Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Br J Anaesth ; 130(2): e351-e360, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36402576

RESUMO

BACKGROUND: It remains controversial whether general anaesthetic drugs contribute to perioperative neurocognitive disorders in adult patients. Preclinical studies have generated conflicting results, likely because of differing animal models, study protocols, and measured outcomes. This scoping review of preclinical studies addressed the question: 'Do general anaesthetic drugs cause cognitive deficits in adult animals that persist after the drugs have been eliminated from the brain?' METHODS: Reports of preclinical studies in the MEDLINE database published from 1953 to 2021 were examined. A structured review process was used to assess original studies of cognitive behaviours, which were measured after treatment (≥24 h) with commonly used general anaesthetic drugs in adult animals. RESULTS: The initial search yielded 380 articles, of which 106 were fully analysed. The most frequently studied animal model was male (81%; n=86/106) rodents (n=106/106) between 2-3 months or 18-20 months of age. Volatile anaesthetic drugs were more frequently studied than injected drugs, and common outcomes were memory behaviours assessed using the Morris water maze and fear conditioning assays. Cognitive deficits were detected in 77% of studies (n=82/106) and were more frequent in studies of older animals (89%), after inhaled anaesthetics, and longer drug treatments. Limitations of the studies included a lack of physiological monitoring, mortality data, and risk of bias attributable to the absence of randomisation and blinding. CONCLUSIONS: Most studies reported cognitive deficits after general anaesthesia, with age, use of volatile anaesthetic drugs, and duration of anaesthesia as risk factors. Recommendations to improve study design and guide future research are presented.


Assuntos
Anestésicos Gerais , Transtornos Cognitivos , Disfunção Cognitiva , Animais , Masculino , Anestesia Geral/efeitos adversos , Transtornos Cognitivos/induzido quimicamente , Disfunção Cognitiva/induzido quimicamente , Anestésicos Gerais/efeitos adversos , Cognição
2.
Anesth Analg ; 136(5): 999-1011, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36469752

RESUMO

BACKGROUND: Perioperative neurocognitive disorders (PNDs) are complex, multifactorial conditions that are associated with poor long-term outcomes. Inflammation and exposure to general anesthetic drugs are likely contributing factors; however, the relative impact of each factor alone versus the combination of these factors remains poorly understood. The goal of this study was to compare the relative impact of inflammation, general anesthesia, and the combination of both factors on memory and executive function. METHODS: To induce neuroinflammation at the time of exposure to an anesthetic drug, adult male mice were treated with lipopolysaccharide (LPS) or vehicle. One day later, they were anesthetized with etomidate (or vehicle). Levels of proinflammatory cytokines were measured in the hippocampus and cortex 24 hours after LPS treatment. Recognition memory and executive function were assessed starting 24 hours after anesthesia using the novel object recognition assay and the puzzle box, respectively. Data are expressed as mean (or median) differences (95% confidence interval). RESULTS: LPS induced neuroinflammation, as indicated by elevated levels of proinflammatory cytokines, including interleukin-1ß (LPS versus control, hippocampus: 3.49 pg/mg [2.06-4.92], P < .001; cortex: 2.60 pg/mg [0.83-4.40], P = .010) and tumor necrosis factor-α (hippocampus: 3.50 pg/mg [0.83-11.82], P = .002; cortex: 2.38 pg/mg [0.44-4.31], P = .021). Recognition memory was impaired in mice treated with LPS, as evinced by a lack of preference for the novel object (novel versus familiar: 1.03 seconds [-1.25 to 3.30], P = .689), but not in mice treated with etomidate alone (novel versus familiar: 2.38 seconds [0.15-4.60], P = .031). Mice cotreated with both LPS and etomidate also exhibited memory deficits (novel versus familiar: 1.40 seconds [-0.83 to 3.62], P = .383). In the puzzle box, mice treated with either LPS or etomidate alone showed no deficits. However, the combination of LPS and etomidate caused deficits in problem-solving tasks (door open task: -0.21 seconds [-0.40 to -0.01], P = .037; plug task: -0.30 seconds [-0.50 to -0.10], P < .001; log values versus control), indicating impaired executive function. CONCLUSIONS: Impairments in recognition memory were driven by inflammation. Deficits in executive function were only observed in mice cotreated with LPS and etomidate. Thus, an interplay between inflammation and etomidate anesthesia led to cognitive deficits that were not observed with either factor alone. These findings suggest that inflammation and anesthetic drugs may interact synergistically, or their combination may unmask covert or latent deficits induced by each factor alone, leading to PNDs.


Assuntos
Etomidato , Função Executiva , Camundongos , Masculino , Animais , Etomidato/efeitos adversos , Lipopolissacarídeos/toxicidade , Doenças Neuroinflamatórias , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Transtornos da Memória/induzido quimicamente , Anestesia Geral/efeitos adversos , Citocinas/metabolismo , Hipocampo/metabolismo , Camundongos Endogâmicos C57BL
3.
Br J Anaesth ; 126(3): 674-683, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33388140

RESUMO

BACKGROUND: Multiple cognitive and psychiatric disorders are associated with an increased tonic inhibitory conductance that is generated by α5 subunit-containing γ-aminobutyric acid type A (α5 GABAA) receptors. Negative allosteric modulators that inhibit α5 GABAA receptors (α5-NAMs) are being developed as treatments for these disorders. The effects of α5-NAMs have been studied on recombinant GABAA receptors expressed in non-neuronal cells; however, no study has compared drug effects on the tonic conductance generated by native GABAA receptors in neurones, which was the goal of this study. METHODS: The effects of five α5-NAMs (basmisanil, Ono-160, L-655,708, α5IA, and MRK-016) on tonic current evoked by a low concentration of GABA were studied using whole-cell recordings in cultured mouse hippocampal neurones. Drug effects on current evoked by a saturating concentration of GABA and on miniature inhibitory postsynaptic currents (mIPSCs) were also examined. RESULTS: The α5-NAMs caused a concentration-dependent decrease in tonic current. The potencies varied as the inhibitory concentration for 50% inhibition (IC50) of basmisanil (127 nM) was significantly higher than those of the other compounds (0.4-0.8 nM). In contrast, the maximal efficacies of the drugs were similar (35.5-51.3% inhibition). The α5-NAMs did not modify current evoked by a saturating GABA concentration or mIPSCs. CONCLUSIONS: Basmisanil was markedly less potent than the other α5-NAMs, an unexpected result based on studies of recombinant α5 GABAA receptors. Studying the effects of α5 GABAA receptor-selective drugs on the tonic inhibitory current in neurones could inform the selection of compounds for future clinical trials.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Antagonistas de Receptores de GABA-A/farmacologia , Hipocampo/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Regulação Alostérica , Animais , Células Cultivadas , Cognição/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hipocampo/metabolismo , Camundongos , Neurônios/metabolismo , Técnicas de Patch-Clamp
4.
Crit Care Med ; 48(4): 533-544, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32205600

RESUMO

OBJECTIVES: Cognitive deficits after traumatic brain injury are a leading cause of disability worldwide, yet no effective pharmacologic treatments exist to improve cognition. Traumatic brain injury increases proinflammatory cytokines, which trigger excess function of α5 subunit-containing γ-aminobutyric acid type A receptors. In several models of brain injury, drugs that inhibit α5 subunit-containing γ-aminobutyric acid type A receptor function improve cognitive performance. Thus, we postulated that inhibiting α5 subunit-containing γ-aminobutyric acid type A receptors would improve cognitive performance after traumatic brain injury. In addition, because traumatic brain injury reduces long-term potentiation in the hippocampus, a cellular correlate of memory, we studied whether inhibition of α5 subunit-containing γ-aminobutyric acid type A receptors attenuated deficits in long-term potentiation after traumatic brain injury. DESIGN: Experimental animal study. SETTING: Research laboratory. SUBJECTS: Adult male mice and hippocampal brain slices. INTERVENTIONS: Anesthetized mice were subjected to traumatic brain injury with a closed-head, free-weight drop method. One week later, the mice were treated with L-655,708 (0.5 mg/kg), an inhibitor that is selective for α5 subunit-containing γ-aminobutyric acid type A receptors, 30 minutes before undergoing behavioral testing. Problem-solving abilities were assessed using the puzzle box assay, and memory performance was studied with novel object recognition and object place recognition assays. In addition, hippocampal slices were prepared 1 week after traumatic brain injury, and long-term potentiation was studied using field recordings in the cornu Ammonis 1 region of slices that were perfused with L-655,708 (100 nM). MEASUREMENTS AND MAIN RESULTS: Traumatic brain injury increased the time required to solve difficult but not simple tasks in the puzzle box assay and impaired memory in the novel object recognition and object place recognition assays. L-655,708 improved both problem solving and memory in the traumatic brain injury mice. Traumatic brain injury reduced long-term potentiation in the hippocampal slices, and L-655,708 attenuated this reduction. CONCLUSIONS: Pharmacologic inhibition of α5 subunit-containing γ-aminobutyric acid type A receptors attenuated cognitive deficits after traumatic brain injury and enhanced synaptic plasticity in hippocampal slices. Collectively, these results suggest that α5 subunit-containing γ-aminobutyric acid type A receptors are novel targets for pharmacologic treatment of traumatic brain injury-induced persistent cognitive deficits.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Imidazóis/farmacologia , Memória de Curto Prazo/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Cognição/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Camundongos , Modelos Animais
5.
Anesthesiology ; 129(3): 477-489, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29889105

RESUMO

WHAT WE ALREADY KNOW ABOUT THIS TOPIC: WHAT THIS ARTICLE TELLS US THAT IS NEW: BACKGROUND:: Postoperative delirium is associated with poor long-term outcomes and increased mortality. General anesthetic drugs may contribute to delirium because they increase cell-surface expression and function of α5 subunit-containing γ-aminobutyric acid type A receptors, an effect that persists long after the drugs have been eliminated. Dexmedetomidine, an α2 adrenergic receptor agonist, prevents delirium in patients and reduces cognitive deficits in animals. Thus, it was postulated that dexmedetomidine prevents excessive function of α5 γ-aminobutyric acid type A receptors. METHODS: Injectable (etomidate) and inhaled (sevoflurane) anesthetic drugs were studied using cultured murine hippocampal neurons, cultured murine and human cortical astrocytes, and ex vivo murine hippocampal slices. γ-Aminobutyric acid type A receptor function and cell-signaling pathways were studied using electrophysiologic and biochemical methods. Memory and problem-solving behaviors were also studied. RESULTS: The etomidate-induced sustained increase in α5 γ-aminobutyric acid type A receptor cell-surface expression was reduced by dexmedetomidine (mean ± SD, etomidate: 146.4 ± 51.6% vs. etomidate + dexmedetomidine: 118.4 ± 39.1% of control, n = 8 each). Dexmedetomidine also reduced the persistent increase in tonic inhibitory current in hippocampal neurons (etomidate: 1.44 ± 0.33 pA/pF, n = 10; etomidate + dexmedetomidine: 1.01 ± 0.45 pA/pF, n = 9). Similarly, dexmedetomidine prevented a sevoflurane-induced increase in the tonic current. Dexmedetomidine stimulated astrocytes to release brain-derived neurotrophic factor, which acted as a paracrine factor to reduce excessive α5 γ-aminobutyric acid type A receptor function in neurons. Finally, dexmedetomidine attenuated memory and problem-solving deficits after anesthesia. CONCLUSIONS: Dexmedetomidine prevented excessive α5 γ-aminobutyric acid type A receptor function after anesthesia. This novel α2 adrenergic receptor- and brain-derived neurotrophic factor-dependent pathway may be targeted to prevent delirium.


Assuntos
Anestésicos Intravenosos/farmacologia , Dexmedetomidina/farmacologia , Etomidato/farmacologia , Hipnóticos e Sedativos/farmacologia , Receptores de GABA-A/fisiologia , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Células Cultivadas , Técnicas de Cocultura , Função Executiva/efeitos dos fármacos , Função Executiva/fisiologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Anesth Analg ; 127(6): 1406-1413, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30303868

RESUMO

As part of the American Society of Anesthesiology Brain Health Initiative goal of improving perioperative brain health for older patients, over 30 experts met at the fifth International Perioperative Neurotoxicity Workshop in San Francisco, CA, in May 2016, to discuss best practices for optimizing perioperative brain health in older adults (ie, >65 years of age). The objective of this workshop was to discuss and develop consensus solutions to improve patient management and outcomes and to discuss what older adults should be told (and by whom) about postoperative brain health risks. Thus, the workshop was provider and patient oriented as well as solution focused rather than etiology focused. For those areas in which we determined that there were limited evidence-based recommendations, we identified knowledge gaps and the types of scientific knowledge and investigations needed to direct future best practice. Because concerns about perioperative neurocognitive injury in pediatric patients are already being addressed by the SmartTots initiative, our workshop discussion (and thus this article) focuses specifically on perioperative cognition in older adults. The 2 main perioperative cognitive disorders that have been studied to date are postoperative delirium and cognitive dysfunction. Postoperative delirium is a syndrome of fluctuating changes in attention and level of consciousness that occurs in 20%-40% of patients >60 years of age after major surgery and inpatient hospitalization. Many older surgical patients also develop postoperative cognitive deficits that typically last for weeks to months, thus referred to as postoperative cognitive dysfunction. Because of the heterogeneity of different tools and thresholds used to assess and define these disorders at varying points in time after anesthesia and surgery, a recent article has proposed a new recommended nomenclature for these perioperative neurocognitive disorders. Our discussion about this topic was organized around 4 key issues: preprocedure consent, preoperative cognitive assessment, intraoperative management, and postoperative follow-up. These 4 issues also form the structure of this document. Multiple viewpoints were presented by participants and discussed at this in-person meeting, and the overall group consensus from these discussions was then drafted by a smaller writing group (the 6 primary authors of this article) into this manuscript. Of course, further studies have appeared since the workshop, which the writing group has incorporated where appropriate. All participants from this in-person meeting then had the opportunity to review, edit, and approve this final manuscript; 1 participant did not approve the final manuscript and asked for his/her name to be removed.


Assuntos
Encéfalo/fisiologia , Síndromes Neurotóxicas/diagnóstico , Complicações Pós-Operatórias/diagnóstico , Complicações Pós-Operatórias/prevenção & controle , Idoso , Anestesia/efeitos adversos , Anestesiologia/métodos , Cognição , Transtornos Cognitivos/etiologia , Delírio , Esquema de Medicação , Eletroencefalografia , Humanos , Testes Neuropsicológicos , Síndromes Neurotóxicas/terapia , Assistência Perioperatória , Período Perioperatório , Período Pós-Operatório , Fatores de Risco , Sociedades Médicas , Estados Unidos
8.
Ann Neurol ; 79(1): 18-26, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26580862

RESUMO

Antifibrinolytic drugs are routinely used worldwide to reduce the bleeding that results from a wide range of hemorrhagic conditions. The most commonly used antifibrinolytic drug, tranexamic acid, is associated with an increased incidence of postoperative seizures. The reported increase in the frequency of seizures is alarming, as these events are associated with adverse neurological outcomes, longer hospital stays, and increased in-hospital mortality. However, many clinicians are unaware that tranexamic acid causes seizures. The goal of this review is to summarize the incidence, risk factors, and clinical features of these seizures. This review also highlights several clinical and preclinical studies that offer mechanistic insights into the potential causes of and treatments for tranexamic acid-associated seizures. This review will aid the medical community by increasing awareness about tranexamic acid-associated seizures and by translating scientific findings into therapeutic interventions for patients.


Assuntos
Antifibrinolíticos/efeitos adversos , Convulsões/induzido quimicamente , Ácido Tranexâmico/efeitos adversos , Animais , Antifibrinolíticos/farmacocinética , Humanos , Convulsões/tratamento farmacológico , Ácido Tranexâmico/farmacocinética , Ácido Tranexâmico/farmacologia
9.
Anesthesiology ; 126(4): 666-677, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27984263

RESUMO

BACKGROUND: The "dissociative " general anesthetic ketamine is a well-known N-methyl-D-aspartate receptor antagonist. However, whether ketamine, at clinically relevant concentrations, increases the activity of inhibitory γ-aminobutyric acid (GABA) receptor type A (GABAA) receptors in different brain regions remains controversial. Here, the authors studied the effects of ketamine on synaptic and extrasynaptic GABAA receptors in hippocampal neurons. Ketamine modulation of extrasynaptic GABAA receptors in cortical neurons was also examined. METHODS: Whole cell currents were recorded from cultured murine neurons. Current evoked by exogenous GABA, miniature inhibitory postsynaptic currents, and currents directly activated by ketamine were studied. RESULTS: Ketamine did not alter the amplitude, frequency, or kinetics of postsynaptic currents but increased a tonic inhibitory current generated by extrasynaptic GABAA receptors in hippocampal neurons. For example, ketamine (100 µM) increased the tonic current by 33.6 ± 6.5% (mean ± SEM; 95% CI, 18.2 to 48.9; n = 8, P < 0.001). Ketamine shifted the GABA concentration-response curve to the left, but only when GABAA receptors were activated by low concentrations of GABA (n = 6). The selective increase in tonic current was attributed to ketamine increasing the apparent potency of GABA at high-affinity extrasynaptic GABAA receptors. Ketamine also increased a tonic current in cortical neurons (n = 11). Ketamine directly gated the opening of GABAA receptors, but only at high concentrations that are unlikely to occur during clinical use. CONCLUSIONS: Clinically relevant concentrations of ketamine increased the activity of high-affinity extrasynaptic GABAA receptors in the hippocampus and cortex, an effect that likely contributes to ketamine's neurodepressive properties.


Assuntos
Anestésicos Dissociativos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Ketamina/farmacologia , Neurônios/efeitos dos fármacos , Receptores de GABA/efeitos dos fármacos , Animais , Células Cultivadas , Córtex Cerebral/fisiologia , Feminino , Hipocampo/fisiologia , Masculino , Camundongos , Modelos Animais , Neurônios/fisiologia , Receptores de GABA/fisiologia
10.
Anesthesiology ; 127(1): 89-97, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28475556

RESUMO

BACKGROUND: The antifibrinolytic drug tranexamic acid is structurally similar to the amino acid glycine and may cause seizures and myoclonus by acting as a competitive antagonist of glycine receptors. Glycine is an obligatory co-agonist of the N-methyl-D-aspartate (NMDA) subtype of glutamate receptors. Thus, it is plausible that tranexamic acid inhibits NMDA receptors by acting as a competitive antagonist at the glycine binding site. The aim of this study was to determine whether tranexamic acid inhibits NMDA receptors, as well as α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and kainate subtypes of ionotropic glutamate receptors. METHODS: Tranexamic acid modulation of NMDA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and kainate receptors was studied using whole cell voltage-clamp recordings of current from cultured mouse hippocampal neurons. RESULTS: Tranexamic acid rapidly and reversibly inhibited NMDA receptors (half maximal inhibitory concentration = 241 ± 45 mM, mean ± SD; 95% CI, 200 to 281; n = 5) and shifted the glycine concentration-response curve for NMDA-evoked current to the right. Tranexamic acid also inhibited α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (half maximal inhibitory concentration = 231 ± 91 mM; 95% CI, 148 to 314; n = 5 to 6) and kainate receptors (half maximal inhibitory concentration = 90 ± 24 mM; 95% CI, 68 to 112; n = 5). CONCLUSIONS: Tranexamic acid inhibits NMDA receptors likely by reducing the binding of the co-agonist glycine and also inhibits α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and kainate receptors. Receptor blockade occurs at high millimolar concentrations of tranexamic acid, similar to the concentrations that occur after topical application to peripheral tissues. Glutamate receptors in tissues including bone, heart, and nerves play various physiologic roles, and tranexamic acid inhibition of these receptors may contribute to adverse drug effects.


Assuntos
Antifibrinolíticos/farmacologia , Receptores Ionotrópicos de Glutamato/efeitos dos fármacos , Ácido Tranexâmico/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Camundongos , Modelos Animais , Neurônios/efeitos dos fármacos
11.
Anesthesiology ; 124(2): 417-27, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26566283

RESUMO

BACKGROUND: Critically ill patients with severe inflammation often exhibit heightened sensitivity to general anesthetics; however, the underlying mechanisms remain poorly understood. Inflammation increases the number of γ-aminobutyric acid type A (GABAA) receptors expressed on the surface of neurons, which supports the hypothesis that inflammation increases up-regulation of GABAA receptor activity by anesthetics, thereby enhancing the behavioral sensitivity to these drugs. METHODS: To mimic inflammation in vitro, cultured hippocampal and cortical neurons were pretreated with interleukin (IL)-1ß. Whole cell patch clamp methods were used to record currents evoked by γ-aminobutyric acid (GABA) (0.5 µM) in the absence and presence of etomidate or isoflurane. To mimic inflammation in vivo, mice were treated with lipopolysaccharide, and several anesthetic-related behavioral endpoints were examined. RESULTS: IL-1ß increased the amplitude of current evoked by GABA in combination with clinically relevant concentrations of either etomidate (3 µM) or isoflurane (250 µM) (n = 5 to 17, P < 0.05). Concentration-response plots for etomidate and isoflurane showed that IL-1ß increased the maximal current 3.3-fold (n = 5 to 9) and 1.5-fold (n = 8 to 11), respectively (P < 0.05 for both), whereas the half-maximal effective concentrations were unchanged. Lipopolysaccharide enhanced the hypnotic properties of both etomidate and isoflurane. The immobilizing properties of etomidate, but not isoflurane, were also increased by lipopolysaccharide. Both lipopolysaccharide and etomidate impaired contextual fear memory. CONCLUSIONS: These results provide proof-of-concept evidence that inflammation increases the sensitivity of neurons to general anesthetics. This increase in anesthetic up-regulation of GABAA receptor activity in vitro correlates with enhanced sensitivity for GABAA receptor-dependent behavioral endpoints in vivo.


Assuntos
Anestésicos Gerais/farmacologia , Inflamação/fisiopatologia , Neurônios/efeitos dos fármacos , Anestésicos Inalatórios/farmacologia , Animais , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Etomidato/farmacologia , Hipnóticos e Sedativos/farmacologia , Isoflurano/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Receptores de GABA-A/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Ácido gama-Aminobutírico/efeitos dos fármacos
12.
Anesth Analg ; 123(5): 1247-1252, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27464975

RESUMO

BACKGROUND: Extrasynaptic γ-aminobutyric acid type A (GABAA) receptors that contain the δ subunit (δGABAA receptors) contribute to memory performance. Dysregulation of δGABAA receptor expression, which occurs in some neurological disorders, is associated with memory impairment. Mice lacking δGABAA receptors (Gabrd) exhibit deficits in their ability to distinguish between similar memories, a process which is referred to as pattern separation. The CA3 and dentate gyrus subfields of the hippocampus regulate pattern separation, raising the possibility that synaptic plasticity is impaired in these regions in Gabrd mice. Although long-term potentiation (LTP), the most widely studied form of synaptic plasticity, is normal in the dentate gyrus of Gabrd mice, LTP in the CA3 subfield has not been studied. Here, we tested the hypothesis that LTP is reduced in the CA3 subfield of Gabrd mice. METHODS: LTP of extracellular field postsynaptic potentials was studied in the mossy fiber (MF)-CA3 pathway using hippocampal slices from Gabrd and wild-type (WT) mice. We also examined paired pulse responses and input-output relationships at MF-CA3 synapses. RESULTS: MF-CA3 LTP was reduced in Gabrd mice, as evidenced by decreased potentiation of field postsynaptic potentials (WT: 178.3% ± 16.1% versus Gabrd: 126.3% ± 6.9%; P = 0.0091). Thus, the deletion of δGABAA receptors is associated with impaired plasticity. Bicuculline (BIC), a GABAA receptor antagonist, reduced plasticity in WT but not in Gabrd mice (WT + BIC: 123.9% ± 7.6% versus Gabrd + BIC: 136.5% ± 7.0%). Paired pulse responses and input-output relationships did not differ between the genotypes (all Ps > 0.05). CONCLUSIONS: Both genetic deletion and pharmacological blockade of δGABAA receptors impair MF-CA3 LTP, suggesting that δGABAA receptors are necessary for synaptic plasticity in the CA3 subfield. Drugs that enhance δGABAA receptor function may reverse deficits in synaptic plasticity in the CA3 subfield and improve pattern separation in neurological disorders.


Assuntos
Região CA3 Hipocampal/fisiologia , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Receptores de GABA-A/deficiência , Receptores de GABA-A/genética , Animais , Região CA3 Hipocampal/efeitos dos fármacos , Antagonistas GABAérgicos/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Memória/efeitos dos fármacos , Camundongos , Camundongos Knockout , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Cultura de Órgãos
13.
J Neurosci ; 34(32): 10624-34, 2014 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-25100596

RESUMO

Hydrogen peroxide (H2O2), a key reactive oxygen species, is produced at low levels during normal cellular metabolism and at higher concentrations under pathological conditions such as ischemia-reperfusion injury. The mechanisms by which H2O2 contributes to physiological and pathological processes in the brain remain poorly understood. Inhibitory GABA type A (GABAA) receptors critically regulate brain function by generating tonic and synaptic currents; however, it remains unknown whether H2O2 directly modulates GABAA receptor function. Here, we performed patch-clamp recordings, together with pharmacological and genetic approaches, to investigate the effects of H2O2 on GABAA receptor-mediated tonic and synaptic currents recorded in cultured mouse hippocampal neurons and CA1 pyramidal neurons in hippocampal slices. We found that H2O2 caused a dramatic increase in tonic current, whereas synaptic currents were unaffected. This increase in tonic current resulted from an extracellular oxidative reaction, which increased the potency of GABA, but only when GABAA receptors were activated by low concentrations of GABA. Oxygen-glucose deprivation, which produces high endogenous levels of H2O2, similarly increased the tonic current. These results suggest that GABAA receptor-mediated tonic current, which is potentiated by H2O2, might contribute to H2O2-induced brain dysfunction.


Assuntos
Hipocampo/citologia , Peróxido de Hidrogênio/farmacologia , Neurônios/efeitos dos fármacos , Oxidantes/farmacologia , Receptores de GABA-A/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Glucose/deficiência , Hipóxia/patologia , Hipóxia/fisiopatologia , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Espécies Reativas de Oxigênio/metabolismo , Receptores de GABA-A/genética
16.
Ann Neurol ; 74(4): 611-21, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23686887

RESUMO

OBJECTIVE: Extrasynaptic γ-aminobutyric acid type A receptors that contain the δ subunit (δGABAA receptors) are highly expressed in the dentate gyrus (DG) subfield of the hippocampus, where they generate a tonic conductance that regulates neuronal activity. GABAA receptor-dependent signaling regulates memory and also facilitates postnatal neurogenesis in the adult DG; however, the role of the δGABAA receptors in these processes is unclear. Accordingly, we sought to determine whether δGABAA receptors regulate memory behaviors, as well as neurogenesis in the DG. METHODS: Memory and neurogenesis were studied in wild-type (WT) mice and transgenic mice that lacked δGABAA receptors (Gabrd(-/-)). To pharmacologically increase δGABAA receptor activity, mice were treated with the δGABAA receptor-preferring agonist 4,5,6,7-tetrahydroisoxazolo(5,4-c)pyridin-3-ol (THIP). Behavioral assays including recognition memory, contextual discrimination, and fear extinction were used. Neurogenesis was studied by measuring the proliferation, survival, migration, maturation, and dendritic complexity of adult-born neurons in the DG. RESULTS: Gabrd(-/-) mice exhibited impaired recognition memory and contextual discrimination relative to WT mice. Fear extinction was also impaired in Gabrd(-/-) mice, although the acquisition of fear memory was enhanced. Neurogenesis was disrupted in Gabrd(-/-) mice as the migration, maturation, and dendritic development of adult-born neurons were impaired. Long-term treatment with THIP facilitated learning and neurogenesis in WT but not Gabrd(-/-) mice. INTERPRETATION: δGABAA receptors promote the performance of certain DG-dependent memory behaviors and facilitate neurogenesis. Furthermore, δGABAA receptors can be pharmacologically targeted to enhance these processes.


Assuntos
Giro Denteado/fisiologia , Memória/fisiologia , Neurogênese/genética , Receptores de GABA-A/metabolismo , Análise de Variância , Animais , Discriminação Psicológica/fisiologia , Eletrochoque/efeitos adversos , Comportamento Exploratório/fisiologia , Agonistas GABAérgicos/farmacologia , Isoxazóis/farmacologia , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de GABA-A/genética , Reconhecimento Psicológico/fisiologia
17.
Anesthesiology ; 130(4): 618-619, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30870211
18.
Transl Res ; 267: 39-53, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38042478

RESUMO

General anesthetic drugs cause cognitive deficits that persist after the drugs have been eliminated. Astrocytes may contribute to such cognition-impairing effects through the release of one or more paracrine factors that increase a tonic inhibitory conductance generated by extrasynaptic γ-aminobutyric acid type A (GABAA) receptors in hippocampal neurons. The mechanisms underlying this astrocyte-to-neuron crosstalk remain unknown. Interestingly, astrocytes express anesthetic-sensitive GABAA receptors. Here, we tested the hypothesis that anesthetic drugs activate astrocytic GABAA receptors to initiate crosstalk leading to a persistent increase in extrasynaptic GABAA receptor function in neurons. We also investigated the signaling pathways in neurons and aimed to identify the paracrine factors released from astrocytes. Astrocytes and neurons from mice were grown in primary cell cultures and studied using in vitro electrophysiological and biochemical assays. We discovered that the commonly used anesthetics etomidate (injectable) and sevoflurane (inhaled) stimulated astrocytic GABAA receptors, which in turn promoted the release paracrine factors, that increased the tonic current in neurons via a p38 MAPK-dependent signaling pathway. The increase in tonic current was mimicked by exogenous IL-1ß and abolished by blocking IL-1 receptors; however, unexpectedly, IL-1ß and other cytokines were not detected in astrocyte-conditioned media. In summary, we have identified a novel form of crosstalk between GABAA receptors in astrocytes and neurons that engages a p38 MAPK-dependent pathway. Brief commentary BACKGROUND: Many older patients experience cognitive deficits after surgery. Anesthetic drugs may be a contributing factor as they cause a sustained increase in the function of "memory blocking" extrasynaptic GABAA receptors in neurons. Interestingly, astrocytes are required for this increase; however, the mechanisms underlying the astrocyte-to-neuron crosstalk remain unknown. TRANSLATIONAL SIGNIFICANCE: We discovered that commonly used general anesthetic drugs stimulate GABAA receptors in astrocytes, which in turn release paracrine factors that trigger a persistent increase in extrasynaptic GABAA receptor function in neurons via p38 MAPK. This novel form of crosstalk may contribute to persistent cognitive deficits after general anesthesia and surgery.


Assuntos
Anestésicos Gerais , Receptores de GABA-A , Humanos , Camundongos , Animais , Receptores de GABA-A/metabolismo , Astrócitos/metabolismo , Neurônios , Anestésicos Gerais/farmacologia , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
BJA Open ; 6: 100143, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37588178

RESUMO

Background: Inflammation and general anaesthesia likely contribute to perioperative neurocognitive disorders, possibly by causing a neuronal imbalance of excitation and inhibition. We showed previously that treatment with lipopolysaccharide (LPS) and sevoflurane causes a sustained increase in a tonic inhibitory conductance in the hippocampus; however, whether excitatory neurotransmission is also altered remains unknown. The goal of this study was to examine excitatory synaptic currents in the hippocampus after treatment with LPS and sevoflurane. Synaptic plasticity in the hippocampus, a cellular correlate of learning and memory, was also studied. Methods: Mice were injected with vehicle or LPS (1 mg kg-1 i.p.), and after 24 h they were then exposed to vehicle or sevoflurane (2.3%; 2 h). Hippocampal slices were prepared 48 h later. Excitatory synaptic currents were recorded from pyramidal neurones. Long-term potentiation (LTP) and long-term depression (LTD) were studied in the Schaffer collateral-cornu ammonis 1 pathway. Results: The amplitude of miniature excitatory postsynaptic currents (EPSCs) was reduced after LPS+sevoflurane (P<0.001), whereas that of spontaneous EPSCs was unaltered, as evidenced by cumulative distribution plots. The frequency, area, and kinetics of both miniature and spontaneous EPSCs were unchanged, as were LTP and LTD. Conclusions: The reduced amplitude of miniature EPSCs, coupled with the previously reported increase in tonic inhibition, indicates that the combination of LPS and sevoflurane markedly disrupts the balance of excitation and inhibition. Restoring this balance by pharmacologically enhancing excitatory neurotransmission and inhibiting the tonic current may represent an effective therapeutic option for perioperative neurocognitive disorders.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA