Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 536
Filtrar
1.
CNS Neurosci Ther ; 30(5): e14716, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38698533

RESUMO

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.


Assuntos
Anestésicos Inalatórios , Hipocampo , Transtornos da Memória , Receptores de GABA-A , Sevoflurano , Sevoflurano/toxicidade , Animais , Camundongos , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/metabolismo , Anestésicos Inalatórios/toxicidade , Receptores de GABA-A/metabolismo , Receptores de GABA-A/biossíntese , Receptores de GABA-A/genética , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia
2.
J Neuroinflammation ; 18(1): 204, 2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34530841

RESUMO

BACKGROUND: Perioperative neurocognitive disorder (PND) is a long-term postoperative complication in elderly surgical patients. The underlying mechanism of PND is unclear, and no effective therapies are currently available. It is believed that neuroinflammation plays an important role in triggering PND. The secreted glycoprotein myeloid differentiation factor 2 (MD2) functions as an activator of the Toll-like receptor 4 (TLR4) inflammatory pathway, and α5GABAA receptors (α5GABAARs) are known to play a key role in regulating inflammation-induced cognitive deficits. Thus, in this study, we aimed to investigate the role of MD2 in PND and determine whether α5GABAARs are involved in the function of MD2. METHODS: Eighteen-month-old C57BL/6J mice were subjected to laparotomy under isoflurane anesthesia to induce PND. The Barnes maze was used to assess spatial reference learning and memory, and the expression of hippocampal MD2 was assayed by western blotting. MD2 expression was downregulated by bilateral injection of AAV-shMD2 into the hippocampus or tail vein injection of the synthetic MD2 degrading peptide Tat-CIRP-CMA (TCM) to evaluate the effect of MD2. Primary cultured neurons from brain tissue block containing cortices and hippocampus were treated with Tat-CIRP-CMA to investigate whether downregulating MD2 expression affected the expression of α5GABAARs. Electrophysiology was employed to measure tonic currents. For α5GABAARs intervention experiments, L-655,708 and L-838,417 were used to inhibit or activate α5GABAARs, respectively. RESULTS: Surgery under inhaled isoflurane anesthesia induced cognitive impairments and elevated the expression of MD2 in the hippocampus. Downregulation of MD2 expression by AAV-shMD2 or Tat-CIRP-CMA improved the spatial reference learning and memory in animals subjected to anesthesia and surgery. Furthermore, Tat-CIRP-CMA treatment decreased the expression of membrane α5GABAARs and tonic currents in CA1 pyramidal neurons in the hippocampus. Inhibition of α5GABAARs by L-655,708 alleviated cognitive impairments after anesthesia and surgery. More importantly, activation of α5GABAARs by L-838,417 abrogated the protective effects of Tat-CIRP-CMA against anesthesia and surgery-induced spatial reference learning and memory deficits. CONCLUSIONS: MD2 contributes to the occurrence of PND by regulating α5GABAARs in aged mice, and Tat-CIRP-CMA is a promising neuroprotectant against PND.


Assuntos
Envelhecimento/metabolismo , Antígeno 96 de Linfócito/biossíntese , Transtornos Neurocognitivos/metabolismo , Complicações Pós-Operatórias/metabolismo , Receptores de GABA-A/biossíntese , Envelhecimento/efeitos dos fármacos , Animais , Células Cultivadas , Feminino , Agonistas GABAérgicos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos Neurocognitivos/etiologia , Transtornos Neurocognitivos/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Período Perioperatório/efeitos adversos , Período Perioperatório/tendências , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/prevenção & controle , Gravidez
3.
Biochimie ; 191: 51-61, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34454977

RESUMO

Methylphenidate (MPD), a psychostimulant, is a prescription medicine for treating attention deficit hyperactivity disorder (ADHD). Previously we have shown that moderate doses of MPD enhanced learning and memory while higher doses impaired it. To understand neurochemical mechanisms and receptors involved in memory enhancing and impairing effects of MPD, the present study concerns the effects of these doses of MPD on serotonin, 5-HT1A, GABA, and NMDA receptor mRNA expression in the prefrontal cortex (PFC). We found that low doses (2.5 mg/kg) of MPD improved performance in the water-maze test but higher doses (5 mg/kg) impaired memory retention. Animals showing improved performance had high 5-HT metabolism in the PFC while these levels were not affected in the group treated with higher MPD doses and exhibiting impaired memory. There was downregulation of 5-HT1A receptors in the PFC of rats treated with higher dose MPD, which didn't occur in low dose of MPD treated animals. Further, a decrease in GABAAreceptor mRNA expression occurred in low doses of MPD treated animals and GluN2A expression was reduced in higher doses of MPD treated animals. The findings suggest that memory enhancing doses of MPD increase 5-HT and reduce GABAA receptor mRNA expression in the PFC to release excitatory glutamate neurons from the inhibitory influence of GABA. Conversely, higher dose of MPD downregulates 5-HT1A receptor mRNA expression to enhance inhibitory GABA influence on glutamate neurons and impair cognitive performance. The findings show an important role of 5-HT1A heteroreceptors in the PFC for improving therapeutic use of MPD and developing novel cognitive enhancers.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Memória/efeitos dos fármacos , Metilfenidato/farmacologia , Córtex Pré-Frontal/metabolismo , Receptor 5-HT1A de Serotonina/biossíntese , Receptores de GABA-A/biossíntese , Receptores de N-Metil-D-Aspartato/biossíntese , Serotonina/metabolismo , Animais , Masculino , Ratos , Ratos Wistar
4.
Mol Neurobiol ; 58(11): 5876-5889, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34417725

RESUMO

Following stroke, attenuation of detrimental inflammatory pathways might be a promising strategy to improve long-term outcome. In particular, cascades driven by pro-inflammatory chemokines interact with neurotransmitter systems such as the GABAergic system. This crosstalk might be of relevance for mechanisms of neuronal plasticity, however, detailed studies are lacking. The purpose of this study was to determine if treatment with 1,1'-[1,4-phenylenebis(methylene)]bis[1,4,8,11-tetraazacyclotetradecane] (AMD3100), an antagonist to the C-X-C chemokine receptor type 4 (CXCR4) and partial allosteric agonist to CXCR7 (AMD3100) alone or in combination with C-X3-C chemokine receptor type 1 (CX3CR1) deficiency, affect the expression of GABAA subunits and glutamate decarboxylase (GAD) isoforms. Heterozygous, CX3CR1-deficient mice and wild-type littermates were subjected to photothrombosis (PT). Treatment with AMD3100 (0.5 mg/kg twice daily i.p.) was administered starting from day 2 after induction of PT until day 14 after the insult. At this time point, GABAA receptor subunits (α3, ß3, δ), GAD65 and GAD67, and CXCR4 were analyzed from the peri-infarct tissue and homotypic brain regions of the contralateral hemisphere by quantitative real-time PCR and Western Blot. Fourteen days after PT, CX3CR1 deficiency resulted in a significant decrease of the three GABAA receptor subunits in both the lesioned and the contralateral hemisphere compared to sham-operated mice. Treatment with AMD3100 promoted the down-regulation of GABAA subunits and GAD67 in the ipsilateral peri-infarct area, while the ß3 subunit and the GAD isoforms were up-regulated in homotypic regions of the contralateral cortex. Changes in GABAA receptor subunits and GABA synthesis suggest that the CXCR4/7 and CX3CR1 signaling pathways are involved in the regulation of GABAergic neurotransmission in the post-ischemic brain.


Assuntos
Anti-Inflamatórios/uso terapêutico , Benzilaminas/uso terapêutico , Receptor 1 de Quimiocina CX3C/deficiência , Ciclamos/uso terapêutico , Glutamato Descarboxilase/biossíntese , Trombose Intracraniana/tratamento farmacológico , Doenças Neuroinflamatórias/tratamento farmacológico , Receptores de GABA-A/biossíntese , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Genes Reporter , Glutamato Descarboxilase/genética , Trombose Intracraniana/genética , Trombose Intracraniana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Doenças Neuroinflamatórias/etiologia , Doenças Neuroinflamatórias/genética , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Subunidades Proteicas , Receptores CXCR , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Receptores de GABA-A/genética
5.
Brain Res Bull ; 174: 84-91, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34090935

RESUMO

Hypertension is the most common chronic disease accompanied by cognitive decline and anxiety-like behavior. Angiotensin II (Ang II) induces hypertension by activating angiotensin II receptor subtype 1 (AT1R). The purpose of the study was to examine the potential underlying mechanism of alterations in cognition and anxiety-like behavior induced by Ang II. Adult C57 mice were intraperitoneal injected with either 1 mg/kg/d Ang II or saline individually for 14 consecutive days. Ang II resulted in cognitive decline and anxious like behavior in C57 mice. Moreover, Ang II disturbed bidirectional synaptic plasticity and neural oscillation coupling between high theta and gamma on PP (perforant pathway)-DG (dentate gyrus) pathway. In addition, Ang II decreased the expression of N-methyl-d-aspartate receptor (NR) 2A and NR 2B and increased the expression of GABAAR α1. The data suggest that Ang II disturb neural oscillations via altering excitatory and inhibitory (E/I) balance and eventually damage cognition and anxiety-like behavior in mice.


Assuntos
Angiotensina II/toxicidade , Ansiedade/induzido quimicamente , Ansiedade/patologia , Comportamento Animal/efeitos dos fármacos , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/psicologia , Ritmo Gama/efeitos dos fármacos , Ritmo Teta/efeitos dos fármacos , Animais , Giro Denteado/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Receptores de GABA-A/biossíntese , Receptores de GABA-A/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/biossíntese , Reconhecimento Psicológico/efeitos dos fármacos
6.
Shock ; 55(5): 620-629, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32433203

RESUMO

BACKGROUND: Knowledge about the neuroinflammatory state during months after sudden cardiac arrest is scarce. Neuroinflammation is mediated by cells that express the 18 kDa translocator protein (TSPO). We determined the time course of TSPO-expressing cells in a rat model of sudden cardiac arrest using longitudinal in vivo positron emission tomography (PET) imaging with the TSPO-specific tracer [18F]DAA1106 over a period of 6 months. METHODS: Five male Sprague Dawley rats were resuscitated from 6 min sudden cardiac arrest due to ventricular fibrillation, three animals served as shams. PET measurements were performed on day 5, 8, 14, 90, and 180 after intervention. Magnetic resonance imaging was performed on day 140. Imaging was preceded by Barnes Maze spatial memory testing on day 3, 13, 90, and 180. Specificity of [18F]DAA1106 binding was confirmed by Iba-1 immunohistochemistry. RESULTS: [18F]DAA1106 accumulated bilaterally in the dorsal hippocampus of all sudden cardiac arrest animals on all measured time points. Immunohistochemistry confirmed Iba-1 expressing cells in the hippocampal CA1 region. The number of Iba-1-immunoreactive objects per mm2 was significantly correlated with [18F]DAA1106 uptake. Additionally, two of the five sudden cardiac arrest rats showed bilateral TSPO-expression in the striatum that persisted until day 180. In Barnes Maze, the relative time spent in the target quadrant negatively correlates with dorsal hippocampal [18F]DAA1106 uptake on day 14 and 180. CONCLUSIONS: After sudden cardiac arrest, TSPO remains expressed over the long-term. Sustainable treatment options for neuroinflammation may be considered to improve cognitive functions after sudden cardiac arrest.


Assuntos
Proteínas de Transporte/biossíntese , Parada Cardíaca/diagnóstico por imagem , Parada Cardíaca/metabolismo , Tomografia por Emissão de Pósitrons , Receptores de GABA-A/biossíntese , Acetamidas , Animais , Masculino , Éteres Fenílicos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
7.
Cereb Cortex ; 31(4): 2026-2037, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33279960

RESUMO

Visuospatial working memory (vsWM) requires information transfer among multiple cortical regions, from primary visual (V1) to prefrontal (PFC) cortices. This information is conveyed via layer 3 glutamatergic neurons whose activity is regulated by gamma-aminobutyric acid (GABA)ergic interneurons. In layer 3 of adult human neocortex, molecular markers of glutamate neurotransmission were lowest in V1 and highest in PFC, whereas GABA markers had the reverse pattern. Here, we asked if these opposite V1-visual association cortex (V2)-posterior parietal cortex (PPC)-PFC gradients across the vsWM network are present in layer 3 of monkey neocortex, when they are established during postnatal development, and if they are specific to this layer. We quantified transcript levels of glutamate and GABA markers in layers 3 and 6 of four vsWM cortical regions in a postnatal developmental series of 30 macaque monkeys. In adult monkeys, glutamate transcript levels in layer 3 increased across V1-V2-PPC-PFC regions, whereas GABA transcripts showed the opposite V1-V2-PPC-PFC gradient. Glutamate transcripts established adult-like expression patterns earlier during postnatal development than GABA transcripts. These V1-V2-PPC-PFC gradients and developmental patterns were less evident in layer 6. These findings demonstrate that expression of glutamate and GABA transcripts differs across cortical regions and layers during postnatal development, revealing potential molecular substrates for vsWM functional maturation.


Assuntos
Ácido Glutâmico/biossíntese , Lobo Parietal/metabolismo , Córtex Pré-Frontal/metabolismo , Transcrição Gênica/fisiologia , Córtex Visual/metabolismo , Ácido gama-Aminobutírico/biossíntese , Fatores Etários , Animais , Transportador 2 de Aminoácido Excitatório/biossíntese , Transportador 2 de Aminoácido Excitatório/genética , Feminino , Neurônios GABAérgicos/metabolismo , Expressão Gênica , Ácido Glutâmico/genética , Macaca mulatta , Lobo Parietal/crescimento & desenvolvimento , Córtex Pré-Frontal/crescimento & desenvolvimento , Receptores de GABA-A/biossíntese , Receptores de GABA-A/genética , Córtex Visual/crescimento & desenvolvimento , Ácido gama-Aminobutírico/genética
8.
Mol Biol Rep ; 47(8): 5699-5710, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32803504

RESUMO

The pivotal role played by ion-channel dysregulations in the pathogenesis of epilepsy has always garnered much attention. Since mutation of ion-channel proteins CACNA1A and GABRD have been associated with epilepsy, it is important to determine the post-traumatic epilepsy-associated changes in expression levels of these ion channel proteins. Additionally, curcumin is already known for its antiepileptic and neuroprotective potential in FeCl3-induced model of post-traumatic epilepsy. Thus, we investigated FeCl3-induced epilepsy mediated differential expression of CACNA1A and GABRD in the cortical region of the rat brain. Furthermore, we investigated the effect of curcumin on the expression of both proteins. For this, epilepsy was induced by intracortical FeCl3 injection (5 µl of 100 mM). Additionally, curcumin (conc. 1000 ppm; 75 mg/kg of b.wt.; for 14 and 28 days) was administered, mixed with normal food pellets. Results obtained from EEG-MUA and Morris water maze assay demonstrate the progression of epilepsy after FeCl3 injection. Additionally, western blotting and histological studies show the downregulation of CACNA1A and GABRD during epileptogenesis. It was observed that epilepsy-associated decline in learning and memory of animals might be linked with the dysregulation of both proteins. Results also demonstrated that curcumin administration ameliorated epilepsy-associated change in expression of both CACNA1A and GABRD proteins. In conclusion, the neuroprotective effect of curcumin against iron-induced epilepsy might be accompanied by the alleviated upregulation of these channel proteins.


Assuntos
Canais de Cálcio Tipo N/biossíntese , Curcumina/farmacologia , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Fármacos Neuroprotetores/farmacologia , Receptores de GABA-A/biossíntese , Animais , Antineoplásicos/farmacologia , Canais de Cálcio Tipo N/genética , Canais de Cálcio Tipo N/metabolismo , Cloretos/administração & dosagem , Modelos Animais de Doenças , Eletroencefalografia/métodos , Epilepsia/induzido quimicamente , Epilepsia/patologia , Compostos Férricos/administração & dosagem , Masculino , Teste do Labirinto Aquático de Morris , Ratos , Ratos Wistar , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo
9.
J Neurosci ; 40(29): 5518-5530, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32513829

RESUMO

GABAA receptors (GABAARs) are profoundly important for controlling neuronal excitability. Spontaneous and familial mutations to these receptors feature prominently in excitability disorders and neurodevelopmental deficits following disruption to GABA-mediated inhibition. Recent genotyping of an individual with severe epilepsy and Williams-Beuren syndrome identified a frameshifting de novo variant in a major GABAAR gene, GABRA1 This truncated the α1 subunit between the third and fourth transmembrane domains and introduced 24 new residues forming the mature protein, α1Lys374Serfs*25 Cell surface expression of mutant murine GABAARs is severely impaired compared with WT, due to retention in the endoplasmic reticulum. Mutant receptors were differentially coexpressed with ß3, but not with ß2, subunits in mammalian cells. Reduced surface expression was reflected by smaller IPSCs, which may underlie the induction of seizures. The mutant does not have a dominant-negative effect on native neuronal GABAAR expression since GABA current density was unaffected in hippocampal neurons, although mutant receptors exhibited limited GABA sensitivity. To date, the underlying mechanism is unique for epileptogenic variants and involves differential ß subunit expression of GABAAR populations, which profoundly affected receptor function and synaptic inhibition.SIGNIFICANCE STATEMENT GABAARs are critical for controlling neural network excitability. They are ubiquitously distributed throughout the brain, and their dysfunction underlies many neurologic disorders, especially epilepsy. Here we report the characterization of an α1-GABAAR variant that results in severe epilepsy. The underlying mechanism is structurally unusual, with the loss of part of the α1 subunit transmembrane domain and part-replacement with nonsense residues. This led to compromised and differential α1 subunit cell surface expression with ß subunits resulting in severely reduced synaptic inhibition. Our study reveals that disease-inducing variants can affect GABAAR structure, and consequently subunit assembly and cell surface expression, critically impacting on the efficacy of synaptic inhibition, a property that will orchestrate the extent and duration of neuronal excitability.


Assuntos
Epilepsia/metabolismo , Receptores de GABA-A/biossíntese , Síndrome de Williams/metabolismo , Animais , Epilepsia/genética , Feminino , Células HEK293 , Hipocampo/metabolismo , Humanos , Lactente , Masculino , Neurônios/metabolismo , Ratos Sprague-Dawley , Receptores de GABA-A/fisiologia , Síndrome de Williams/complicações , Síndrome de Williams/genética , Xenopus laevis
10.
Brain Res Bull ; 162: 1-10, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32428626

RESUMO

OBJECTIVE: To observe the impacts of electroacupuncture (EA) stimulation at "Zusanli and Kunlun Points" on spinal dorsal horn microglia activation in L5 spinal nerve ligation (SNL) rats and BNDF, P2 × 4 and GABAAγ2, and the changes in spinal dorsal horn synaptic plasticity in model rats. METHODS: Adult male SD rats (180-220 g) were selected and randomly divided into 6 groups, including the sham group, the SNL group, the SNL + EA group, the SNL+5-BDBD group, the SNL + EA + 5-BDBD group and the SNL + FEA group. The changes in the Iba-1, BDNF, P2 × 4 and GABAAγ2 in the spinal cord of rats were observed by Western blotting, immunofluorescence, RT-PCR and other techniques; the long-term changes in the potential after the excitatory synapse of the spinal dorsal horn in rats were observed by in vivo electrophysiological technique. RESULTS: After 7 days of intervention, the fluorescence intensity (FI) of P2 × 4 and Iba-1 in the SNL + EA group was lower than that in the SNL group and higher than that in the sham group(P < 0.01), but the FI of GABAAγ2 was higher than that in the SNL group(P < 0.01); the expression of Iba-1, BDNF and P2 × 4 proteins in the SNL + EA group, the SNL+5-BDBD group and the SNL + EA + 5-BDBD group was significantly lower than that in the SNL + FEA group(P < 0.05), but the expression of GABAAγ2 protein was higher (P < 0.05); after treatment with EA, the expression levels of Iba-1 mRNA and P2 × 4 mRNA in the SNL + EA group were lower than those in the SNL group(P < 0.01), but the expression levels of GABAAγ2 mRNA were higher (P < 0.01). Meanwhile, long-term potentiation changes could not be induced in the SNL + EA group. CONCLUSION: The EA stimulation at "Zusanli" and "Kunlun" points can improve the pain threshold of rats with neuropathic pain (NP), inhibit the excitatory postsynaptic potential (EPSP), and weaken the excitatory transmission efficiency between synapses during NP.


Assuntos
Eletroacupuntura/métodos , Potenciação de Longa Duração/fisiologia , Neuralgia/metabolismo , Receptores de GABA-A/biossíntese , Receptores Purinérgicos P2X4/biossíntese , Medula Espinal/metabolismo , Animais , Expressão Gênica , Masculino , Neuralgia/genética , Neuralgia/terapia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/genética , Receptores Purinérgicos P2X4/genética , Resultado do Tratamento
11.
J Comp Neurol ; 528(15): 2551-2568, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32220012

RESUMO

GABAA receptors are composed of five subunits arranged around a central chloride channel. Their subunits originate from different genes or gene families. The majority of GABAA receptors in the mammalian brain consist of two α-, two ß- and one γ- or δ-subunit. This subunit organization crucially determines the physiological and pharmacological properties of the GABAA receptors. Using immunohistochemistry, we investigated the distribution of 10 GABAA receptor subunits (α1, α2, α3, α4, α5, ß1, ß2, ß3, γ2, and δ) in the fore brain of three female rhesus monkeys (Macaca mulatta). Within the cerebral cortex, subunits α1, α5, ß2, ß3, and γ2 were found in all layers, α2, α3, and ß1 were more concentrated in the inner and outer layers. The caudate/putamen was rich in α1, α2, α5, all three ß-subunits, γ2, and δ. Subunits α3 and α5 were more concentrated in the caudate than in the putamen. In contrast, α1, α2, ß1, ß2, γ2, and δ were highest in the pallidum. Most dorsal thalamic nuclei contained subunits α1, α2, α4, ß2, ß3, and γ2, whereas α1, α3, ß1, and γ2 were most abundant in the reticular nucleus. Within the amygdala, subunits α1, α2, α5, ß1, ß3, γ2, and δ were concentrated in the cortical nucleus, whereas in the lateral and basolateral amygdala α1, α2, α5, ß1, ß3, and δ, and in the central amygdala α1, α2, ß3, and γ2 were most abundant. Interestingly, subunit α3-IR outlined the intercalated nuclei of the amygdala. In the hippocampus, subunits α1, α2, α5, ß2, ß3, γ2, and δ were highly expressed in the dentate molecular layer, whereas α1, α2, α3, α5, ß1, ß2, ß3, and γ2 were concentrated in sector CA1 and the subiculum. The distribution of GABAA receptor subunits in the rhesus monkey was highly heterogeneous indicating a high number of differently assembled receptors. In most areas investigated, notably in the striatum/pallidum, amygdaloid nuclei and in the hippocampus it was more diverse than in the rat and mouse indicating a more heterogeneous and less defined receptor assembly in the monkey than in rodent brain.


Assuntos
Prosencéfalo/química , Prosencéfalo/metabolismo , Subunidades Proteicas/biossíntese , Receptores de GABA-A/biossíntese , Fatores Etários , Sequência de Aminoácidos , Animais , Feminino , Imuno-Histoquímica , Macaca mulatta , Subunidades Proteicas/análise , Subunidades Proteicas/genética , Receptores de GABA-A/análise , Receptores de GABA-A/genética
12.
Curr Mol Pharmacol ; 13(3): 251-260, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31894752

RESUMO

BACKGROUND: Epilepsy remains challenging to treat still no etiologic treatment has been identified, however, some antiepileptic drugs (AEDs) are able to modify the pathogenesis of the disease. Lacosamide (LCM) has been shown to possess complex anticonvulsant and neuroprotective actions, being an enhancer of the slow inactivation of voltage-gated sodium channels, and it has the potential to prevent epileptogenesis. Recent evidence has shown that LCM indirectly improves the function of GABAA receptors. Receptors at most GABAergic synapses involve the gamma-2 subunit, which contributes to both phasic and tonic inhibition, and its presence assures benzodiazepine sensitivity. Moreover, mutant gamma-2 subunits were associated with generalized epilepsy syndromes. In animal models, the expression of the gamma-2 subunit of the gamma-aminobutyric acid A receptor (GABAAg2) was shown to be increased in pentylenetetrazole (PTZ)-induced chemical kindling in Wistar rats. OBJECTIVE: This study hypothesized that LCM might affect the kindling process by influencing the expression of GABAA receptors in the hippocampus. METHODS: The gene and protein expression levels of the GABAAg2 were studied using RT-qPCR and immunofluorescent staining. RESULTS: It was found that LCM treatment (10 mg/kg i.p. daily for 57 days) reduced the maximal intensity of the PTZ-induced seizures but did not prevent kindling. On the other hand, LCM treatment reverted the increase of mRNA expression of GABAAg2 in the hippocampus and prevented the decrease of GABAAg2 protein in the hippocampal CA1 region. CONCLUSION: LCM could exhibit modulatory effects on the GABAergic system of the hippocampus that may be independent of the anticonvulsant action.


Assuntos
Anticonvulsivantes/farmacologia , Excitação Neurológica/genética , Lacosamida/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Receptores de GABA-A/biossíntese , Convulsões/prevenção & controle , Animais , Anticonvulsivantes/uso terapêutico , Convulsivantes/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Lacosamida/uso terapêutico , Masculino , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , Pentilenotetrazol/toxicidade , Subunidades Proteicas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Receptores de GABA-A/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico
13.
Alcohol Clin Exp Res ; 44(2): 423-434, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31840824

RESUMO

BACKGROUND: Alcohol exposure alters the expression of a large number of genes, resulting in neuronal adaptions and neuronal loss, but the underlying mechanisms are largely unknown. miRNAs are gene repressors that are abundant in the brain. A recent study identified ~ 35 miRNAs that are up-regulated in the prefrontal cortex of human alcoholics and predicted to target genes that are down-regulated in the same region. Although interactions between alcohol-responsive miRNAs and their target genes have been predicted, few studies have validated these predictions. METHODS: We measured the expression of GABAA α5 mRNA in the prefrontal and motor cortices of human alcoholics and matched controls using real-time PCR. The expression of miR-203 was measured in a subset of these cases. The predicted interaction of miR-203 and GABRA5 was validated for miR-203 using a luciferase reporter assay. RESULTS: In both frontal and motor cortices, the expression of GABAA α5 was significantly lower in cirrhotic alcoholics compared with controls. Further, the pattern of expression between the groups was significantly different between males and females. The expression of miR-203 was higher in the prefrontal cortex of cirrhotic alcoholics compared with controls and uncomplicated alcoholics. These differences were particularly marked in female cases. Cotransfection of GABRA5 with miR-203 in HEK293T cells reduced luciferase reporter activity. CONCLUSION: There are sex differences in the expression of GABAA α5 and miR-203 in the brain of human alcoholics which are particularly marked in alcoholics with cirrhosis of the liver. Further, miR-203 may mediate the changes in expression of this GABAA receptor isoform that is brought about by alcohol exposure.


Assuntos
Alcoólicos , Alcoolismo/metabolismo , Cirrose Hepática Alcoólica/metabolismo , Receptores de GABA-A/biossíntese , Caracteres Sexuais , Adulto , Idoso , Alcoolismo/epidemiologia , Alcoolismo/genética , Estudos de Coortes , Feminino , Expressão Gênica , Células HEK293 , Humanos , Cirrose Hepática Alcoólica/epidemiologia , Cirrose Hepática Alcoólica/genética , Masculino , Pessoa de Meia-Idade , Córtex Motor/metabolismo , Córtex Pré-Frontal/metabolismo , Receptores de GABA-A/genética
14.
Biomed Pharmacother ; 121: 109588, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31707350

RESUMO

Translocator protein (TSPO) is highly expressed in the cardiovascular system, exerting crucial effects on both myocardial damage and protection. However, the role and mechanism of TSPO in myocardial ischemia/reperfusion (I/R) injury remains elusive. In the current study, we subjected H9c2 cardiomyocytes to anoxia/reoxygenation (A/R) and knocked down TSPO expression by RNA interference to investigate the possible mechanism of TSPO on I/R injury. TSPO expression in cardiomyocytes was up-regulated when exposed to A/R, but normal in anoxic preconditioned (APC) cardiomyocytes. Moreover, A/R also led to an increase in reactive oxygen species (ROS), oxidative stress aggravation, mitochondrial membrane potential collapse, mitochondrial permeability transition pore (mPTP) opening, and cell apoptosis. However, these events were completely compensated by downregulating TSPO expression. TSPO-downregulated cardiomyocytes produced lesser lactate dehydrogenase (LDH) and creatine phosphokinase (CPK), and showed lesser cytosol malondialdehyde (MDA) accumulation than normal cells after A/R injury. On the other hand, the TSPO- downregulated cells showed higher activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), mitochondrial function stabilization, resulting in less cell apoptosis and damage in case of A/R condition. In conclusion, TSPO expression is up-regulated under A/R injury, whereas repression of TSPO improves the endurance of cardiomyocytes against A/R injury by reducing oxidative stress, mitochondrial damage and cell apoptosis.


Assuntos
Proteínas de Transporte/biossíntese , Regulação para Baixo/fisiologia , Homeostase/fisiologia , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/fisiologia , Receptores de GABA-A/biossíntese , Animais , Proteínas de Transporte/genética , Hipóxia Celular/fisiologia , Linhagem Celular , Expressão Gênica , Ratos , Espécies Reativas de Oxigênio/metabolismo , Receptores de GABA-A/genética
15.
Auton Neurosci ; 221: 102579, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31445405

RESUMO

Lifelong functional plasticity of the gastrointestinal (GI) tract is essential for health, yet the underlying molecular mechanisms are poorly understood. The enteric nervous system (ENS) regulates all aspects of the gut function, via a range of neurotransmitter pathways, one of which is the GABA-GABAA receptor (GABAAR) system. We have previously shown that GABAA receptor subunits are differentially expressed within the ENS and are involved in regulating various GI functions. We have also shown that these receptors are involved in mediating stress-induced colonic inflammation. However, the expression and function of intestinal GABAARs, at different ages, is largely unexplored and was the focus of this study. Here we show that the impact of GABAAR activation on colonic contractility changes from early postnatal period through to late adulthood, in an age-dependant manner. We also show that the highest levels of expression for all GABAAR subunits is evident at postnatal day (P) 10 apart from the α3 subunit which increased with age. This increase in the α3 subunit expression in late adulthood (18 months old) is accompanied by an increase in the expression of inflammatory markers within the mouse colon. Finally, we demonstrate that the deletion of the α3 subunit prevents the increase in the expression of colonic inflammatory markers associated with healthy ageing. Collectively, the data provide the first demonstration of the molecular and functional plasticity of the GI GABAAR system over the course of a lifetime, and its possible role in mediating the age-induced colonic inflammation associated with healthy ageing.


Assuntos
Envelhecimento/fisiologia , Colo/fisiologia , Sistema Nervoso Entérico/fisiologia , Motilidade Gastrointestinal/fisiologia , Doenças Inflamatórias Intestinais/fisiopatologia , Plasticidade Neuronal/fisiologia , Receptores de GABA-A/fisiologia , Alprazolam/farmacologia , Animais , Colo/crescimento & desenvolvimento , Colo/inervação , Moduladores GABAérgicos/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peristaltismo/efeitos dos fármacos , Peristaltismo/fisiologia , Subunidades Proteicas , Receptores de GABA-A/biossíntese , Receptores de GABA-A/deficiência , Receptores de GABA-A/genética
16.
Brain ; 142(7): 1938-1954, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31056671

RESUMO

We performed next generation sequencing on 1696 patients with epilepsy and intellectual disability using a gene panel with 480 epilepsy-related genes including all GABAA receptor subunit genes (GABRs), and we identified six de novo GABR mutations, two novel GABRA5 mutations (c.880G>T, p.V294F and c.1238C>T, p.S413F), two novel GABRA1 mutations (c.778C>T, p.P260S and c.887T>C, p.L296S/c.944G>T, p.W315L) and two known GABRA1 mutations (c.335G>A, p.R112Q and c.343A>G, p.N115D) in six patients with intractable early onset epileptic encephalopathy. The α5(V294F and S413F) and α1(P260S and L296S/W315L) subunit residue substitutions were all in transmembrane domains, while the α1(R112Q and N115R) subunit residue substitutions were in the N-terminal GABA binding domain. Using multidisciplinary approaches, we compared effects of mutant GABAA receptor α5 and α1 subunits on the properties of recombinant α5ß3γ2 and α1ß3γ2 GABAA receptors in both neuronal and non-neuronal cells and characterized their effects on receptor clustering, biogenesis and channel function. GABAA receptors containing mutant α5 and α1 subunits all had reduced cell surface and total cell expression with altered endoplasmic reticulum processing, impaired synaptic clustering, reduced GABAA receptor function and decreased GABA binding potency. Our study identified GABRA5 as a causative gene for early onset epileptic encephalopathy and expands the mutant GABRA1 phenotypic spectrum, supporting growing evidence that defects in GABAergic neurotransmission contribute to early onset epileptic encephalopathy phenotypes.


Assuntos
Epilepsia/genética , Deficiência Intelectual/genética , Receptores de GABA-A/genética , Sinapses/genética , Criança , Pré-Escolar , Epilepsia/complicações , Feminino , Predisposição Genética para Doença/genética , Humanos , Deficiência Intelectual/complicações , Masculino , Potenciais da Membrana/fisiologia , Potenciais Pós-Sinápticos em Miniatura/fisiologia , Mutação , Cultura Primária de Células , Receptores de GABA-A/biossíntese , Receptores de GABA-A/metabolismo , Receptores de GABA-A/fisiologia , Sinapses/fisiologia , Adulto Jovem , Ácido gama-Aminobutírico/metabolismo
17.
Int J Neurosci ; 129(10): 986-994, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30957600

RESUMO

Background: Numerous experimental studies show that anesthetics are potentially toxic to the immature brain. Even though benzodiazepines are widely used in pediatric anesthesia and intensive care medicine, only a few studies examine the effects of these drugs on immature neurons. Methods: Hippocampal neuronal cell cultures of embryonic Wistar rats (15 days in culture) were incubated with midazolam 100 or 300 nM for either 30 min or 4 h. The time course of the mRNA expression of the glutamate receptors subunits NR1, NR2A and NR2B of the NMDA receptor, the GluA-1 and A-2 subunits of the AMPA receptor as well as the alpha 1 subunit of the GABAA receptor were examined by PCR. Apoptosis was detected using Western blot analysis for BAX, Bcl-2 and Caspase-3. Results: Midazolam at 100 and 300 nM applied for 30 min and 100 nM for 4 h affected glutamate receptor and GABAA receptor subunit expression. However, these effects were reversible within 72 h following washout. When 300 nM midazolam was applied for 4 h a significant increase in the NR 1 and NR 2A mRNA subunit expression could be detected. The increase in NR 2B receptor subunit expression as well as the GluA1 subunit expression was not reversible within 72 h following washout. This increase in mRNA glutamate receptor subunit expression was associated with a significant increase in neuronal apoptosis. Conclusion: In immature neurons midazolam altered GABA and glutamate mRNA receptor subunit expression. Prolonged increase in midazolam-induced glutamate receptor expression was associated with apoptosis.


Assuntos
Moduladores GABAérgicos/farmacologia , Hipocampo/metabolismo , Midazolam/farmacologia , Neurônios/metabolismo , RNA Mensageiro/biossíntese , Receptores de GABA-A/biossíntese , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/embriologia , Neurônios/efeitos dos fármacos , RNA Mensageiro/genética , Ratos , Ratos Wistar , Receptores de GABA-A/genética
18.
J Pharmacol Exp Ther ; 368(1): 100-105, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30389723

RESUMO

Central α4ßδ receptors are the most abundant isoform of δ subunit-containing extrasynaptic GABAA receptors that mediate tonic inhibition. Although the amplitude of GABA-activated currents through α4ßδ receptors is modulated by multiple general anesthetics, the effects of general anesthetics on desensitization and deactivation of α4ßδ receptors remain unknown. In the current study, we investigated the effect of etomidate, a potent general anesthetic, on the kinetics and the pseudo steady-state current amplitude of α4ß3δ receptors inducibly expressed in human embryonic kidney 293 TetR cells. Etomidate directly activates α4ß3δ receptors in a concentration-dependent manner. Etomidate at a clinically relevant concentration (3.2 µM) enhances maximal response without altering the EC50 of GABA concentration response. Etomidate also increases the extent of desensitization and prolongs the deactivation of α4ß3δ receptors in the presence of maximally activating concentrations of GABA (1 mM). To mimic the modulatory effect of etomidate on tonic currents, long pulses (30-60 seconds) of a low GABA concentration (1 µM) were applied to activate α4ß3δ receptors in the absence and presence of etomidate. Although etomidate increases the desensitization of α4ß3δ receptors, the pseudo steady-state current amplitude at 1 µM GABA is augmented by etomidate. Our data demonstrate that etomidate enhances the pseudo steady-state current of α4ß3δ receptors evoked by a GABA concentration comparable to an ambient GABA level, suggesting that α4ß3δ receptors may mediate etomidate's anesthetic effect in the brain.


Assuntos
Etomidato/farmacologia , Agonistas GABAérgicos/farmacologia , Hipnóticos e Sedativos/farmacologia , Receptores de GABA-A/biossíntese , Relação Dose-Resposta a Droga , Expressão Gênica , Células HEK293 , Humanos , Receptores de GABA-A/genética , Ácido gama-Aminobutírico/farmacologia
19.
Br J Pharmacol ; 176(2): 297-316, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30318707

RESUMO

BACKGROUND AND PURPOSE: Altered function or expression of GABAA receptors contributes to anxiety disorders. Benzodiazepines are widely prescribed for the treatment of anxiety. However, the long-term use of benzodiazepines increases the risk of developing drug dependence and tolerance. Thus, it is urgent to explore new therapeutic approaches. Metformin is widely used to treat Type 2 diabetes and other metabolic syndromes. However, the role of metformin in psychiatric disorders, especially anxiety, remains largely unknown. EXPERIMENTAL APPROACH: We examined the effects of metformin on anxiety-like behaviour of rats in open field test and elevated plus maze test. We also observed the effect of metformin (10 µM, in vitro; 100 mg·kg-1 , in vivo) on the trafficking of GABAA receptors, as mechanisms underlying the anxiolytic effects of metformin. KEY RESULTS: Metformin (100 mg·kg-1 , i.p. 30 min) displayed a robust and rapid anxiolytic effect, without tolerance. Metformin up-regulated the surface expression of GABAA receptors and increased miniature inhibitory postsynaptic currents (mIPSCs). AMP-activated protein kinase (AMPK) activated by metformin-induced stimulation of forkhead box O3a (FoxO3a) transcriptional activity, followed by increased expression of GABAA receptor-associated protein (GABARAP) and its binding to GABAA receptors finally resulted in the membrane insertion of GABAA receptors. CONCLUSIONS AND IMPLICATIONS: Metformin increased mIPSCs by up-regulating the membrane insertion of GABAA receptors, via a pathway involving AMPK, FoxO3a, and the GABAA receptor-associated protein. Thus metformin has a potential new use in the treatment of anxiety disorders.


Assuntos
Ansiolíticos/farmacologia , Ansiedade/tratamento farmacológico , Metformina/farmacologia , Receptores de GABA-A/biossíntese , Animais , Ansiolíticos/administração & dosagem , Ansiedade/metabolismo , Glicemia/análise , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Proteína Forkhead Box O3/antagonistas & inibidores , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Inativação Gênica/efeitos dos fármacos , Injeções Intraperitoneais , Masculino , Metformina/administração & dosagem , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/genética , Relação Estrutura-Atividade , Regulação para Cima/efeitos dos fármacos
20.
Protein Expr Purif ; 154: 7-15, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30248449

RESUMO

Owing to their therapeutic relevance, considerable efforts are devoted to the structural characterisation of membrane proteins. Such studies are limited by the availability of high quality protein due to the difficulty of overexpression in recombinant mammalian systems. We sought to systematically optimise multiple aspects in the process of transiently transfecting HEK293 cells, to allow the rapid expression of membrane proteins, without the lengthy process of stable clone formation. We assessed the impact of medium formulation, cell line, and harvest time on the expression of GABAA receptors, as determined by [3H]muscimol binding in cell membranes. Furthermore, transfection with the use of calcium phosphate/polyethyleneimine multishell nanoparticles was optimised, and a dual vector system utilising viral enhancing elements was designed and implemented. These efforts resulted in a 40-fold improvement in GABAA α1ß3 receptor expression, providing final yields of 22 fmol/cm2. The findings from this work provide a guide to the optimisation of transient expression of proteins in mammalian cells and should assist in the structural characterisation of membrane proteins.


Assuntos
Membrana Celular/metabolismo , Expressão Gênica , Receptores de GABA-A , Transfecção , Adesão Celular , Membrana Celular/genética , Células HEK293 , Humanos , Muscimol/farmacologia , Receptores de GABA-A/biossíntese , Receptores de GABA-A/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...