Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Eur J Pharmacol ; 982: 176911, 2024 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-39179091

RESUMEN

The coronaridine congeners catharanthine and 18-methoxycoronaridine (18-MC) display sedative, anxiolytic, and antidepressant properties by acting on mechanisms involving GABAergic and/or monoaminergic transmissions. Here, we expanded their pharmacological properties by studying their anticonvulsant activity in male and female mice using the pentylenetetrazole (PTZ)-induced seizure test. To determine potential neurochemical mechanisms, the effect of congeners on monoamine content and kainic acid (KA)-induced epileptiform discharge was studied in the hippocampus. The behavioral results showed that coronaridine congeners induce acute anticonvulsant activity in a dose-dependent but sex-independent manner. Repeated treatment with a subthreshold dose (20 mg/kg) of each congener produced anticonvulsant activity in a sex-independent manner, but was significantly higher in male mice when compared to its acute effect. Using a behaviourally relevant regimen, we found that PTZ increased dopamine metabolites and serotonin tissue content. Coronaridine congeners, which induced distinct effects on monoamines, blunted the effect of PTZ instead of potentiating it, suggesting the existence of another mechanism in their anticonvulsant activity. The electrophysiological results indicated that both congeners inhibit KA-induced epileptiform discharges in hippocampal slices. A key aspect of this study is that the activity of both congeners was observed only in the presence of GABA, supporting the notion that hippocampal GABAAR potentiation plays an important role. Our study showed that coronaridine congeners induce acute anticonvulsant activity in a sex-independent manner. However, a comparatively higher susceptibility was observed in male mice after repeated treatment. The underlying hippocampal mechanisms mainly involve GABAAR potentiation, whereas monoamines play a minor role in the anticonvulsive action.


Asunto(s)
Anticonvulsivantes , Hipocampo , Receptores de GABA-A , Convulsiones , Animales , Masculino , Anticonvulsivantes/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Femenino , Receptores de GABA-A/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Convulsiones/fisiopatología , Pentilenotetrazol , Ibogaína/análogos & derivados , Ibogaína/farmacología , Relación Dosis-Respuesta a Droga
2.
Int J Mol Sci ; 25(10)2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38791206

RESUMEN

Dementia exists as a 'progressive clinical syndrome of deteriorating mental function significant enough to interfere with activities of daily living', with the most prevalent type of dementia being Alzheimer's disease (AD), accounting for about 80% of diagnosed cases. AD is associated with an increased risk of comorbidity with other clinical conditions such as hypertension, diabetes, and neuropsychiatric symptoms (NPS) including, agitation, anxiety, and depression as well as increased mortality in late life. For example, up to 70% of patients diagnosed with AD are affected by anxiety. As aging is the major risk factor for AD, this represents a huge global burden in ageing populations. Over the last 10 years, significant efforts have been made to recognize the complexity of AD and understand the aetiology and pathophysiology of the disease as well as biomarkers for early detection. Yet, earlier treatment options, including acetylcholinesterase inhibitors and glutamate receptor regulators, have been limited as they work by targeting the symptoms, with only the more recent FDA-approved drugs being designed to target amyloid-ß protein with the aim of slowing down the progression of the disease. However, these drugs may only help temporarily, cannot stop or reverse the disease, and do not act by reducing NPS associated with AD. The first-line treatment options for the management of NPS are selective serotonin reuptake inhibitors/selective noradrenaline reuptake inhibitors (SSRIs/SNRIs) targeting the monoaminergic system; however, they are not rational drug choices for the management of anxiety disorders since the GABAergic system has a prominent role in their development. Considering the overall treatment failures and side effects of currently available medication, there is an unmet clinical need for rationally designed therapies for anxiety disorders associated with AD. In this review, we summarize the current status of the therapy of AD and aim to highlight novel angles for future drug therapy in our ongoing efforts to alleviate the cognitive deficits and NPS associated with this devastating disease.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/etiología , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Animales , Inhibidores de la Colinesterasa/uso terapéutico
3.
Brain Commun ; 6(2): fcae110, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38650830

RESUMEN

We have previously characterized the molecular mechanisms for variants in γ-aminobutyric acid transporter 1-encoding solute carrier family 6-member 1 (SLC6A1) in vitro and concluded that a partial or complete loss of γ-aminobutyric acid uptake due to impaired protein trafficking is the primary aetiology. Impairment of γ-aminobutyric acid transporter 1 function could cause compensatory changes in the expression of γ-aminobutyric acid receptors, which, in turn, modify disease pathophysiology and phenotype. Here we used different approaches including radioactive 3H γ-aminobutyric acid uptake in cells and synaptosomes, immunohistochemistry and confocal microscopy as well as brain slice surface protein biotinylation to characterize Slc6a1+/A288V and Slc6a1+/S295L mice, representative of a partial or a complete loss of function of SLC6A1 mutations, respectively. We employed the γ-aminobutyric acid transporter 1-specific inhibitor [3H]tiagabine binding and GABAA receptor subunit-specific radioligand binding to profile the γ-aminobutyric acid transporter 1 and GABAA receptor expression in major brain regions such as cortex, cerebellum, hippocampus and thalamus. We also determined the total and surface expression of γ-aminobutyric acid transporter 1, γ-aminobutyric acid transporter 3 and expression of GABAA receptor in the major brain regions in the knockin mice. We found that γ-aminobutyric acid transporter 1 protein was markedly reduced in cortex, hippocampus, thalamus and cerebellum in both mutant mouse lines. Consistent with the findings of reduced γ-aminobutyric acid uptake for both γ-aminobutyric acid transporter 1(A288V) and γ-aminobutyric acid transporter 1(S295L), both the total and the γ-aminobutyric acid transporter 1-mediated 3H γ-aminobutyric acid reuptake was reduced. We found that γ-aminobutyric acid transporter 3 is only abundantly expressed in the thalamus and there was no compensatory increase of γ-aminobutyric acid transporter 3 in either of the mutant mouse lines. γ-Aminobutyric acid transporter 1 was reduced in both somatic regions and nonsomatic regions in both mouse models, in which a ring-like structure was identified only in the Slc6a1+/A288V mouse, suggesting more γ-aminobutyric acid transporter 1 retention inside endoplasmic reticulum in the Slc6a1+/A288V mouse. The [3H]tiagabine binding was similar in both mouse models despite the difference in γ-aminobutyric acid uptake function and γ-aminobutyric acid transporter 1 protein expression for both mutations. There were no differences in GABAA receptor subtype expression, except for a small increase in the expression of α5 subunits of GABAA receptor in the hippocampus of Slc6a1S295L homozygous mice, suggesting a potential interaction between the expression of this GABAA receptor subtype and the mutant γ-aminobutyric acid transporter 1. The study provides the first comprehensive characterization of the SLC6A1 mutations in vivo in two representative mouse models. Because both γ-aminobutyric acid transporter 1 and GABAA receptors are targets for anti-seizure medications, the findings from this study can help guide tailored treatment options based on the expression and function of γ-aminobutyric acid transporter 1 and GABAA receptor in SLC6A1 mutation-mediated neurodevelopmental and epileptic encephalopathies.

4.
Biomolecules ; 13(3)2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36979350

RESUMEN

Febrile seizures (FS) are the most common form of epilepsy in children between six months and five years of age. FS is a self-limited type of fever-related seizure. However, complicated prolonged FS can lead to complex partial epilepsy. We found that among the GABAA receptor subunit (GABR) genes, most variants associated with FS are harbored in the γ2 subunit (GABRG2). Here, we characterized the effects of eight variants in the GABAA receptor γ2 subunit on receptor biogenesis and channel function. Two-thirds of the GABRG2 variants followed the expected autosomal dominant inheritance in FS and occurred as missense and nonsense variants. The remaining one-third appeared as de novo in the affected probands and occurred only as missense variants. The loss of GABAA receptor function and dominant negative effect on GABAA receptor biogenesis likely caused the FS phenotype. In general, variants in the GABRG2 result in a broad spectrum of phenotypic severity, ranging from asymptomatic, FS, genetic epilepsy with febrile seizures plus (GEFS+), and Dravet syndrome individuals. The data presented here support the link between FS, epilepsy, and GABRG2 variants, shedding light on the relationship between the variant topological occurrence and disease severity.


Asunto(s)
Epilepsias Mioclónicas , Epilepsia , Convulsiones Febriles , Humanos , Convulsiones Febriles/genética , Receptores de GABA-A/genética , Epilepsias Mioclónicas/genética , Epilepsia/genética , Mutación Missense , Mutación
5.
Neuron ; 111(7): 1104-1117.e6, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36681074

RESUMEN

Addictive drugs increase ventral tegmental area (VTA) dopamine (DA) neuron activity through distinct cellular mechanisms, one of which involves disinhibition of DA neurons by inhibiting local GABA neurons. How drugs regulate VTA GABA neuron activity and drive addictive behaviors remains poorly understood. Here, we show that astrocytes control VTA GABA neuron activity in cocaine reward via tonic inhibition in mice. Repeated cocaine exposure potentiates astrocytic tonic GABA release through volume-regulated anion channels (VRACs) and augments tonic inhibition of VTA GABA neurons, thus downregulating their activities and disinhibiting nucleus accumbens (NAc) projecting DA neurons. Attenuation of tonic inhibition by either deleting Swell1 (Lrrc8a), the obligatory subunit of VRACs, in VTA astrocytes or disrupting δ subunit of GABAA receptors in VTA GABA neurons reduces cocaine-evoked changes in neuron activity, locomotion, and reward behaviors in mice. Together, our findings reveal the critical role of astrocytes in regulating the VTA local circuit and cocaine reward.


Asunto(s)
Cocaína , Ratones , Animales , Cocaína/farmacología , Área Tegmental Ventral/fisiología , Astrocitos , Neuronas Dopaminérgicas , Receptores de GABA-A , Ácido gamma-Aminobutírico , Recompensa , Proteínas de la Membrana
6.
Front Psychiatry ; 14: 1296527, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38268565

RESUMEN

Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system. In the mature brain, inhibitory GABAergic signaling is critical in maintaining neuronal homeostasis and vital human behaviors such as cognition, emotion, and motivation. While classically known to inhibit neuronal function under physiological conditions, previous research indicates a paradoxical switch from inhibitory to excitatory GABAergic signaling that is implicated in several neurological disorders. Various mechanisms have been proposed to contribute to the excitatory switch such as chloride ion dyshomeostasis, alterations in inhibitory receptor expression, and modifications in GABAergic synaptic plasticity. Of note, the hypothesized mechanisms underlying excitatory GABAergic signaling are highlighted in a number of neurodevelopmental, substance use, stress, and neurodegenerative disorders. Herein, we present an updated review discussing the presence of excitatory GABAergic signaling in various neurological disorders, and their potential contributions towards disease pathology.

7.
Mol Neurobiol ; 59(10): 6049-6061, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35849280

RESUMEN

Chronic neuropathic pain often leads to cognitive impairment, but the exact mechanism remains unclear. Gamma-aminobutyric acid A receptors (GABAARs) are the major inhibitory receptors in the brain, of which the α5-containing GABAARs (GABAARs-α5) are implicated in a range of neuropsychiatric disorders with cognitive deficits. However, whether GABAARs-α5 are involved in chronic neuropathic pain-related cognitive impairment remains unknown. In this study, the rats with chronic neuropathic pain induced by right sciatic nerve ligation injury (SNI) exhibited cognitive impairment with declined spontaneous alternation in Y-maze test and discrimination index in novel object recognition test. The GABAARs-α5 expressing on parvalbumin and somatostatin interneurons increased remarkably in hippocampus, resulting in decreased mean frequency of spontaneous inhibitory postsynaptic currents in hippocampal pyramidal neurons. Significantly, antagonizing the GABAARs-α5 by L655708 rescued weakened inhibitory synaptic transmission and cognitive impairment induced by chronic neuropathic pain. Taken together, these data suggest that the GABAARs-α5 play a crucial role in chronic neuropathic pain-induced cognitive impairment by weakening inhibitory synaptic transmission, which may provide insights into the pharmacologic treatment of chronic neuropathic pain-related cognitive impairment.


Asunto(s)
Disfunción Cognitiva , Neuralgia , Animales , Emparejamiento Cromosómico , Disfunción Cognitiva/complicaciones , Hipocampo/metabolismo , Neuralgia/complicaciones , Ratas , Receptores de GABA , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico
8.
Front Behav Neurosci ; 16: 893835, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35755407

RESUMEN

Gamma-aminobutyric acid type B receptor (GABABR) has been extensively involved in alcohol use disorders; however, the mechanisms by which this receptor modulates alcohol drinking behavior remain murky. In this study, we investigate alcohol consumption and preference in mice lacking functional GABABR using the 2-bottle choice paradigm. We found that GABAB(1), knockout (KO), and heterozygous (HZ) mice drank higher amounts of an alcoholic solution, preferred alcohol to water, and reached higher blood alcohol concentrations (BACs) compared to wild-type (WT) littermates. The GABABR agonist GHB significantly reduced alcohol consumption in the GABAB(1) HZ and WT but not in the KO mice. Next, because of a functional crosstalk between GABABR and δ-containing GABAA receptor (δ-GABA A R), we profiled δ subunit mRNA expression levels in brain regions in which the crosstalk was characterized. We found a loss of the alcohol-sensitive GABAAR δ subunit in the hippocampus of the GABAB(1) KO alcohol-naïve mice that was associated with increased É£2 subunit abundance. Electrophysiological recordings revealed that these molecular changes were associated with increased phasic inhibition, suggesting a potential gain of synaptic GABAAR responsiveness to alcohol that has been previously described in an animal model of excessive alcohol drinking. Interestingly, voluntary alcohol consumption did not revert the dramatic loss of hippocampal δ-GABAAR occurring in the GABAB(1) KO mice but rather exacerbated this condition. Finally, we profiled hippocampal neuroactive steroids levels following acute alcohols administration in the GABAB(1) KO and WT mice because of previous involvement of GABABR in the regulation of cerebral levels of these compounds. We found that systemic administration of alcohol (1.5 g/kg) did not produce alcohol-induced neurosteroid response in the GABAB(1) KO mice but elicited an expected increase in the hippocampal level of progesterone and 3α,5α-THP in the WT controls. In conclusion, we show that genetic ablation of the GABAB(1) subunit results in increased alcohol consumption and preference that were associated with functional changes in hippocampal GABAAR, suggesting a potential mechanism by which preference for alcohol consumption is maintained in the GABAB(1) KO mice. In addition, we documented that GABAB(1) deficiency results in lack of alcohol-induced neurosteroids, and we discussed the potential implications of this finding in the context of alcohol drinking and dependence.

9.
Schizophr Res ; 249: 93-97, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-34916095

RESUMEN

BACKGROUND: Accumulating evidence suggests that the pathology of some psychiatric symptoms may relate to autoantibodies against various neuronal surface antigens, such as NMDA receptors (NMDARs) or inhibitory GABAA receptors (GABAARs). However, it is unclear whether the plasma of patients with schizophrenia contains autoantibodies targeting to NMDARs or GABAARs. METHODS: Serum samples of 293 patients with schizophrenia were analyzed using a combination of live-cell-based assay (CBA) and immunostaining on primary neurons to quantify the positive rate of autoantibodies targeting NMDARs or GABAARs. RESULTS: Only one sample was found positive for anti-NMDAR autoantibodies, and no surface autoantibodies against GABAARs were found. No obvious difference in clinical manifestations was observed between the patients with positive and negative anti-NMDAR autoantibodies. CONCLUSIONS: Our results suggest that autoantibodies against NMDARs or GABAARs may affect only a small group of patients with schizophrenia, and the rates of these autoantibodies are lower than reported in prior work. It would be interesting to perform studies with psychotic disorder instead of schizophrenia to determine whether NMDAR or GABAAR autoantibody can be used as a biomarker to provide a new avenue for immunomodulatory therapy.


Asunto(s)
Esquizofrenia , Humanos , Autoanticuerpos , Receptores de GABA-A , Receptores de N-Metil-D-Aspartato , Ácido gamma-Aminobutírico
10.
Neuroscience ; 449: 147-156, 2020 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-32926954

RESUMEN

Dendrite-targeting somatostatin-expressing interneurons (SST-INs) powerfully control signal integration and synaptic plasticity in pyramidal dendrites during cortical development. We previously showed that synaptic transmission from SST-INs to pyramidal cells (PCs) (SST-IN → PC) in the mouse visual cortex suddenly declined at around the second postnatal week. However, it is unclear what specific postsynaptic mechanisms underlie this developmental change. Using multiple whole-cell patch-clamp recordings, we found that application of an α5-GABAA receptor-selective inverse agonist, alpha5IA, significantly weakened SST-IN → PC unitary inhibitory postsynaptic currents (uIPSCs) in layer 2/3 of the mouse visual cortex, but had no effect on uIPSCs from SST-INs to other types of interneurons. The extent of alpha5IA-induced reduction of SST-IN → PC synaptic transmission was significantly larger at postnatal days 11-13 (P11-13) than P14-17. Moreover, α5-subunit-containing GABAA receptors (α5-GABAARs)-mediated uIPSCs had slow rise and decay kinetics. Apart from pharmacological test, we observed that SST-IN → PC synapses did indeed contain α5-GABAARs by immunogold labeling for electron microscopy. More importantly, coinciding with the weakening of SST-IN → PC synaptic transmission, the number of α5-GABAAR particles in SST-IN → PC synapses significantly decreased at around the second postnatal week. Together, these data indicate that α5-GABAARs are involved in synaptic transmission from SST-INs to PCs in the neocortex, and are significantly diminished around the second postnatal week.


Asunto(s)
Receptores de GABA-A , Corteza Visual , Animales , Interneuronas/metabolismo , Ratones , Neuronas/metabolismo , Células Piramidales/metabolismo , Receptores de GABA-A/metabolismo , Somatostatina , Transmisión Sináptica , Corteza Visual/metabolismo , Ácido gamma-Aminobutírico
11.
Cell Rep ; 31(12): 107785, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32579917

RESUMEN

Molecular mechanisms underlying plasticity at brain inhibitory synapses remain poorly characterized. Increased postsynaptic clustering of GABAA receptors (GABAARs) rapidly strengthens inhibition during inhibitory long-term potentiation (iLTP). However, it is unclear how synaptic GABAAR clustering is maintained to sustain iLTP. Here, we identify a role for miR376c in regulating the translation of mRNAs encoding the synaptic α1 and γ2 GABAAR subunits, GABRA1 and GABRG2, respectively. Following iLTP induction, transcriptional repression of miR376c is induced through a calcineurin-NFAT-HDAC signaling pathway and promotes increased translation and clustering of synaptic GABAARs. This pathway is essential for the long-term expression of iLTP and is blocked by miR376c overexpression, specifically impairing inhibitory synaptic strength. Finally, we show that local de novo synthesis of synaptic GABAARs occurs exclusively in dendrites and in a miR376c-dependent manner following iLTP. Together, this work describes a local post-transcriptional mechanism that regulates inhibitory synaptic plasticity via miRNA control of dendritic protein synthesis.


Asunto(s)
Potenciación a Largo Plazo/genética , MicroARNs/genética , Biosíntesis de Proteínas/genética , Receptores de GABA-A/genética , Animales , Secuencia de Bases , Calcineurina/metabolismo , Dendritas/metabolismo , Regulación de la Expresión Génica , Silenciador del Gen , Células HEK293 , Humanos , MicroARNs/metabolismo , Factores de Transcripción NFATC/metabolismo , Inhibición Neural , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de GABA-A/metabolismo , Sinapsis/metabolismo , Transcripción Genética
12.
Int J Mol Sci ; 21(3)2020 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-31979108

RESUMEN

γ-Aminobutyric acid type A receptors (GABAARs) are the main inhibitory mediators in the central nervous system (CNS). GABAARs are pentameric ligand gated ion channels, and the main subunit composition is usually 2α2ßγ, with various isotypes assembled within a set of 19 different subunits. The inhibitory function is mediated by chloride ion movement across the GABAARs, activated by synaptic GABA release, reducing neuronal excitability in the adult CNS. Several studies highlighted the importance of GABA-mediated transmission during neuro-development, and its involvement in different neurological and neurodevelopmental diseases, from anxiety to epilepsy. However, while it is well known how different classes of drugs are able to modulate the GABAARs function (benzodiazepines, barbiturates, neurosteroids, alcohol), up to now little is known about GABAARs and cannabinoids interaction in the CNS. Endocannabinoids and phytocannabinoids are lately emerging as a new class of promising drugs for a wide range of neurological conditions, but their safety as medication, and their mechanisms of action are still to be fully elucidated. In this review, we will focus our attention on two of the most promising molecules (Δ9-tetrahydrocannabinol; Δ9-THC and cannabidiol; CBD) of this new class of drugs and their possible mechanism of action on GABAARs.


Asunto(s)
Cannabinoides/farmacología , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/metabolismo , Receptores de GABA-A/metabolismo , Animales , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Humanos
13.
Neuroscience ; 439: 125-136, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31356900

RESUMEN

To be highly reliable, synaptic transmission needs postsynaptic receptors (Rs) in precise apposition to the presynaptic release sites. At inhibitory synapses, the postsynaptic protein gephyrin self-assembles to form a scaffold that anchors glycine and GABAARs to the cytoskeleton, thus ensuring the accurate accumulation of postsynaptic receptors at the right place. This protein undergoes several post-translational modifications which control protein-protein interaction and downstream signaling pathways. In addition, through the constant exchange of scaffolding elements and receptors in and out of synapses, gephyrin dynamically regulates synaptic strength and plasticity. The aim of the present review is to highlight recent findings on the functional role of gephyrin at GABAergic inhibitory synapses. We will discuss different approaches used to interfere with gephyrin in order to unveil its function. In addition, we will focus on the impact of gephyrin structure and distribution at the nanoscale level on the functional properties of inhibitory synapses as well as the implications of this scaffold protein in synaptic plasticity processes. Finally, we will emphasize how gephyrin genetic mutations or alterations in protein expression levels are implicated in several neuropathological disorders, including autism spectrum disorders, schizophrenia, temporal lobe epilepsy and Alzheimer's disease, all associated with severe deficits of GABAergic signaling. This article is part of a Special Issue entitled: Honoring Ricardo Miledi - outstanding neuroscientist of XX-XXI centuries.


Asunto(s)
Proteínas de la Membrana , Sinapsis , Proteínas Portadoras , Humanos , Proteínas de la Membrana/metabolismo , Receptores de GABA-A , Sinapsis/metabolismo
14.
Cell Rep ; 29(3): 671-684.e6, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31618635

RESUMEN

Precisely controlling the excitatory and inhibitory balance is crucial for the stability and information-processing ability of neuronal networks. However, the molecular mechanisms maintaining this balance during ongoing sensory experiences are largely unclear. We show that Nogo-A signaling reciprocally regulates excitatory and inhibitory transmission. Loss of function for Nogo-A signaling through S1PR2 rapidly increases GABAAR diffusion, thereby decreasing their number at synaptic sites and the amplitude of GABAergic mIPSCs at CA3 hippocampal neurons. This increase in GABAAR diffusion rate is correlated with an increase in Ca2+ influx and requires the calcineurin-mediated dephosphorylation of the γ2 subunit at serine 327. These results suggest that Nogo-A signaling rapidly strengthens inhibitory GABAergic transmission by restricting the diffusion dynamics of GABAARs. Together with the observation that Nogo-A signaling regulates excitatory transmission in an opposite manner, these results suggest a crucial role for Nogo-A signaling in modulating the excitation and inhibition balance to restrict synaptic plasticity.


Asunto(s)
Proteínas Nogo/metabolismo , Receptores de GABA-A/metabolismo , Animales , Anticuerpos Bloqueadores/inmunología , Calcineurina/metabolismo , Calcio/metabolismo , Células Cultivadas , Femenino , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nogo/inmunología , Técnicas de Placa-Clamp , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Wistar , Receptores de GABA-A/genética , Transducción de Señal , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Receptores de Esfingosina-1-Fosfato/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica
15.
Biomed Pharmacother ; 118: 109239, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31351431

RESUMEN

Diazepam could regulate immune system and inflammation, which might be a potential therapeutic agent for pulmonary fibrosis in clinic. This study showed that diazepam reversed LPS-induced inhibition of cell proliferation and promotion of cell apoptosis. Of note, LPS specifically induced Caspase-11 dependent cell pyroptosis, which were significantly attenuated by diazepam or pyroptosis inhibitor necrosulfonamide (NSA) treatment. In addition, α4- and α5-subunits of GABAARs were highly expressed in human bronchial 16HBE cells, human pulmonary epithelial cells (BEAS-2B) and pulmonary epithelial cells isolated from mice (mPECs). Further results showed that only knock-down of α4-GABAARs abrogated the effects of diazepam on LPS induced cell pyroptosis, apoptosis and proliferation. Similiarly, either diazepam or NSA treatment could alleviate development of LPS induced inflammatory reactions and pulmonary fibrosis in mice, which were abrogated by synergistically knocking down α4-GABAARs. Taken together, diazepam alleviated LPS-induced cell pyroptosis and development of pulmonary fibrosis in mice by activating α4-GABAARs.


Asunto(s)
Diazepam/uso terapéutico , Fibrosis Pulmonar/tratamiento farmacológico , Piroptosis , Receptores de GABA-A/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Diazepam/farmacología , Modelos Animales de Enfermedad , Humanos , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Lipopolisacáridos , Ratones Endogámicos C57BL , Fibrosis Pulmonar/patología , Piroptosis/efectos de los fármacos
16.
Neurotoxicol Teratol ; 74: 106812, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31251980

RESUMEN

BACKGROUND: General anesthetics (GAs) may exert harmful effects on the developing brain by disrupting neuronal circuit formation. Anesthetics that act on γ-aminobutyric acid (GABA) receptors can interfere with axonal growth cone guidance, a critical process in the assembly of neuronal circuitry. Here we investigate the mechanism by which isoflurane prevents sensing of the repulsive guidance cue, Semaphorin 3A (Sema3A). METHODS: Growth cone sensing was assayed by measuring growth cone collapse in dissociated neocortical cultures exposed to recombinant Sema3A in the presence or absence of isoflurane and/or a panel of reagents with specific actions on components of the GABA receptor and chloride ion systems. RESULTS: Isoflurane exposure prevents Sema3A induced growth cone collapse. A GABAA α2 specific agonist replicates this effect (36.83 ±â€¯3.417% vs 70.82 ±â€¯2.941%, in the Sema3A induced control group, p < 0.0001), but an α1-specific agonist does not. Both a Na-K-Cl cotransporter 1 antagonism (bumetanide, BUM) and a chloride ionophore (IONO) prevent isoflurane from disrupting growth cone sensing of Sema3A. (65.67 ±â€¯3.775% in Iso + BUM group vs 67.45 ±â€¯3.624% in Sema3A induced control group, 65.34 ±â€¯1.678% in Iso + IONO group vs 68.71 ±â€¯2.071% in Sema3A induced control group, no significant difference) (n = 96 growth cones per group). CONCLUSION: Our data suggest that the effects of isoflurane on growth cone sensing are mediated by the α2 subunit of the GABAA receptor and also that they are dependent on the developmental chloride gradient, in which Cl- exhibits a depolarizing effect. These findings provide a rationale for why immature neurons are particularly susceptible to anesthetic toxicity.


Asunto(s)
Anestésicos por Inhalación/farmacología , Orientación del Axón/efectos de los fármacos , Cloruros/metabolismo , Conos de Crecimiento/efectos de los fármacos , Isoflurano/farmacología , Receptores de GABA-A/metabolismo , Semaforina-3A/metabolismo , Animales , Conos de Crecimiento/metabolismo , Cultivo Primario de Células , Ratas Sprague-Dawley
17.
Artículo en Inglés | MEDLINE | ID: mdl-30951784

RESUMEN

Temporal lobe epilepsy (TLE), which is one of the most common neurological diseases, is accompanied by a high incidence of psychiatric disorders. Among these psychiatric disorders, anxiety is one of the major psychiatric comorbidities in epilepsy patients. However, anxiety in epilepsy patients often remains unrecognized and untreated. It is believed that the inhibitory networks of γ-aminobutyric acid (GABA) neurotransmission play pivotal roles in the modulation of emotion and mood responses in both physiological and pathological conditions. The impairment of neurotransmission mediated by GABAergic signaling is related to the pathophysiology of anxiety. However, it remains unclear whether and how GABAergic signaling modulates anxiety responses in the context of an epileptic brain. In the present study, we sought to determine the role of inhibitory networks of GABAergic signaling in the anxiety-like behavior of epileptic mice. Our results show epileptic mice exhibited increased anxiety-like behavior, and this increased anxiety-like behavior was accompanied by a decrease in GABAergic interneurons and an increase in GABA type A receptor (GABAAR) ß3 subunit (GABRB3) expression in the hippocampus. Furthermore, the activation of GABAARs produced an anxiolytic-like effect, while the inhibition of GABAARs elicited an anxiogenic-like effect in the epileptic mice, suggesting that the alteration of GABAergic signaling is associated with anxiety-like behavior in epileptic mice. Thus, targeting GABAergic signaling in the epileptic brain may provide an effective anxiolytic treatment in epilepsy patients.


Asunto(s)
Ansiedad/etiología , Epilepsia del Lóbulo Temporal/complicaciones , Neuronas GABAérgicas/fisiología , Animales , Ansiedad/metabolismo , Ansiedad/fisiopatología , Western Blotting , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/fisiopatología , Neuronas GABAérgicas/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiopatología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Muscimol/farmacología , Picrotoxina/farmacología , Pilocarpina/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de GABA-A/metabolismo
18.
Biochem Biophys Res Commun ; 511(2): 356-362, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30791981

RESUMEN

The endoplasmic reticulum-Golgi intermediate compartment protein-53 (ERGIC-53, aka LMAN1), which cycles between the endoplasmic reticulum (ER) and Golgi, is a known cargo receptor for a number of soluble proteins. However, whether LMAN1 plays a role as a trafficking factor in the central nervous system is largely unknown. Here, we determined the role of LMAN1 on endogenous protein levels of the Cys-loop superfamily of neuroreceptors, including gamma-aminobutyric acid type A receptors (GABAARs), 5-hydroxytryptamine (serotonin) type 3 (5-HT3) receptors, and nicotinic acetylcholine receptors (nAChRs). Knockdown of LMAN1 reduces the surface trafficking of endogenous ß3 subunits of GABAARs in mouse hypothalamic GT1-7 neurons. Furthermore, Western blot analysis of brain homogenates from LMAN1 knockout mice demonstrated that loss of LMAN1 decreases the total protein levels of 5HT3A receptors and γ2 subunits of GABAARs. LMAN1 knockout regulates the ER proteostasis network by upregulating ERP44 without changing calnexin levels. Interestingly, despite the critical role of the glycan-binding function of LMAN1 in its other known cargo clients, LMAN1 interacts with GABAARs in a glycan-independent manner. In summary, LMAN1 is a trafficking factor for certain neuroreceptors in the central nervous system. This is the first report of LMAN1 function in membrane protein trafficking.


Asunto(s)
Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de GABA-A/metabolismo , Receptores de Serotonina 5-HT3/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular , Humanos , Lectinas de Unión a Manosa/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Transporte de Proteínas
19.
J Biomol Struct Dyn ; 37(12): 3065-3080, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30608219

RESUMEN

The γ-amino butyric acid type A receptors (GABAA-Rs) are GABA-gated chloride ion channels that mediate fast inhibitory neurotransmissions. Due to their essential role in normal brain function, neuromodulatory therapies are targeted at them for restoring GABA-mediated inhibition. The receptor modulation by benzodiazepine (BZD) shows therapeutically useful actions. The mechanisms, by which BZD-site performs selective transduction while modulating GABAA-Rs, and its correlation with the occurrence of sedation is not fully known. In pursuance, we performed a computational study starting from modeling of α2-subtype GABAA-R, docking of α1/2-selective ligands followed by molecular dynamics simulations of the obtained complexes. The results show that during early stages of activation, a) allosteric binding initiate structural changes through BZD-site for GABA-elicited activation; b) selective BZD-binders positively modulate orthosteric GABA-bound site with fin-like C- and F-loop movements, which supports twisting of inner and outer ß-barrel; c) modulation by α1/2-selective ligands was only evident at site 1, mimicking mandatory doubly bound state; d) strength of allosteric communication was prominent for α2-modulators, however, the basic nature of allosteric-orthosteric site cross-talk remains same for both α1/2-modulators; and e) ratio of hydrophobic:hydrophilic ligand contact surface decides α2-selectivity, less value of ratio favors it. These insights would enable us to design better α2-selective modulator/s. Altogether our computational study reveals early stages of allosteric modulation, highlighting subtype selective activation and pathways recommending GABA binding sites during selective modulation. Communicated by Ramaswamy H. Sarma.


Asunto(s)
Regulación Alostérica/fisiología , Sitio Alostérico/fisiología , Sitios de Unión/fisiología , Unión Proteica/fisiología , Receptores de GABA/metabolismo , Benzodiazepinas/metabolismo , Humanos , Ligandos , Simulación de Dinámica Molecular , Subunidades de Proteína/metabolismo , Receptores de GABA-A/metabolismo , Relación Estructura-Actividad
20.
J Biomol Struct Dyn ; 37(6): 1597-1615, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29633901

RESUMEN

γ-aminobutyric acid-type A (GABAA) receptors mediate fast synaptic inhibition in the central nervous system of mammals. They are modulated via several sites by numerous compounds, which include GABA, benzodiazepines, ethanol, neurosteroids and anaesthetics among others. Due to their potential as targets of novel drugs, a detailed knowledge of their structure-function relationships is needed. Here, we present the model of the α1ß2γ2 subtype GABAA receptor in the APO state and in complex with selected ligands, including agonists, antagonists and allosteric modulators. The model is based on the crystallographic structure of the human ß3 homopentamer GABAA receptor. The complexes were refined using atomistic molecular dynamics simulations. This allowed a broad description of the binding modes and the detection of important interactions in agreement with experimental information. From the best of our knowledge, this is the only model of the α1ß2γ2 GABAA receptor that represents altogether the desensitized state of the channel and comprehensively describes the interactions of ligands of the orthosteric and benzodiazepines binding sites in agreement with the available experimental data. Furthermore, it is able to explain small differences regarding the binding of a variety of chemically divergent ligands. Finally, this new model may pave the way for the design of focused experimental studies that will allow a deeper description of the receptor.


Asunto(s)
Benzodiazepinas/química , Antagonistas de Receptores de GABA-A/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Receptores de GABA-A/química , Secuencia de Aminoácidos , Benzodiazepinas/farmacología , Sitios de Unión , Descubrimiento de Drogas/métodos , Antagonistas de Receptores de GABA-A/farmacología , Enlace de Hidrógeno , Ligandos , Reproducibilidad de los Resultados , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA