Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 650
Filtrar
1.
Curr Neuropharmacol ; 22(2): 217-240, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37563812

RESUMO

Rhythmic eupneic breathing in mammals depends on the coordinated activities of the neural system that sends cranial and spinal motor outputs to respiratory muscles. These outputs modulate lung ventilation and adjust respiratory airflow, which depends on the upper airway patency and ventilatory musculature. Anesthetics are widely used in clinical practice worldwide. In addition to clinically necessary pharmacological effects, respiratory depression is a critical side effect induced by most general anesthetics. Therefore, understanding how general anesthetics modulate the respiratory system is important for the development of safer general anesthetics. Currently used volatile anesthetics and most intravenous anesthetics induce inhibitory effects on respiratory outputs. Various general anesthetics produce differential effects on respiratory characteristics, including the respiratory rate, tidal volume, airway resistance, and ventilatory response. At the cellular and molecular levels, the mechanisms underlying anesthetic-induced breathing depression mainly include modulation of synaptic transmission of ligand-gated ionotropic receptors (e.g., γ-aminobutyric acid, N-methyl-D-aspartate, and nicotinic acetylcholine receptors) and ion channels (e.g., voltage-gated sodium, calcium, and potassium channels, two-pore domain potassium channels, and sodium leak channels), which affect neuronal firing in brainstem respiratory and peripheral chemoreceptor areas. The present review comprehensively summarizes the modulation of the respiratory system by clinically used general anesthetics, including the effects at the molecular, cellular, anatomic, and behavioral levels. Specifically, analgesics, such as opioids, which cause respiratory depression and the "opioid crisis", are discussed. Finally, underlying strategies of respiratory stimulation that target general anesthetics and/or analgesics are summarized.


Assuntos
Anestésicos Gerais , Receptores Nicotínicos , Insuficiência Respiratória , Animais , Humanos , Anestésicos Gerais/farmacologia , Anestésicos Gerais/uso terapêutico , Analgésicos , Sistema Nervoso , Canais de Potássio , Sódio , Mamíferos
3.
J Phys Chem B ; 127(28): 6306-6315, 2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37432029

RESUMO

General anesthetics are indispensable in modern medicine because they induce a reversible loss of consciousness and sensation in humans. On the other hand, their molecular mechanisms of action have not yet been elucidated. Several studies have identified the main targets of some general anesthetics. The structures of γ-aminobutyric acid A (GABAA) receptors with the intravenous anesthetics such as propofol and etomidate have recently been determined. Although these anesthetic binding structures provide essential insights into the mechanism of action of anesthetics, the detailed molecular mechanism of how the anesthetic binding affects the Cl- permeability of GABAA receptors remains to be elucidated. In this study, we performed coarse-grained molecular dynamics simulations for GABAA receptors and analyzed the resulting simulation trajectories to investigate the effects of anesthetic binding on the motion of GABAA receptors. The results showed large structural fluctuations in GABAA receptors, correlations of motion between the amino acid residues, large amplitude motion, and autocorrelated slow motion, which were obtained by advanced statistical analyses. In addition, a comparison of the resulting trajectories in the presence or absence of the anesthetic molecules revealed a characteristic pore motion related to the gate-opening motion of GABAA receptors.


Assuntos
Anestésicos Gerais , Propofol , Humanos , Receptores de GABA-A/química , Simulação de Dinâmica Molecular , Anestésicos Intravenosos/farmacologia , Propofol/farmacologia , Propofol/química , Anestésicos Gerais/farmacologia , Ácido gama-Aminobutírico
4.
J Phys Chem B ; 127(27): 6078-6090, 2023 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-37368412

RESUMO

General anesthesia can be caused by various, chemically very different molecules, while several other molecules, many of which are structurally rather similar to them, do not exhibit anesthetic effects at all. To understand the origin of this difference and shed some light on the molecular mechanism of general anesthesia, we report here molecular dynamics simulations of the neat dipalmitoylphosphatidylcholine (DPPC) membrane as well as DPPC membranes containing the anesthetics diethyl ether and chloroform and the structurally similar non-anesthetics n-pentane and carbon tetrachloride, respectively. To also account for the pressure reversal of anesthesia, these simulations are performed both at 1 bar and at 600 bar. Our results indicate that all solutes considered prefer to stay both in the middle of the membrane and close to the boundary of the hydrocarbon domain, at the vicinity of the crowded region of the polar headgroups. However, this latter preference is considerably stronger for the (weakly polar) anesthetics than for the (apolar) non-anesthetics. Anesthetics staying in this outer preferred position increase the lateral separation between the lipid molecules, giving rise to a decrease of the lateral density. The lower lateral density leads to an increased mobility of the DPPC molecules, a decreased order of their tails, an increase of the free volume around this outer preferred position, and a decrease of the lateral pressure at the hydrocarbon side of the apolar/polar interface, a change that might well be in a causal relation with the occurrence of the anesthetic effect. All these changes are clearly reverted by the increase of pressure. Furthermore, non-anesthetics occur in this outer preferred position in a considerably smaller concentration and hence either induce such changes in a much weaker form or do not induce them at all.


Assuntos
Anestesia , Anestésicos Gerais , Anestésicos Gerais/farmacologia , Bicamadas Lipídicas/química , Membranas , Clorofórmio/química , 1,2-Dipalmitoilfosfatidilcolina/química
5.
Nat Commun ; 14(1): 3169, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37264005

RESUMO

General anesthetics and neuromuscular blockers are used together during surgery to stabilize patients in an unconscious state. Anesthetics act mainly by potentiating inhibitory ion channels and inhibiting excitatory ion channels, with the net effect of dampening nervous system excitability. Neuromuscular blockers act by antagonizing nicotinic acetylcholine receptors at the motor endplate; these excitatory ligand-gated ion channels are also inhibited by general anesthetics. The mechanisms by which anesthetics and neuromuscular blockers inhibit nicotinic receptors are poorly understood but underlie safe and effective surgeries. Here we took a direct structural approach to define how a commonly used anesthetic and two neuromuscular blockers act on a muscle-type nicotinic receptor. We discover that the intravenous anesthetic etomidate binds at an intrasubunit site in the transmembrane domain and stabilizes a non-conducting, desensitized-like state of the channel. The depolarizing neuromuscular blocker succinylcholine also stabilizes a desensitized channel but does so through binding to the classical neurotransmitter site. Rocuronium binds in this same neurotransmitter site but locks the receptor in a resting, non-conducting state. Together, this study reveals a structural mechanism for how general anesthetics work on excitatory nicotinic receptors and further rationalizes clinical observations in how general anesthetics and neuromuscular blockers interact.


Assuntos
Anestésicos Gerais , Anestésicos , Etomidato , Receptores Nicotínicos , Humanos , Receptores Nicotínicos/metabolismo , Anestésicos Intravenosos/farmacologia , Anestésicos Gerais/farmacologia , Etomidato/farmacologia , Músculos/metabolismo
6.
Neuroendocrinology ; 113(10): 1008-1023, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37271138

RESUMO

INTRODUCTION: Despite the widespread use of general anaesthetics, the mechanisms mediating their effects are still not understood. Although suppressed in most parts of the brain, neuronal activity, as measured by FOS activation, is increased in the hypothalamic supraoptic nucleus (SON) by numerous general anaesthetics, and evidence points to this brain region being involved in the induction of general anaesthesia (GA) and natural sleep. Posttranslational modifications of proteins, including changes in phosphorylation, enable fast modulation of protein function which could be underlying the rapid effects of GA. In order to identify potential phosphorylation events in the brain-mediating GA effects, we have explored the phosphoproteome responses in the rat SON and compared these to cingulate cortex (CC) which displays no FOS activation in response to general anaesthetics. METHODS: Adult Sprague-Dawley rats were treated with isoflurane for 15 min. Proteins from the CC and SON were extracted and processed for nano-LC mass spectrometry (LC-MS/MS). Phosphoproteomic determinations were performed by LC-MS/MS. RESULTS: We found many changes in the phosphoproteomes of both the CC and SON in response to 15 min of isoflurane exposure. Pathway analysis indicated that proteins undergoing phosphorylation adaptations are involved in cytoskeleton remodelling and synaptic signalling events. Importantly, changes in protein phosphorylation appeared to be brain region specific suggesting that differential phosphorylation adaptations might underlie the different neuronal activity responses to GA between the CC and SON. CONCLUSION: In summary, these data suggest that rapid posttranslational modifications in proteins involved in cytoskeleton remodelling and synaptic signalling events might mediate the central mechanisms mediating GA.


Assuntos
Anestésicos Gerais , Isoflurano , Ratos , Animais , Núcleo Supraóptico/metabolismo , Isoflurano/farmacologia , Isoflurano/metabolismo , Cromatografia Líquida , Ratos Sprague-Dawley , Proteínas Proto-Oncogênicas c-fos/metabolismo , Espectrometria de Massas em Tandem , Hipotálamo/metabolismo , Anestésicos Gerais/metabolismo , Anestésicos Gerais/farmacologia , Núcleo Hipotalâmico Paraventricular/metabolismo
8.
Naunyn Schmiedebergs Arch Pharmacol ; 396(2): 375-381, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36385685

RESUMO

General anesthetic drugs have been associated with various unwanted effects including an interference with mitochondrial function. We had previously observed increases of lactate formation in the mouse brain during anesthesia with volatile anesthetic agents. In the present work, we used mitochondria that were freshly isolated from mouse brain to test mitochondrial respiration and ATP synthesis in the presence of six common anesthetic drugs. The volatile anesthetics isoflurane, halothane, and (to a lesser extent) sevoflurane caused an inhibition of complex I of the electron transport chain in a dose-dependent manner. Significant effects were seen at concentrations that are reached under clinical conditions (< 0.5 mM). Pentobarbital and propofol also inhibited complex I but at concentrations that were two-fold higher than clinical EC50 values. Only propofol caused an inhibition of complex II. Complex IV respiration was not affected by either agent. Ketamine did not affect mitochondrial respiration. Similarly, all anesthetic agents except ketamine suppressed ATP production at high concentrations. Only halothane increased cytochrome c release indicating damage of the mitochondrial membrane. In summary, volatile general anesthetic agents as well as pentobarbital and propofol dose-dependently inhibit mitochondrial respiration. This action may contribute to depressive actions of the drugs in the brain.


Assuntos
Anestésicos Gerais , Isoflurano , Ketamina , Propofol , Camundongos , Animais , Halotano/farmacologia , Ketamina/farmacologia , Propofol/farmacologia , Pentobarbital , Anestésicos Gerais/farmacologia , Isoflurano/farmacologia , Mitocôndrias , Complexo I de Transporte de Elétrons , Trifosfato de Adenosina
9.
CNS Neurosci Ther ; 29(1): 378-389, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36353753

RESUMO

AIMS: The claustrum has long been regarded as a vital center for conscious control. Electrical stimulation or damage to the claustrum can result in decreased awareness or loss of consciousness, suggesting that the claustrum may be a target for the action of general anesthetics. This study aimed to determine the role of the claustrum in propofol anesthesia. METHODS: We first applied a fiber photometry calcium signal recording system to record the claustral neuronal activity during the entire process of propofol anesthesia. Chemogenetic activation of claustral neurones was then performed to verify their role in anesthesia. Finally, muscimol (GABAa receptor agonist) and gabazine (GABAa receptor antagonist) were microinjected into the claustrum to determine whether their GABAa receptors were involved in modulating propofol anesthesia. EEG and behavioral indicators, such as anesthetic sensitivity and efficacy, were recorded and analyzed. RESULTS: An evident anesthesia-related change in claustrum neuronal activity was suppressed during propofol-induced unconsciousness and restored following recovery from anesthesia. Chemogenetic activation of claustrum neurons results in attenuated propofol sensitivity, a shorter anesthesia duration, and an EEG shift toward wakefulness. Manipulation of GABAa receptors in the claustrum showed bidirectional control of propofol sensitivity, as activation decreases anesthesia efficiency while inactivation augments it. Additionally, inhibiting claustrum GABAa receptors increases cortical EEG slow waves. CONCLUSIONS: Claustrum neurones and their GABAa receptors are implicated in the modulation of propofol anesthesia in both behavioral and EEG assessments. Our findings create scope to reveal the brain targets of anesthetic action further and add to the existing evidence on the consciousness-modulating role of the claustrum.


Assuntos
Anestesia , Anestésicos Gerais , Claustrum , Propofol , Propofol/farmacologia , Receptores de GABA-A , Anestésicos Gerais/farmacologia , Eletroencefalografia
10.
Br J Anaesth ; 129(5): 703-715, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36031420

RESUMO

BACKGROUND: Hypersensitivity to general anaesthetics predicts adverse postoperative outcomes in patients. Hypoxia exerts extensive pathophysiological effects on the brain; however, whether hypoxia influences sevoflurane sensitivity and its underlying mechanisms remain poorly understood. METHODS: Mice were acclimated to hypoxia (oxygen 10% for 8 h day-1) for 28 days and anaesthetised with sevoflurane; the effective concentrations for 50% of the animals (EC50) showing loss of righting reflex (LORR) and loss of tail-pinch withdrawal response (LTWR) were determined. Positron emission tomography-computed tomography, O-glycoproteomics, seahorse analysis, carbon-13 tracing, site-specific mutagenesis, and electrophysiological techniques were performed to explore the underlying mechanisms. RESULTS: Compared with the control group, the hypoxia-acclimated mice required higher concentrations of sevoflurane to present LORR and LTWR (EC50LORR: 1.61 [0.03]% vs 1.46 [0.04]%, P<0.01; EC50LTWR: 2.46 [0.14]% vs 2.22 [0.06]%, P<0.01). Hypoxia-induced reduction in sevoflurane sensitivity was correlated with elevation of protein O-linked N-acetylglucosamine (O-GlcNAc) modification in brain, especially in the thalamus, and could be abolished by 6-diazo-5-oxo-l-norleucine, a glutamine fructose-6-phosphate amidotransferase inhibitor, and mimicked by thiamet-G, a selective O-GlcNAcase inhibitor. Mechanistically, O-GlcNAcylation drives de novo synthesis of glutamine from glucose in astrocytes and promotes the glutamate-glutamine cycle, partially via glycolytic flux and activation of glutamine synthetase. CONCLUSIONS: Intermittent hypoxia exposure decreased mouse sensitivity to sevoflurane anaesthesia through enhanced O-GlcNAc-dependent modulation of the glutamate-glutamine cycle in the brain.


Assuntos
Acetilglucosamina , Anestésicos Gerais , Animais , Camundongos , Acetilglucosamina/metabolismo , Acetilglucosamina/farmacologia , Sevoflurano/farmacologia , Glutamina/farmacologia , Diazo-Oxo-Norleucina/farmacologia , Glutamato-Amônia Ligase/metabolismo , Glutamato-Amônia Ligase/farmacologia , Encéfalo , Hipóxia , Glucose/metabolismo , Anestésicos Gerais/farmacologia , Oxigênio/farmacologia , Glutamatos/farmacologia
11.
Brain Res ; 1792: 148018, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35850186

RESUMO

Propofol is well known to cause amnesia independent of its sedative effect. Memory consolidation processes in the hippocampus have been proposed as a target - however the neural substrates for propofol's amnesic actions remain understudied and poorly described. In particular, the potential role of the cerebral cortex has not been investigated. As an in vitro experimental model of cortical memory consolidation, potentiated cerebral cortex evoked responses were generated in mouse neocortical slices using high frequency (20 Hz) stimulation to layer IV cortical grey matter or subcortical white matter. In separate experiments, slices were pretreated with propofol at two concentrations, 2 µg/mL and 4 µg/mL, to determine the effect of clinically relevant propofol levels on the potentiation response. Only grey matter stimulation induced a significant and lasting increase in cortical evoked potential amplitude in the drug-free condition. Propofol at 2 µg/mL completely inhibited cortical evoked response potentiation, while the 4 µg/mL concentration caused a small but significant depressant effect consequent to the high frequency stimulation. These findings support the hypothesis that propofol disrupts memory consolidation and actively facilitates memory decay in the cerebral cortex. The results further highlight the importance of the cerebral cortex in the early phase of long term memory consolidation.


Assuntos
Anestésicos Gerais , Neocórtex , Propofol , Anestésicos Gerais/farmacologia , Anestésicos Intravenosos/farmacologia , Animais , Hipocampo , Camundongos , Propofol/farmacologia
12.
Neuron ; 110(12): 1891-1893, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35709695

RESUMO

In this issue of Neuron, Bharioke et al. (2022) demonstrate that diverse general anesthetic regimens all reversibly and selectively synchronize spontaneous activity of pyramidal neurons in layer 5 of mouse cortex. We discuss the implications of these findings for the mechanism of consciousness and anesthetic-induced unconsciousness.


Assuntos
Anestésicos Gerais , Estado de Consciência , Anestesia Geral , Anestésicos Gerais/farmacologia , Animais , Estado de Consciência/fisiologia , Eletroencefalografia , Camundongos , Neurônios/fisiologia , Inconsciência/induzido quimicamente
13.
PLoS Comput Biol ; 18(6): e1009743, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35737717

RESUMO

General anesthetics work through a variety of molecular mechanisms while resulting in the common end point of sedation and loss of consciousness. Generally, the administration of common anesthetics induces reduction in synaptic excitation while promoting synaptic inhibition. Exogenous modulation of the anesthetics' synaptic effects can help determine the neuronal pathways involved in anesthesia. For example, both animal and human studies have shown that exogenously induced increases in acetylcholine in the brain can elicit wakeful-like behavior despite the continued presence of the anesthetic. However, the underlying mechanisms of anesthesia reversal at the cellular level have not been investigated. Here we apply a computational model of a network of excitatory and inhibitory neurons to simulate the network-wide effects of anesthesia, due to changes in synaptic inhibition and excitation, and their reversal by cholinergic activation through muscarinic receptors. We use a differential evolution algorithm to fit model parameters to match measures of spiking activity, neuronal connectivity, and network dynamics recorded in the visual cortex of rodents during anesthesia with desflurane in vivo. We find that facilitating muscarinic receptor effects of acetylcholine on top of anesthetic-induced synaptic changes predicts the reversal of anesthetic suppression of neurons' spiking activity, functional connectivity, as well as pairwise and population interactions. Thus, our model predicts a specific neuronal mechanism for the cholinergic reversal of anesthesia consistent with experimental behavioral observations.


Assuntos
Anestesia , Anestésicos Gerais , Acetilcolina/metabolismo , Acetilcolina/farmacologia , Anestésicos Gerais/farmacologia , Animais , Córtex Cerebral/fisiologia , Colinérgicos/farmacologia
14.
Neuron ; 110(12): 2024-2040.e10, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35452606

RESUMO

General anesthetics induce loss of consciousness, a global change in behavior. However, a corresponding global change in activity in the context of defined cortical cell types has not been identified. Here, we show that spontaneous activity of mouse layer 5 pyramidal neurons, but of no other cortical cell type, becomes consistently synchronized in vivo by different general anesthetics. This heightened neuronal synchrony is aperiodic, present across large distances, and absent in cortical neurons presynaptic to layer 5 pyramidal neurons. During the transition to and from anesthesia, changes in synchrony in layer 5 coincide with the loss and recovery of consciousness. Activity within both apical and basal dendrites is synchronous, but only basal dendrites' activity is temporally locked to somatic activity. Given that layer 5 is a major cortical output, our results suggest that brain-wide synchrony in layer 5 pyramidal neurons may contribute to the loss of consciousness during general anesthesia.


Assuntos
Anestésicos Gerais , Células Piramidais , Anestesia Geral , Anestésicos Gerais/farmacologia , Animais , Dendritos/fisiologia , Camundongos , Células Piramidais/fisiologia , Inconsciência
15.
Int J Mol Sci ; 23(4)2022 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-35216466

RESUMO

General anesthetics mainly act by modulating synaptic inhibition on the one hand (the potentiation of GABA transmission) or synaptic excitation on the other (the inhibition of NMDA receptors), but they can also have effects on numerous other proteins, receptors, and channels. The effects of general anesthetics on ion channels have been the subject of research since the publication of reports of direct actions of these drugs on ion channel proteins. In particular, there is considerable interest in T-type voltage-gated calcium channels that are abundantly expressed in the thalamus, where they control patterns of cellular excitability and thalamocortical oscillations during awake and sleep states. Here, we summarized and discussed our recent studies focused on the CaV3.1 isoform of T-channels in the nonspecific thalamus (intralaminar and midline nuclei), which acts as a key hub through which natural sleep and general anesthesia are initiated. We used mouse genetics and in vivo and ex vivo electrophysiology to study the role of thalamic T-channels in hypnosis induced by a standard general anesthetic, isoflurane, as well as novel neuroactive steroids. From the results of this study, we conclude that CaV3.1 channels contribute to thalamocortical oscillations during anesthetic-induced hypnosis, particularly the slow-frequency range of δ oscillations (0.5-4 Hz), by generating "window current" that contributes to the resting membrane potential. We posit that the role of the thalamic CaV3.1 isoform of T-channels in the effects of various classes of general anesthetics warrants consideration.


Assuntos
Anestésicos Gerais/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Hipnóticos e Sedativos/farmacologia , Neurônios/metabolismo , Animais , Humanos , Potenciais da Membrana , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia
16.
Curr Neuropharmacol ; 20(1): 27-54, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34344292

RESUMO

General anesthetics depress excitatory and/or enhance inhibitory synaptic transmission principally by modulating the function of glutamatergic or GABAergic synapses, respectively, with relative anesthetic agent-specific mechanisms. Synaptic signaling proteins, including ligand- and voltage-gated ion channels, are targeted by general anesthetics to modulate various synaptic mechanisms, including presynaptic neurotransmitter release, postsynaptic receptor signaling, and dendritic spine dynamics to produce their characteristic acute neurophysiological effects. As synaptic structure and plasticity mediate higher-order functions such as learning and memory, long-term synaptic dysfunction following anesthesia may lead to undesirable neurocognitive consequences depending on the specific anesthetic agent and the vulnerability of the population. Here we review the cellular and molecular mechanisms of transient and persistent general anesthetic alterations of synaptic transmission and plasticity.


Assuntos
Anestésicos Gerais , Anestésicos Gerais/farmacologia , Humanos , Aprendizagem , Sinapses , Transmissão Sináptica
17.
Curr Neuropharmacol ; 20(1): 55-71, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34503426

RESUMO

Although general anesthetics have been used in the clinic for more than 170 years, the ways in which they induce amnesia, unconsciousness, analgesia, and immobility remain elusive. Modulations of various neural nuclei and circuits are involved in the actions of general anesthetics. The expression of the immediate-early gene c-fos and its nuclear product, c-fos protein, can be induced by neuronal depolarization; therefore, c-fos staining is commonly used to identify the activated neurons during sleep and/or wakefulness, as well as in various physiological conditions in the central nervous system. Identifying c-fos expression is also a direct and convenient method to explore the effects of general anesthetics on the activity of neural nuclei and circuits. Using c-fos staining, general anesthetics have been found to interact with sleep- and wakefulness-promoting systems throughout the brain, which may explain their ability to induce unconsciousness and emergence from general anesthesia. This review summarizes the actions of general anesthetics on neural nuclei and circuits based on a c-fos expression.


Assuntos
Anestésicos Gerais , Anestesia Geral , Anestésicos Gerais/farmacologia , Encéfalo , Humanos , Neurônios , Sono
18.
J Neurophysiol ; 126(5): 1660-1669, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34644166

RESUMO

Studies of in vivo neuronal responses to auditory inputs in the superior olive complex (SOC) are usually done under anesthesia. However, little attention has been paid to the effect of anesthesia itself on response properties. Here, we assessed the effect of anesthesia depth under ketamine-xylazine anesthetics on auditory evoked response properties of lateral SOC neurons. Anesthesia depth was tracked by monitoring EEG spectral peak frequencies. An increase in anesthesia depth led to a decrease of spontaneous discharge activities and an elevated response threshold. The temporal responses to suprathreshold tones were also affected, with adapted responses reduced but peak responses unaffected. Deepening the anesthesia depth also increased first spike latency. However, spike jitter was not affected. Auditory brainstem responses to clicks confirmed that ketamine-xylazine anesthesia depth affects auditory neuronal activities and the effect on spike rate and spike timing persists through the auditory pathway. We concluded from those observations that ketamine-xylazine affects lateral SOC response properties depending on the anesthesia depth.NEW & NOTEWORTHY We studied how the depth of ketamine-xylazine anesthesia altered response properties of lateral superior olive complex neurons, and auditory brainstem evoked responses. Our results provide direct evidence that anesthesia depth affects auditory neuronal responses and reinforce the notion that both the anesthetics and the anesthesia depth should be considered when interpreting/comparing in vivo neuronal recordings.


Assuntos
Anestesia , Anestésicos Gerais/farmacologia , Percepção Auditiva/efeitos dos fármacos , Ketamina/farmacologia , Complexo Olivar Superior/efeitos dos fármacos , Xilazina/farmacologia , Animais , Eletroencefalografia/efeitos dos fármacos , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Feminino , Gerbillinae , Masculino
19.
J Neurophysiol ; 126(2): 668-679, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34259043

RESUMO

Activation of renal sensory nerves by chemo- and mechanosensitive stimuli produces changes in efferent sympathetic nerve activity (SNA) and arterial blood pressure (ABP). Anesthesia and sex influence autonomic function and cardiovascular hemodynamics, but it is unclear to what extent anesthesia and sex impact SNA and ABP responses to renal sensory stimuli. We measured renal, splanchnic, and lumbar SNA and ABP in male and female Sprague-Dawley rats during contralateral renal infusion of capsaicin and bradykinin or during elevation in renal pelvic pressure. Responses were evaluated with a decerebrate preparation or Inactin, urethane, or isoflurane anesthesia. Intrarenal arterial infusion of capsaicin (0.1-30.0 µM) increased renal SNA, splanchnic SNA, or ABP but decreased lumbar SNA in the Inactin group. Intrarenal arterial infusion of bradykinin (0.1-30.0 µM) increased renal SNA, splanchnic SNA, and ABP but decreased lumbar SNA in the Inactin group. Elevated renal pelvic pressure (0-20 mmHg, 30 s) significantly increased renal SNA and splanchnic SNA but not lumbar SNA in the Inactin group. In marked contrast, SNA and ABP responses to every renal stimulus were severely blunted in the urethane and decerebrate groups and absent in the isoflurane group. In the Inactin group, the magnitude of SNA responses to chemo- and mechanosensory stimuli were not different between male and female rats. Thus, chemo- and mechanosensitive stimuli produce differential changes in renal, splanchnic, and lumbar SNA. Experimentally, future investigations should consider Inactin anesthesia to examine sympathetic and hemodynamic responses to renal sensory stimuli.NEW & NOTEWORTHY The findings highlight the impact of anesthesia, and to a lesser extent sex, on sympathetic efferent and hemodynamic responses to chemosensory and mechanosensory renal stimuli. Sympathetic nerve activity (SNA) and arterial blood pressure (ABP) responses were present in Inactin-anesthetized rats but largely absent in decerebrate, isoflurane, or urethane preparations. Renal chemosensory stimuli differentially changed SNA: renal and splanchnic SNA increased, but lumbar SNA decreased. Future investigations should consider Inactin anesthesia to study SNA and hemodynamic responses to renal sensory nerve activation.


Assuntos
Anestésicos Gerais/farmacologia , Hemodinâmica , Rim/inervação , Neurônios Eferentes/fisiologia , Sistema Nervoso Simpático/fisiologia , Animais , Capsaicina/farmacologia , Feminino , Isoflurano/farmacologia , Rim/efeitos dos fármacos , Rim/fisiologia , Masculino , Neurônios Eferentes/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fármacos do Sistema Sensorial/farmacologia , Fatores Sexuais , Sistema Nervoso Simpático/efeitos dos fármacos , Tiopental/análogos & derivados , Tiopental/farmacologia , Tato , Uretana/farmacologia
20.
AANA J ; 89(3): 213-220, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34042572

RESUMO

Carotid endarterectomy (CEA) has a 1-5% risk of periprocedural stroke. The ability to emerge patients from anesthesia quickly to detect neurological abnormalities immediately after surgery is vital in this patient population. The objective of this pilot study was to assess if any of three general anesthetic techniques for CEA are associated with a shorter time to a reliable postoperative neurological exam. Secondary objectives were to assess postoperative cognitive dysfunction (POCD), postoperative delirium (POD), and hemodynamic stability. Twenty-one patients undergoing CEA were enrolled and randomized to different combinations of inhalational and intravenous anesthesia: Group A: propofol, remifentanil, and desflurane; Group B: dexmedetomidine, remifentanil, and desflurane; Group C: remifentanil and desflurane. Anesthetic depth was titrated using a bispectral index (BIS) monitor to a goal of 50-60. Time was recorded from surgery end to first meaningful neurological exam. Neurocognitive testing was completed preoperatively and up to 1 week postoperatively to assess POD (3D-CAM) and POCD (Short Blessed Test). Time to first reliable neurological exam was 2 minutes longer in group A (9 min ± 4 min) compared to group B and group C (7 min ± 3 min; 7 min ± 4 min), although this was not statistically significant. In addition, extubation time was significantly longer in group A (11 min) compared to group B and group C (5 min; 6 min) (P = 0.03). 3D -CAM and Short Blessed Test data along with hemodynamics did not differ significantly between the groups. Time to first useful neurologic exam and hemodynamics did not differ between the groups. However, extubation time was significantly prolonged in patients who received propofol, but not dexmedetomidine, as part of their anesthetic for CEA. These findings are best verified in an adequately powered prospective randomized study.


Assuntos
Anestésicos Gerais , Endarterectomia das Carótidas , Propofol , Anestesia Geral , Anestésicos Gerais/farmacologia , Anestésicos Intravenosos , Hemodinâmica/efeitos dos fármacos , Humanos , Projetos Piloto , Piperidinas/farmacologia , Estudos Prospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...