Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.339
Filter
Add more filters

Publication year range
1.
Cell ; 166(2): 299-313, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27293187

ABSTRACT

Patients with autism spectrum disorders (ASDs) commonly experience aberrant tactile sensitivity, yet the neural alterations underlying somatosensory dysfunction and the extent to which tactile deficits contribute to ASD characteristics are unknown. We report that mice harboring mutations in Mecp2, Gabrb3, Shank3, and Fmr1 genes associated with ASDs in humans exhibit altered tactile discrimination and hypersensitivity to gentle touch. Deletion of Mecp2 or Gabrb3 in peripheral somatosensory neurons causes mechanosensory dysfunction through loss of GABAA receptor-mediated presynaptic inhibition of inputs to the CNS. Remarkably, tactile defects resulting from Mecp2 or Gabrb3 deletion in somatosensory neurons during development, but not in adulthood, cause social interaction deficits and anxiety-like behavior. Restoring Mecp2 expression exclusively in the somatosensory neurons of Mecp2-null mice rescues tactile sensitivity, anxiety-like behavior, and social interaction deficits, but not lethality, memory, or motor deficits. Thus, mechanosensory processing defects contribute to anxiety-like behavior and social interaction deficits in ASD mouse models. PAPERCLIP.


Subject(s)
Autism Spectrum Disorder/physiopathology , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Behavior, Animal , Disease Models, Animal , Interpersonal Relations , Methyl-CpG-Binding Protein 2/genetics , Mice , Receptors, GABA-A/genetics , Sensory Receptor Cells , Spinal Cord Dorsal Horn/metabolism , Synapses/metabolism , Touch
2.
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
3.
Proc Natl Acad Sci U S A ; 121(26): e2315100121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38889143

ABSTRACT

Synapses containing γ-aminobutyric acid (GABA) constitute the primary centers for inhibitory neurotransmission in our nervous system. It is unclear how these synaptic structures form and align their postsynaptic machineries with presynaptic terminals. Here, we monitored the cellular distribution of several GABAergic postsynaptic proteins in a purely glutamatergic neuronal culture derived from human stem cells, which virtually lacks any vesicular GABA release. We found that several GABAA receptor (GABAAR) subunits, postsynaptic scaffolds, and major cell-adhesion molecules can reliably coaggregate and colocalize at even GABA-deficient subsynaptic domains, but remain physically segregated from glutamatergic counterparts. Genetic deletions of both Gephyrin and a Gephyrin-associated guanosine di- or triphosphate (GDP/GTP) exchange factor Collybistin severely disrupted the coassembly of these postsynaptic compositions and their proper apposition with presynaptic inputs. Gephyrin-GABAAR clusters, developed in the absence of GABA transmission, could be subsequently activated and even potentiated by delayed supply of vesicular GABA. Thus, molecular organization of GABAergic postsynapses can initiate via a GABA-independent but Gephyrin-dependent intrinsic mechanism.


Subject(s)
Carrier Proteins , Membrane Proteins , Presynaptic Terminals , Receptors, GABA-A , Synapses , gamma-Aminobutyric Acid , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , gamma-Aminobutyric Acid/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Presynaptic Terminals/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Rho Guanine Nucleotide Exchange Factors/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics
4.
Proc Natl Acad Sci U S A ; 121(24): e2311570121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38830095

ABSTRACT

Even a transient period of hearing loss during the developmental critical period can induce long-lasting deficits in temporal and spectral perception. These perceptual deficits correlate with speech perception in humans. In gerbils, these hearing loss-induced perceptual deficits are correlated with a reduction of both ionotropic GABAA and metabotropic GABAB receptor-mediated synaptic inhibition in auditory cortex, but most research on critical period plasticity has focused on GABAA receptors. Therefore, we developed viral vectors to express proteins that would upregulate gerbil postsynaptic inhibitory receptor subunits (GABAA, Gabra1; GABAB, Gabbr1b) in pyramidal neurons, and an enzyme that mediates GABA synthesis (GAD65) presynaptically in parvalbumin-expressing interneurons. A transient period of developmental hearing loss during the auditory critical period significantly impaired perceptual performance on two auditory tasks: amplitude modulation depth detection and spectral modulation depth detection. We then tested the capacity of each vector to restore perceptual performance on these auditory tasks. While both GABA receptor vectors increased the amplitude of cortical inhibitory postsynaptic potentials, only viral expression of postsynaptic GABAB receptors improved perceptual thresholds to control levels. Similarly, presynaptic GAD65 expression improved perceptual performance on spectral modulation detection. These findings suggest that recovering performance on auditory perceptual tasks depends on GABAB receptor-dependent transmission at the auditory cortex parvalbumin to pyramidal synapse and point to potential therapeutic targets for developmental sensory disorders.


Subject(s)
Auditory Cortex , Gerbillinae , Hearing Loss , Animals , Auditory Cortex/metabolism , Auditory Cortex/physiopathology , Hearing Loss/genetics , Hearing Loss/physiopathology , Receptors, GABA-B/metabolism , Receptors, GABA-B/genetics , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/genetics , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Parvalbumins/metabolism , Parvalbumins/genetics , Auditory Perception/physiology , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Genetic Vectors/genetics
5.
J Neurosci ; 44(20)2024 May 15.
Article in English | MEDLINE | ID: mdl-38604778

ABSTRACT

The reversal potential refers to the membrane potential at which the net current flow through a channel reverses direction. The reversal potential is determined by transmembrane ion gradients and, in turn, determines how the channel's activity will affect the membrane potential. Traditional investigation into the reversal potential of inhibitory ligand-gated ion channels (EInh) has relied upon the activation of endogenous receptors, such as the GABA-A receptor (GABAAR). There are, however, challenges associated with activating endogenous receptors, including agonist delivery, isolating channel responses, and the effects of receptor saturation and desensitization. Here, we demonstrate the utility of using a light-gated anion channel, stGtACR2, to probe EInh in the rodent brain. Using mice of both sexes, we demonstrate that the properties of this optically activated channel make it a suitable proxy for studying GABAAR receptor-mediated inhibition. We validate this agonist-independent optogenetic strategy in vitro and in vivo and further show how it can accurately capture differences in EInh dynamics following manipulations of endogenous ion fluxes. This allows us to explore distinct resting EInh differences across genetically defined neuronal subpopulations. Using this approach to challenge ion homeostasis mechanisms in neurons, we uncover cell-specific EInh dynamics that are supported by the differential expression of endogenous ion handling mechanisms. Our findings therefore establish an effective optical strategy for revealing novel aspects of inhibitory reversal potentials and thereby expand the repertoire of optogenetics.


Subject(s)
Membrane Potentials , Optogenetics , Animals , Optogenetics/methods , Mice , Male , Female , Membrane Potentials/physiology , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Neurons/physiology , Neurons/metabolism , Mice, Inbred C57BL , Neural Inhibition/physiology , Ligand-Gated Ion Channels/metabolism , Ligand-Gated Ion Channels/genetics , Mice, Transgenic
6.
J Neurosci ; 44(32)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38942471

ABSTRACT

The mechanisms utilized by neurons to regulate the efficacy of phasic and tonic inhibition and their impacts on synaptic plasticity and behavior are incompletely understood. Cleft lip and palate transmembrane protein 1 (Clptm1) is a membrane-spanning protein that interacts with multiple γ-aminobutyric acid type A receptor (GABAAR) subunits, trapping them in the endoplasmic reticulum and Golgi network. Overexpression and knock-down studies suggest that Clptm1 modulates GABAAR-mediated phasic inhibition and tonic inhibition as well as activity-induced inhibitory synaptic homeostasis in cultured hippocampal neurons. To investigate the role of Clptm1 in the modulation of GABAARs in vivo, we generated Clptm1 knock-out (KO) mice. Here, we show that genetic KO of Clptm1 elevated phasic and tonic inhibitory transmission in both male and female heterozygous mice. Although basal excitatory synaptic transmission was not affected, Clptm1 haploinsufficiency significantly blocked high-frequency stimulation-induced long-term potentiation (LTP) in hippocampal CA3→CA1 synapses. In the hippocampus-dependent contextual fear-conditioning behavior task, both male and female Clptm1 heterozygous KO mice exhibited impairment in contextual fear memory. In addition, LTP and contextual fear memory were rescued by application of L-655,708, a negative allosteric modulator of the extrasynaptic GABAAR α5 subunit. These results suggest that haploinsufficiency of Clptm1 contributes to cognitive deficits through altered synaptic transmission and plasticity by elevation of inhibitory neurotransmission, with tonic inhibition playing a major role.


Subject(s)
Haploinsufficiency , Membrane Proteins , Mice, Knockout , Neuronal Plasticity , Receptors, GABA-A , Synaptic Transmission , Animals , Mice , Male , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Female , Membrane Proteins/genetics , Membrane Proteins/metabolism , Synaptic Transmission/physiology , Neuronal Plasticity/physiology , Neuronal Plasticity/genetics , Mice, Inbred C57BL , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/drug effects , Long-Term Potentiation/physiology , Long-Term Potentiation/genetics , Hippocampus/metabolism , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/physiopathology , Fear/physiology , Inhibitory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/drug effects , Memory/physiology , Neural Inhibition/physiology
7.
Hum Mol Genet ; 32(19): 2929-2939, 2023 09 16.
Article in English | MEDLINE | ID: mdl-37498167

ABSTRACT

Human disease-associated genetic variations often map to long non-coding RNA (lncRNA) genes; however, elucidation of their functional impact is challenging. We previously identified a new genetic variant rs4454083 (A/G) residing in exon of an uncharacterized lncRNA ARBAG that strongly associates with plasma levels of C-peptide, a hormone that regulates insulin bioavailability. On the opposite strand, rs4454083 also corresponds to an intron of a cerebellum-specific GABA receptor subunit gene GABRA6 that mediates strengthening of inhibitory synapses by insulin. Here, we show that alleles of rs4454083 modulate transcript levels of the antisense gene, ARBAG, which then controls the expression of the sense gene, GABRA6. Predisposing to low C-peptide, GG (a minor allele genotype across ethnicities) stabilizes ARBAG lncRNA causing higher transcript levels in cerebellum. ARBAG lncRNA abundance leads to cleavage of GABRA6 mRNA at the complementary region, resulting in a dysfunctional GABRA6 protein that would not be recruited for synapse strengthening. Together, our findings in human cerebellar cell-line and induced Pluripotent Stem Cells (iPSCs) demonstrate biological role of a novel lncRNA in determining the ratio of mRNA isoforms of a protein-coding gene and the ability of an embedded variant in modulating lncRNA stability leading to inter-individual differences in protein expression.


Subject(s)
RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , C-Peptide/genetics , C-Peptide/metabolism , Genome-Wide Association Study , Cerebellum/metabolism , RNA, Antisense/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism
8.
Brain ; 147(1): 224-239, 2024 01 04.
Article in English | MEDLINE | ID: mdl-37647766

ABSTRACT

Genetic variants associated with developmental and epileptic encephalopathies have been identified in the GABRB3 gene that encodes the ß3 subunit of GABAA receptors. Typically, variants alter receptor sensitivity to GABA resulting in either gain- or loss-of-function, which correlates with patient phenotypes. However, it is unclear how another important receptor property, desensitization, contributes to the greater clinical severity of gain-of-function variants. Desensitization properties of 20 gain-of-function GABRB3 variant receptors were evaluated using two-electrode voltage-clamp electrophysiology. The parameters measured included current decay rates and steady-state currents. Selected variants with increased or reduced desensitization were also evaluated using whole-cell electrophysiology in transfected mammalian cell lines. Of the 20 gain-of-function variants assessed, 13 were found to alter receptor desensitization properties. Seven variants reduced desensitization at equilibrium, which acts to worsen gain-of-function traits. Six variants accelerated current decay kinetics, which limits gain-of-function traits. All affected patients displayed severe clinical phenotypes with intellectual disability and difficult-to-treat epilepsy. Nevertheless, variants that reduced desensitization at equilibrium were associated with more severe clinical outcomes. This included younger age of first seizure onset (median 0.5 months), movement disorders (dystonia and dyskinesia), epilepsy of infancy with migrating focal seizures (EIMFS) and risk of early mortality. Variants that accelerated current decay kinetics were associated with slightly milder phenotypes with later seizure onset (median 4 months), unclassifiable developmental and epileptic encephalopathies or Lennox-Gastaut syndrome and no movement disorders. Our study reveals that gain-of-function GABRB3 variants can increase or decrease receptor desensitization properties and that there is a correlation with the degree of disease severity. Variants that reduced the desensitization at equilibrium were clustered in the transmembrane regions that constitute the channel pore and correlated with greater disease severity, while variants that accelerated current decay were clustered in the coupling loops responsible for receptor activation and correlated with lesser severity.


Subject(s)
Epilepsy, Generalized , Epilepsy , Movement Disorders , Animals , Humans , Infant, Newborn , Gain of Function Mutation , Mutation/genetics , Epilepsy/genetics , Seizures , Mammals/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism
9.
Differentiation ; 138: 100790, 2024.
Article in English | MEDLINE | ID: mdl-38908344

ABSTRACT

Mutation of the GABRA1 gene is associated with neurodevelopmental defects and epilepsy. GABRA1 encodes for the α1 subunit of the γ-aminobutyric acid type A receptor (GABAAR), which regulates the fast inhibitory impulses of the nervous system. Multiple model systems have been developed to understand the function of GABRA1, but these models have produced complex and, at times, incongruent data. Thus, additional model systems are required to validate and substantiate previous results. We sought to provide initial phenotypic analysis of a novel germline mutant allele. Our analysis provides a solid foundation for the future use of this allele to characterize gabra1 functionally and pharmacologically using zebrafish. We investigated the behavioral swim patterns associated with a nonsense mutation of the zebrafish gabra1 (sa43718 allele) gene. The sa43718 allele causes a decrease in gabra1 mRNA expression, which is associated with light induced hypermotility, one phenotype previously associated with seizure like behavior in zebrafish. Mutation of gabra1 was accompanied by decreased mRNA expression of gabra2, gabra3, and gabra5, indicating a reduction in the expression of additional α sub-units of the GABAAR. Although multiple sub-units were decreased, larvae continued to respond to pentylenetetrazole (PTZ), indicating that a residual GABAAR exists in the sa43718 allele. Proteomics analysis demonstrated that mutation of gabra1 is associated with abnormal expression of proteins that regulate synaptic vesicle fusion, vesicle transport, synapse development, and mitochondrial protein complexes. These data support previous studies performed in a zebrafish nonsense allele created by CRISPR/Cas9 and validate that loss of function mutations in the gabra1 gene result in seizure-like phenotypes with abnormal development of the GABA synapse. Our results add to the existing body of knowledge as to the function of GABRA1 during development and validate that zebrafish can be used to provide complete functional characterization of the gene.


Subject(s)
Alleles , Receptors, GABA-A , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/growth & development , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Loss of Function Mutation , Codon, Nonsense/genetics , Germ-Line Mutation , Phenotype , Seizures/genetics , Seizures/pathology
10.
Pharmacol Rev ; 74(1): 238-270, 2022 01.
Article in English | MEDLINE | ID: mdl-35017178

ABSTRACT

GABAA receptors containing the α6 subunit are highly expressed in cerebellar granule cells and less abundantly in many other neuronal and peripheral tissues. Here, we for the first time summarize their importance for the functions of the cerebellum and the nervous system. The cerebellum is not only involved in motor control but also in cognitive, emotional, and social behaviors. α6ßγ2 GABAA receptors located at cerebellar Golgi cell/granule cell synapses enhance the precision of inputs required for cerebellar timing of motor activity and are thus involved in cognitive processing and adequate responses to our environment. Extrasynaptic α6ßδ GABAA receptors regulate the amount of information entering the cerebellum by their tonic inhibition of granule cells, and their optimal functioning enhances input filtering or contrast. The complex roles of the cerebellum in multiple brain functions can be compromised by genetic or neurodevelopmental causes that lead to a hypofunction of cerebellar α6-containing GABAA receptors. Animal models mimicking neuropsychiatric phenotypes suggest that compounds selectively activating or positively modulating cerebellar α6-containing GABAA receptors can alleviate essential tremor and motor disturbances in Angelman and Down syndrome as well as impaired prepulse inhibition in neuropsychiatric disorders and reduce migraine and trigeminal-related pain via α6-containing GABAA receptors in trigeminal ganglia. Genetic studies in humans suggest an association of the human GABAA receptor α6 subunit gene with stress-associated disorders. Animal studies support this conclusion. Neuroimaging and post-mortem studies in humans further support an involvement of α6-containing GABAA receptors in various neuropsychiatric disorders, pointing to a broad therapeutic potential of drugs modulating α6-containing GABAA receptors. SIGNIFICANCE STATEMENT: α6-Containing GABAA receptors are abundantly expressed in cerebellar granule cells, but their pathophysiological roles are widely unknown, and they are thus out of the mainstream of GABAA receptor research. Anatomical and electrophysiological evidence indicates that these receptors have a crucial function in neuronal circuits of the cerebellum and the nervous system, and experimental, genetic, post-mortem, and pharmacological studies indicate that selective modulation of these receptors offers therapeutic prospects for a variety of neuropsychiatric disorders and for stress and its consequences.


Subject(s)
Cerebellum , Receptors, GABA-A , Animals , Cerebellum/metabolism , Humans , Neurons/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid
11.
Biophys J ; 123(14): 2085-2096, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38400541

ABSTRACT

GABAA receptors (GABAARs) are neurotransmitter-gated ion channels critical for inhibitory synaptic transmission as well as the molecular target for benzodiazepines (BZDs), one of the most widely prescribed class of psychotropic drugs today. Despite structural insight into the conformations underlying functional channel states, the detailed molecular interactions involved in conformational transitions and the physical basis for their modulation by BZDs are not fully understood. We previously identified that alanine substitution at the central residue in the α1 subunit M2-M3 linker (V279A) enhances the efficiency of linkage between the BZD site and the pore gate. Here, we expand on this work by investigating the effect of alanine substitutions at the analogous positions in the M2-M3 linkers of ß2 (I275A) and γ2 (V290A) subunits, which together with α1 comprise typical heteromeric α1ß2γ2 synaptic GABAARs. We find that these mutations confer subunit-specific effects on the intrinsic pore closed-open equilibrium and its modulation by the BZD diazepam (DZ). The mutations α1(V279A) or γ2(V290A) bias the channel toward a closed conformation, whereas ß2(I275A) biases the channel toward an open conformation to the extent that the channel becomes leaky and opens spontaneously in the absence of agonist. In contrast, only α1(V279A) enhances the efficiency of DZ-to-pore linkage, whereas mutations in the other two subunits have no effect. These observations show that the central residue in the M2-M3 linkers of distinct subunits in synaptic α1ß2γ2 GABAARs contribute asymmetrically to the intrinsic closed-open equilibrium and its modulation by DZ.


Subject(s)
Diazepam , Ion Channel Gating , Protein Subunits , Receptors, GABA-A , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Diazepam/pharmacology , Diazepam/chemistry , Ion Channel Gating/drug effects , Protein Subunits/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , Humans , Animals , Mutation , HEK293 Cells
12.
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
13.
Hum Mol Genet ; 31(19): 3216-3230, 2022 09 29.
Article in English | MEDLINE | ID: mdl-34957497

ABSTRACT

Mutations in the γ-aminobutyric acid type A (GABAA) receptor γ2 subunit gene, GABRG2, have been associated with a variety of epilepsy syndromes. A de novo mutation (c.T1027C, p.F343L) in GABRG2 was identified in a patient with early onset epileptic encephalopathy. Zebrafish overexpressing mutant human GABRG2 (F343L) subunits displayed spontaneous seizure activity and convulsive behaviors. In this study, we demonstrated that Tg (hGABRG2F343L) zebrafish displayed hyperactivity during light phase with normal circadian rhythm, as well as increased drug-induced locomotor activity. Real-time quantitative PCR, whole mount in situ hybridization and western blotting showed that Tg(hGABRG2F343L) zebrafish had altered expression of GABAA receptor subunits. Furthermore, investigation of synaptic protein expression and synapse ultrastructure uncovered a robust synaptic phenotype that is causally linked to GABRG2(F343L) mutation. Strikingly, Tg(hGABRG2F343L) zebrafish not only had postsynaptic defects, but also displayed an unanticipated deficit at the presynaptic level. Overall, our Tg(hGABRG2F343L) overexpression zebrafish model has expanded the GABAergic paradigm in epileptic encephalopathy from channelopathy to synaptopathy.


Subject(s)
Brain Diseases , Receptors, GABA-A , Animals , Humans , Mutation , Mutation, Missense/genetics , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Seizures , Zebrafish/genetics , Zebrafish/metabolism , gamma-Aminobutyric Acid/genetics
14.
Eur J Neurosci ; 60(4): 4393-4408, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38858171

ABSTRACT

Animal studies and clinical trials suggest that maintenance of gamma-aminobutyric acid (GABA)-ergic activity may be crucial in coping with stressful conditions, anxiety and mood disorders. Drugs highly efficient in promoting anxiolysis were shown to activate this system, particularly via the α2-subunit of type A receptors (GABAA α2). Given the high expression of GABAA α2 in the dentate gyrus (DG) sub-field of the hippocampus, we sought to examine whether manipulation of the α2 subunit in this area will evoke changes in emotional behaviour, memory and learning as well as in synaptic plasticity. We found that knockdown of GABAAα2 receptor specifically in the dorsal DG of rats caused increased anxiety without affecting locomotor activity. Spatial memory and learning in the Morris water maze were also impaired in GABAAα2 receptor knocked down rats, an effect accompanied by alterations in synaptic plasticity, as assessed by long-term potentiation in the DG. Our findings provide further support to the notion that emotional information processing in the hippocampus may be controlled, at least in part, via the inhibitory GABAA α2 receptor subunit, opening a potential avenue for early interventions from pre- puberty into adulthood, as a strategy for controlling anxiety-related psychopathology.


Subject(s)
Anxiety , Dentate Gyrus , Neuronal Plasticity , Receptors, GABA-A , Animals , Dentate Gyrus/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Anxiety/metabolism , Neuronal Plasticity/physiology , Male , Rats , Memory Disorders/metabolism , Memory Disorders/genetics , Maze Learning/physiology , Gene Knockdown Techniques/methods , Long-Term Potentiation/physiology , Rats, Wistar
15.
J Transl Med ; 22(1): 767, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143639

ABSTRACT

Genetic epilepsy with febrile seizures plus (GEFS+) is a genetic epilepsy syndrome characterized by a marked hereditary tendency inherited as an autosomal dominant trait. Patients with GEFS+ may develop typical febrile seizures (FS), while generalized tonic-clonic seizures (GTCSs) with fever commonly occur between 3 months and 6 years of age, which is generally followed by febrile seizure plus (FS+), with or without absence seizures, focal seizures, or GTCSs. GEFS+ exhibits significant genetic heterogeneity, with polymerase chain reaction, exon sequencing, and single nucleotide polymorphism analyses all showing that the occurrence of GEFS+ is mainly related to mutations in the gamma-aminobutyric acid type A receptor gamma 2 subunit (GABRG2) gene. The most common mutations in GABRG2 are separated in large autosomal dominant families, but their pathogenesis remains unclear. The predominant types of GABRG2 mutations include missense (c.983A → T, c.245G → A, p.Met199Val), nonsense (R136*, Q390*, W429*), frameshift (c.1329delC, p.Val462fs*33, p.Pro59fs*12), point (P83S), and splice site (IVS6+2T → G) mutations. All of these mutations types can reduce the function of ion channels on the cell membrane; however, the degree and mechanism underlying these dysfunctions are different and could be linked to the main mechanism of epilepsy. The γ2 subunit plays a special role in receptor trafficking and is closely related to its structural specificity. This review focused on investigating the relationship between GEFS+ and GABRG2 mutation types in recent years, discussing novel aspects deemed to be great significance for clinically accurate diagnosis, anti-epileptic treatment strategies, and new drug development.


Subject(s)
Mutation , Receptors, GABA-A , Seizures, Febrile , Humans , Receptors, GABA-A/genetics , Seizures, Febrile/genetics , Mutation/genetics , Epilepsy/genetics , Animals
16.
Synapse ; 78(2): e22289, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38436644

ABSTRACT

Epileptic seizures are seen as a result of changing excitability balance depending on the deterioration in synaptic plasticity in the brain. Neuroplastin, and its related molecules which are known to play a role in synaptic plasticity, neurotransmitter activities that provide balance of excitability and, different neurological diseases, have not been studied before in epilepsy. In this study, a total of 34 Sprague-Dawley male and female rats, 2 months old, weighing 250-300 g were used. The epilepsy model in rats was made via pentylenetetrazole (PTZ). After the completion of the experimental procedure, the brain tissue of the rats were taken and the histopathological changes in the hippocampus and cortex parts and the brain stem were investigated, as well as the immunoreactivity of the proteins related to the immunohistochemical methods. As a result of the histopathological evaluation, it was determined that neuron degeneration and the number of dilated blood vessels in the hippocampus, frontal cortex, and brain stem were higher in the PTZ status epilepticus (SE) groups than in the control groups. It was observed that neuroplastin and related proteins TNF receptor-associated factor 6 (TRAF6), Gamma amino butyric acid type A receptors [(GABA(A)], and plasma membrane Ca2+ ATPase (PMCA) protein immunoreactivity levels increased especially in the male hippocampus, and only AMPA receptor subunit type 1 (GluA1) immunoreactivity decreased, unlike other proteins. We believe this may be caused by a problem in the mechanisms regulating the interaction of neuroplastin and GluA1 and may cause problems in synaptic plasticity in the experimental epilepsy model. It may be useful to elucidate this mechanism and target GluA1 when determining treatment strategies.


Subject(s)
Epilepsy , Animals , Female , Male , Rats , Brain Stem/metabolism , Epilepsy/chemically induced , Epilepsy/genetics , Hippocampus/metabolism , Pentylenetetrazole , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , TNF Receptor-Associated Factor 6/genetics , Plasma Membrane Calcium-Transporting ATPases/genetics , Receptors, AMPA/genetics , Cerebral Cortex/metabolism
17.
Epilepsia ; 65(1): 204-217, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37746768

ABSTRACT

OBJECTIVE: γ-Aminobutyric acid type A (GABAA ) receptor subunit gene mutations are major causes of various epilepsy syndromes, including severe kinds such as Dravet syndrome. Although the GABAA receptor is a major target for antiseizure medications, treating GABAA receptor mutations with receptor channel modulators is ineffective. Here, we determined the effect of a novel treatment with 4-phenylbutyrate (PBA) in Gabrg2+/Q390X knockin mice associated with Dravet syndrome. METHODS: We used biochemistry in conjunction with differential tagging of the wild-type and the mutant alleles, live brain slice surface biotinylation, microsome isolation, patch-clamp whole-cell recordings, and video-monitoring synchronized electroencephalographic (EEG) recordings in Gabrg2+/Q390X mice to determine the effect of PBA in vitro with recombinant GABAA receptors and in vivo with knockin mice. RESULTS: We found that PBA reduced the mutant γ2(Q390X) subunit protein aggregates, enhanced the wild-type GABAA receptor subunits' trafficking, and increased the membrane expression of the wild-type receptors. PBA increased the current amplitude of GABA-evoked current in human embryonic kidney 293T cells and the neurons bearing the γ2(Q390X) subunit protein. PBA also proved to reduce endoplasmic reticulum (ER) stress caused by the mutant γ2(Q390X) subunit protein, as well as mitigating seizures and EEG abnormalities in the Gabrg2+/Q390X mice. SIGNIFICANCE: This research has unveiled a promising and innovative approach for treating epilepsy linked to GABAA receptor mutations through an unconventional antiseizure mechanism. Rather than directly modulating the affected mutant channel, PBA facilitates the folding and transportation of wild-type receptor subunits to the cell membrane and synapse. Combining these findings with our previous study, which demonstrated PBA's efficacy in restoring GABA transporter 1 (encoded by SLC6A1) function, we propose that PBA holds significant potential for a wide range of genetic epilepsies. Its ability to target shared molecular pathways involving mutant protein ER retention and impaired protein membrane trafficking suggests broad application in treating such conditions.


Subject(s)
Epilepsies, Myoclonic , Epilepsy , Phenylbutyrates , Mice , Humans , Animals , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, GABA/metabolism , Epilepsies, Myoclonic/drug therapy , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/complications , Seizures/complications , Epilepsy/genetics , gamma-Aminobutyric Acid , Endoplasmic Reticulum Stress/genetics
18.
Cell Mol Life Sci ; 80(10): 280, 2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37684532

ABSTRACT

Nuclear Cyclin D1 (Ccnd1) is a main regulator of cell cycle progression and cell proliferation. Interestingly, Ccnd1 moves to the cytoplasm at the onset of differentiation in neuronal precursors. However, cytoplasmic functions and targets of Ccnd1 in post-mitotic neurons are unknown. Here we identify the α4 subunit of gamma-aminobutyric acid (GABA) type A receptors (GABAARs) as an interactor and target of Ccnd1-Cdk4. Ccnd1 binds to an intracellular loop in α4 and, together with Cdk4, phosphorylates the α4 subunit at threonine 423 and serine 431. These modifications upregulate α4 surface levels, increasing the response of α4-containing GABAARs, measured in whole-cell patch-clamp recordings. In agreement with this role of Ccnd1-Cdk4 in neuronal signalling, inhibition of Cdk4 or expression of the non-phosphorylatable α4 decreases synaptic and extra-synaptic currents in the hippocampus of newborn rats. Moreover, according to α4 functions in synaptic pruning, CCND1 knockout mice display an altered pattern of dendritic spines that is rescued by the phosphomimetic α4. Overall, our findings molecularly link Ccnd1-Cdk4 to GABAARs activity in the central nervous system and highlight a novel role for this G1 cyclin in neuronal signalling.


Subject(s)
Cyclin D1 , Cyclin-Dependent Kinase 4 , Receptors, GABA-A , Animals , Mice , Rats , Cyclin D1/genetics , gamma-Aminobutyric Acid , Mice, Knockout , Neurons , Phosphorylation , Receptors, GABA-A/genetics , Cyclin-Dependent Kinase 4/genetics
19.
Clin Exp Pharmacol Physiol ; 51(3): e13840, 2024 03.
Article in English | MEDLINE | ID: mdl-38302076

ABSTRACT

Remimazolam is a newly developed ultra-short-acting benzodiazepine that exerts sedative effects. This study aimed to clarify the effects of remimazolam on cardiac contractility. In a randomised-parallel group trial, haemodynamic parameters were compared between propofol (n = 11) and remimazolam (n = 12) groups during the induction of general anaesthesia in patients undergoing non-cardiac surgery. In a preclinical study, the direct effects of remimazolam on cardiac contractility were also evaluated using isolated rat hearts. RNA sequence data obtained from rat and human hearts were analysed to assess the expression patterns of the cardiac γ-aminobutyric acid type A (GABAA ) receptor subunits. In a clinical study, the proportional change of the maximum rate of arterial pressure rise was milder during the study period in the remimazolam group (propofol: -52.6 [10.2] (mean [standard deviation])% vs. remimazolam: -39.7% [10.5%], p = 0.007). In a preclinical study, remimazolam did not exert a negative effect on left ventricle developed pressure, whereas propofol did exert a negative effect after bolus administration of a high dose (propofol: -26.9% [3.5%] vs. remimazolam: -1.1 [6.9%], p < 0.001). Analysis of the RNA sequence revealed a lack of γ subunits, which are part of the major benzodiazepine binding site of the GABAA receptor, in rat and human hearts. These results indicate that remimazolam does not have a direct negative effect on cardiac contractility, which might contribute to its milder effect on cardiac contractility during the induction of general anaesthesia. The expression patterns of cardiac GABAA receptor subunits might be associated with the unique pharmacokinetics of benzodiazepines in the heart.


Subject(s)
Propofol , Humans , Animals , Rats , Propofol/pharmacology , Receptors, GABA-A/genetics , Benzodiazepines/pharmacology , gamma-Aminobutyric Acid
20.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33972423

ABSTRACT

GABAergic neurotransmission constitutes a major inhibitory signaling mechanism that plays crucial roles in central nervous system physiology and immune cell immunomodulation. However, its roles in innate immunity remain unclear. Here, we report that deficiency in the GABAergic neuromuscular junctions (NMJs) of Caenorhabditis elegans results in enhanced resistance to pathogens, whereas pathogen infection enhances the strength of GABAergic transmission. GABAergic synapses control innate immunity in a manner dependent on the FOXO/DAF-16 but not the p38/PMK-1 pathway. Our data reveal that the insulin-like peptide INS-31 level was dramatically decreased in the GABAergic NMJ GABAAR-deficient unc-49 mutant compared with wild-type animals. C. elegans with ins-31 knockdown or loss of function exhibited enhanced resistance to Pseudomonas aeruginosa PA14 exposure. INS-31 may act downstream of GABAergic NMJs and in body wall muscle to control intestinal innate immunity in a cell-nonautonomous manner. Our results reveal a signaling axis of synapse-muscular insulin-intestinal innate immunity in vivo.


Subject(s)
Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans/immunology , Immunity, Innate/immunology , Insulin/immunology , Intestines/immunology , Receptors, GABA-A/immunology , Synapses/immunology , Adult , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , GABAergic Neurons/immunology , GABAergic Neurons/metabolism , GABAergic Neurons/microbiology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Insulin/metabolism , Intestines/microbiology , Intestines/physiology , Mutation , Neuromuscular Junction/immunology , Neuromuscular Junction/microbiology , Neuromuscular Junction/physiology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/physiology , Receptors, GABA-A/genetics , Receptors, GABA-A/physiology , Signal Transduction/immunology , Synapses/microbiology , Synapses/physiology , Synaptic Transmission/genetics , Synaptic Transmission/immunology , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL