Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.348
Filter
1.
Acta Neuropathol ; 147(1): 80, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714540

ABSTRACT

GABAergic interneurons play a critical role in maintaining neural circuit balance, excitation-inhibition regulation, and cognitive function modulation. In tuberous sclerosis complex (TSC), GABAergic neuron dysfunction contributes to disrupted network activity and associated neurological symptoms, assumingly in a cell type-specific manner. This GABAergic centric study focuses on identifying specific interneuron subpopulations within TSC, emphasizing the unique characteristics of medial ganglionic eminence (MGE)- and caudal ganglionic eminence (CGE)-derived interneurons. Using single-nuclei RNA sequencing in TSC patient material, we identify somatostatin-expressing (SST+) interneurons as a unique and immature subpopulation in TSC. The disrupted maturation of SST+ interneurons may undergo an incomplete switch from excitatory to inhibitory GABAergic signaling during development, resulting in reduced inhibitory properties. Notably, this study reveals markers of immaturity specifically in SST+ interneurons, including an abnormal NKCC1/KCC2 ratio, indicating an imbalance in chloride homeostasis crucial for the postsynaptic consequences of GABAergic signaling as well as the downregulation of GABAA receptor subunits, GABRA1, and upregulation of GABRA2. Further exploration of SST+ interneurons revealed altered localization patterns of SST+ interneurons in TSC brain tissue, concentrated in deeper cortical layers, possibly linked to cortical dyslamination. In the epilepsy context, our research underscores the diverse cell type-specific roles of GABAergic interneurons in shaping seizures, advocating for precise therapeutic considerations. Moreover, this study illuminates the potential contribution of SST+ interneurons to TSC pathophysiology, offering insights for targeted therapeutic interventions.


Subject(s)
GABAergic Neurons , Interneurons , Tuberous Sclerosis , Interneurons/pathology , Interneurons/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis/metabolism , Humans , GABAergic Neurons/pathology , GABAergic Neurons/metabolism , Male , Female , Median Eminence/pathology , Median Eminence/metabolism , Somatostatin/metabolism , Child , Child, Preschool , Receptors, GABA-A/metabolism , Adolescent , Ganglionic Eminence
2.
Molecules ; 29(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731442

ABSTRACT

Two series, "a" and "b", each consisting of nine chemical compounds, with 2,3-disubstituted quinazolin-4(3H)-one scaffold, were synthesized and evaluated for their anticonvulsant activity. They were investigated as dual potential positive allosteric modulators of the GABAA receptor at the benzodiazepine binding site and inhibitors of carbonic anhydrase II. Quinazolin-4(3H)-one derivatives were evaluated in vivo (D1-3 = 50, 100, 150 mg/kg, administered intraperitoneally) using the pentylenetetrazole (PTZ)-induced seizure model in mice, with phenobarbital and diazepam, as reference anticonvulsant agents. The in silico studies suggested the compounds act as anticonvulsants by binding on the allosteric site of GABAA receptor and not by inhibiting the carbonic anhydrase II, because the ligands-carbonic anhydrase II predicted complexes were unstable in the molecular dynamics simulations. The mechanism targeting GABAA receptor was confirmed through the in vivo flumazenil antagonism assay. The pentylenetetrazole experimental anticonvulsant model indicated that the tested compounds, 1a-9a and 1b-9b, present a potential anticonvulsant activity. The evaluation, considering the percentage of protection against PTZ, latency until the onset of the first seizure, and reduction in the number of seizures, revealed more favorable results for the "b" series, particularly for compound 8b.


Subject(s)
Anticonvulsants , Pentylenetetrazole , Receptors, GABA-A , Seizures , Anticonvulsants/pharmacology , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Animals , Mice , Seizures/drug therapy , Seizures/chemically induced , Receptors, GABA-A/metabolism , Quinazolinones/pharmacology , Quinazolinones/chemistry , Quinazolinones/chemical synthesis , Molecular Docking Simulation , Male , Structure-Activity Relationship , Molecular Dynamics Simulation , Computer Simulation , Disease Models, Animal , Molecular Structure , Allosteric Site
3.
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
4.
CNS Neurosci Ther ; 30(5): e14716, 2024 05.
Article in English | MEDLINE | ID: mdl-38698533

ABSTRACT

BACKGROUND: Sevoflurane is a superior agent for maintaining anesthesia during surgical procedures. However, the neurotoxic mechanisms of clinical concentration remain poorly understood. Sevoflurane can interfere with the normal function of neurons and synapses and impair cognitive function by acting on α5-GABAAR. METHODS: Using MWM test, we evaluated cognitive abilities in mice following 1 h of anesthesia with 2.7%-3% sevoflurane. Based on hippocampal transcriptome analysis, we analyzed the differential genes and IL-6 24 h post-anesthesia. Western blot and RT-PCR were performed to measure the levels of α5-GABAAR, Radixin, P-ERM, P-Radixin, Gephyrin, IL-6, and ROCK. The spatial distribution and expression of α5-GABAAR on neuronal somata were analyzed using histological and three-dimensional imaging techniques. RESULTS: MWM test indicated that partial long-term learning and memory impairment. Combining molecular biology and histological analysis, our studies have demonstrated that sevoflurane induces immunosuppression, characterized by reduced IL-6 expression levels, and that enhanced Radixin dephosphorylation undermines the microstructural stability of α5-GABAAR, leading to its dissociation from synaptic exterior and resulting in a disordered distribution in α5-GABAAR expression within neuronal cell bodies. On the synaptic cleft, the expression level of α5-GABAAR remained unchanged, the spatial distribution became more compact, with an increased fluorescence intensity per voxel. On the extra-synaptic space, the expression level of α5-GABAAR decreased within unchanged spatial distribution, accompanied by an increased fluorescence intensity per voxel. CONCLUSION: Dysregulated α5-GABAAR expression and distribution contributes to sevoflurane-induced partial long-term learning and memory impairment, which lays the foundation for elucidating the underlying mechanisms in future studies.


Subject(s)
Anesthetics, Inhalation , Hippocampus , Memory Disorders , Receptors, GABA-A , Sevoflurane , Sevoflurane/toxicity , Animals , Mice , Male , Memory Disorders/chemically induced , Memory Disorders/metabolism , Anesthetics, Inhalation/toxicity , Receptors, GABA-A/metabolism , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/genetics , Hippocampus/metabolism , Hippocampus/drug effects , Mice, Inbred C57BL , Maze Learning/drug effects , Maze Learning/physiology
5.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38695719

ABSTRACT

Microglia sense the changes in their environment. How microglia actively translate these changes into suitable cues to adapt brain physiology is unknown. We reveal an activity-dependent regulation of cortical inhibitory synapses by microglia, driven by purinergic signaling acting on P2RX7 and mediated by microglia-derived TNFα. We demonstrate that sleep induces microglia-dependent synaptic enrichment of GABAARs in a manner dependent on microglial TNFα and P2RX7. We further show that microglia-specific depletion of TNFα alters slow waves during NREM sleep and blunt memory consolidation in sleep-dependent learning tasks. Together, our results reveal that microglia orchestrate sleep-intrinsic plasticity of synaptic GABAARs, sculpt sleep slow waves, and support memory consolidation.


Subject(s)
Microglia , Receptors, GABA-A , Sleep, Slow-Wave , Synapses , Tumor Necrosis Factor-alpha , Animals , Male , Mice , Memory Consolidation , Mice, Inbred C57BL , Microglia/metabolism , Neuronal Plasticity/physiology , Receptors, GABA-A/metabolism , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics , Signal Transduction , Sleep/physiology , Synapses/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731820

ABSTRACT

A significant number of patients with genetic epilepsy do not obtain seizure freedom, despite developments in new antiseizure drugs, suggesting a need for novel therapeutic approaches. Many genetic epilepsies are associated with misfolded mutant proteins, including GABRG2(Q390X)-associated Dravet syndrome, which we have previously shown to result in intracellular accumulation of mutant GABAA receptor γ2(Q390X) subunit protein. Thus, a potentially promising therapeutic approach is modulation of proteostasis, such as increasing endoplasmic reticulum (ER)-associated degradation (ERAD). To that end, we have here identified an ERAD-associated E3 ubiquitin ligase, HRD1, among other ubiquitin ligases, as a strong modulator of wildtype and mutant γ2 subunit expression. Overexpressing HRD1 or knockdown of HRD1 dose-dependently reduced the γ2(Q390X) subunit. Additionally, we show that zonisamide (ZNS)-an antiseizure drug reported to upregulate HRD1-reduces seizures in the Gabrg2+/Q390X mouse. We propose that a possible mechanism for this effect is a partial rescue of surface trafficking of GABAA receptors, which are otherwise sequestered in the ER due to the dominant-negative effect of the γ2(Q390X) subunit. Furthermore, this partial rescue was not due to changes in ER chaperones BiP and calnexin, as total expression of these chaperones was unchanged in γ2(Q390X) models. Our results here suggest that leveraging the endogenous ERAD pathway may present a potential method to degrade neurotoxic mutant proteins like the γ2(Q390X) subunit. We also demonstrate a pharmacological means of regulating proteostasis, as ZNS alters protein trafficking, providing further support for the use of proteostasis regulators for the treatment of genetic epilepsies.


Subject(s)
Endoplasmic Reticulum , Epilepsies, Myoclonic , Proteolysis , Receptors, GABA-A , Epilepsies, Myoclonic/metabolism , Epilepsies, Myoclonic/genetics , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Animals , Endoplasmic Reticulum/metabolism , Mice , Humans , Seizures, Febrile/metabolism , Seizures, Febrile/genetics , Endoplasmic Reticulum-Associated Degradation , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Mutation , HEK293 Cells , Endoplasmic Reticulum Chaperone BiP/metabolism
7.
PLoS One ; 19(4): e0298065, 2024.
Article in English | MEDLINE | ID: mdl-38626211

ABSTRACT

Anoxia in the mammalian brain leads to hyper-excitability and cell death; however, this cascade of events does not occur in the anoxia-tolerant brain of the western painted turtle, Chrysemys picta belli. The painted turtle has become an important anoxia-tolerant model to study brain, heart, and liver function in the absence of oxygen, but being anoxia-tolerant likely means that decapitation alone is not a suitable method of euthanasia. Many anesthetics have long-term effects on ion channels and are not appropriate for same day experimentation. Using whole-cell electrophysiological techniques, we examine the effects of the anesthetic, Alfaxalone, on pyramidal cell action potential amplitude, threshold, rise and decay time, width, frequency, whole cell conductance, and evoked GABAA receptors currents to determine if any of these characteristics are altered with the use of Alfaxalone for animal sedation. We find that Alfaxalone has no long-term impact on action potential parameters or whole-cell conductance. When acutely applied to naïve tissue, Alfaxalone did lengthen GABAA receptor current decay rates by 1.5-fold. Following whole-animal sedation with Alfaxalone, evoked whole cell GABAA receptor current decay rates displayed an increasing trend with 1 and 2 hours after brain sheet preparation, but showed no significant change after a 3-hour washout period. Therefore, we conclude that Alfaxalone is a suitable anesthetic for same day use in electrophysiological studies in western painted turtle brain tissue.


Subject(s)
Anesthetics , Hypoxia, Brain , Pregnanediones , Turtles , Animals , Turtles/physiology , Receptors, GABA-A/metabolism , Pyramidal Cells/metabolism , Hypoxia/metabolism , Anesthetics/pharmacology , Mammals
8.
J Neurosci ; 44(19)2024 May 08.
Article in English | MEDLINE | ID: mdl-38565288

ABSTRACT

Excitotoxicity and the concurrent loss of inhibition are well-defined mechanisms driving acute elevation in excitatory/inhibitory (E/I) balance and neuronal cell death following an ischemic insult to the brain. Despite the high prevalence of long-term disability in survivors of global cerebral ischemia (GCI) as a consequence of cardiac arrest, it remains unclear whether E/I imbalance persists beyond the acute phase and negatively affects functional recovery. We previously demonstrated sustained impairment of long-term potentiation (LTP) in hippocampal CA1 neurons correlating with deficits in learning and memory tasks in a murine model of cardiac arrest/cardiopulmonary resuscitation (CA/CPR). Here, we use CA/CPR and an in vitro ischemia model to elucidate mechanisms by which E/I imbalance contributes to ongoing hippocampal dysfunction in male mice. We reveal increased postsynaptic GABAA receptor (GABAAR) clustering and function in the CA1 region of the hippocampus that reduces the E/I ratio. Importantly, reduced GABAAR clustering observed in the first 24 h rebounds to an elevation of GABAergic clustering by 3 d postischemia. This increase in GABAergic inhibition required activation of the Ca2+-permeable ion channel transient receptor potential melastatin-2 (TRPM2), previously implicated in persistent LTP and memory deficits following CA/CPR. Furthermore, we find Ca2+-signaling, likely downstream of TRPM2 activation, upregulates Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity, thereby driving the elevation of postsynaptic inhibitory function. Thus, we propose a novel mechanism by which inhibitory synaptic strength is upregulated in the context of ischemia and identify TRPM2 and CaMKII as potential pharmacological targets to restore perturbed synaptic plasticity and ameliorate cognitive function.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Mice, Inbred C57BL , Signal Transduction , TRPM Cation Channels , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Male , Mice , TRPM Cation Channels/metabolism , Signal Transduction/physiology , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Receptors, GABA-A/metabolism , Hippocampus/metabolism , Neural Inhibition/physiology , GABAergic Neurons/metabolism , Heart Arrest/complications , Heart Arrest/metabolism
9.
Neurosci Biobehav Rev ; 161: 105651, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38579901

ABSTRACT

GABA is the primary inhibitory neurotransmitter in the adult brain and through its actions on GABAARs, it protects against excitotoxicity and seizure activity, ensures temporal fidelity of neurotransmission, and regulates concerted rhythmic activity of neuronal populations. In the developing brain, the development of GABAergic neurons precedes that of glutamatergic neurons and the GABA system serves as a guide and framework for the development of other brain systems. Despite this early start, the maturation of the GABA system also continues well into the early postnatal period. In this review, we organize evidence around two scenarios based on the essential and protracted nature of GABA system development: 1) disruptions in the development of the GABA system can lead to large scale disruptions in other developmental processes (i.e., GABA as the cause), 2) protracted maturation of this system makes it vulnerable to the effects of developmental insults (i.e., GABA as the effect). While ample evidence supports the importance of GABA/GABAAR system in both scenarios, large gaps in existing knowledge prevent strong mechanistic conclusions.


Subject(s)
Brain , gamma-Aminobutyric Acid , gamma-Aminobutyric Acid/metabolism , Humans , Animals , Brain/growth & development , Brain/metabolism , Receptors, GABA-A/metabolism , GABAergic Neurons/physiology , GABAergic Neurons/metabolism
10.
Eur J Pharmacol ; 974: 176616, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38679122

ABSTRACT

The purpose of this study was to investigate the mechanisms underlying sex differences in the role of spinal α6-subunit containing GABAA (α6GABAA) receptors in rats with neuropathic pain. Intrathecal 2,5-dihydro-7-methoxy-2-(4-methoxyphenyl)-3H-pyrazolo [4,3-c] quinoline-3-one (PZ-II-029, positive allosteric modulator of α6GABAA receptors) reduced tactile allodynia in female but not in male rats with neuropathic pain. PZ-II-029 was also more effective in females than males in inflammatory and nociplastic pain. Ovariectomy abated the antiallodynic effect of PZ-II-029 in neuropathic rats, whereas 17ß-estradiol or 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol (PPT), estradiol receptor-α agonist, restored the effect of PZ-II-029 in ovariectomized rats. Blockade of estradiol receptor-α, using MPP (1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy) phenol]-1H-pyrazole dihydrochloride), prevented the effect of 17ß-estradiol on PZ-II-029-induced antiallodynia in ovariectomized neuropathic females. Nerve injury reduced α6GABAA receptor protein expression at the dorsal root ganglia (DRG) and spinal cord of intact and ovariectomized female rats. In this last group, reconstitution with 17ß-estradiol fully restored its expression in DRG and spinal cord. In male rats, nerve injury reduced α6GABAA receptor protein expression only at the spinal cord. Nerve injury enhanced estradiol receptor-α protein expression at the DRG in intact non-ovariectomized rats. However, ovariectomy decreased estradiol receptor-α protein expression at the DRG. In the spinal cord there were no changes in estradiol receptor-α protein expression. 17ß-estradiol restored estradiol receptor-α protein expression at the DRG and increased it at the spinal cord of neuropathic rats. These data suggest that 17ß-estradiol modulates the expression and function of the α6GABAA receptor through its interaction with estradiol receptor-α in female rats.


Subject(s)
Estradiol , Neuralgia , Receptors, GABA-A , Spinal Cord , Animals , Female , Estradiol/pharmacology , Receptors, GABA-A/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Rats , Male , Spinal Cord/drug effects , Spinal Cord/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Ovariectomy , Rats, Sprague-Dawley , Sex Characteristics , Estrogen Receptor alpha/metabolism , Pyrazoles/pharmacology
11.
J Cell Mol Med ; 28(9): e18352, 2024 May.
Article in English | MEDLINE | ID: mdl-38685685

ABSTRACT

Gliomas, the most lethal tumours in brain, have a poor prognosis despite accepting standard treatment. Limited benefits from current therapies can be attributed to genetic, epigenetic and microenvironmental cues that affect cell programming and drive tumour heterogeneity. Through the analysis of Hi-C data, we identified a potassium-chloride co-transporter SLC12A5 associated with disrupted topologically associating domain which was downregulated in tumour tissues. Multiple independent glioma cohorts were included to analyse the characterization of SLC12A5 and found it was significantly associated with pathological features, prognostic value, genomic alterations, transcriptional landscape and drug response. We constructed two SLC12A5 overexpression cell lines to verify the function of SLC12A5 that suppressed tumour cell proliferation and migration in vitro. In addition, SLC12A5 was also positively associated with GABAA receptor activity and negatively associated with pro-tumour immune signatures and immunotherapy response. Collectively, our study provides a comprehensive characterization of SLC12A5 in glioma and supports SLC12A5 as a potential suppressor of disease progression.


Subject(s)
Brain Neoplasms , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioma , K Cl- Cotransporters , Symporters , Humans , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Cell Proliferation/genetics , Cell Line, Tumor , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Symporters/genetics , Symporters/metabolism , Cell Movement/genetics , Prognosis , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics
12.
Medicina (Kaunas) ; 60(4)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38674283

ABSTRACT

Background and Objectives: Drug resistant epilepsy (DRE) is a major hurdle in epilepsy, which hinders clinical care, patients' management and treatment outcomes. DRE may partially result from genetic variants that alter proteins responsible for drug targets and drug transporters in the brain. We aimed to examine the relationship between SCN1A, GABRA1 and ABCB1 polymorphism and drug response in epilepsy children in Vietnam. Materials and Methods: In total, 213 children diagnosed with epilepsy were recruited in this study (101 were drug responsive and 112 were drug resistant). Sanger sequencing had been performed in order to detect six single nucleotide polymorphisms (SNPs) belonging to SCN1A (rs2298771, rs3812718, rs10188577), GABRA1 (rs2279020) and ABCB1 (rs1128503, rs1045642) in study group. The link between SNPs and drug response status was examined by the Chi-squared test or the Fisher's exact test. Results: Among six investigated SNPs, two SNPs showed significant difference between the responsive and the resistant group. Among those, heterozygous genotype of SCN1A rs2298771 (AG) were at higher frequency in the resistant patients compared with responsive patients, playing as risk factor of refractory epilepsy. Conversely, the heterozygous genotype of SCN1A rs3812718 (CT) was significantly lower in the resistant compared with the responsive group. No significant association was found between the remaining four SNPs and drug response. Conclusions: Our study demonstrated a significant association between the SCN1A genetic polymorphism which increased risk of drug-resistant epilepsy in Vietnamese epileptic children. This important finding further supports the underlying molecular mechanisms of SCN1A genetic variants in the pathogenesis of drug-resistant epilepsy in children.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Anticonvulsants , Epilepsy , NAV1.1 Voltage-Gated Sodium Channel , Polymorphism, Single Nucleotide , Receptors, GABA-A , Humans , NAV1.1 Voltage-Gated Sodium Channel/genetics , Vietnam , Male , Female , Child , ATP Binding Cassette Transporter, Subfamily B/genetics , Child, Preschool , Epilepsy/genetics , Epilepsy/drug therapy , Receptors, GABA-A/genetics , Anticonvulsants/therapeutic use , Drug Resistant Epilepsy/genetics , Drug Resistant Epilepsy/drug therapy , Infant , Genotype , Adolescent , Southeast Asian People
13.
Exp Neurol ; 376: 114775, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38604438

ABSTRACT

OBJECTIVE: Sleep-related hypermotor epilepsy (SHE) is a focal epilepsy syndrome characterized by seizures that predominantly occur during sleep. The pathogenesis of these seizures remains unclear. We previously detected rare variants in GABRG2, which encodes the γ2 subunit of γ-aminobutyric acid type A receptor (GABAAR), in patients with SHE and demonstrated that these variants impaired GABAAR function in vitro. However, the mechanisms by which GABRG2 variants contribute to seizure attacks during sleep remain unclear. METHODS: In this study, we designed a knock-in (KI) mouse expressing the mouse Gabrg2 T316N variant, corresponding to human GABRG2 T317N variant, using CRISPR/Cas9. Continuous video-electroencephalogram monitoring and in vivo multichannel electrophysiological recordings were performed to explore seizure susceptibility to pentylenetetrazol (PTZ), alterations in the sleep-wake cycle, spontaneous seizure patterns, and synchronized activity in the motor thalamic nuclei (MoTN) and secondary motor cortex (M2). Circadian variations in the expression of total, membrane-bound, and synaptic GABAAR subunits were also investigated. RESULTS: No obvious changes in gross morphology were detected in Gabrg2T316N/+ mice compared to their wild-type (Gabrg2+/+) littermates. Gabrg2T316N/+ mice share key phenotypes with patients, including sleep fragmentation and spontaneous seizures during sleep. Gabrg2T316N/+ mice showed increased susceptibility to PTZ-induced seizures and higher mortality after seizures. Synchronization of the local field potentials between the MoTN and M2 was abnormally enhanced in Gabrg2T316N/+ mice during light phase, when sleep dominates, accompanied by increased local activities in the MoTN and M2. Interestingly, in Gabrg2+/+ mice, GABAAR γ2 subunits showed a circadian increase on the neuronal membrane and synaptosomes in the transition from dark phase to light phase, which was absent in Gabrg2T316N/+ mice. CONCLUSION: We generated a new SHE mouse model and provided in vivo evidence that rare variants of GABRG2 contribute to seizure attacks during sleep in SHE.


Subject(s)
Receptors, GABA-A , Animals , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Mice , Phenotype , Sleep/physiology , Sleep/genetics , Male , Mice, Transgenic , Thalamus/metabolism , Thalamus/pathology , Mice, Inbred C57BL , Electroencephalography , Gene Knock-In Techniques , Epilepsy/genetics , Epilepsy/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Female
14.
Eur J Pharmacol ; 972: 176561, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38580182

ABSTRACT

Neuronal depression in the thalamus underlies anesthetic-induced loss of consciousness, while the precise sub-thalamus nuclei and molecular targets involved remain to be elucidated. The present study investigated the role of extrasynaptic GABAA receptors in the central medial thalamic nucleus (CM) in anesthesia induced by gaboxadol (THIP) and diazepam (DZP) in rats. Local lesion of the CM led to a decrease in the duration of loss of righting reflex induced by THIP and DZP. CM microinjection of THIP but not DZP induced anesthesia. The absence of righting reflex in THIP-treated rats was consistent with the increase of low frequency oscillations in the delta band in the medial prefrontal cortex. CM microinjection of GABAA receptor antagonist SR95531 significantly attenuated the anesthesia induced by systemically-administered THIP, but not DZP. Moreover, the rats with declined expression of GABAA receptor δ-subunit in the CM were less responsive to THIP or DZP. These findings explained a novel mechanism of THIP-induced loss of consciousness and highlighted the role of CM extrasynaptic GABAA receptors in mediating anesthesia.


Subject(s)
Anesthesia , Isoxazoles , Receptors, GABA-A , Animals , Receptors, GABA-A/metabolism , Male , Rats , Isoxazoles/pharmacology , Diazepam/pharmacology , Rats, Sprague-Dawley , Mediodorsal Thalamic Nucleus/drug effects , Mediodorsal Thalamic Nucleus/metabolism , Mediodorsal Thalamic Nucleus/physiology , Reflex, Righting/drug effects , Synapses/drug effects , Synapses/metabolism , Thalamus/drug effects , Thalamus/metabolism
15.
Proc Natl Acad Sci U S A ; 121(15): e2318041121, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38568976

ABSTRACT

Stable matching of neurotransmitters with their receptors is fundamental to synapse function and reliable communication in neural circuits. Presynaptic neurotransmitters regulate the stabilization of postsynaptic transmitter receptors. Whether postsynaptic receptors regulate stabilization of presynaptic transmitters has received less attention. Here, we show that blockade of endogenous postsynaptic acetylcholine receptors (AChR) at the neuromuscular junction destabilizes the cholinergic phenotype in motor neurons and stabilizes an earlier, developmentally transient glutamatergic phenotype. Further, expression of exogenous postsynaptic gamma-aminobutyric acid type A receptors (GABAA receptors) in muscle cells stabilizes an earlier, developmentally transient GABAergic motor neuron phenotype. Both AChR and GABAA receptors are linked to presynaptic neurons through transsynaptic bridges. Knockdown of specific components of these transsynaptic bridges prevents stabilization of the cholinergic or GABAergic phenotypes. Bidirectional communication can enforce a match between transmitter and receptor and ensure the fidelity of synaptic transmission. Our findings suggest a potential role of dysfunctional transmitter receptors in neurological disorders that involve the loss of the presynaptic transmitter.


Subject(s)
Receptors, Cholinergic , Synapses , Synapses/metabolism , Receptors, Cholinergic/metabolism , Synaptic Transmission/physiology , Motor Neurons/metabolism , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/metabolism , Neurotransmitter Agents/metabolism , Cholinergic Agents , Receptors, Presynaptic
16.
Biochem Pharmacol ; 223: 116183, 2024 May.
Article in English | MEDLINE | ID: mdl-38580167

ABSTRACT

In this study, we have investigated the pharmacological activity and structural interaction of two novel psychoplastogens, tabernanthalog (TBG) and ibogainalog (IBG) at heterologously-expressed rat (r) and human (h) nicotinic acetylcholine receptors (nAChRs), the rα1ß2γ2L γ-aminobutyric acid type A receptor (GABAAR), and the human voltage-gated N-type calcium channel (CaV2.2 channel). Both compounds inhibited the nAChRs with the following receptor selectivity: α9α10 > α7 > α3ß2 â‰… α3ß4, indicating that ß2/ß4 subunits are relatively less important for their activity. The potencies of TBG and IBG were comparable at hα7 and hα9α10 subtypes, and comparable to their rat counterparts. TBG- and IBG-induced inhibition of rα7 was ACh concentration-independent and voltage-dependent, whereas rα9α10 inhibition was ACh concentration-dependent and voltage-independent, suggesting that they interact with the α7 ion channel pore and α9α10 orthosteric ligand binding site, respectively. These results were supported by molecular docking studies showing that at the α7 model TBG forms stable interactions with luminal rings at 9', 13', and 16', whereas IBG mostly interacts with the extracellular-transmembrane junction. In the α9α10 model, however, these compounds interacted with several residues from the principal (+) and complementary (-) sides in the transmitter binding site. Ibogaminalog (DM506) also interacted with a non-luminal site at α7, and one α9α10 orthosteric site. TBG and IBG inhibited the GABAAR and CaV2.2 channels with 10 to 30-fold lower potencies. In sum, we show that TBG and IBG inhibit the α7 and α9α10 nAChRs by noncompetitive and competitive mechanisms, respectively, and with higher potency than the GABAAR and CaV2.2 channel.


Subject(s)
Receptors, Nicotinic , Rats , Animals , Humans , Receptors, Nicotinic/metabolism , Receptors, GABA-A/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Molecular Docking Simulation , gamma-Aminobutyric Acid
17.
Brain Res ; 1835: 148929, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38599510

ABSTRACT

Temporal order memory is impaired in autism spectrum disorder (ASD) and schizophrenia (SCZ). These disorders, more prevalent in males, result in abnormal dendritic spine pruning during adolescence in layer 3 (L3) medial prefrontal cortex (mPFC), yielding either too many (ASD) or too few (SCZ) spines. Here we tested whether altering spine density in neural circuits including the mPFC could be associated with impaired temporal order memory in male mice. We have shown that α4ßδ GABAA receptors (GABARs) emerge at puberty on spines of L5 prelimbic mPFC (PL) where they trigger pruning. We show here that α4ßδ receptors also increase at puberty in L3 PL (P < 0.0001) and used these receptors as a target to manipulate spine density here. Pubertal injection (14 d) of the GABA agonist gaboxadol, at a dose (3 mg/kg) selective for α4ßδ, reduced L3 spine density by half (P < 0.0001), while α4 knock-out increased spine density âˆ¼ 40 % (P < 0.0001), mimicking spine densities in SCZ and ASD, respectively. In both cases, performance on the mPFC-dependent temporal order recognition task was impaired, resulting in decreases in the discrimination ratio which assesses preference for the novel object: -0.39 ± 0.15, gaboxadol versus 0.52 ± 0.09, vehicle; P = 0.0002; -0.048 ± 0.10, α4 KO versus 0.49 ± 0.04, wild-type; P < 0.0001. In contrast, the number of approaches was unaltered, reflecting unchanged locomotion. These data suggest that altering α4ßδ GABAR expression/activity alters spine density in L3 mPFC and impairs temporal order memory to mimic changes in ASD and SCZ. These findings may provide insight into these disorders.


Subject(s)
Dendritic Spines , Prefrontal Cortex , Receptors, GABA-A , Schizophrenia , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Animals , Receptors, GABA-A/metabolism , Male , Schizophrenia/metabolism , Mice , Dendritic Spines/metabolism , Dendritic Spines/drug effects , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Mice, Inbred C57BL , Isoxazoles/pharmacology , Autistic Disorder/metabolism , Autistic Disorder/pathology , GABA-A Receptor Agonists/pharmacology , Autism Spectrum Disorder/metabolism , Recognition, Psychology/physiology , Recognition, Psychology/drug effects
18.
Proc Natl Acad Sci U S A ; 121(18): e2314541121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38657049

ABSTRACT

Recent evidence has demonstrated that the transsynaptic nanoscale organization of synaptic proteins plays a crucial role in regulating synaptic strength in excitatory synapses. However, the molecular mechanism underlying this transsynaptic nanostructure in inhibitory synapses still remains unclear and its impact on synapse function in physiological or pathological contexts has not been demonstrated. In this study, we utilized an engineered proteolysis technique to investigate the effects of acute cleavage of neuroligin-2 (NL2) on synaptic transmission. Our results show that the rapid cleavage of NL2 led to impaired synaptic transmission by reducing both neurotransmitter release probability and quantum size. These changes were attributed to the dispersion of RIM1/2 and GABAA receptors and a weakened spatial alignment between them at the subsynaptic scale, as observed through superresolution imaging and model simulations. Importantly, we found that endogenous NL2 undergoes rapid MMP9-dependent cleavage during epileptic activities, which further exacerbates the decrease in inhibitory transmission. Overall, our study demonstrates the significant impact of nanoscale structural reorganization on inhibitory transmission and unveils ongoing modulation of mature GABAergic synapses through active cleavage of NL2 in response to hyperactivity.


Subject(s)
Cell Adhesion Molecules, Neuronal , Nerve Tissue Proteins , Synapses , Synaptic Transmission , Animals , Mice , Cell Adhesion Molecules, Neuronal/metabolism , Epilepsy/metabolism , Epilepsy/physiopathology , Epilepsy/pathology , Hippocampus/metabolism , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Proteolysis , Receptors, GABA-A/metabolism , Synapses/metabolism , Synaptic Transmission/physiology
19.
Article in English | MEDLINE | ID: mdl-38681506

ABSTRACT

Background: Essential tremor patients may find that low alcohol amounts suppress tremor. A candidate mechanism is modulation of α6ß3δ extra-synaptic GABAA receptors, that in vitro respond to non-intoxicating alcohol levels. We previously found that low-dose alcohol reduces harmaline tremor in wild-type mice, but not in littermates lacking δ or α6 subunits. Here we addressed whether low-dose alcohol requires the ß3 subunit for tremor suppression. Methods: We tested whether low-dose alcohol suppresses tremor in cre-negative mice with intact ß3 exon 3 flanked by loxP, and in littermates in which this region was excised by cre expressed under the α6 subunit promotor. Tremor in the harmaline model was measured as a percentage of motion power in the tremor bandwidth divided by overall motion power. Results: Alcohol, 0.500 and 0.575 g/kg, reduced harmaline tremor compared to vehicle-treated controls in floxed ß3 cre- mice, but had no effect on tremor in floxed ß3 cre+ littermates that have ß3 knocked out. This was not due to potential interference of α6 expression by the insertion of the cre gene into the α6 gene since non-floxed ß3 cre+ and cre- littermates exhibited similar tremor suppression by alcohol. Discussion: As α6ß3δ GABAA receptors are sensitive to low-dose alcohol, and cerebellar granule cells express ß3 and are the predominant brain site for α6 and δ expression together, our overall findings suggest alcohol acts to suppress tremor by modulating α6ß3δ GABAA receptors on these cells. Novel drugs that target this receptor may potentially be effective and well-tolerated for essential tremor. Highlights: We previously found with the harmaline essential tremor model that GABAA receptors containing α6 and δ subunits mediate tremor suppression by alcohol. We now show that ß3 subunits in α6-expressing cells, likely cerebellar granule cells, are also required, indicating that alcohol suppresses tremor by modulating α6ß3δ extra-synaptic GABAA receptors.


Subject(s)
Essential Tremor , Ethanol , Harmaline , Receptors, GABA-A , Animals , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Harmaline/pharmacology , Essential Tremor/drug therapy , Essential Tremor/genetics , Mice , Ethanol/pharmacology , Central Nervous System Depressants/pharmacology , Disease Models, Animal , Male , Mice, Knockout
20.
Adv Med Sci ; 69(1): 176-189, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38561071

ABSTRACT

PURPOSE: Metabolic syndrome (MetS) is a common disorder associated with disturbed neurotransmitter homeostasis. Memantine, an N-methyl-d-aspartate receptor (NMDAR) antagonist, was first used in Alzheimer's disease. Allopregnanolone (Allo), a potent positive allosteric modulator of the Gamma-Amino-Butyric Acid (GABA)-A receptors, decreases in neurodegenerative diseases. The study investigated the impact of Memantine versus Allo administration on the animal model of MetS to clarify whether the mechanism of abnormalities is related more to excitatory or inhibitory neurotransmitter dysfunction. MATERIALS AND METHODS: Fifty-six male rats were allocated into 7 groups: 4 control groups, 1 MetS group, and 2 treated MetS groups. They underwent assessment of cognition-related behavior by open field and forced swimming tests, electroencephalogram (EEG) recording, serum markers confirming the establishment of MetS model and hippocampal Glial Fibrillary Acidic Protein (GFAP) and Brain-Derived Neurotrophic Factor (BDNF). RESULTS: Allo improved anxiety-like behavior and decreased grooming frequency compared to Memantine. Both drugs increased GFAP and BDNF expression, improving synaptic plasticity and cognition-related behaviors. The therapeutic effect of Allo was more beneficial regarding lipid profile and anxiety. We reported progressive slowing of EEG waves in the MetS group with Memantine and Allo treatment with increased relative theta and decreased relative delta rhythms. CONCLUSIONS: Both Allo and Memantine boosted the outcome parameters in the animal model of MetS. Allo markedly improved the anxiety-like behavior in the form of significantly decreased grooming frequency compared to the Memantine-treated groups. Both drugs were associated with increased hippocampal GFAP and BDNF expression, indicating an improvement in synaptic plasticity and so, cognition-related behaviors.


Subject(s)
Memantine , Metabolic Syndrome , Neuronal Plasticity , Receptors, GABA-A , Receptors, N-Methyl-D-Aspartate , Animals , Neuronal Plasticity/drug effects , Male , Rats , Metabolic Syndrome/metabolism , Metabolic Syndrome/drug therapy , Receptors, N-Methyl-D-Aspartate/metabolism , Memantine/pharmacology , Receptors, GABA-A/metabolism , Brain/metabolism , Brain/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Pregnanolone/pharmacology , Pregnanolone/metabolism , Rats, Wistar , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL