Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.962
Filter
Add more filters

Publication year range
1.
Nature ; 622(7981): 195-201, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37730991

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 α1ß2γ2 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/ß 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.


Subject(s)
Cryoelectron Microscopy , Neurosteroids , Receptors, GABA-A , gamma-Aminobutyric Acid , Animals , Mice , Binding Sites/drug effects , Depression, Postpartum/drug therapy , Flurazepam/pharmacology , gamma-Aminobutyric Acid/metabolism , Hypnotics and Sedatives/pharmacology , Ion Channel Gating/drug effects , Neurosteroids/metabolism , Neurosteroids/pharmacology , Photobleaching , Pregnanolone/pharmacology , Protein Conformation/drug effects , Protein Subunits/chemistry , Protein Subunits/drug effects , Protein Subunits/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Receptors, GABA-A/ultrastructure , Sleep Initiation and Maintenance Disorders/drug therapy , Zolpidem/pharmacology
2.
J Neurophysiol ; 132(1): 177-183, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38836296

ABSTRACT

The reliable induction of long-term potentiation (LTP) in the dentate gyrus (DG) in vitro requires the blockade of the γ-aminobutyric acid A (GABAA) receptor. In these studies we examined the effectiveness of the specific GABAA receptor antagonist bicuculline methiodide (BMI) in facilitating LTP in the DG from hippocampal slices obtained from either C57Bl/6 mice or Sprague-Dawley rats, two species commonly used for electrophysiology. In the C57Bl/6 mice, maximal short-term potentiation and LTP in the DG were produced with a concentration of 5 µM BMI. In contrast, a concentration of 10 µM BMI was required to produce maximal short-term potentiation and LTP in the DG of Sprague-Dawley rats. These results reveal that there are species differences in the optimal amount of BMI required to produce robust and reliable LTP in the rodent DG in vitro and highlight the need to take consideration of the species being used when choosing concentrations of pharmacological agents to employ for electrophysiological use.NEW & NOTEWORTHY In this report we provide specific neurophysiological evidence for concentrations of GABAA antagonist required to study long-term potentiation in the medial perforant pathway of the dentate gyrus. Two commonly used species, Sprague-Dawley rats and C57Bl/6 mice, require different concentrations of bicuculline methiodide to induce optimal short-term and long-term potentiation.


Subject(s)
Bicuculline , Dentate Gyrus , GABA-A Receptor Antagonists , Long-Term Potentiation , Mice, Inbred C57BL , Rats, Sprague-Dawley , Animals , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Dentate Gyrus/drug effects , Dentate Gyrus/physiology , Bicuculline/pharmacology , Bicuculline/analogs & derivatives , GABA-A Receptor Antagonists/pharmacology , Mice , Rats , Male , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Receptors, GABA-A/physiology , Species Specificity
3.
Anesthesiology ; 140(6): 1192-1200, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38624275

ABSTRACT

Tonic inhibition in mouse hippocampal CA1 pyramidal neurons is mediated by α5 subunit-containing γ-aminobutyric acid type A receptors. By Caraiscos VB, Elliott EM, You-Ten KE, Cheng VY, Belelli D, Newell JG, Jackson MF, Lambert JJ, Rosahl TW, Wafford KA, MacDonald JF, Orser BA. Proc Natl Acad Sci U S A 2004; 101:3662-7. Reprinted with permission. In this Classic Paper Revisited, the author recounts the scientific journey leading to a report published in the Proceedings of the National Academy of Sciences (PNAS) and shares several personal stories from her formative years and "research truths" that she has learned along the way. Briefly, the principal inhibitory neurotransmitter in the brain, γ-aminobutyric acid (GABA), was conventionally thought to regulate cognitive processes by activating synaptic GABA type A (GABAA) receptors and generating transient inhibitory synaptic currents. However, the author's laboratory team discovered a novel nonsynaptic form of tonic inhibition in hippocampal pyramidal neurons, mediated by extrasynaptic GABAA receptors that are pharmacologically distinct from synaptic GABAA receptors. This tonic current is highly sensitive to most general anesthetics, including sevoflurane and propofol, and likely contributes to the memory-blocking properties of these drugs. Before the publication in PNAS, the subunit composition of GABAA receptors that generate the tonic current was unknown. The team's research showed that GABAA receptors containing the α5 subunit (α5GABAARs) generated the tonic inhibitory current in hippocampal neurons. α5GABAARs are highly sensitive to GABA, desensitize slowly, and are thus well suited for detecting low, persistent, ambient concentrations of GABA in the extracellular space. Interest in α5GABAARs has surged since the PNAS report, driven by their pivotal roles in cognitive processes and their potential as therapeutic targets for treating various neurologic disorders.


Subject(s)
Receptors, GABA-A , Animals , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Mice , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Pyramidal Cells/metabolism , Humans , Synapses/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , gamma-Aminobutyric Acid/metabolism
4.
Pharmacol Res ; 208: 107356, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39216838

ABSTRACT

Recent advances in genetic diagnosis identified variants in genes encoding GABAA receptors as causative for genetic epilepsy. Here, we selected eight disease-associated variants in the α1 subunit of GABAA receptors causing mild to severe clinical phenotypes and showed that they are loss of function, mainly by reducing the folding and surface trafficking of the α1 protein. Furthermore, we sought client protein-specific pharmacological chaperones to restore the function of pathogenic receptors. Applications of positive allosteric modulators, including Hispidulin and TP003, increase the functional surface expression of the α1 variants. Mechanism of action study demonstrated that they enhance the folding, assembly, and trafficking and reduce the degradation of GABAA variants without activating the unfolded protein response in HEK293T cells and human iPSC-derived neurons. Since these compounds cross the blood-brain barrier, such a pharmacological chaperoning strategy holds great promise to treat genetic epilepsy in a GABAA receptor-specific manner.


Subject(s)
Epilepsy , Proteostasis , Receptors, GABA-A , Humans , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Proteostasis/drug effects , Epilepsy/drug therapy , Epilepsy/genetics , Epilepsy/metabolism , HEK293 Cells , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism
5.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33593898

ABSTRACT

Tethered photoswitches are molecules with two photo-dependent isomeric forms, each with different actions on their biological targets. They include reactive chemical groups capable of covalently binding to their target. Our aim was to develop a ß-subunit-tethered propofol photoswitch (MAP20), as a tool to better study the mechanism of anesthesia through the GABAA α1ß3γ2 receptor. We used short spacers between the tether (methanethiosulfonate), the photosensitive moiety (azobenzene), and the ligand (propofol), to allow a precise tethering adjacent to the putative propofol binding site at the ß+α- interface of the receptor transmembrane helices (TMs). First, we used molecular modeling to identify possible tethering sites in ß3TM3 and α1TM1, and then introduced cysteines in the candidate positions. Two mutant subunits [ß3(M283C) and α1(V227C)] showed photomodulation of GABA responses after incubation with MAP20 and illumination with lights at specific wavelengths. The α1ß3(M283C)γ2 receptor showed the greatest photomodulation, which decreased as GABA concentration increased. The location of the mutations that produced photomodulation confirmed that the propofol binding site is located in the ß+α- interface close to the extracellular side of the transmembrane helices. Tethering the photoswitch to cysteines introduced in the positions homologous to ß3M283 in two other subunits (α1W288 and γ2L298) also produced photomodulation, which was not entirely reversible, probably reflecting the different nature of each interface. The results are in agreement with a binding site in the ß+α- interface for the anesthetic propofol.


Subject(s)
Anesthetics, Intravenous/pharmacology , Cell Membrane/metabolism , Light , Oocytes/metabolism , Propofol/pharmacology , Receptors, GABA-A/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/radiation effects , Humans , Oocytes/drug effects , Oocytes/radiation effects , Protein Conformation , Protein Domains , Receptors, GABA-A/chemistry , Receptors, GABA-A/drug effects , Receptors, GABA-A/radiation effects , Xenopus laevis , gamma-Aminobutyric Acid
6.
Altern Ther Health Med ; 30(5): 136-140, 2024 May.
Article in English | MEDLINE | ID: mdl-38743887

ABSTRACT

Objective: To investigate the molecular mechanism of sevoflurane affecting the development of the offspring's nervous system through the GABAAR/Sirt 1 pathway. Methods: Pregnant rats were obtained by mating females and males, and were randomly divided into 3 h sevoflurane (2.3% sevoflurane anesthesia for 3 h), 6 h sevoflurane (2.3% sevoflurane anesthesia for 6 h), Sirt-1 activator-SRT1720 (10 mg/kg SRT1720), 6 h sevoflurane+SRT1720 (10 mg/kg SRT1720) and control groups) group and control group, 31-day-old littermates were taken out and their learning and memory functions were examined by the water maze experiment; the heads were severed to remove the brains, and the kits were used to detect the levels of 5-HT and Ach in the brain tissue; the hippocampal tissues of the littermates were isolated, and neuronal damage in the hippocampal tissues was assessed by Nissen staining; neuronal apoptosis in the hippocampal tissues was detected by TUNEL staining; and GABAAR in the hippocampal tissues was detected by Western blot. GABAAR, Sirt-1, and apoptosis-related proteins (Caspase-3, BCL-2, BAX) in hippocampal tissue. Results: Compared with the control group, the 3 h sevoflurane group and the 6 h sevoflurane group neurons were arranged sparsely, the cells appeared to be swollen, the evasion latency, the apoptosis rate of neurons, the expression of Caspase-3, and BAX increased significantly, and the number of crossing the plateau, the level of 5-HT and Ach in the brain tissues, and the expression of GABAAR, Sirt-1, and BCL-2 were decreased significantly, and the differences existed between the groups (P < .5); compared with the 6 h sevoflurane group, neuronal morphological changes in the hippocampal tissue of the 6 h sevoflurane+SRT1720 group were improved, with a significant decrease in the evasion latency, neuronal apoptosis rate, expression of Caspase-3 and BAX, and a significant increase in the number of traversing platforms, brain tissue 5-HT, Ach level, GABAAR, Sirt-1, and BCL-2 expression (P < .5); compared with the SRT1720 group, the neurons in the 6 h sevoflurane + SRT1720 group were sparsely arranged, with a significant increase in evasion latency, neuronal apoptosis rate, caspase-3, BAX expression, and a significant decrease in the number of traversing platforms, brain tissue 5-HT, Ach level, GABAAR, Sirt-1, and BCL-2 expression (P < .5 ). Conclusion: Sevoflurane can affect the neurological development of rat offspring, which may be related to the inhibition of Sirt-1 expression.


Subject(s)
Sevoflurane , Sirtuin 1 , Sevoflurane/pharmacology , Animals , Sirtuin 1/metabolism , Rats , Female , Pregnancy , Anesthetics, Inhalation/pharmacology , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Male , Rats, Sprague-Dawley , Hippocampus/drug effects , Hippocampus/metabolism , Apoptosis/drug effects , Signal Transduction/drug effects
7.
Acta Med Okayama ; 78(3): 227-235, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38902210

ABSTRACT

Zolpidem, a non-benzodiazepine hypnotic, is primarily used to treat insomnia. In a previous study, pior treatment with non-benzodiazepine receptor agonists was associated with inflammation. The present study aimed to clarify the association between the effects of zolpidem and inflammation in mice treated with lipopolysaccharide (LPS), a known model of inflammation. We assessed the zolpidem-induced loss of righting reflex (LORR) duration 24 h after LPS treatment in mice. Additionally, the expressions of γ-aminobutyric acid (GABA)A receptor subunit and K+-Cl- cotransporter isoform 2 (KCC2) mRNA in the hippocampus and frontal cortex were examined in LPS-treated mice. Pretreatment with LPS was associated with significantly prolonged duration of zolpidem-induced LORR compared to control mice. This effect was significantly attenuated by administering bicuculline, a GABAA receptor antagonist, or flumazenil, a benzodiazepine receptor antagonist, in LPS-treated mice. Compared to controls, LPS-treated mice showed no significant change in the expression of GABAA receptor subunits in the hippocampus or frontal cortex. Bumetanide, an Na+-K+-2Cl- cotransporter isoform 1 blocker, attenuated the extended duration of zolpidem-induced LORR observed in LPS-treated mice. LPS significantly decreased Kcc2 mRNA expression in the hippocampus and the frontal cortex. These findings suggest that inflammation increases zolpidem-induced LORR, possibly through a reduction in KCC2 expression.


Subject(s)
Lipopolysaccharides , Pyridines , Receptors, GABA-A , Reflex, Righting , Symporters , Zolpidem , Animals , Zolpidem/pharmacology , Mice , Pyridines/pharmacology , Male , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Symporters/genetics , Symporters/metabolism , Reflex, Righting/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , K Cl- Cotransporters , Hypnotics and Sedatives/pharmacology , Inflammation/chemically induced , Frontal Lobe/drug effects , Frontal Lobe/metabolism
8.
Int J Urol ; 31(9): 1052-1060, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38884570

ABSTRACT

OBJECTIVES: To investigate roles of brain carbon monoxide (CO), an endogenous gasotransmitter, in regulation of the rat micturition reflex. METHODS: In urethane-anesthetized (0.8 g/kg, ip) male rats, evaluation of urodynamic parameters was started 1 h before intracerebroventricular administration of CORM-3 (CO donor) or ZnPP (non-selective inhibitor of heme oxygenase, a CO producing enzyme) and continued for 2 h after the administration. We also investigated effects of centrally pretreated SR95531 (GABAA receptor antagonist) or SCH50911 (GABAB receptor antagonist) on the CORM-3-induced response. RESULTS: CORM-3 significantly prolonged intercontraction intervals (ICIs) without changing maximal voiding pressure (MVP), while ZnPP significantly shortened ICI and reduced single-voided volume and bladder capacity without affecting MVP, post-voided residual volume, or voiding efficiency. The ZnPP-induced ICI shortening was reversed by CORM-3. The CORM-3-induced ICI prolongation was significantly attenuated by centrally pretreated SR95531 or SCH50911, respectively. CONCLUSIONS: Brain CO can suppress the rat micturition reflex through brain γ-aminobutyric acid (GABA) receptors.


Subject(s)
Brain , Carbon Monoxide , Rats, Sprague-Dawley , Urinary Bladder , Urination , Animals , Male , Urination/drug effects , Rats , Carbon Monoxide/pharmacology , Brain/drug effects , Brain/physiology , Urinary Bladder/drug effects , Urinary Bladder/physiology , Reflex/drug effects , Organometallic Compounds/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Urodynamics/drug effects , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Receptors, GABA/drug effects , Receptors, GABA/metabolism
9.
Arch Pharm (Weinheim) ; 357(10): e2400357, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38943436

ABSTRACT

Anticonvulsant drug discovery has achieved significant progress; however, pharmacotherapy of epilepsy continues to be a challenge for modern medicine and pharmacy. To expand the chemical space of heterocycles as potential antiepileptic agents, herein we report on the synthesis and evaluation of anticonvulsant properties of a series of thiopyrano[2,3-d]thiazoles. The studied heterocycles are characterized by satisfactory drug-likeness and pharmacokinetics properties, calculated in silico using SwissADME. The anticonvulsant activity of thiopyrano[2,3-d]thiazole derivatives was evaluated in vivo using the subcutaneous pentylenetetrazole test. Three hits, that is, compounds 12, 14, and 16, that caused a pronounced anticonvulsant effect were identified. Derivatives 12, 14, and 16 positively affected the latent period of onset of clonic seizures, number of seizures, mortality rate, and duration of the seizure period of animals under experimental conditions. The anticonvulsant properties of compound 14 were equivalent to the effect of the reference drug, sodium valproate. All hit compounds are characterized by satisfying toxicity properties in the human lymphocytes and HEK293 cell line. The most active hit 14 possesses a potential affinity with the GABAA receptor in the molecular docking study and forms a stable complex in the molecular dynamics experiments equal to diazepam. Preliminary SAR results were obtained and discussed based on screening data.


Subject(s)
Anticonvulsants , Molecular Docking Simulation , Seizures , Thiazoles , Anticonvulsants/pharmacology , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Humans , Thiazoles/pharmacology , Thiazoles/chemical synthesis , Thiazoles/chemistry , Animals , Structure-Activity Relationship , Seizures/drug therapy , Seizures/chemically induced , Mice , HEK293 Cells , Molecular Structure , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Male , Pentylenetetrazole , Dose-Response Relationship, Drug
10.
J Headache Pain ; 25(1): 75, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38724972

ABSTRACT

BACKGROUND: GABA, a key inhibitory neurotransmitter, has synaptic and extrasynaptic receptors on the postsynaptic neuron. Background GABA, which spills over from the synaptic cleft, acts on extrasynaptic delta subunit containing GABAA receptors. The role of extrasynaptic GABAergic input in migraine is unknown. We investigated the susceptibility to valid migraine-provoking substances with clinically relevant behavioral readouts in Genetic Absence Epilepsy of Rats Strasbourg (GAERS), in which the GABAergic tonus was altered. Subsequently, we screened relevant GABAergic mechanisms in Wistar rats by pharmacological means to identify the mechanisms. METHODS: Wistar and GAERS rats were administered nitroglycerin (10 mg/kg) or levcromakalim (1 mg/kg). Mechanical allodynia and photophobia were assessed using von Frey monofilaments and a dark-light box. Effects of GAT-1 blocker tiagabine (5 mg/kg), GABAB receptor agonist baclofen (2 mg/kg), synaptic GABAA receptor agonist diazepam (1 mg/kg), extrasynaptic GABAA receptor agonists gaboxadol (4 mg/kg), and muscimol (0.75 mg/kg), T-type calcium channel blocker ethosuximide (100 mg/kg) or synaptic GABAA receptor antagonist flumazenil (15 mg/kg) on levcromakalim-induced migraine phenotype were screened. RESULTS: Unlike Wistar rats, GAERS exhibited no reduction in mechanical pain thresholds or light aversion following nitroglycerin or levcromakalim injection. Ethosuximide did not reverse the resistant phenotype in GAERS, excluding the role of T-type calcium channel dysfunction in this phenomenon. Tiagabine prevented levcromakalim-induced mechanical allodynia in Wistar rats, suggesting a key role in enhanced GABA spillover. Baclofen did not alleviate mechanical allodynia. Diazepam failed to mitigate levcromakalim-induced migraine phenotype. Additionally, the resistant phenotype in GAERS was not affected by flumazenil. Extrasynaptic GABAA receptor agonists gaboxadol and muscimol inhibited periorbital allodynia in Wistar rats. CONCLUSION: Our study introduced a rat strain resistant to migraine-provoking agents and signified a critical involvement of extrasynaptic δGABAergic receptors. Extrasynaptic δ GABAA receptors, by mediating constant background inhibition on the excitability of neurons, stand as a novel drug target with a therapeutic potential in migraine.


Subject(s)
Migraine Disorders , Phenotype , Rats, Wistar , Receptors, GABA-A , Animals , Migraine Disorders/metabolism , Migraine Disorders/drug therapy , Migraine Disorders/physiopathology , Rats , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Male , Disease Models, Animal , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Epilepsy, Absence/drug therapy , Epilepsy, Absence/physiopathology , Nitroglycerin/pharmacology , Nitroglycerin/toxicity , Photophobia/etiology , Photophobia/physiopathology
11.
J Cogn Neurosci ; 35(6): 976-989, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36976900

ABSTRACT

Animals need to cope with abundant sensory information, and one strategy is to selectively direct attention to only the most relevant part of the environment. Although the cortical networks of selective attention have been studied extensively, its underlying neurotransmitter systems, especially the role of the inhibitory neurotransmitter gamma-aminobutyric acid (GABA), remain less well understood. Increased GABAA receptor activity because of administration of benzodiazepines such as lorazepam is known to slow reactions in cognitive tasks. However, there is limited knowledge about GABAergic involvement in selective attention. Particularly, it is unknown whether increased GABAA receptor activity slows the build-up of selectivity or generally widens attentional focus. To address this question, participants (n = 29) received 1 mg lorazepam and placebo (within-subjects, double-blind) and performed an extended version of the flanker task. The spatial distribution of selective attention was studied by systematically manipulating number and position of incongruent flankers; the temporal build-up was characterized using delta plots. An online task version was presented to an independent, unmedicated sample (n = 25) to verify task effects. Under placebo and in the unmedicated sample, only the number of incongruent flankers, but not their position, influenced RTs. Incongruent flankers impaired RTs more strongly under lorazepam than placebo, especially when adjacent to the target. Delta plot analyses of RT showed that this effect persisted even when participants reacted slowly, indicating that lorazepam-induced impairments in selective attention do not result from simply slowed down build-up of selectivity. Instead, our data indicate that increased GABAA receptor activity widens the attentional focus.


Subject(s)
Attention , GABA Modulators , Receptors, GABA-A , Double-Blind Method , Lorazepam/pharmacology , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Humans , Attention/drug effects , Attention/physiology , GABA Modulators/pharmacology
12.
Int J Mol Sci ; 24(17)2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37685955

ABSTRACT

Acid-sensing ion channels (ASICs) are proton-gated ion channels that mediate nociception in the peripheral nervous system and contribute to fear and learning in the central nervous system. Sevanol was reported previously as a naturally-occurring ASIC inhibitor from thyme with favorable analgesic and anti-inflammatory activity. Using electrophysiological methods, we found that in the high micromolar range, the compound effectively inhibited homomeric ASIC1a and, in sub- and low-micromolar ranges, positively modulated the currents of α1ß2γ2 GABAA receptors. Next, we tested the compound in anxiety-related behavior models using a targeted delivery into the hippocampus with parallel electroencephalographic measurements. In the open field, 6 µM sevanol reduced both locomotor and θ-rhythmic activity similar to GABA, suggesting a primary action on the GABAergic system. At 300 µM, sevanol markedly suppressed passive avoidance behavior, implying alterations in conditioned fear memory. The observed effects could be linked to distinct mechanisms involving GABAAR and ASIC1a. These results elaborate the preclinical profile of sevanol as a candidate for drug development and support the role of ASIC channels in fear-related functions of the hippocampus.


Subject(s)
Thymus Plant , Acid Sensing Ion Channels , Fear/drug effects , gamma-Aminobutyric Acid , Hippocampus/drug effects , Receptors, GABA-A/drug effects , Thymus Plant/chemistry
13.
Turk J Med Sci ; 53(5): 1058-1066, 2023.
Article in English | MEDLINE | ID: mdl-38813010

ABSTRACT

Background/aim: Propofol is a positive allosteric modulator of GABAA receptor (GABAAR) and has potent antioxidant activity. The aim of this study was to investigate the effect of propofol on damage to the cerebral cortex and hippocampus in a lithium chloride (LiCl)-pilocarpine animal model of status epilepticus (SE). Materials and methods: Adult male Sprague Dawley rats were injected with LiCl-pilocarpine to induce SE. They were then randomized and injected 30 min later with vehicle saline (SE+saline), propofol (SE+PPF, 50 mg/kg), Diazepam (SE+DZP, 10 mg/kg), Scopolamine (SE+SCOP, 10 mg/kg), or MK-801 (SE+MK-801, 2 mg/kg). Another group of rats received saline only and served as the naïve control (BLK). The levels of superoxide dismutase (SOD), glutathione (GSH) and malondialdehyde (MDA) in the serum, cortex and hippocampus were analyzed 2 and 24 h posttreatment. The degree of tissue damage in the cortex and hippocampus of individual rats was assessed 24 h posttreatment, together with expression of the GABAAR α1 subunit. Results: The propofol group showed reduced levels of tissue damage in the cerebral cortex and hippocampus, decreased levels of MDA, and increased levels of GSH compared to the SE+saline group. No changes in SOD level were observed in serum and tissue samples from the cortex and hippocampus of SE+saline rats. Immunohistochemistry and Western blot assays showed that propofol treatment significantly increased the expression of GABAAR α1 subunit in the cortical and hippocampal tissues of SE rats. Conclusion: Propofol treatment protected against SE-induced tissue injury in the cortex and hippocampus of rats. This was due at least in part to its antioxidant activity and to its induction of GABAAR α1 subunit expression in the brain.


Subject(s)
Disease Models, Animal , Lithium Chloride , Oxidative Stress , Pilocarpine , Propofol , Rats, Sprague-Dawley , Receptors, GABA-A , Status Epilepticus , Animals , Propofol/pharmacology , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Status Epilepticus/drug therapy , Pilocarpine/toxicity , Male , Lithium Chloride/pharmacology , Oxidative Stress/drug effects , Rats , Hippocampus/metabolism , Hippocampus/drug effects , Brain Injuries/metabolism , Brain Injuries/drug therapy , Brain Injuries/chemically induced , Malondialdehyde/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects
14.
J Neurosci ; 41(33): 7148-7159, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34210784

ABSTRACT

Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.


Subject(s)
Diazepam Binding Inhibitor/therapeutic use , GABA-A Receptor Agonists/therapeutic use , Neurons/drug effects , Neuropeptides/therapeutic use , Peptide Fragments/therapeutic use , Receptors, GABA-A/drug effects , Stroke/drug therapy , Adult , Animals , Astrocytes/metabolism , Cortical Spreading Depression/physiology , Diazepam Binding Inhibitor/deficiency , Diazepam Binding Inhibitor/physiology , Drug Implants , Evoked Potentials, Somatosensory , Female , GABA-A Receptor Agonists/pharmacology , Humans , Hydrogels , Infarction, Middle Cerebral Artery/drug therapy , Intracranial Thrombosis/drug therapy , Intracranial Thrombosis/etiology , Light , Mice , Mice, Inbred C57BL , N-Methylaspartate/toxicity , Neurons/physiology , Neuropeptides/deficiency , Neuropeptides/physiology , Patch-Clamp Techniques , Peptide Fragments/deficiency , Peptide Fragments/physiology , Rats , Rose Bengal/radiation effects , Rose Bengal/toxicity , Single-Blind Method , Stroke/etiology
15.
Pharm Biol ; 60(1): 1341-1348, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35840545

ABSTRACT

CONTEXT: The sleep-promoting activity of Nelumbo nucifera Gaertn. (Nymphaeaceae) alkaloids in leaves or seeds are well known. However, the sleep-promoting activity of the lotus rhizome (LE), which is used mainly as food, has not yet been evaluated. OBJECTIVE: We investigated the sleep-promoting activity of LE water extract. MATERIALS AND METHODS: Institute of Cancer Research (ICR) mice (n = 8) were subject to a pentobarbital-induced sleep test to assess changes in sleep latency and duration following the administration of LE (80-150 mg/kg). In addition, electroencephalography analysis was performed to determine the sleep quality after LE treatment as well as the sleep recovery effect of LE using a caffeine-induced insomnia SD rat model. Real-time PCR and western blot analysis were performed to investigate the expression of neurotransmitter receptors, and the GABAA receptor antagonists were used for receptor binding analysis. RESULTS: An oral administration of 150 mg/kg LE significantly increased sleep duration by 24% compared to the control. Furthermore, LE increased nonrapid eye movement (NREM) sleep by increasing theta and delta powers. In the insomnia model, LE increased sleep time by increasing NREM sleep. Moreover, treatment with picrotoxin and flumazenil decreased the sleep time by 33% and 23%, respectively, indicating an involvement of the GABAA receptor in the sleep-enhancing activity of LE. The expression of GABAA receptors and the concentration of GABA in the brain were increased by LE. DISCUSSION AND CONCLUSIONS: The results suggest that the sleep-promoting activity of LE was via the GABAA receptor. Collectively, these data show that LE may promote sleep.


Subject(s)
Lotus , Nelumbo , Plant Extracts , Receptors, GABA-A , Sleep Initiation and Maintenance Disorders , Animals , Mice , Nelumbo/metabolism , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Rhizome/chemistry , Sleep/drug effects , Sleep Initiation and Maintenance Disorders/drug therapy , Water/pharmacology , gamma-Aminobutyric Acid/pharmacology
16.
Mol Pharmacol ; 99(6): 426-434, 2021 06.
Article in English | MEDLINE | ID: mdl-33766924

ABSTRACT

The anesthetic etomidate modulates synaptic α1ß2/3γ2 GABAA receptors via binding sites located in transmembrane ß+/α- interfaces. Various approaches indicate that etomidate binds near ß2/3M286 side chains, including recent cryogenic electron microscopy images in α1ß2γ2L receptors under nonphysiologic conditions with ∼3.5-Å resolution. We hypothesized that substituted cysteine modification and protection experiments using variably sized n-alkyl-methanethiosulfonate (MTS) reagents could precisely estimate the distance between bound etomidate and ß3M286 side chains in activated functional receptors. Using voltage-clamp electrophysiology in Xenopus oocytes expressing α1ß3M286Cγ2L GABAA receptors, we measured functional changes after exposing GABA-activated receptors to n-alkyl-MTS reagents, from methyl-MTS to n-decyl-MTS. Based on previous studies using a large sulfhydryl reagent, we anticipated that cysteine modifications large enough to overlap etomidate sites would cause persistently increased GABA sensitivity and decreased etomidate modulation and that etomidate would hinder these modifications, reducing effects. Based on altered GABA or etomidate sensitivity, ethyl-MTS and larger n-alkyl-MTS reagents modified GABA-activated α1ß3M286Cγ2L GABAA receptors. Receptor modification by n-propyl-MTS or larger reagents caused persistently increased GABA sensitivity and decreased etomidate modulation. Receptor-bound etomidate blocked ß3M286C modification by n-propyl-MTS, n-butyl-MTS, and n-hexyl-MTS. In contrast, GABA sensitivity was unaltered by receptor exposure to methyl-MTS or ethyl-MTS, and ethyl-MTS modification uniquely increased etomidate modulation. These results reveal a "cut-on" between ethyl-MTS and n-propyl-MTS, from which we infer that -S-(n-propyl) is the smallest ß3M286C appendage that overlaps with etomidate sites. Molecular models of the native methionine and -S-ethyl and -S-(n-propyl) modified cysteines suggest that etomidate is located between 1.7 and 3.0 Å from the ß3M286 side chain. SIGNIFICANCE STATEMENT: Precise spatial relationships between drugs and their receptor sites are essential for mechanistic understanding and drug development. This study combined electrophysiology, a cysteine substitution, and n-alkyl-methanethiosulfonate modifiers, creating a precise molecular ruler to estimate the distance between a α1ß3γ2L GABA type A receptor residue and etomidate bound in the transmembrane ß+/α- interface.


Subject(s)
Anesthetics, Intravenous/pharmacology , Cysteine/chemistry , Etomidate/pharmacology , Indicators and Reagents/chemistry , Mesylates/chemistry , Receptors, GABA-A/drug effects , Animals , Female , Humans , Xenopus laevis , gamma-Aminobutyric Acid/pharmacology
17.
J Neurophysiol ; 126(4): 1090-1100, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34406874

ABSTRACT

The general anesthetic etomidate, which acts through γ-aminobutyric acid type A (GABAA) receptors, impairs the formation of new memories under anesthesia. This study addresses the molecular and cellular mechanisms by which this occurs. Here, using a new line of genetically engineered mice carrying the GABAA receptor (GABAAR) ß2-N265M mutation, we tested the roles of receptors that incorporate GABAA receptor ß2 versus ß3 subunits to suppression of long-term potentiation (LTP), a cellular model of learning and memory. We found that brain slices from ß2-N265M mice resisted etomidate suppression of LTP, indicating that the ß2-GABAARs are an essential target in this model. As these receptors are most heavily expressed by interneurons in the hippocampus, this finding supports a role for interneuron modulation in etomidate control of synaptic plasticity. Nevertheless, ß2 subunits are also expressed by pyramidal neurons, so they might also contribute. Therefore, using a previously established line of ß3-N265M mice, we also examined the contributions of ß2- versus ß3-GABAARs to GABAA,slow dendritic inhibition, because dendritic inhibition is particularly well suited to controlling synaptic plasticity. We also examined their roles in long-lasting suppression of population activity through feedforward and feedback inhibition. We found that both ß2- and ß3-GABAARs contribute to GABAA,slow inhibition and that both ß2- and ß3-GABAARs contribute to feedback inhibition, whereas only ß3-GABAARs contribute to feedforward inhibition. We conclude that modulation of ß2-GABAARs is essential to etomidate suppression of LTP. Furthermore, to the extent that this occurs through GABAARs on pyramidal neurons, it is through modulation of feedback inhibition.NEW & NOTEWORTHY Etomidate exerts its anesthetic actions through GABAA receptors. However, the mechanism remains unknown. Here, using a hippocampal brain slice model, we show that ß2-GABAARs are essential to this effect. We also show that these receptors contribute to long-lasting dendritic inhibition in feedback but not feedforward inhibition of pyramidal neurons. These findings hold implications for understanding how anesthetics block memory formation and, more generally, how inhibitory circuits control learning and memory.


Subject(s)
Anesthetics, Intravenous/pharmacology , Etomidate/pharmacology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Neural Inhibition/drug effects , Pyramidal Cells/drug effects , Receptors, GABA-A/drug effects , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic
18.
J Neurophysiol ; 126(4): 1209-1220, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34406887

ABSTRACT

Presympathetic neurons in the paraventricular nucleus of the hypothalamus (PVN) play a key role in cardiovascular regulation. We have previously shown that brain-derived neurotrophic factor (BDNF), acting in the PVN, increases sympathetic activity and blood pressure and serves as a key regulator of stress-induced hypertensive responses. BDNF is known to alter glutamatergic and GABA-ergic signaling broadly in the central nervous system, but whether BDNF has similar actions in the PVN remains to be investigated. Here, we tested the hypothesis that increased BDNF expression in the PVN elevates blood pressure by enhancing N-methyl-d-aspartate (NMDA) receptor (NMDAR)- and inhibiting GABAA receptor (GABAAR)-mediated signaling. Sprague-Dawley rats received bilateral PVN injections of AAV2 viral vectors expressing green fluorescent protein (GFP) or BDNF. Three weeks later, cardiovascular responses to PVN injections of NMDAR and GABAAR agonists and antagonists were recorded under α-chloralose-urethane anesthesia. In addition, expressions of excitatory and inhibitory signaling components in the PVN were assessed using immunofluorescence. Our results showed that NMDAR inhibition led to a greater decrease in blood pressure in the BDNF vs. GFP group, while GABAAR inhibition led to greater increases in blood pressure in the GFP group compared to BDNF. Conversely, GABAAR activation decreased blood pressure significantly more in GFP vs. BDNF rats. In addition, immunoreactivity of NMDAR1 was upregulated, while GABAAR-α1 and K+/Cl- cotransporter 2 were downregulated by BDNF overexpression in the PVN. In summary, our findings indicate that hypertensive actions of BDNF within the PVN are mediated, at least in part, by augmented NMDAR and reduced GABAAR signaling.NEW & NOTEWORTHY We have shown that BDNF, acting in the PVN, elevates blood pressure in part by augmenting NMDA receptor-mediated excitatory input and by diminishing GABAA receptor-mediated inhibitory input to PVN neurons. In addition, we demonstrate that elevated BDNF expression in the PVN upregulates NMDA receptor immunoreactivity and downregulates GABAA receptor as well as KCC2 transporter immunoreactivity.


Subject(s)
Blood Pressure/physiology , Brain-Derived Neurotrophic Factor/metabolism , Electrophysiological Phenomena/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Sympathetic Nervous System/physiology , Animals , Blood Pressure/drug effects , Brain-Derived Neurotrophic Factor/pharmacology , Electrophysiological Phenomena/drug effects , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Sympathetic Nervous System/drug effects
19.
Neurobiol Dis ; 150: 105244, 2021 03.
Article in English | MEDLINE | ID: mdl-33385516

ABSTRACT

There is a growing body of evidence demonstrating the significant involvement of the sigma-1 chaperone protein in the modulation of seizures. Several sigma-1 receptor (Sig1R) ligands have been demonstrated to regulate the seizure threshold in acute and chronic seizure models. However, the mechanism by which Sig1R modulates the excitatory and inhibitory pathways in the brain has not been elucidated. The aim of this study was to compare the susceptibility to seizures of wild type (WT) and Sig1R knockout (Sig1R-/-) mice in intravenous pentylenetetrazol (PTZ) and (+)-bicuculline (BIC) infusion-induced acute seizure and Sig1R antagonist NE-100-induced seizure models. To determine possible molecular mechanisms, we used quantitative PCR, Western blotting and immunohistochemistry to assess the possible involvement of several seizure-related genes and proteins. Peripheral tissue contractile response of WT and Sig1R-/- mice was studied in an isolated vasa deferentia model. The most important finding was the significantly decreased expression of the R2 subunit of the GABA-B receptor in the hippocampus and habenula of Sig1R-/- mice. Our results demonstrated that Sig1R-/- mice have decreased thresholds for PTZ- and BIC-induced tonic seizures. In the NE-100-induced seizure model, Sig1R-/- animals demonstrated lower seizure scores, shorter durations and increased latency times of seizures compared to WT mice. Sig1R-independent activities of NE-100 included downregulation of the gene expression of iNOS and GABA-A γ2 and inhibition of KCl-induced depolarization in both WT and Sig1R-/- animals. In conclusion, the results of this study indicate that the lack of Sig1R resulted in decreased expression of the R2 subunit of the GABA-B receptor and increased susceptibility to seizures. Our results confirm that Sig1R is a significant molecular target for seizure modulation and warrants further investigation for the development of novel anti-seizure drugs.


Subject(s)
Convulsants/toxicity , Habenula/metabolism , Hippocampus/metabolism , Receptors, GABA-B/genetics , Receptors, sigma/genetics , Seizures/genetics , Animals , Anisoles/toxicity , Bicuculline/toxicity , Gene Expression/drug effects , Gene Expression/genetics , Genetic Predisposition to Disease , Habenula/drug effects , Hippocampus/drug effects , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/genetics , Pentylenetetrazole/toxicity , Propylamines/toxicity , Receptors, GABA-A/drug effects , Receptors, GABA-A/genetics , Receptors, GABA-B/metabolism , Seizures/chemically induced , Sigma-1 Receptor
20.
Neurobiol Learn Mem ; 183: 107459, 2021 09.
Article in English | MEDLINE | ID: mdl-34015441

ABSTRACT

Alcohol use disorder (AUD) frequently co-occurs with dissociative disorders and disorders with dissociative symptoms, suggesting a common neurobiological basis. It has been proposed that facilitated information processing under the influence of alcohol, resulting in the formation of dissociated memories, might be an important factor controlling alcohol use. Access to such memories is facilitated under the effect of alcohol, thus further reinforcing alcohol use. To interrogate possible mechanisms associated with these phenotypes, we used a mouse model of dissociative amnesia, combined with a high-alcohol preferring (HAP) model of AUD. Dissociated memory was induced by activation of hippocampal extrasynaptic GABA type A receptor delta subunits (GABAAR-δ), which control tonic inhibition and to which ethanol binds with high affinity. Increased ethanol preference was associated with increased propensity to form dissociated memories dependent on GABAAR-δ in the dorsal hippocampus (DH). Furthermore, the DH level of GABAAR-δ protein, but not mRNA, was increased in HAP mice, and was inversely correlated to the level of miR-365-3p, suggesting an miRNA-mediated post-transcriptional mechanism contributing to elevated GABAAR-δ. The observed changes of DH GABAAR-δ were associated with a severe reduction of excitatory projections stemming from GABAAR-δ-containing pyramidal neurons in the subiculum and terminating in the mammillary body. These results suggest that both molecular and circuit dysfunction involving hippocampal GABAAR-δ receptors might contribute to the co-occurrence of ethanol preference and dissociated information processing.


Subject(s)
Amnesia/metabolism , Central Nervous System Depressants/administration & dosage , Choice Behavior/physiology , Ethanol/administration & dosage , Hippocampus/metabolism , Memory/physiology , Pyramidal Cells/metabolism , Receptors, GABA-A/metabolism , Amnesia/physiopathology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Choice Behavior/drug effects , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Fear , GABA Agonists/pharmacology , Hippocampus/physiopathology , Isoxazoles/pharmacology , Mammillary Bodies/metabolism , Mammillary Bodies/physiopathology , Memory/drug effects , Memory, Episodic , Mice , MicroRNAs/drug effects , MicroRNAs/metabolism , Neural Inhibition , Neural Pathways , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, GABA-A/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL