Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.517
Filter
1.
Nat Commun ; 15(1): 7795, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242530

ABSTRACT

ρ-type γ-aminobutyric acid-A (GABAA) receptors are widely distributed in the retina and brain, and are potential drug targets for the treatment of visual, sleep and cognitive disorders. Endogenous neuroactive steroids including ß-estradiol and pregnenolone sulfate negatively modulate the function of ρ1 GABAA receptors, but their inhibitory mechanisms are not clear. By combining five cryo-EM structures with electrophysiology and molecular dynamics simulations, we characterize binding sites and negative modulation mechanisms of ß-estradiol and pregnenolone sulfate at the human ρ1 GABAA receptor. ß-estradiol binds in a pocket at the interface between extracellular and transmembrane domains, apparently specific to the ρ subfamily, and disturbs allosteric conformational transitions linking GABA binding to pore opening. In contrast, pregnenolone sulfate binds inside the pore to block ion permeation, with a preference for activated structures. These results illuminate contrasting mechanisms of ρ1 inhibition by two different neuroactive steroids, with potential implications for subtype-specific gating and pharmacological design.


Subject(s)
Cryoelectron Microscopy , Estradiol , Molecular Dynamics Simulation , Pregnenolone , Receptors, GABA-A , Humans , Pregnenolone/metabolism , Pregnenolone/pharmacology , Pregnenolone/chemistry , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Binding Sites , Estradiol/metabolism , Estradiol/pharmacology , HEK293 Cells
2.
J Mol Neurosci ; 74(3): 83, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39230641

ABSTRACT

The soy isoflavone daidzin (DZN) has been considered a hopeful bioactive compound having diverse biological activities, including anxiolytic, memory-enhancing, and antiepileptic effects, in experimental animals. However, its sedative and hypnotic effects are yet to be discovered. This study aimed to evaluate its sedative/hypnotic effect on Swiss mice. Additionally, in silico studies were also performed to see the possible molecular mechanisms behind the tested neurological effect. For this, male Swiss albino mice were treated with DZN (5, 10, or 20 mg/kg) intraperitoneally (i.p.) with or without the standard GABAergic medication diazepam (DZP) and/or flumazenil (FLU) and checked for the onset and duration of sleeping time using thiopental sodium-induced as well as DZP-induced sleeping tests. A molecular docking study was also performed to check its interaction capacity with the α1 and ß2 subunits of the GABAA receptor. Findings suggest that DZN dose-dependently and significantly reduced the latency while increasing the duration of sleep in animals. In combination therapy, DZN shows synergistic effects with the DZP-2 and DZP-2 + FLU-0.01 groups, resulting in significantly (p < 0.05) reduced latency and increased sleep duration. Further, molecular docking studies demonstrate that DZN has a strong binding affinity of - 7.2 kcal/mol, which is closer to the standard ligand DZP (- 8.3 kcal/mol) against the GABAA (6X3X) receptor. Molecular dynamic simulations indicated stability and similar binding locations for DZP and DZN with 6X3X. In conclusion, DZN shows sedative effects on Swiss mice, possibly through the GABAA receptor interaction pathway.


Subject(s)
Hypnotics and Sedatives , Molecular Docking Simulation , Receptors, GABA-A , Animals , Receptors, GABA-A/metabolism , Mice , Male , Hypnotics and Sedatives/pharmacology , Sleep/drug effects , Flumazenil/pharmacology , Diazepam/pharmacology , Molecular Dynamics Simulation
3.
Drug Dev Res ; 85(6): e22250, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39154218

ABSTRACT

Insomnia is a sleep disorder in which you have trouble falling and/or staying asleep. This research aims to evaluate the sedative effects of fraxin (FX) on sleeping mice induced by thiopental sodium (TS). In addition, a molecular docking study was conducted to investigate the molecular processes underlying these effects. The study used adult male Swiss albino mice and administered FX (10 and 20 mg/kg, i.p.) and diazepam (DZP) (2 mg/kg) either separately or in combination within the different groups to examine their modulatory effects. After a period of 30 min, the mice that had been treated were administered (TS: 20 mg/kg, i.p.) to induce sleep. The onset of sleep for the mice and the length of their sleep were manually recorded. Additionally, a computational analysis was conducted to predict the role of gamma-aminobutyric acid (GABA) receptors in the sleep process and evaluate their pharmacokinetics and toxicity. The outcomes indicated that FX extended the length of sleep and reduced the time it took to fall asleep. When the combined treatment of FX and DZP showed synergistic sedative action. Also, FX had a binding affinity of -7.2 kcal/mol, while DZP showed -8.4 kcal/mol. The pharmacokinetic investigation of FX demonstrated favorable drug-likeness and strong pharmacokinetic characteristics. Ultimately, FX demonstrated a strong sedative impact in the mouse model, likely via interacting with the GABAA receptor pathways.


Subject(s)
Diazepam , Hypnotics and Sedatives , Molecular Docking Simulation , Sleep , Animals , Male , Mice , Hypnotics and Sedatives/pharmacology , Diazepam/pharmacology , Sleep/drug effects , Receptors, GABA/metabolism , Receptors, GABA-A/metabolism
4.
Development ; 151(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39190555

ABSTRACT

Terminal selectors are transcription factors that control neuronal identity by regulating expression of key effector molecules, such as neurotransmitter biosynthesis proteins and ion channels. Whether and how terminal selectors control neuronal connectivity is poorly understood. Here, we report that UNC-30 (PITX2/3), the terminal selector of GABA nerve cord motor neurons in Caenorhabditis elegans, is required for neurotransmitter receptor clustering, a hallmark of postsynaptic differentiation. Animals lacking unc-30 or madd-4B, the short isoform of the motor neuron-secreted synapse organizer madd-4 (punctin/ADAMTSL), display severe GABA receptor type A (GABAAR) clustering defects in postsynaptic muscle cells. Mechanistically, UNC-30 acts directly to induce and maintain transcription of madd-4B and GABA biosynthesis genes (e.g. unc-25/GAD, unc-47/VGAT). Hence, UNC-30 controls GABAA receptor clustering in postsynaptic muscle cells and GABA biosynthesis in presynaptic cells, transcriptionally coordinating two crucial processes for GABA neurotransmission. Further, we uncover multiple target genes and a dual role for UNC-30 as both an activator and a repressor of gene transcription. Our findings on UNC-30 function may contribute to our molecular understanding of human conditions, such as Axenfeld-Rieger syndrome, caused by PITX2 and PITX3 gene variants.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Motor Neurons , Transcription Factors , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , gamma-Aminobutyric Acid/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Motor Neurons/metabolism , Nerve Tissue Proteins , Neurotransmitter Agents/metabolism , Receptors, GABA/metabolism , Receptors, GABA/genetics , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Synapses/metabolism , Synaptic Transmission , Transcription Factors/metabolism , Transcription Factors/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
5.
J Physiol ; 602(17): 4195-4213, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39141819

ABSTRACT

The subiculum is a key region of the brain involved in the initiation of pathological activity in temporal lobe epilepsy, and local GABAergic inhibition is essential to prevent subicular-originated epileptiform discharges. Subicular pyramidal cells may be easily distinguished into two classes based on their different firing patterns. Here, we have compared the strength of the GABAa receptor-mediated inhibitory postsynaptic currents received by regular- vs. burst-firing subicular neurons and their dynamic modulation by the activation of µ opioid receptors. We have taken advantage of the sequential re-patching of the same cell to initially classify pyramidal neurons according to their firing patters, and then to measure GABAergic events triggered by the optogenetic stimulation of parvalbumin- and somatostatin-expressing interneurons. Activation of parvalbumin-expressing cells generated larger responses in postsynaptic burst-firing neurons whereas the opposite was observed for currents evoked by the stimulation of somatostatin-expressing interneurons. In all cases, events depended critically on ω-agatoxin IVA- but not on ω-conotoxin GVIA-sensitive calcium channels. Optogenetic GABAergic input originating from both parvalbumin- and somatostatin-expressing cells was reduced in amplitude following the exposure to a µ opioid receptor agonist. The kinetics of this pharmacological sensitivity was different in regular- vs. burst-firing neurons, but only when responses were evoked by the activation of parvalbumin-expressing neurons, whereas no differences were observed when somatostatin-expressing cells were stimulated. In conclusion, our results show that a high degree of complexity regulates the organizing principles of subicular GABAergic inhibition, with the interaction of pre- and postsynaptic diversity at multiple levels. KEY POINTS: Optogenetic stimulation of parvalbumin- and somatostatin-expressing interneurons (PVs and SOMs) triggers inhibitory postsynaptic currents (IPSCs) in both regular- and burst-firing (RFs and BFs) subicular pyramidal cells. The amplitude of optogenetically evoked IPSCs from PVs (PV-opto IPSCs) is larger in BFs whereas IPSCs generated by the light activation of SOMs (SOM-opto IPSCs) are larger in RFs. Both PV- and SOM-opto IPSCs critically depend on ω-agatoxin IVA-sensitive P/Q type voltage-gated calcium channels, whereas no major effects are observed following exposure to ω-conotoxin GVIA, suggesting no significant involvement of N-type channels. The amplitude of both PV- and SOM-opto IPSCs is reduced by the probable pharmacological activation of presynaptic µ opioid receptors, with a faster kinetics of the effect observed in PV-opto IPSCs from RFs vs. BFs, but not in SOM-opto IPSCs. These results help us understand the complex interactions between different layers of diversity regulating GABAergic input onto subicular microcircuits.


Subject(s)
Parvalbumins , Pyramidal Cells , Somatostatin , Animals , Pyramidal Cells/physiology , Mice , Somatostatin/metabolism , Parvalbumins/metabolism , Interneurons/physiology , Inhibitory Postsynaptic Potentials , Male , GABAergic Neurons/physiology , GABAergic Neurons/metabolism , Hippocampus/physiology , Hippocampus/cytology , Optogenetics , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/physiology , Mice, Inbred C57BL , Female , Receptors, GABA-A/metabolism , Receptors, GABA-A/physiology
6.
PLoS One ; 19(8): e0307668, 2024.
Article in English | MEDLINE | ID: mdl-39186592

ABSTRACT

The majority of somatosensory DRG neurons express GABAA receptors (GABAAR) and depolarise in response to its activation based on the high intracellular chloride concentration maintained by the Na-K-Cl cotransporter type 1 (NKCC1). The translation of this response to peripheral nerve terminals in people is so far unclear. We show here that GABA (EC50 = 16.67µM) acting via GABAAR produces an influx of extracellular calcium in approximately 20% (336/1720) of isolated mouse DRG neurons. In contrast, upon injection into forearm skin of healthy volunteers GABA (1mM, 100µl) did not induce any overt sensations nor a specific flare response and did not sensitize C-nociceptors to slow depolarizing electrical sinusoidal stimuli. Block of the inward chloride transporter NKCC1 by furosemide (1mg/100µl) did not reduce electrically evoked pain ratings nor did repetitive GABA stimulation in combination with an inhibited NKCC1 driven chloride replenishment by furosemide. Finally, we generated a sustained period of C-fiber firing by iontophoretically delivering codeine or histamine to induce tonic itch. Neither the intensity nor the duration of histamine or codeine itch was affected by prior injection of furosemide. We conclude that although GABA can evoke calcium transients in a proportion of isolated mouse DRG neurons, it does not induce or modify pain or itch ratings in healthy human skin even when chloride gradients are altered by inhibition of the sodium coupled NKCC1 transporter.


Subject(s)
Furosemide , Ganglia, Spinal , Healthy Volunteers , Hyperalgesia , Solute Carrier Family 12, Member 2 , gamma-Aminobutyric Acid , Humans , Animals , Mice , gamma-Aminobutyric Acid/metabolism , Male , Adult , Furosemide/pharmacology , Solute Carrier Family 12, Member 2/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Hyperalgesia/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Female , Neurons/metabolism , Neurons/drug effects , Acute Pain/metabolism , Acute Pain/physiopathology , Calcium/metabolism , Receptors, GABA-A/metabolism , Pruritus/chemically induced , Pruritus/metabolism , Pruritus/physiopathology , Young Adult
7.
Proc Natl Acad Sci U S A ; 121(33): e2400420121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39106304

ABSTRACT

Brain rhythms provide the timing for recruitment of brain activity required for linking together neuronal ensembles engaged in specific tasks. The γ-oscillations (30 to 120 Hz) orchestrate neuronal circuits underlying cognitive processes and working memory. These oscillations are reduced in numerous neurological and psychiatric disorders, including early cognitive decline in Alzheimer's disease (AD). Here, we report on a potent brain-permeable small molecule, DDL-920 that increases γ-oscillations and improves cognition/memory in a mouse model of AD, thus showing promise as a class of therapeutics for AD. We employed anatomical, in vitro and in vivo electrophysiological, and behavioral methods to examine the effects of our lead therapeutic candidate small molecule. As a novel in central nervous system pharmacotherapy, our lead molecule acts as a potent, efficacious, and selective negative allosteric modulator of the γ-aminobutyric acid type A receptors most likely assembled from α1ß2δ subunits. These receptors, identified through anatomical and pharmacological means, underlie the tonic inhibition of parvalbumin (PV) expressing interneurons (PV+INs) critically involved in the generation of γ-oscillations. When orally administered twice daily for 2 wk, DDL-920 restored the cognitive/memory impairments of 3- to 4-mo-old AD model mice as measured by their performance in the Barnes maze. Our approach is unique as it is meant to enhance cognitive performance and working memory in a state-dependent manner by engaging and amplifying the brain's endogenous γ-oscillations through enhancing the function of PV+INs.


Subject(s)
Alzheimer Disease , Cognition , Disease Models, Animal , Gamma Rhythm , Animals , Alzheimer Disease/drug therapy , Mice , Cognition/drug effects , Gamma Rhythm/drug effects , Memory/drug effects , Receptors, GABA-A/metabolism , Mice, Transgenic , Humans , Male , Memory, Short-Term/drug effects , Brain/drug effects , Brain/metabolism , Alanine/analogs & derivatives , Azepines
8.
J Med Chem ; 67(14): 12349-12365, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39013072

ABSTRACT

Multitarget strategies are essential in addressing complex diseases, yet developing multitarget-directed ligands (MTDLs) is particularly challenging when aiming to engage multiple therapeutic targets across different tissues. Here, we present a molecular transformer strategy, enhancing traditional MTDLs. By utilizing esterase-driven hydrolysis, this approach mimics the adaptive nature of transformers for enabling molecules to modify their pharmacological effects in response to the biological milieu. By virtual screening and biological evaluation, we identified KGP-25, a novel compound initially targeting the voltage-gated sodium channel 1.8 (Nav1.8) in the peripheral nervous system (PNS) for analgesia, and later the γ-aminobutyric acid subtype A receptor (GABAA) in the central nervous system (CNS) for general anesthesia. Our findings confirm KGP-25's dual efficacy in cellular and animal models, effectively reducing opioid-related side effects. This study validates the molecular transformer approach in drug design and highlights its potential to overcome the limitations of conventional MTDLs, paving new avenues in innovative therapeutic strategies.


Subject(s)
Analgesics , Esterases , Ligands , Analgesics/pharmacology , Analgesics/chemistry , Animals , Humans , Esterases/metabolism , Anesthetics/pharmacology , Anesthetics/chemistry , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Drug Design , Mice , Male
9.
J Neurophysiol ; 132(2): 501-513, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38958282

ABSTRACT

Neuromodulation in the retina is crucial for effective processing of retinal signal at different levels of illuminance. Intrinsically photosensitive retinal ganglion cells (ipRGCs), the neurons that drive nonimage-forming visual functions, express a variety of neuromodulatory receptors that tune intrinsic excitability as well as synaptic inputs. Past research has examined actions of neuromodulators on light responsiveness of ipRGCs, but less is known about how neuromodulation affects synaptic currents in ipRGCs. To better understand how neuromodulators affect synaptic processing in ipRGC, we examine actions of opioid and dopamine agonists have on inhibitory synaptic currents in ipRGCs. Although µ-opioid receptor (MOR) activation had no effect on γ-aminobutyric acid (GABA) currents, dopamine [via the D1-type dopamine receptor (D1R)]) amplified GABAergic currents in a subset of ipRGCs. Furthermore, this D1R-mediated facilitation of the GABA conductance in ipRGCs was mediated by a cAMP/PKA-dependent mechanism. Taken together, these findings reinforce the idea that dopamine's modulatory role in retinal adaptation affects both nonimage-forming and image-forming visual functions.NEW & NOTEWORTHY Neuromodulators such as dopamine are important regulators of retinal function. Here, we demonstrate that dopamine increases inhibitory inputs to intrinsically photosensitive retinal ganglion cells (ipRGCs), in addition to its previously established effect on intrinsic light responsiveness. This indicates that dopamine, in addition to its ability to intrinsically modulate ipRGC activity, can also affect synaptic inputs to ipRGCs, thereby tuning retina circuits involved in nonimage-forming visual functions.


Subject(s)
Dopamine , Receptors, GABA-A , Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Dopamine/metabolism , Dopamine/pharmacology , Receptors, GABA-A/metabolism , Mice , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/antagonists & inhibitors , Mice, Inbred C57BL , Receptors, Opioid, mu/metabolism , Male , Inhibitory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/drug effects , Female , Dopamine Agonists/pharmacology
10.
Eur J Med Chem ; 276: 116602, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38971049

ABSTRACT

Zuranolone (SAGE-217) is a neuroactive steroid (γ-aminobutyric acid)A (GABAA) receptor positive allosteric modulator (PAM) as the first oral drug approved by the FDA in 2023, which is used to treat patients with postpartum depression (PPD). SAGE-217 has a "black box" warning with impairing ability to drive or engage in other potentially hazardous activities. In addition, SAGE-217 can cause CNS depressant effects such as somnolence and confusion, suicidal thoughts and behavior and embryo-fetal toxicity. Based on the structure-activity relationship (SAR) of SAGE-217, a total of 28 neuroactive steroids with novel pharmacophore at C-21 modulated SAGE-217 derivatives were designed and synthesized. The biological activities were evaluated by both synaptic α1ß2γ2 GABAA receptor and extrasynaptic α4ß3δ GABAA receptor cell assays. The optimal compound S28 exhibited much more potent potency and similar efficacy at extrasynaptic GABAA receptor than SAGE-217. Different from above, compound S28 exhibited similar potency and lower efficacy at synaptic GABAA receptor than SAGE-217, which were consistent with the analysis of molecular docking and dynamics simulation results. The appropriate lower efficacy at synaptic GABAA receptor of compound S28 might contribute to reduce the side effects of excessive sedation. Furthermore, compound S28 was demonstrated to have excellent in vivo pharmacokinetic (PK) parameters, robust in vivo pharmacodynamic (PD) effects and good safety profiles. Therefore, compound S28 represents a potentially promising treatment of PPD candidate that warrants further investigation.


Subject(s)
Receptors, GABA-A , Receptors, GABA-A/metabolism , Structure-Activity Relationship , Humans , Animals , Molecular Structure , Dose-Response Relationship, Drug , Mice , Neurosteroids/pharmacology , Neurosteroids/metabolism , Neurosteroids/chemical synthesis , Neurosteroids/chemistry , Molecular Docking Simulation , Allosteric Regulation/drug effects , Male , GABA Modulators/pharmacology , GABA Modulators/chemical synthesis , GABA Modulators/chemistry , Pharmacophore , Pregnanolone , Pyrazoles
11.
Thorac Cancer ; 15(24): 1822-1824, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38984426

ABSTRACT

Anti-gamma-aminobutyric acid receptor type A (GABAA) encephalitis is a relatively rare autoimmune encephalitis, and often associated with thymoma. Here, a 44-year-old female was diagnosed as having a thymoma with autoimmune encephalitis. At 4-month follow-up she was without recurrence of symptoms after treatment with methylprednisolone pulse therapy and immunotherapy. This case report provides a reference for the identification of this type of paraneoplastic encephalitis and for a therapeutic schedule. It also highlights that conservative treatment may be effective for patients with a tumor and GABAA encephalitis.


Subject(s)
Thymoma , Humans , Female , Adult , Thymoma/complications , Thymoma/drug therapy , Encephalitis/drug therapy , Conservative Treatment/methods , Receptors, GABA-A/metabolism , Thymus Neoplasms/complications , Thymus Neoplasms/drug therapy , Thymus Neoplasms/pathology
12.
Pestic Biochem Physiol ; 203: 105972, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39084765

ABSTRACT

The Drosophila melanogaster MD-RR strain contains an Rdl mutation (A301S) resulting in resistance to several insecticide classes viz. phenyl pyrazoles (e.g., fipronil), cyclodienes (e.g., dieldrin), and chlorinated aliphatic hydrocarbons (e.g., lindane). Fitness costs are commonly observed with resistant insect populations as side effects of the genetic change conferring the resistant phenotype. Because of fitness costs, reversion from the resistant to susceptible genotype and phenotype is common. However, the Rdl genotype in D. melanogaster appears to allow the flies to maintain the resistant genotype/phenotype without selective pressure and with minimal fitness costs. We provide evidence that compensation for the Rdl mutation influences the cholinergic system, where an increase in acetylcholinesterase gene expression and enzyme activity results in neurophysiological changes and cross resistance to a carbamate insecticide (propoxur oral resistance ratio (RR) of 63) and an organophosphate insecticide (dichlorvos oral RR of 7). Such cross resistance was not previously reported with the initial collection and testing of this strain. In addition to acetylcholinesterase, the Rdl mutation influences the expression of the muscarinic acetylcholine receptor subtype-B, resulting in resistance to non-selective muscarinic compounds (pilocarpine and atropine). Collectively, these results indicate that the Rdl mutation (A301S) at GABA-gated ionophore complex influences the physiology of the cholinergic system, leading to resistance to established insecticide classes. Additionally, this mutation may impact the effectiveness of insecticides targeting novel sites, like muscarinic receptors.


Subject(s)
Acetylcholinesterase , Chloride Channels , Drosophila Proteins , Drosophila melanogaster , Insecticide Resistance , Receptors, GABA-A , Animals , Acetylcholinesterase/genetics , Acetylcholinesterase/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/drug effects , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Insecticide Resistance/genetics , Insecticides , Mutation , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism
13.
Neurochem Int ; 178: 105804, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002759

ABSTRACT

Anxiety is a commonly prevailing psychological disorder that requires effective treatment, wherein phytopharmaceuticals and nutraceuticals could offer a desirable therapeutic profile. Hybanthus enneaspermus (L.) F. Muell. is a powerful medicinal herb, reportedly effective against several ailments, including psychological disorders. The current research envisaged evaluating the anxiolytic potential of the ethanolic extract of Hybanthus enneaspermus (EEHE) and its toluene insoluble biofraction (ITHE) employing experimental and computational approaches. Elevated Plus Maze, Light and Dark Transition, Mirror Chamber, Hole board and Open field tests were used as screening models to assess the antianxiety potential of 100, 200 and 400 mg/kg body weight of EEHE and ITHE in rats subjected to social isolation, using Diazepam as standard. The brains of rats exhibiting significant anxiolytic activity were dissected for histopathological and biochemical studies. Antioxidant enzymes like catalase, superoxide dismutase, glutathione-S-transferase, glutathione peroxidase, glutathione reductase; and neurotransmitters viz. monoamines (serotonin, noradrenaline, dopamine), Gamma-aminobutyric acid (GABA), and glutamate were quantified in the different regions of rats' brain (cortex, hippocampus, pons, medulla oblongata, cerebellum). Chromatographic techniques were used to isolate phytoconstituents from the fraction exhibiting significant activity that were characterized by spectroscopic methods and subjected to in silico molecular docking. ITHE at 400 mg/kg body weight significantly mitigated anxiety in all the screening models (p < 0.05), reduced the inflammatory vacuoles and necrosis (p < 0.05) and potentiated the antioxidant enzymes (p < 0.05). It enhanced the monoamines and GABA levels while attenuating glutamate levels (p < 0.01) in the brain. Three significant flavonoids viz. Quercitrin, Rutin and Hesperidin were isolated from ITHE. In silico docking studies of these flavonoids revealed that the compounds exhibited substantial binding to the GABAA receptor. ITHE displayed a promising pharmacological profile in combating anxiety and modulating oxidative stress, attributing its therapeutic virtues to the flavonoids present.


Subject(s)
Anti-Anxiety Agents , Anxiety , Plant Extracts , Rats, Wistar , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/isolation & purification , Anti-Anxiety Agents/therapeutic use , Anti-Anxiety Agents/chemistry , Rats , Plant Extracts/pharmacology , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Plant Extracts/chemistry , Anxiety/drug therapy , Anxiety/metabolism , Male , Molecular Docking Simulation , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Brain/drug effects , Brain/metabolism , Maze Learning/drug effects
14.
Proc Natl Acad Sci U S A ; 121(31): e2400339121, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39047036

ABSTRACT

The mammalian circadian clock located in the suprachiasmatic nucleus (SCN) produces robust daily rhythms including rest-wake. SCN neurons synthesize and respond to γ-aminobutyric acid (GABA), but its role remains unresolved. We tested the hypothesis that γ2- and δ-subunits of the GABAA receptor in the SCN differ in their regulation of synchrony among circadian cells. We used two approaches: 1) shRNA to knock-down (KD) the expression of either γ2 or δ subunits in the SCN or 2) knock-in mice harboring a point mutation in the M2 domains of the endogenous GABAA γ2 or δ subunits. KD of either γ2 or δ subunits in the SCN increased daytime running and reduced nocturnal running by reducing their circadian amplitude by a third. Similarly, δ subunit knock-in mice showed decreased circadian amplitude, increased duration of daily activity, and decreased total daily activity. Reduction, or mutation of either γ2 or δ subunits halved the synchrony among, and amplitude of, circadian SCN cells as measured by firing rate or expression of the PERIOD2 protein, in vitro. Surprisingly, overexpression of the γ2 subunit rescued these phenotypes following KD or mutation of the δ subunit, and overexpression of the δ subunit rescued deficiencies due to γ2 subunit KD or mutation. We conclude that γ2 and δ GABAA receptor subunits play similar roles in maintaining circadian synchrony in the SCN and amplitude of daily rest-wake rhythms, but that modulation of their relative densities can change the duration and amplitude of daily activities.


Subject(s)
Circadian Rhythm , Receptors, GABA-A , Suprachiasmatic Nucleus , Animals , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Circadian Rhythm/physiology , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/physiology , Mice , Male , Wakefulness/physiology , Wakefulness/genetics , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology
15.
Elife ; 132024 Jul 04.
Article in English | MEDLINE | ID: mdl-38963323

ABSTRACT

Protein homeostasis (proteostasis) deficiency is an important contributing factor to neurological and metabolic diseases. However, how the proteostasis network orchestrates the folding and assembly of multi-subunit membrane proteins is poorly understood. Previous proteomics studies identified Hsp47 (Gene: SERPINH1), a heat shock protein in the endoplasmic reticulum lumen, as the most enriched interacting chaperone for gamma-aminobutyric acid type A (GABAA) receptors. Here, we show that Hsp47 enhances the functional surface expression of GABAA receptors in rat neurons and human HEK293T cells. Furthermore, molecular mechanism study demonstrates that Hsp47 acts after BiP (Gene: HSPA5) and preferentially binds the folded conformation of GABAA receptors without inducing the unfolded protein response in HEK293T cells. Therefore, Hsp47 promotes the subunit-subunit interaction, the receptor assembly process, and the anterograde trafficking of GABAA receptors. Overexpressing Hsp47 is sufficient to correct the surface expression and function of epilepsy-associated GABAA receptor variants in HEK293T cells. Hsp47 also promotes the surface trafficking of other Cys-loop receptors, including nicotinic acetylcholine receptors and serotonin type 3 receptors in HEK293T cells. Therefore, in addition to its known function as a collagen chaperone, this work establishes that Hsp47 plays a critical and general role in the maturation of multi-subunit Cys-loop neuroreceptors.


Subject(s)
Endoplasmic Reticulum , Receptors, GABA-A , Animals , Humans , Rats , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , HEK293 Cells , Neurons/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics
16.
Sci Rep ; 14(1): 17461, 2024 07 29.
Article in English | MEDLINE | ID: mdl-39075105

ABSTRACT

GABAergic transmission is influenced by post-translational modifications, like phosphorylation, impacting channel conductance, allosteric modulator sensitivity, and membrane trafficking. O-GlcNAcylation is a post-translational modification involving the O-linked attachment of ß-N-acetylglucosamine on serine/threonine residues. Previously we reported an acute increase in O-GlcNAcylation elicits a long-term depression of evoked GABAAR inhibitory postsynaptic currents (eIPSCs) onto hippocampal principal cells. Importantly, O-GlcNAcylation and phosphorylation can co-occur or compete for the same residue; whether they interact in modulating GABAergic IPSCs is unknown. We tested this by recording IPSCs from hippocampal principal cells and pharmacologically increased O-GlcNAcylation, before or after increasing serine phosphorylation using the adenylate cyclase activator, forskolin. Although forskolin had no significant effect on baseline eIPSC amplitude, we found that a prior increase in O-GlcNAcylation unmasks a forskolin-dependent increase in eIPSC amplitude, reversing the O-GlcNAc-induced eIPSC depression. Inhibition of adenylate cyclase or protein kinase A did not prevent the potentiating effect of forskolin, indicating serine phosphorylation is not the mechanism. Surprisingly, increasing O-GlcNAcylation also unmasked a potentiating effect of the neurosteroids 5α-pregnane-3α,21-diol-20-one (THDOC) and progesterone on eIPSC amplitude in about half of the recorded cells, mimicking forskolin. Our findings show that under conditions of heightened O-GlcNAcylation, the neurosteroid site on synaptic GABAARs is possibly accessible to agonists, permitting strengthening of synaptic inhibition.


Subject(s)
Colforsin , Hippocampus , Receptors, GABA-A , Synapses , Colforsin/pharmacology , Animals , Hippocampus/metabolism , Hippocampus/drug effects , Receptors, GABA-A/metabolism , Synapses/metabolism , Synapses/drug effects , Phosphorylation/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Rats , Neurosteroids/metabolism , Neurosteroids/pharmacology , Protein Processing, Post-Translational/drug effects , Male , Synaptic Transmission/drug effects , Rats, Sprague-Dawley
17.
Neuropharmacology ; 258: 110055, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-38950692

ABSTRACT

Sleep disturbances and persistent pain conditions are public health challenges worldwide. Although it is well-known that sleep deficit increases pain sensitivity, the underlying mechanisms remain elusive. We have recently demonstrated the involvement of nucleus accumbens (NAc) and anterior cingulate cortex (ACC) in the pronociceptive effect of sleep restriction. In this study, we found that sleep restriction increases c-Fos expression in NAc and ACC, suggesting hyperactivation of these regions during prolonged wakefulness in male Wistar rats. Blocking adenosine A2A receptors in the NAc or GABAA receptors in the ventral tegmental area (VTA), dorsal raphe nucleus (DRN), or locus coeruleus (LC) effectively mitigated the pronociceptive effect of sleep restriction. In contrast, the blockade of GABAA receptors in each of these nuclei only transiently reduced carrageenan-induced hyperalgesia. Pharmacological activation of dopamine D2, serotonin 5-HT1A and noradrenaline alpha-2 receptors within the ACC also prevented the pronociceptive effect of sleep restriction. While pharmacological inhibition of these same monoaminergic receptors in the ACC restored the pronociceptive effect which had been prevented by the GABAergic disinhibition of the of the VTA, DRN or LC. Overall, these findings suggest that the pronociceptive effect of sleep restriction relies on increased adenosinergic activity on NAc, heightened GABAergic activity in VTA, DRN, and LC, and reduced inhibitory monoaminergic activity on ACC. These findings advance our understanding of the interplay between sleep and pain, shedding light on potential NAc-brainstem-ACC mechanisms that could mediate increased pain sensitivity under conditions of sleep impairment.


Subject(s)
Nucleus Accumbens , Rats, Wistar , Sleep Deprivation , Ventral Tegmental Area , Animals , Male , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Rats , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Receptor, Adenosine A2A/metabolism , Hyperalgesia/metabolism , Dorsal Raphe Nucleus/metabolism , Dorsal Raphe Nucleus/drug effects , Gyrus Cinguli/metabolism , Gyrus Cinguli/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Brain Stem/metabolism , Brain Stem/drug effects , Locus Coeruleus/metabolism , Locus Coeruleus/drug effects , Carrageenan , Receptors, GABA-A/metabolism , Receptors, Dopamine D2/metabolism , Adenosine A2 Receptor Antagonists/pharmacology
18.
Neurotoxicology ; 104: 85-94, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39079579

ABSTRACT

Exposure to inorganic arsenic (iAs) detrimentally affects the structure and function of the central nervous system. In-utero and postnatal exposure to iAs has been connected to adverse effects on cognitive development. Therefore, this investigation explores neurobehavioral and neurochemical effects of 0.05 and 0.10 mg/L iAs exposure during gestation and lactation periods on 90-day-old female offspring rats. The assessment of anxiety- and depressive-like behaviors was conducted through the application of an elevated plus maze and a forced swim test. The neurochemical changes were evaluated in the prefrontal cortex (PFC) through the determination of enzyme activities and α1 GABAA subunit expression levels. Our findings revealed a notable impact of iAs exposure on anxiety and the induction of depressive-like behavior in 90-day-old female offspring. Furthermore, the antioxidant status within the PFC exhibited discernible alterations in exposed rats. Notably, the activities of acetylcholinesterase and glutamate pyruvate transaminase demonstrated an increase, while glutamate oxaloacetate transaminase activity displayed a decrease within the PFC due to the iAs treatment. Additionally, a distinct downregulation in the mRNA expression of the α1GABAA receptor was observed in this neuronal region. These findings strongly suggest that iAs exposure during early stages of rat development causes significant modifications in brain oxidative stress markers and perturbs the activity of enzymes associated with cholinergic and glutamatergic systems. In parallel, it elicits a discernible reduction in the level of GABA receptors within the PFC. These molecular alterations may play a role in the diminished anxiety levels and the depressive-like behavior outlined in the current investigation.


Subject(s)
Anxiety , Depression , Prefrontal Cortex , Prenatal Exposure Delayed Effects , Rats, Wistar , Animals , Female , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Anxiety/chemically induced , Prenatal Exposure Delayed Effects/chemically induced , Depression/chemically induced , Depression/metabolism , Pregnancy , Rats , Receptors, GABA-A/metabolism , Oxidative Stress/drug effects , Behavior, Animal/drug effects , Arsenic/toxicity , Male
19.
Biomed Res ; 45(4): 151-161, 2024.
Article in English | MEDLINE | ID: mdl-39010191

ABSTRACT

Linalool and linalyl acetate are major components of lavender essential oil. These substances possess many biological activities, such as anti-inflammatory activity, analgesic and anxiolytic effects, and anticonvulsant properties, and they also induce modulation of neuronal activity in the autonomic nervous system. However, there are no reports of the direct effects of linalool on respiratory activity. In the present study, we analyzed the effects of linalool and linalyl acetate on central respiratory activity in the brainstem-spinal cord preparation isolated from newborn rats. Linalool dose-dependently decreased the rate of respiratory activity. This effect was reversed by bicuculline, suggesting that linalool enhanced inhibitory synaptic connections via GABAA receptors. In addition, linalool reduced the coefficient of variation of inspiratory burst intervals and thus could work to stabilize the respiratory rhythm. Linalyl acetate did not cause inhibitory effects as observed in linalool treatment. Linalool depressed burst activity of pre-inspiratory neurons in the medullary respiratory networks and increased the amplitude of inspiratory inhibitory postsynaptic potentials of pre-inspiratory neurons. We concluded that linalool caused inhibitory effects on respiratory rhythm generation mainly through activation of presynaptic GABAA receptors of pre-inspiratory neurons.


Subject(s)
Acyclic Monoterpenes , Animals, Newborn , Brain Stem , Monoterpenes , Neurons , Spinal Cord , Animals , Acyclic Monoterpenes/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Rats , Monoterpenes/pharmacology , Neurons/drug effects , Neurons/metabolism , Brain Stem/drug effects , Brain Stem/physiology , Receptors, GABA-A/metabolism , Respiration/drug effects , Bicuculline/pharmacology
20.
Biochem Pharmacol ; 227: 116449, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39053637

ABSTRACT

Mutations in the γ-amino butyric acid type A (GABAA) receptor γ2 subunit gene, GABRG2, have been associated with refractory epilepsy. Increasing evidence indicates that suberoylanilide hydroxamic acid (SAHA), a broad-spectrum histone acetyltransferases (HDACs) inhibitor, can inhibit seizure onset. However, the mechanisms involved remains unknown. The present study aimed to explore the anti-epileptic effect and underlying mechanisms of SAHA in the treatment of refractory epilepsy induced by GABRG2 mutation. In the zebrafish line expressing human mutant GABRG2(F343L), Tg(hGABRG2F343L), SAHA was found to reduce seizure onset, swimming activity, and neuronal activity. In both Tg(hGABRG2F343L) zebrafish and HEK293T cells transfected with GABAA receptor subunits, SAHA could improve the pan-acetylation level and reduce the expression of HDAC1/10. The decreased expressions of GABAA receptor subunits could be rescued by SAHA treatment both in vivo and in vitro, which might be the result of increased gene transcription and protein trafficking. The up-regulated acetylation of histone H3 and H4 as well as Bip expression might be involved in the process. Taken together, our data proved that both histone and non-histone acetylation might contribute to the anti-epileptic effect of SAHA in refractory epilepsy caused by GABRG2(F343L) mutation, demonstrating SAHA as a promising therapeutic agent for refractory epilepsy.


Subject(s)
Mutation , Receptors, GABA-A , Vorinostat , Zebrafish , Animals , Humans , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , HEK293 Cells , Vorinostat/pharmacology , Vorinostat/therapeutic use , Drug Resistant Epilepsy/drug therapy , Drug Resistant Epilepsy/genetics , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Animals, Genetically Modified
SELECTION OF CITATIONS
SEARCH DETAIL