharmacyNantong UniversityNantongJiangsuChina
| | - Yipeng Gu
- School of PharmacyNantong UniversityNantongJiangsuChina
| | - Weiyi Su
- School of PharmacyNantong UniversityNantongJiangsuChina
| | - Jie‐Jia Li
- Affiliated Hospital 2 of Nantong UniversityNantongJiangsuChina
- State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacauChina
| | - Yue Xu
- State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacauChina
| | - Hongyan Zhu
- School of PharmacyNantong UniversityNantongJiangsuChina
| | - Junxu Li
- School of PharmacyNantong UniversityNantongJiangsuChina
| | - Qing Zhu
- School of PharmacyNantong UniversityNantongJiangsuChina
- Provincial Key Laboratory of Inflammation and Molecular Drug TargetNantongJiangsuChina
| |
Collapse
|
2
|
Junkes L, Mendlowicz MV, Shader R, Nardi AE. Leo Sternbach and the benzodiazepines 60 years on: A revolutionary treatment for anxiety disorders. Pharmacol Res 2024; 207:107310. [PMID: 39059612 DOI: 10.1016/j.phrs.2024.107310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Affiliation(s)
- Larissa Junkes
- Institute of Psychiatry (IPUB), Federal University of Rio de Janeiro (UFRJ), Venceslau Brás Avenue, 71, Botafogo, Rio de Janeiro 22290-140, Brazil.
| | - Mauro V Mendlowicz
- Federal Fluminense University (UFF), Marquês de Paraná Avenue, 303, Downtown, Niteroi 24030-215, Brazil
| | - Richard Shader
- Tufts University, Graduate School of Biomedical Sciences, Pharmacology Program, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Antonio E Nardi
- Institute of Psychiatry (IPUB), Federal University of Rio de Janeiro (UFRJ), Venceslau Brás Avenue, 71, Botafogo, Rio de Janeiro 22290-140, Brazil
| |
Collapse
|
3
|
Bampali K, Koniuszewski F, Vogel FD, Fabjan J, Andronis C, Lekka E, Virvillis V, Seidel T, Delaunois A, Royer L, Rolf MG, Giuliano C, Traebert M, Roussignol G, Fric-Bordat M, Mazelin-Winum L, Bryant SD, Langer T, Ernst M. GABA A receptor-mediated seizure liabilities: a mixed-methods screening approach. Cell Biol Toxicol 2023; 39:2793-2819. [PMID: 37093397 PMCID: PMC10693519 DOI: 10.1007/s10565-023-09803-y] [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: 12/08/2022] [Accepted: 03/09/2023] [Indexed: 04/25/2023]
Abstract
GABAA receptors, members of the pentameric ligand-gated ion channel superfamily, are widely expressed in the central nervous system and mediate a broad range of pharmaco-toxicological effects including bidirectional changes to seizure threshold. Thus, detection of GABAA receptor-mediated seizure liabilities is a big, partly unmet need in early preclinical drug development. This is in part due to the plethora of allosteric binding sites that are present on different subtypes of GABAA receptors and the critical lack of screening methods that detect interactions with any of these sites. To improve in silico screening methods, we assembled an inventory of allosteric binding sites based on structural data. Pharmacophore models representing several of the binding sites were constructed. These models from the NeuroDeRisk IL Profiler were used for in silico screening of a compiled collection of drugs with known GABAA receptor interactions to generate testable hypotheses. Amoxapine was one of the hits identified and subjected to an array of in vitro assays to examine molecular and cellular effects on neuronal excitability and in vivo locomotor pattern changes in zebrafish larvae. An additional level of analysis for our compound collection is provided by pharmacovigilance alerts using FAERS data. Inspired by the Adverse Outcome Pathway framework, we postulate several candidate pathways leading from specific binding sites to acute seizure induction. The whole workflow can be utilized for any compound collection and should inform about GABAA receptor-mediated seizure risks more comprehensively compared to standard displacement screens, as it rests chiefly on functional data.
Collapse
Affiliation(s)
- Konstantina Bampali
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Filip Koniuszewski
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Florian D Vogel
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Jure Fabjan
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | | | | | | | - Thomas Seidel
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Josef-Holaubek-Platz 2, 1090, Vienna, Austria
| | - Annie Delaunois
- UCB Biopharma SRL, Chemin du Foriest, Braine-L'Alleud, Belgium
| | - Leandro Royer
- UCB Biopharma SRL, Chemin du Foriest, Braine-L'Alleud, Belgium
| | - Michael G Rolf
- R&D Biopharmaceuticals, Astra Zeneca, Pepparedsleden 1, 431 83, Mölndal, Sweden
| | - Chiara Giuliano
- R&D Biopharmaceuticals, Astra Zeneca, Fleming Building (B623), Babraham Research Park, Babraham, Cambridgeshire, CB22 3AT, UK
| | - Martin Traebert
- Novartis Institutes for Biomedical Research, Fabrikstrasse 2, CH-4056, Basel, Switzerland
| | | | | | | | - Sharon D Bryant
- Inte:Ligand GmbH, Mariahilferstrasse 74B/11, 1070, Vienna, Austria
| | - Thierry Langer
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, University of Vienna, Josef-Holaubek-Platz 2, 1090, Vienna, Austria
| | - Margot Ernst
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| |
Collapse
|
4
|
Ali AB, Islam A, Constanti A. The fate of interneurons, GABA A receptor sub-types and perineuronal nets in Alzheimer's disease. Brain Pathol 2022; 33:e13129. [PMID: 36409151 PMCID: PMC9836378 DOI: 10.1111/bpa.13129] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurological disease, which is associated with gradual memory loss and correlated with synaptic hyperactivity and abnormal oscillatory rhythmic brain activity that precedes phenotypic alterations and is partly responsible for the spread of the disease pathology. Synaptic hyperactivity is thought to be because of alteration in the homeostasis of phasic and tonic synaptic inhibition, which is orchestrated by the GABAA inhibitory system, encompassing subclasses of interneurons and GABAA receptors, which play a vital role in cognitive functions, including learning and memory. Furthermore, the extracellular matrix, the perineuronal nets (PNNs) which often go unnoticed in considerations of AD pathology, encapsulate the inhibitory cells and neurites in critical brain regions and have recently come under the light for their crucial role in synaptic stabilisation and excitatory-inhibitory balance and when disrupted, serve as a potential trigger for AD-associated synaptic imbalance. Therefore, in this review, we summarise the current understanding of the selective vulnerability of distinct interneuron subtypes, their synaptic and extrasynaptic GABAA R subtypes as well as the changes in PNNs in AD, detailing their contribution to the mechanisms of disease development. We aim to highlight how seemingly unique malfunction in each component of the interneuronal GABA inhibitory system can be tied together to result in critical circuit dysfunction, leading to the irreversible symptomatic damage observed in AD.
Collapse
|
5
|
Kaplanian A, Vinos M, Skaliora I. GABAb- and GABAa- mediated regulation of Up and Down states across development. J Physiol 2022; 600:2401-2427. [PMID: 35365894 DOI: 10.1113/jp282736] [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: 12/15/2021] [Accepted: 03/18/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Slow oscillations (SOs), the EEG hallmark of non-REM sleep, and their cellular counterpart, Up-and-Down states (UDSs), are considered the default activity of the cerebral cortex and reflect the underlying neural connectivity. GABAb- and GABAa- receptor-mediated inhibition play a major role in regulating UDS activity. Although SOs and UDSs exhibit significant alterations as a function of age, it is unknown how developmental changes in inhibition contribute to the developmental profile of this activity. In this study, we reveal for the first time, age-dependent effects of GABAb and GABAa signalling on UDSs. We also document the differential subunit composition of postsynaptic GABAa receptors in young and adult animals, highlighting the α1-subunit as a major component of the age-differentiated regulation of UDSs. These findings help clarify the mechanisms that underlie the maturation of cortical network activity, and enhance our understanding regarding the emergence of neurodevelopmental disorders. ABSTRACT Slow oscillations, the hallmark of non-REM sleep, and their cellular counterpart, Up-and-Down states (UDSs), are considered a signature of cortical dynamics that reflect the intrinsic network organization. Although previous studies have explored the role of inhibition in regulating UDSs, little is known about whether this role changes with maturation. This is surprising since both slow oscillations and UDSs exhibit significant age-dependent alterations. To elucidate the developmental impact of GABAb and GABAa receptors on UDS activity, we conducted simultaneous LFP and intracellular recordings ex vivo, in brain slices of young and adult male mice, using selective blockers, CGP and non-saturating concentration of gabazine, respectively. Blockade of both GABAb- and GABAa- signalling showed age-differentiated functions. CGP caused an increase in Down state duration in young animals, but a decrease in adults. Gabazine evoked Spike-and-Wave-Discharges in both ages; however, while young networks became completely epileptic, adults maintained the ability to generate UDSs. Furthermore, voltage clamp recordings of mIPSCs revealed that gabazine selectively blocks phasic currents, particularly involving postsynaptic mechanisms. The latter exhibit clear maturational changes, suggesting a different subunit composition of GABAa receptors in young vs. adult animals. Indeed, subsequent LFP recordings under diazepam (nanomolar or micromolar concentrations) revealed that mechanisms engaging the drug's classical-binding-site, mediated by α1-subunit containing GABAa receptors, have a bigger contribution in Up state initiation in young networks compared to adults. Taken together, these findings help clarify the mechanisms that underlie the maturation of cortical network activity and enhance our understanding regarding the emergence of neurodevelopmental disorders. Abstract figure legend GABAb receptors' participation in Up state termination mechanisms is well-conserved across development. However, regulation of Down-to-Up transitions is age-dependent; GABAb receptors promote them in young while preventing them in adults. Up state maintenance is determined by age-dependent synaptic GABAa receptors' subunit composition and kinetics; α1-GABAa receptors dominate in young while non-α1-GABAa receptors dominate in adults. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ani Kaplanian
- Center for Basic Research, Biomedical Research Foundation of Academy of Athens (BRFAA), Athens, 11527, Greece.,Department of Biology, University of Patras, Rio, 26504, Greece
| | - Michael Vinos
- Center for Basic Research, Biomedical Research Foundation of Academy of Athens (BRFAA), Athens, 11527, Greece.,Department of History and Philosophy of Science, University of Athens, Athens, 15771, Greece
| | - Irini Skaliora
- Center for Basic Research, Biomedical Research Foundation of Academy of Athens (BRFAA), Athens, 11527, Greece.,Department of History and Philosophy of Science, University of Athens, Athens, 15771, Greece
| |
Collapse
|
6
|
Crocetti L, Guerrini G, Cantini N, Vergelli C, Melani F, Mascia MP, Giovannoni MP. 'Proximity frequencies' a new parameter to evaluate the profile of GABA AR modulators. Bioorg Med Chem Lett 2021; 34:127755. [PMID: 33359444 DOI: 10.1016/j.bmcl.2020.127755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022]
Abstract
We reported the synthesis of new 8-methoxypyrazolo[1,5-a]quinazolines bearing an amide fragment at the 3-position. The final compounds, as aromatic (2a-i) and 4,5-dihydro derivatives (3a-i), have been evaluated in vitrofor their ability to modulate the chlorine current on recombinant GABAA receptors of the α1β2γ2L type (expressed in frog oocytes of the Xenopus laevis species). From electrophysiological test two groups of compounds emerged: positive modulators agonist (2e, h, i and 3e, h) and null modulators antagonist (2a, b, d, f, g and 3a-d, f, g) of GABAA subtype receptor. Using a set of compounds (new derivatives, known products and GABAA subtype receptor ligands from our library) we identify the amino acids at the α+/γ- interface, which could be involved in the agonist or antagonist profile, using the 'Proximity Frequencies', namely the frequencies with which a ligand intercepts two or more binding-site amino acids during the molecular dynamic simulation. The linear discriminant analysis (LDA) evidences that the combination of amino acids αVAL203- γTHR142 and αTYR 160- γTYR 58 allowed to collocate 70.6% of agonists and 72.7% of antagonists in their respective class.
Collapse
Affiliation(s)
- Letizia Crocetti
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy.
| | - Gabriella Guerrini
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy.
| | - Niccolò Cantini
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy
| | - Claudia Vergelli
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy.
| | - Fabrizio Melani
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy
| | - Maria Paola Mascia
- CNR - Institute of Neuroscience, Cittadella Unversitaria, Cagliari, Italy
| | - Maria Paola Giovannoni
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy.
| |
Collapse
|
7
|
Design, synthesis, and pharmacological evaluation of novel 1,2,4-triazol-3-amine derivatives as potential agonists of GABAA subtype receptors with anticonvulsant and hypnotic effects. Bioorg Chem 2020; 104:104212. [DOI: 10.1016/j.bioorg.2020.104212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/24/2020] [Indexed: 01/10/2023]
|
8
|
Crocetti L, Guerrini G. GABA A receptor subtype modulators in medicinal chemistry: an updated patent review (2014-present). Expert Opin Ther Pat 2020; 30:409-432. [PMID: 32200689 DOI: 10.1080/13543776.2020.1746764] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Introduction: Ligands at the benzodiazepine binding site of the GABAA receptor (GABAAR) act by modulating the effect of GABA (γ-aminobutyric acid). The benzodiazepine drugs are conventionally categorized as positive allosteric modulators enhancing the chloride ion current GABA-induced. In literature there are also reported ligands that act as negative allosteric modulators, reducing chloride ion current, and silent allosteric modulators not influencing the chloride ion flux.Areas covered: This review covers patents published from 2014 to present on ligands for the benzodiazepine binding site of the GABAARs. Patents filed from different companies and research groups report many compounds that may be used in the treatment/prevention of a large variety of diseases.Expert opinion: Since the discovery of the first benzodiazepine about 60 years have passed and about 50 years since the identification of their target, GABAA receptor. Even if benzodiazepines are the most popular anxiolytic drugs, the research in this field is still very active. From patents/application analysis arises that most of them claim methods for alleviating specific symptoms in different neurodegenerative diseases and their related memory deficits. Noteworthy is the presence of the α4- and α5-GABAA receptor subtype ligands as new pharmacological tools for airway hyperresponsiveness, inflammation diseases, and asthma.
Collapse
Affiliation(s)
- Letizia Crocetti
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy
| | - Gabriella Guerrini
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
9
|
Tonon MC, Vaudry H, Chuquet J, Guillebaud F, Fan J, Masmoudi-Kouki O, Vaudry D, Lanfray D, Morin F, Prevot V, Papadopoulos V, Troadec JD, Leprince J. Endozepines and their receptors: Structure, functions and pathophysiological significance. Pharmacol Ther 2020; 208:107386. [DOI: 10.1016/j.pharmthera.2019.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
|
10
|
Effects of the α2/α3-subtype-selective GABAA receptor positive allosteric modulator KRM-II-81 on pain-depressed behavior in rats: comparison with ketorolac and diazepam. Behav Pharmacol 2020; 30:452-461. [PMID: 30640180 DOI: 10.1097/fbp.0000000000000464] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This study examined effects of the α2/α3-subtype-selective GABAA receptor positive allosteric modulator KRM-II-81 in an assay of pain-related behavioral depression. Adult, male Sprague-Dawley rats responded for electrical brain stimulation in a frequency-rate intracranial self-stimulation (ICSS) procedure. Intraperitoneal injection of 1.8% lactic acid served as an acute noxious stimulus to depress ICSS. Effects of KRM-II-81 were evaluated in the absence and presence of the acid noxious stimulus. The NSAID ketorolac and the benzodiazepine diazepam were tested as comparators. Neither ketorolac nor KRM-II-81 altered ICSS in the absence of the acid noxious stimulus; however, diazepam produced facilitation consistent with its abuse liability. Ketorolac blocked acid-induced depression of ICSS, and effects of 1.0 mg/kg ketorolac lasted for at least 5 h. KRM-II-81 (1.0 mg/kg) produced significant antinociception after 30 min that dissipated by 60 min. Diazepam also attenuated acid-depressed ICSS, but only at doses that facilitated ICSS when administered alone. The lack of ketorolac or KRM-II-81 effects on ICSS in the absence of the acid noxious stimulus suggests low abuse liability for both compounds. The effectiveness of ketorolac to block acid-induced ICSS depression agrees with clinical analgesic efficacy of ketorolac. KRM-II-81 produced significant but less consistent and shorter-acting antinociception than ketorolac.
Collapse
|
11
|
Cao Y, Yan H, Yu G, Su R. Flumazenil-insensitive benzodiazepine binding sites in GABAA receptors contribute to benzodiazepine-induced immobility in zebrafish larvae. Life Sci 2019; 239:117033. [DOI: 10.1016/j.lfs.2019.117033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/16/2019] [Accepted: 10/28/2019] [Indexed: 10/25/2022]
|
12
|
Abstract
As the inhibitory γ-aminobutyric acid-ergic (GABAergic) transmission has a pivotal role in the central nervous system (CNS) and defective forms of its synapses are associated with serious neurological disorders, numerous versions of caged GABA and, more recently, photoswitchable ligands have been developed to investigate such transmission. While the complementary nature of these probes is evident, the mechanisms by which the GABA receptors can be photocontrolled have not been fully exploited. In fact, the ultimate need for specificity is critical for the proper synaptic exploration. No caged allosteric modulators of the GABAA receptor have been reported so far; to introduce such an investigational approach, we exploited the structural motifs of the benzodiazepinic scaffold to develop a photocaged version of diazepam (CD) that was tested on basolateral amygdala (BLa) pyramidal cells in mouse brain slices. CD is devoid of any intrinsic activity toward the GABAA receptor before irradiation. Importantly, CD is a photoreleasable GABAA receptor-positive allosteric modulator that offers a different probing mechanism compared to caged GABA and photoswitchable ligands. CD potentiates the inhibitory signaling by prolonging the decay time of postsynaptic GABAergic currents upon photoactivation. Additionally, no effect on presynaptic GABA release was recorded. We developed a photochemical technology to individually study the GABAA receptor, which specifically expands the toolbox available to study GABAergic synapses.
Collapse
|
13
|
Kwakowsky A, Calvo-Flores Guzmán B, Pandya M, Turner C, Waldvogel HJ, Faull RL. GABA A receptor subunit expression changes in the human Alzheimer's disease hippocampus, subiculum, entorhinal cortex and superior temporal gyrus. J Neurochem 2019; 145:374-392. [PMID: 29485232 DOI: 10.1111/jnc.14325] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/17/2018] [Accepted: 02/12/2018] [Indexed: 12/14/2022]
Abstract
Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system. GABA type A receptors (GABAA Rs) are severely affected in Alzheimer's disease (AD). However, the distribution and subunit composition of GABAA Rs in the AD brain are not well understood. This is the first comprehensive study to show brain region- and cell layer-specific alterations in the expression of the GABAA R subunits α1-3, α5, β1-3 and γ2 in the human AD hippocampus, entorhinal cortex and superior temporal gyrus. In late-stage AD tissue samples using immunohistochemistry we found significant alteration of all investigated GABAA Rs subunits except for α3 and β1 that were well preserved. The most prominent changes include an increase in GABAA R α1 expression associated with AD in all layers of the CA3 region, in the stratum (str.) granulare and hilus of the dentate gyrus. We found a significant increase in GABAA R α2 expression in the str. oriens of the CA1-3, str. radiatum of the CA2,3 and decrease in the str. pyramidale of the CA1 region in AD cases. In AD there was a significant increase in GABAA R α5 subunit expression in str. pyramidale, str. oriens of the CA1 region and decrease in the superior temporal gyrus. We also found a significant decrease in the GABAA R β3 subunit immunoreactivity in the str. oriens of the CA2, str. granulare and str. moleculare of the dentate gyrus. In conclusion, these findings indicate that the expression of the GABAA R subunits shows brain region- and layer-specific alterations in AD, and these changes could significantly influence and alter GABAA R function in the disease. Cover Image for this issue: doi: 10.1111/jnc.14179.
Collapse
Affiliation(s)
- Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Beatriz Calvo-Flores Guzmán
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Madhavi Pandya
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
14
|
Chen X, van Gerven J, Cohen A, Jacobs G. Human pharmacology of positive GABA-A subtype-selective receptor modulators for the treatment of anxiety. Acta Pharmacol Sin 2019; 40:571-582. [PMID: 30518829 DOI: 10.1038/s41401-018-0185-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022]
Abstract
Anxiety disorders arise from disruptions among the highly interconnected circuits that normally serve to process the streams of potentially threatening stimuli. The resulting imbalance among these circuits can cause a fundamental misinterpretation of neural sensory information as threatening and can lead to the inappropriate emotional and behavioral responses observed in anxiety disorders. There is considerable preclinical evidence that the GABAergic system, in general, and its α2- and/or α5-subunit-containing GABA(A) receptor subtypes, in particular, are involved in the pathophysiology of anxiety disorders. However, the clinical efficacy of GABA-A α2-selective agonists for the treatment of anxiety disorders has not been unequivocally demonstrated. In this review, we present several human pharmacological studies that have been performed with the aim of identifying the pharmacologically active doses/exposure levels of several GABA-A subtype-selective novel compounds with potential anxiolytic effects. The pharmacological selectivity of novel α2-subtype-selective GABA(A) receptor partial agonists has been demonstrated by their distinct effect profiles on the neurophysiological and neuropsychological measurements that reflect the functions of multiple CNS domains compared with those of benzodiazepines, which are nonselective, full GABA(A) agonists. Normalizing the undesired pharmacodynamic side effects against the desired on-target effects on the saccadic peak velocity is a useful approach for presenting the pharmacological features of GABA(A)-ergic modulators. Moreover, combining the anxiogenic symptom provocation paradigm with validated neurophysiological and neuropsychological biomarkers may provide further construct validity for the clinical effects of novel anxiolytic agents. In addition, the observed drug effects on serum prolactin levels support the use of serum prolactin levels as a complementary neuroendocrine biomarker to further validate the pharmacodynamic measurements used during the clinical pharmacological study of novel anxiolytic agents.
Collapse
|
15
|
Guerrini G, Crocetti L, Daniele S, Iacovone A, Cantini N, Martini C, Melani F, Vergelli C, Giovannoni MP. New 3,6‐Disubstituted Pyrazolo[1,5‐
a
]quinazolines as Ligands to GABA
A
Receptor Subtype. J Heterocycl Chem 2019. [DOI: 10.1002/jhet.3535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Gabriella Guerrini
- Dipartimento Neurofarba, Sezione Farmaceutica e NutraceuticaUniversità degli Studi di Firenze Via Ugo Schiff 6, Sesto Fiorentino 50019 Florence Italy
| | - Letizia Crocetti
- Dipartimento Neurofarba, Sezione Farmaceutica e NutraceuticaUniversità degli Studi di Firenze Via Ugo Schiff 6, Sesto Fiorentino 50019 Florence Italy
| | - Simona Daniele
- Dipartimento FarmaciaUniversità degli Studi di Pisa Via Bonanno 6 Pisa Italy
| | - Antonella Iacovone
- Dipartimento Neurofarba, Sezione Farmaceutica e NutraceuticaUniversità degli Studi di Firenze Via Ugo Schiff 6, Sesto Fiorentino 50019 Florence Italy
| | - Niccolò Cantini
- Dipartimento Neurofarba, Sezione Farmaceutica e NutraceuticaUniversità degli Studi di Firenze Via Ugo Schiff 6, Sesto Fiorentino 50019 Florence Italy
| | - Claudia Martini
- Dipartimento FarmaciaUniversità degli Studi di Pisa Via Bonanno 6 Pisa Italy
| | - Fabrizio Melani
- Dipartimento Neurofarba, Sezione Farmaceutica e NutraceuticaUniversità degli Studi di Firenze Via Ugo Schiff 6, Sesto Fiorentino 50019 Florence Italy
| | - Claudia Vergelli
- Dipartimento Neurofarba, Sezione Farmaceutica e NutraceuticaUniversità degli Studi di Firenze Via Ugo Schiff 6, Sesto Fiorentino 50019 Florence Italy
| | - Maria Paola Giovannoni
- Dipartimento Neurofarba, Sezione Farmaceutica e NutraceuticaUniversità degli Studi di Firenze Via Ugo Schiff 6, Sesto Fiorentino 50019 Florence Italy
| |
Collapse
|
16
|
The Benzodiazepine Binding Sites of GABAA Receptors. Trends Pharmacol Sci 2018; 39:659-671. [DOI: 10.1016/j.tips.2018.03.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/15/2018] [Accepted: 03/22/2018] [Indexed: 11/24/2022]
|
17
|
Guerrini G, Ciciani G, Daniele S, Martini C, Costagli C, Guarino C, Selleri S. A new class of pyrazolo[5,1-c][1,2,4]triazines as γ-aminobutyric type A (GABAA) receptor subtype ligand: synthesis and pharmacological evaluation. Bioorg Med Chem 2018; 26:2475-2487. [DOI: 10.1016/j.bmc.2018.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/23/2018] [Accepted: 04/03/2018] [Indexed: 12/21/2022]
|
18
|
Everington EA, Gibbard AG, Swinny JD, Seifi M. Molecular Characterization of GABA-A Receptor Subunit Diversity within Major Peripheral Organs and Their Plasticity in Response to Early Life Psychosocial Stress. Front Mol Neurosci 2018; 11:18. [PMID: 29467616 PMCID: PMC5807923 DOI: 10.3389/fnmol.2018.00018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/12/2018] [Indexed: 11/13/2022] Open
Abstract
Gamma aminobutyric acid (GABA) subtype A receptors (GABAARs) are integral membrane ion channels composed of five individual proteins or subunits. Up to 19 different GABAAR subunits (α1–6, β1–3, γ1–3, δ, ε, θ, π, and ρ1–3) have been identified, resulting in anatomically, physiologically, and pharmacologically distinct multiple receptor subtypes, and therefore GABA-mediated inhibition, across the central nervous system (CNS). Additionally, GABAAR-modulating drugs are important tools in clinical medicine, although their use is limited by adverse effects. While significant advances have been made in terms of characterizing the GABAAR system within the brain, relatively less is known about the molecular phenotypes within the peripheral nervous system of major organ systems. This represents a potentially missed therapeutic opportunity in terms of utilizing or repurposing clinically available GABAAR drugs, as well as promising research compounds discarded due to their poor CNS penetrance, for the treatment of peripheral disorders. In addition, a broader understanding of the peripheral GABAAR subtype repertoires will contribute to the design of therapies which minimize peripheral side-effects when treating CNS disorders. We have recently provided a high resolution molecular and function characterization of the GABAARs within the enteric nervous system of the mouse colon. In this study, the aim was to determine the constituent GABAAR subunit expression profiles of the mouse bladder, heart, liver, kidney, lung, and stomach, using reverse transcription polymerase chain reaction and western blotting with brain as control. The data indicate that while some subunits are expressed widely across various organs (α3–5), others are restricted to individual organs (γ2, only stomach). Furthermore, we demonstrate complex organ-specific developmental expression plasticity of the transporters which determine the chloride gradient within cells, and therefore whether GABAAR activation has a depolarizing or hyperpolarizing effect. Finally, we demonstrate that prior exposure to early life psychosocial stress induces significant changes in peripheral GABAAR subunit expression and chloride transporters, in an organ- and subunit-specific manner. Collectively, the data demonstrate the molecular diversity of the peripheral GABAAR system and how this changes dynamically in response to life experience. This provides a molecular platform for functional analyses of the GABA–GABAAR system in health, and in diseases affecting various peripheral organs.
Collapse
Affiliation(s)
- Ethan A Everington
- Institute for Biomedical and Biomolecular Sciences and School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Adina G Gibbard
- Institute for Biomedical and Biomolecular Sciences and School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Jerome D Swinny
- Institute for Biomedical and Biomolecular Sciences and School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Mohsen Seifi
- Institute for Biomedical and Biomolecular Sciences and School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
19
|
Lewter LA, Fisher JL, Siemian JN, Methuku KR, Poe MM, Cook JM, Li JX. Antinociceptive Effects of a Novel α2/α3-Subtype Selective GABA A Receptor Positive Allosteric Modulator. ACS Chem Neurosci 2017; 8:1305-1312. [PMID: 28150939 DOI: 10.1021/acschemneuro.6b00447] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pain remains a challenging clinical condition and spinal GABAA receptors are crucial modulators of pain processing. α2/α3-subtype GABAA receptors mediate the analgesic actions of benzodiazepines. Positive allosteric modulators (PAMs) at α2/α3-subtype GABAA receptors may have analgesic potential. Here we report a new selective α2/α3-subtype GABAA receptor PAM in in vitro and in vivo pain assays. KRM-II-81 demonstrated similar efficacy at α1/α2/α3 GABAA receptors and negligible efficacy at α4/α5/α6 GABAA receptors, with α2 and α3-subtypes being 17- and 28-fold more potent than α1 subtypes in HEK-293T cells expressing GABAA receptors with different α subunits. In contrast, KRM-II-18B showed significant efficacy at α1/α2/α3/ α5 subtypes, with similar potency at α1/α2/α3 subtypes. Both PAMs and morphine dose-dependently decreased 0.6% acetic acid- and 0.32% lactic acid-induced writhing. The effects of both PAMs were reversed by the benzodiazepine receptor antagonist flumazenil, confirming their action at the benzodiazepine binding site of GABAA receptors. Both PAMS and morphine all dose-dependently reversed 0.32% lactic acid (but not 0.6% acetic acid) induced suppression of nesting behavior. Acetaminophen, but not the PAMs, reversed acid-depressed locomotor activity. Combined, these findings suggest that KRM-II-81 is a selective α2/α3 subtype GABAA PAM with significant antinociceptive effects in chemical stimulation-induced pain in mice.
Collapse
Affiliation(s)
- Lakeisha A. Lewter
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14214, United States
| | - Janet L. Fisher
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Justin N. Siemian
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14214, United States
| | - Kashi Reddy Methuku
- Department of Chemistry, University of Wisconsin—Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Michael M. Poe
- Department of Chemistry, University of Wisconsin—Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - James M. Cook
- Department of Chemistry, University of Wisconsin—Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14214, United States
| |
Collapse
|
20
|
Brown LE, Nicholson MW, Arama JE, Mercer A, Thomson AM, Jovanovic JN. γ-Aminobutyric Acid Type A (GABAA) Receptor Subunits Play a Direct Structural Role in Synaptic Contact Formation via Their N-terminal Extracellular Domains. J Biol Chem 2016; 291:13926-13942. [PMID: 27129275 PMCID: PMC4933154 DOI: 10.1074/jbc.m116.714790] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Indexed: 11/17/2022] Open
Abstract
The establishment of cell-cell contacts between presynaptic GABAergic neurons and their postsynaptic targets initiates the process of GABAergic synapse formation. GABAA receptors (GABAARs), the main postsynaptic receptors for GABA, have been recently demonstrated to act as synaptogenic proteins that can single-handedly induce the formation and functional maturation of inhibitory synapses. To establish how the subunit composition of GABAARs influences their ability to induce synaptogenesis, a co-culture model system incorporating GABAergic medium spiny neurons and the HEK293 cells, stably expressing different combinations of receptor subunits, was developed. Analyses of HEK293 cell innervation by medium spiny neuron axons using immunocytochemistry, activity-dependent labeling, and electrophysiology have indicated that the γ2 subunit is required for the formation of active synapses and that its effects are influenced by the type of α/β subunits incorporated into the functional receptor. To further characterize this process, the large N-terminal extracellular domains (ECDs) of α1, α2, β2, and γ2 subunits were purified using the baculovirus/Sf9 cell system. When these proteins were applied to the co-cultures of MSNs and α1/β2/γ2-expressing HEK293 cells, the α1, β2, or γ2 ECD each caused a significant reduction in contact formation, in contrast to the α2 ECD, which had no effect. Together, our experiments indicate that the structural role of GABAARs in synaptic contact formation is determined by their subunit composition, with the N-terminal ECDs of each of the subunits directly participating in interactions between the presynaptic and postsynaptic elements, suggesting the these interactions are multivalent and specific.
Collapse
Affiliation(s)
- Laura E Brown
- Research Department of Pharmacology, UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Martin W Nicholson
- Research Department of Pharmacology, UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Jessica E Arama
- Research Department of Pharmacology, UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Audrey Mercer
- Research Department of Pharmacology, UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Alex M Thomson
- Research Department of Pharmacology, UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Jasmina N Jovanovic
- Research Department of Pharmacology, UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom.
| |
Collapse
|
21
|
Gumireddy K, Li A, Kossenkov AV, Sakurai M, Yan J, Li Y, Xu H, Wang J, Zhang PJ, Zhang L, Showe LC, Nishikura K, Huang Q. The mRNA-edited form of GABRA3 suppresses GABRA3-mediated Akt activation and breast cancer metastasis. Nat Commun 2016; 7:10715. [PMID: 26869349 PMCID: PMC4754346 DOI: 10.1038/ncomms10715] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 01/12/2016] [Indexed: 12/19/2022] Open
Abstract
Metastasis is a critical event affecting breast cancer patient survival. To identify molecules contributing to the metastatic process, we analysed The Cancer Genome Atlas (TCGA) breast cancer data and identified 41 genes whose expression is inversely correlated with survival. Here we show that GABAA receptor alpha3 (Gabra3), normally exclusively expressed in adult brain, is also expressed in breast cancer, with high expression of Gabra3 being inversely correlated with breast cancer survival. We demonstrate that Gabra3 activates the AKT pathway to promote breast cancer cell migration, invasion and metastasis. Importantly, we find an A-to-I RNA-edited form of Gabra3 only in non-invasive breast cancers and show that edited Gabra3 suppresses breast cancer cell invasion and metastasis. A-to-I-edited Gabra3 has reduced cell surface expression and suppresses the activation of AKT required for cell migration and invasion. Our study demonstrates a significant role for mRNA-edited Gabra3 in breast cancer metastasis. GABRA3, a subunit of the GABA receptor, is often highly expressed in brain metastasis and breast cancers. Here, the authors demonstrated that GABRA3 activates AKT to promote breast cancer cell invasion and that the A-to-I edited form of GABRA3, specifically expressed in noninvasive breast cancers, can suppress the function of wild type GABRA3.
Collapse
Affiliation(s)
- Kiranmai Gumireddy
- Department of Tumor Microenvironment and Metastasis, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - Anping Li
- Department of Tumor Microenvironment and Metastasis, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - Andrew V Kossenkov
- Department of Tumor Microenvironment and Metastasis, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - Masayuki Sakurai
- Department of Tumor Microenvironment and Metastasis, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - Jinchun Yan
- University of Washington Medical Center, 1959 N.E. Pacific Street, Seattle, Washington 98195, USA.,Department of Radiation Oncology, Cancer Hospital of Fudan University, 270 Dong An Road, Shanghai 200032, China
| | - Yan Li
- Institute of Cancer Stem Cell, Department of Anatomy, College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, China
| | - Hua Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430030, China
| | - Jian Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Paul J Zhang
- Department of Pathology and Laboratory Medicine, Hospital of The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Louise C Showe
- Department of Tumor Microenvironment and Metastasis, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - Kazuko Nishikura
- Department of Tumor Microenvironment and Metastasis, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - Qihong Huang
- Department of Tumor Microenvironment and Metastasis, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|