Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Br J Anaesth ; 131(2): 348-359, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37268445

RESUMO

BACKGROUND: Anaesthetic-induced unresponsiveness and non-rapid eye movement (NREM) sleep share common neural pathways and neurophysiological features. We hypothesised that these states bear resemblance also at the experiential level. METHODS: We compared, in a within-subject design, the prevalence and content of experiences in reports obtained after anaesthetic-induced unresponsiveness and NREM sleep. Healthy males (N=39) received dexmedetomidine (n=20) or propofol (n=19) in stepwise doses to induce unresponsiveness. Those rousable were interviewed and left unstimulated, and the procedure was repeated. Finally, the anaesthetic dose was increased 50%, and the participants were interviewed after recovery. The same participants (N=37) were also later interviewed after NREM sleep awakenings. RESULTS: Most subjects were rousable, with no difference between anaesthetic agents (P=0.480). Lower drug plasma concentrations were associated with being rousable for both dexmedetomidine (P=0.007) and propofol (P=0.002) but not with recall of experiences in either drug group (dexmedetomidine: P=0.543; propofol: P=0.460). Of the 76 and 73 interviews performed after anaesthetic-induced unresponsiveness and NREM sleep, 69.7% and 64.4% included experiences, respectively. Recall did not differ between anaesthetic-induced unresponsiveness and NREM sleep (P=0.581), or between dexmedetomidine and propofol in any of the three awakening rounds (P>0.05). Disconnected dream-like experiences (62.3% vs 51.1%; P=0.418) and memory incorporation of the research setting (88.7% vs 78.7%; P=0.204) were equally often present in anaesthesia and sleep interviews, respectively, whereas awareness, signifying connected consciousness, was rarely reported in either state. CONCLUSIONS: Anaesthetic-induced unresponsiveness and NREM sleep are characterised by disconnected conscious experiences with corresponding recall frequencies and content. CLINICAL TRIAL REGISTRATION: Clinical trial registration. This study was part of a larger study registered at ClinicalTrials.gov (NCT01889004).


Assuntos
Anestésicos , Dexmedetomidina , Propofol , Humanos , Masculino , Dexmedetomidina/efeitos adversos , Movimentos Oculares , Hipnóticos e Sedativos/efeitos adversos , Propofol/efeitos adversos , Sono
2.
J Neurosci ; 43(26): 4884-4895, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37225435

RESUMO

Establishing the neural mechanisms responsible for the altered global states of consciousness during anesthesia and dissociating these from other drug-related effects remains a challenge in consciousness research. We investigated differences in brain activity between connectedness and disconnectedness by administering various anesthetics at concentrations designed to render 50% of the subjects unresponsive. One hundred and sixty healthy male subjects were randomized to receive either propofol (1.7 µg/ml; n = 40), dexmedetomidine (1.5 ng/ml; n = 40), sevoflurane (0.9% end-tidal; n = 40), S-ketamine (0.75 µg/ml; n = 20), or saline placebo (n = 20) for 60 min using target-controlled infusions or vaporizer with end-tidal monitoring. Disconnectedness was defined as unresponsiveness to verbal commands probed at 2.5-min intervals and unawareness of external events in a postanesthesia interview. High-resolution positron emission tomography (PET) was used to quantify regional cerebral metabolic rates of glucose (CMRglu) utilization. Contrasting scans where the subjects were classified as connected and responsive versus disconnected and unresponsive revealed that for all anesthetics, except S-ketamine, the level of thalamic activity differed between these states. A conjunction analysis across the propofol, dexmedetomidine and sevoflurane groups confirmed the thalamus as the primary structure where reduced metabolic activity was related to disconnectedness. Widespread cortical metabolic suppression was observed when these subjects, classified as either connected or disconnected, were compared with the placebo group, suggesting that these findings may represent necessary but alone insufficient mechanisms for the change in the state of consciousness.SIGNIFICANCE STATEMENT Experimental anesthesia is commonly used in the search for measures of brain function which could distinguish between global states of consciousness. However, most previous studies have not been designed to separate effects related to consciousness from other effects related to drug exposure. We employed a novel study design to disentangle these effects by exposing subjects to predefined EC50 doses of four commonly used anesthetics or saline placebo. We demonstrate that state-related effects are remarkably limited compared with the widespread cortical effects related to drug exposure. In particular, decreased thalamic activity was associated with disconnectedness with all used anesthetics except for S-ketamine.


Assuntos
Anestesia , Anestésicos Inalatórios , Dexmedetomidina , Ketamina , Propofol , Masculino , Humanos , Propofol/farmacologia , Sevoflurano/farmacologia , Ketamina/farmacologia , Dexmedetomidina/farmacologia , Anestésicos Inalatórios/farmacologia , Anestésicos Intravenosos
3.
BJA Open ; 4: 100114, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37588789

RESUMO

Background: This exploratory study aimed to investigate whether dexmedetomidine, propofol, sevoflurane, and S-ketamine affect oxylipins and bile acids, which are functionally diverse molecules with possible connections to cellular bioenergetics, immune modulation, and organ protection. Methods: In this randomised, open-label, controlled, parallel group, Phase IV clinical drug trial, healthy male subjects (n=160) received equipotent doses (EC50 for verbal command) of dexmedetomidine (1.5 ng ml-1; n=40), propofol (1.7 µg ml-1; n=40), sevoflurane (0.9% end-tidal; n=40), S-ketamine (0.75 µg ml-1; n=20), or placebo (n=20). Blood samples for tandem mass spectrometry were obtained at baseline, after study drug administration at 60 and 130 min from baseline; 40 metabolites were analysed. Results: Statistically significant changes vs placebo were observed in 62.5%, 12.5%, 5.0%, and 2.5% of analytes in dexmedetomidine, propofol, sevoflurane, and S-ketamine groups, respectively. Data are presented as standard deviation score, 95% confidence interval, and P-value. Dexmedetomidine induced wide-ranging decreases in oxylipins and bile acids. Amongst others, 9,10-dihydroxyoctadecenoic acid (DiHOME) -1.19 (-1.6; -0.78), P<0.001 and 12,13-DiHOME -1.22 (-1.66; -0.77), P<0.001 were affected. Propofol elevated 9,10-DiHOME 2.29 (1.62; 2.96), P<0.001 and 12,13-DiHOME 2.13 (1.42; 2.84), P<0.001. Analytes were mostly unaffected by S-ketamine. Sevoflurane decreased tauroursodeoxycholic acid (TUDCA) -2.7 (-3.84; -1.55), P=0.015. Conclusions: Dexmedetomidine-induced oxylipin alterations may be connected to pathways associated with organ protection. In contrast to dexmedetomidine, propofol emulsion elevated DiHOMEs, oxylipins associated with acute respiratory distress syndrome, and mitochondrial dysfunction in high concentrations. Further research is needed to establish the behaviour of DIHOMEs during prolonged propofol/dexmedetomidine infusions and to verify the sevoflurane-induced reduction in TUDCA, a suggested neuroprotective agent. Clinical trial registration: NCT02624401.

4.
J Neurosci ; 41(8): 1769-1778, 2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33372062

RESUMO

What happens in the brain when conscious awareness of the surrounding world fades? We manipulated consciousness in two experiments in a group of healthy males and measured brain activity with positron emission tomography. Measurements were made during wakefulness, escalating and constant levels of two anesthetic agents (experiment 1, n = 39), and during sleep-deprived wakefulness and non-rapid eye movement sleep (experiment 2, n = 37). In experiment 1, the subjects were randomized to receive either propofol or dexmedetomidine until unresponsiveness. In both experiments, forced awakenings were applied to achieve rapid recovery from an unresponsive to a responsive state, followed by immediate and detailed interviews of subjective experiences during the preceding unresponsive condition. Unresponsiveness rarely denoted unconsciousness, as the majority of the subjects had internally generated experiences. Unresponsive anesthetic states and verified sleep stages, where a subsequent report of mental content included no signs of awareness of the surrounding world, indicated a disconnected state. Functional brain imaging comparing responsive and connected versus unresponsive and disconnected states of consciousness during constant anesthetic exposure revealed that activity of the thalamus, cingulate cortices, and angular gyri are fundamental for human consciousness. These brain structures were affected independent from the pharmacologic agent, drug concentration, and direction of change in the state of consciousness. Analogous findings were obtained when consciousness was regulated by physiological sleep. State-specific findings were distinct and separable from the overall effects of the interventions, which included widespread depression of brain activity across cortical areas. These findings identify a central core brain network critical for human consciousness.SIGNIFICANCE STATEMENT Trying to understand the biological basis of human consciousness is currently one of the greatest challenges of neuroscience. While the loss and return of consciousness regulated by anesthetic drugs and physiological sleep are used as model systems in experimental studies on consciousness, previous research results have been confounded by drug effects, by confusing behavioral "unresponsiveness" and internally generated consciousness, and by comparing brain activity levels across states that differ in several other respects than only consciousness. Here, we present carefully designed studies that overcome many previous confounders and for the first time reveal the neural mechanisms underlying human consciousness and its disconnection from behavioral responsiveness, both during anesthesia and during normal sleep, and in the same study subjects.


Assuntos
Encéfalo/fisiologia , Estado de Consciência/fisiologia , Hipnóticos e Sedativos/farmacologia , Privação do Sono/fisiopatologia , Sono REM/fisiologia , Vigília/fisiologia , Adulto , Encéfalo/efeitos dos fármacos , Dexmedetomidina/farmacologia , Humanos , Masculino , Tomografia por Emissão de Pósitrons , Propofol/farmacologia , Inconsciência/induzido quimicamente , Inconsciência/fisiopatologia
5.
BMC Anesthesiol ; 19(1): 222, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31805854

RESUMO

BACKGROUND: Surgery and diseases modify inflammatory responses and the immune system. Anesthetic agents also have effects on the human immune system but the responses they induce may be altered or masked by the surgical procedures or underlying illnesses. The aim of this study was to assess how single-drug dexmedetomidine and propofol anesthesia without any surgical intervention alter acute immunological biomarkers in healthy subjects. METHODS: Thirty-five healthy, young male subjects were anesthetized using increasing concentrations of dexmedetomidine (n = 18) or propofol (n = 17) until loss of responsiveness (LOR) was detected. The treatment allocation was randomized. Multi-parametric immunoassays for the detection of 48 cytokines, chemokines and growth factors were used. Concentrations were determined at baseline and at the highest drug concentration for each subject. RESULTS: The changes in the concentration of eotaxin (decrease after dexmedetomidine) and platelet-derived growth factor (PDGF, increase after propofol) were statistically significantly different between the groups. Significant changes were detected within both groups; the concentrations of monocyte chemotactic protein 1, chemokine ligand 27 and macrophage migration inhibitory factor were lower in both groups after the drug administration. Dexmedetomidine decreased the concentration of eotaxin, interleukin-18, interleukin-2Rα, stem cell factor, stem cell growth factor and vascular endothelial growth factor, and propofol decreased significantly the levels of hepatocyte growth factor, IFN-γ-induced protein 10 and monokine induced by IFN-γ, and increased the levels of interleukin-17, interleukin-5, interleukin-7 and PDGF. CONCLUSIONS: Dexmedetomidine seemed to have an immunosuppressive effect on the immune system whereas propofol seemed to induce mixed pro- and anti-inflammatory effects on the immune system. The choice of anesthetic agent could be relevant when treating patients with compromised immunological defense mechanisms. TRIAL REGISTRATION: Before subject enrollment, the study was registered in the European Clinical Trials database (EudraCT number 2013-001496-21, The Neural Mechanisms of Anesthesia and Human Consciousness) and in ClinicalTrials.gov (Principal Investigator: Harry Scheinin, number NCT01889004, The Neural Mechanisms of Anesthesia and Human Consciousness, Part 2, on the 23rd of June 2013).


Assuntos
Citocinas/metabolismo , Dexmedetomidina/farmacologia , Hipnóticos e Sedativos/farmacologia , Propofol/farmacologia , Adulto , Anestésicos Intravenosos/administração & dosagem , Anestésicos Intravenosos/farmacologia , Quimiocinas/metabolismo , Dexmedetomidina/administração & dosagem , Relação Dose-Resposta a Droga , Humanos , Hipnóticos e Sedativos/administração & dosagem , Masculino , Propofol/administração & dosagem , Adulto Jovem
6.
Anesthesiology ; 129(1): 22-36, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29642080

RESUMO

BACKGROUND: Differentiating drug-related changes and state-related changes on the electroencephalogram during anesthetic-induced unconsciousness has remained a challenge. To distinguish these, we designed a rigorous experimental protocol with two drugs known to have distinct molecular mechanisms of action. We hypothesized that drug- and state-related changes can be separated. METHODS: Forty-seven healthy participants were randomized to receive dexmedetomidine (n = 23) or propofol (n = 24) as target-controlled infusions until loss of responsiveness. Then, an attempt was made to arouse the participant to regain responsiveness while keeping the drug infusion constant. Finally, the concentration was increased 1.5-fold to achieve presumable loss of consciousness. We conducted statistical comparisons between the drugs and different states of consciousness for spectral bandwidths, and observed how drug-induced electroencephalogram patterns reversed upon awakening. Cross-frequency coupling was also analyzed between slow-wave phase and alpha power. RESULTS: Eighteen (78%) and 10 (42%) subjects were arousable during the constant drug infusion in the dexmedetomidine and propofol groups, respectively (P = 0.011 between the drugs). Corresponding with deepening anesthetic level, slow-wave power increased, and a state-dependent alpha anteriorization was detected with both drugs, especially with propofol. The slow-wave and frontal alpha activities were momentarily disrupted as the subjects regained responsiveness at awakening. Negative phase-amplitude coupling before and during loss of responsiveness frontally and positive coupling during the highest drug concentration posteriorly were observed in the propofol but not in the dexmedetomidine group. CONCLUSIONS: Electroencephalogram effects of dexmedetomidine and propofol are strongly drug- and state-dependent. Changes in slow-wave and alpha activity seemed to best detect different states of consciousness.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Dexmedetomidina/administração & dosagem , Eletroencefalografia/efeitos dos fármacos , Hipnóticos e Sedativos/administração & dosagem , Propofol/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 2/sangue , Adulto , Anestésicos Intravenosos , Dexmedetomidina/sangue , Eletroencefalografia/métodos , Humanos , Hipnóticos e Sedativos/sangue , Infusões Intravenosas , Masculino , Propofol/sangue , Adulto Jovem
7.
J Neurosci ; 32(14): 4935-43, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22492049

RESUMO

One of the greatest challenges of modern neuroscience is to discover the neural mechanisms of consciousness and to explain how they produce the conscious state. We sought the underlying neural substrate of human consciousness by manipulating the level of consciousness in volunteers with anesthetic agents and visualizing the resultant changes in brain activity using regional cerebral blood flow imaging with positron emission tomography. Study design and methodology were chosen to dissociate the state-related changes in consciousness from the effects of the anesthetic drugs. We found the emergence of consciousness, as assessed with a motor response to a spoken command, to be associated with the activation of a core network involving subcortical and limbic regions that become functionally coupled with parts of frontal and inferior parietal cortices upon awakening from unconsciousness. The neural core of consciousness thus involves forebrain arousal acting to link motor intentions originating in posterior sensory integration regions with motor action control arising in more anterior brain regions. These findings reveal the clearest picture yet of the minimal neural correlates required for a conscious state to emerge.


Assuntos
Estado de Consciência/fisiologia , Lobo Frontal/fisiologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Lobo Parietal/fisiologia , Vigília/fisiologia , Adulto , Anestesia Geral/métodos , Encéfalo/citologia , Encéfalo/fisiologia , Mapeamento Encefálico/métodos , Lobo Frontal/citologia , Humanos , Masculino , Rede Nervosa/citologia , Lobo Parietal/citologia , Adulto Jovem
8.
Anesth Analg ; 112(3): 587-92, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21233497

RESUMO

BACKGROUND: Conventional time and frequency domain measures of heart rate variability (HRV) are strongly influenced by anesthetic drugs, and are therefore not able to detect subtle changes in HRV, even during light anesthesia. Approximate entropy of R-R intervals is an HRV measure that has a tendency to decrease during anesthesia, but it is severely compromised by low-frequency variations of the signal. However, the negative effect of the low-frequency variations can be eliminated by differentiating the R-R interval tachogram before analysis. We designed this study to investigate characteristics of a novel HRV measure, named δ entropy (dEn), during deepening anesthesia. METHODS: Eight healthy subjects were anesthetized with sevoflurane and 8 with propofol in a stepwise manner using 3 escalating concentrations (2%, 3%, and 4% end-tidal concentration and 7.4 ± 1.7, 12.3 ± 2.6, and 18.3 ± 5.0 µg/mL plasma concentration, respectively) at 30-minute intervals. A third group of 8 subjects received a supramaximal IV dose of glycopyrrolate without anesthesia to examine the effect of cardiac vagal activity on dEn. We computed dEn at baseline, during each step of anesthesia and during the anticholinergic blockade. RESULTS: The dEn decreased along with deepening levels of sevoflurane and propofol anesthesia up to 33% (95% confidence interval [CI] 21%-44%) and 38% (95% CI 28%-48%), respectively. At each anesthesia level, dEn differed significantly (P < 0.05) from that measured at the preceding level, similarly in both the sevoflurane and propofol groups. Parasympathetic blockade by glycopyrrolate was found to decrease dEn by 17% (95% CI 6%-28%). CONCLUSIONS: The dEn is a novel HRV measure able to detect subtle sympathetic- and parasympathetic-mediated alterations in HRV both during deepening levels of sevoflurane and propofol anesthesia and during exceedingly deep anesthesia.


Assuntos
Anestesia , Ritmo Delta/fisiologia , Entropia , Frequência Cardíaca/fisiologia , Monitorização Intraoperatória/métodos , Adulto , Anestesia/efeitos adversos , Ritmo Delta/efeitos dos fármacos , Eletrocardiografia/efeitos dos fármacos , Eletrocardiografia/métodos , Frequência Cardíaca/efeitos dos fármacos , Humanos , Masculino , Éteres Metílicos/efeitos adversos , Propofol/efeitos adversos , Sevoflurano , Adulto Jovem
9.
Anesth Analg ; 108(2): 593-600, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19151294

RESUMO

BACKGROUND: General anesthetics can alter the relationship between regional cerebral glucose metabolism (rCMR(glc)) and blood flow (rCBF). In this positron emission tomography study, our aim was to assess both rCMR(glc) and rCBF in the same individuals during xenon anesthesia. METHODS: (18)F-labeled fluorodeoxyglucose and (15)O-labeled water were used to determine rCMR(glc) and rCBF, respectively, in five healthy male subjects at baseline (awake) and during 1 minimum alveolar anesthetic concentration of xenon. Anesthesia was based solely on xenon. Changes in rCMR(glc) and rCBF were quantified using region-of-interest and voxel-based analyses. RESULTS: The mean (sd) xenon concentration during anesthesia was 67.2 (0.8)%. Xenon anesthesia induced a uniform reduction in rCMR(glc), whereas rCBF decreased in 7 of 13 brain regions. The mean decreases in the gray matter were 32.4 (4.0)% (P < 0.001) and 14.8 (5.9)% (P = 0.007) for rCMR(glc) and rCBF, respectively. rCMR(glc) decreased by 10.9 (6.4)% in the white matter (P = 0.030), whereas rCBF increased by 9.2 (7.3)% (P = 0.049). The rCBF/rCMR(glc) ratio was especially increased in the insula, anterior and posterior cingulate, and in the somatosensory cortex. CONCLUSIONS: In general, the magnitude of the decreases in rCMR(glc) during 1 minimum alveolar anesthetic concentration xenon anesthesia exceeded the reductions in rCBF. As a result, the ratio between rCMR(glc) and rCBF was shifted to a higher level. Interestingly, xenon-induced changes in cerebral metabolism and blood flow resemble those induced by volatile anesthetics.


Assuntos
Anestesia por Inalação , Anestésicos Inalatórios , Química Encefálica/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Circulação Cerebrovascular/efeitos dos fármacos , Glucose/metabolismo , Xenônio , Adulto , Anestesia com Circuito Fechado , Glicemia/metabolismo , Fluordesoxiglucose F18 , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Respiração Artificial , Adulto Jovem
10.
Anesth Analg ; 106(1): 129-34, table of contents, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18165567

RESUMO

BACKGROUND: The noble gas xenon acts as an anesthetic with favorable hemodynamic and neuroprotective properties. Based on animal and in vitro data, it is thought to exert its anesthetic effects by inhibiting glutamatergic signaling, but effects on gamma-aminobutyric acid type A (GABA(A)) receptors also have been reported. The mechanism of anesthetic action of xenon in the living human brain still remains to be determined. METHODS: We used the specific GABA(A) receptor benzodiazepine-site ligand 11C-flumazenil and positron emission tomography to study the GABAergic effects of xenon in eight healthy male volunteers. Each subject underwent two dynamic 60-min positron emission tomography studies awake and during approximately one minimum alveolar concentration of xenon (65%). Bispectral index was recorded. Cortical and subcortical gray matter regions were analyzed using both automated regions-of-interest analysis and voxel-based analysis. RESULTS: During anesthesia, the mean +/- sd bispectral index was 23 +/- 7, and there were no significant changes in heart rate or mean arterial blood pressure. Xenon did not significantly affect 11C-flumazenil binding in any brain region. CONCLUSIONS: Xenon did not affect 11C-flumazenil binding in the living human brain, indicating that the anesthetic effect of xenon is not mediated via the GABA(A) receptor system.


Assuntos
Anestésicos/farmacologia , Encéfalo/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Xenônio/farmacologia , Adulto , Anestésicos/administração & dosagem , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Radioisótopos de Carbono , Eletroencefalografia , Flumazenil/metabolismo , Moduladores GABAérgicos/metabolismo , Hemodinâmica/efeitos dos fármacos , Humanos , Ligantes , Masculino , Tomografia por Emissão de Pósitrons , Ligação Proteica , Compostos Radiofarmacêuticos/metabolismo , Receptores de GABA-A/metabolismo , Xenônio/administração & dosagem
11.
Anesthesiology ; 108(1): 63-70, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18156883

RESUMO

BACKGROUND: The aim was to evaluate the performance of anesthesia depth monitors, Bispectral Index (BIS) and Entropy, during single-agent xenon anesthesia in 17 healthy subjects. METHODS: After mask induction with xenon and intubation, anesthesia was continued with xenon only. BIS, State Entropy and Response Entropy, and electroencephalogram were monitored throughout induction, steady-state anesthesia, and emergence. The performance of BIS, State Entropy, and Response Entropy were evaluated with prediction probability, sensitivity, and specificity analyses. The power spectrum of the raw electroencephalogram signal was calculated. RESULTS: The mean (SD) xenon concentration during anesthesia was 66.4% (2.4%). BIS, State Entropy, and Response Entropy demonstrated low prediction probability values at loss of response (0.455, 0.656, and 0.619) but 1 min after that the values were high (0.804, 0.941, and 0.929). Thereafter, equally good performance was demonstrated for all indices. At emergence, the prediction probability values to distinguish between steady-state anesthesia and return of response for BIS, State Entropy, and Response Entropy were 0.988, 0.892, and 0.992. No statistical differences between the performances of the monitors were observed. Quantitative electroencephalogram analyses showed generalized increase in total power (P < 0.001), delta (P < 0.001) and theta activity (P < 0.001), and increased alpha activity (P = 0.003) in the frontal brain regions. CONCLUSIONS: Electroencephalogram-derived depth of sedation indices BIS and Entropy showed a delay to detect loss of response during induction of xenon anesthesia. Both monitors performed well in distinguishing between conscious and unconscious states during steady-state anesthesia. Xenon-induced changes in electroencephalogram closely resemble those induced by propofol.


Assuntos
Anestesia por Inalação/métodos , Eletroencefalografia/efeitos dos fármacos , Eletroencefalografia/métodos , Entropia , Xenônio , Adulto , Humanos , Masculino
12.
Anesthesiology ; 106(6): 1128-33, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17525587

RESUMO

BACKGROUND: Animal studies have demonstrated a strong neuroprotective property of xenon. Its usefulness in patients with cerebral pathology could be compromised by deleterious effects on regional cerebral blood flow (rCBF). METHODS: 15O-labeled water was used to determine rCBF in nine healthy male subjects at baseline and during 1 minimum alveolar concentration (MAC) of xenon (63%). Anesthesia was based solely on xenon. Absolute changes in rCBF were quantified using region-of-interest analysis and voxel-based analysis. RESULTS: Mean arterial blood pressure and arterial partial pressure for carbon dioxide remained unchanged. The mean (+/-SD) xenon concentration during anesthesia was 65.2+/-2.3%. Xenon anesthesia decreased absolute rCBF by 34.7+/-9.8% in the cerebellum (P<0.001), by 22.8+/-10.4% in the thalamus (P=0.001), and by 16.2+/-6.2% in the parietal cortex (P<0.001). On average, xenon anesthesia decreased absolute rCBF by 11.2+/-8.6% in the gray matter (P=0.008). A 22.1+/-13.6% increase in rCBF was detected in the white matter (P=0.001). Whole-brain voxel-based analysis revealed widespread cortical reductions and increases in rCBF in the precentral and postcentral gyri. CONCLUSIONS: One MAC of xenon decreased rCBF in several areas studied. The greatest decreases were detected in the cerebellum, the thalamus and the cortical areas. Increases in rCBF were observed in the white matter and in the pre- and postcentral gyri. These results are in clear contradiction with ketamine, another N-methyl-D-aspartate antagonist and neuroprotectant, which induces a general increase in cerebral blood flow at anesthetic concentrations.


Assuntos
Anestésicos Inalatórios/farmacologia , Circulação Cerebrovascular/efeitos dos fármacos , Tomografia por Emissão de Pósitrons , Xenônio/farmacologia , Adulto , Humanos , Masculino , Fluxo Sanguíneo Regional/efeitos dos fármacos
13.
Anesth Analg ; 101(3): 722-725, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16115981

RESUMO

Positron emission tomography (PET) studies suggest that propofol and inhaled anesthetics increase (11)C-flumazenil binding in the living human brain, thus supporting the involvement of gamma-aminobutyric acid type A (GABA(A)) receptors in the mechanism of action of these drugs. Ketamine produces its anesthetic effects primarily by N-methyl-d-aspartate receptor antagonism, but it may also have GABA(A) receptor agonistic properties. By using PET, we studied the cerebral (11)C-flumazenil binding in 10 healthy subjects before and during a subanesthetic racemic ketamine infusion reaching a serum concentration of 350 +/- 42 ng/mL. Ketamine did not affect (11)C-flumazenil binding to GABA(A) receptor in the brain, indicating that this mechanism is of minor importance in the actions of subanesthetic ketamine.


Assuntos
Encéfalo/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Flumazenil/farmacocinética , Moduladores GABAérgicos/farmacocinética , Ketamina/farmacologia , Adulto , Encéfalo/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Humanos , Infusões Intravenosas , Ketamina/administração & dosagem , Masculino , Tomografia por Emissão de Pósitrons , Receptores de GABA-A/efeitos dos fármacos
14.
Anesthesiology ; 103(2): 258-68, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16052107

RESUMO

BACKGROUND: Animal studies have demonstrated neuroprotective properties of S-ketamine, but its effects on cerebral blood flow (CBF), metabolic rate of oxygen (CMRO2), and glucose metabolic rate (GMR) have not been comprehensively studied in humans. METHODS: Positron emission tomography was used to quantify CBF and CMRO2 in eight healthy male volunteers awake and during S-ketamine infusion targeted to subanesthetic (150 ng/ml) and anesthetic (1,500-2,000 ng/ml) concentrations. In addition, subjects' GMRs were assessed awake and during anesthesia. Whole brain estimates for cerebral blood volume were obtained using kinetic modeling. RESULTS: The mean +/- SD serum S-ketamine concentration was 159 +/- 21 ng/ml at the subanesthetic and 1,959 +/- 442 ng/ml at the anesthetic levels. The total S-ketamine dose was 10.4 mg/kg. S-ketamine increased heart rate (maximally by 43.5%) and mean blood pressure (maximally by 27.0%) in a concentration-dependent manner (P = 0.001 for both). Subanesthetic S-ketamine increased whole brain CBF by 13.7% (P = 0.035). The greatest regional CBF increase was detected in the anterior cingulate (31.6%; P = 0.010). No changes were detected in CMRO2. Anesthetic S-ketamine increased whole brain CBF by 36.4% (P = 0.006) but had no effect on whole brain CMRO2 or GMR. Regionally, CBF was increased in nearly all brain structures studied (greatest increase in the insula 86.5%; P < 0.001), whereas CMRO2 increased only in the frontal cortex (by 15.7%; P = 0.007) and GMR increased only in the thalamus (by 11.7%; P = 0.010). Cerebral blood volume was increased by 51.9% (P = 0.011) during anesthesia. CONCLUSIONS: S-ketamine-induced CBF increases exceeded the minor changes in CMRO2 and GMR during anesthesia.


Assuntos
Anestesia , Encéfalo/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Ketamina/farmacologia , Adulto , Pressão Sanguínea/efeitos dos fármacos , Encéfalo/metabolismo , Glucose/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Humanos , Masculino , Consumo de Oxigênio , Tomografia por Emissão de Pósitrons
15.
Anesth Analg ; 99(5): 1420-1426, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15502041

RESUMO

Based on in vitro studies and animal data, most anesthetics are supposed to act via gamma-aminobutyric acid type A (GABA(A)) receptors. However, this fundamental characteristic has not been extensively investigated in humans. We studied (11)C-flumazenil binding to GABA(A) receptors during sevoflurane and propofol anesthesia in the living human brain using positron emission tomography (PET). Fourteen healthy male subjects underwent 2 60-min dynamic PET studies with (11)C-labeled flumazenil, awake and during anesthesia. Anesthesia was maintained with 2% end-tidal sevoflurane (n = 7) or propofol at a target plasma concentration of 9.0 +/- 3.0 (mean +/- sd) microg/mL (n = 7). The depth of anesthesia was measured with bispectral index (BIS). Values of regional distribution volumes (DV) of (11)C-flumazenil were calculated in several brain areas using metabolite-corrected arterial plasma curves and a two-compartment model. Separate voxel-based statistical analysis using parametric DV images was performed for detailed visualization. The average BIS index was 35 +/- 6 in the sevoflurane group and 28 +/- 8 in the propofol group (P = 0.02). Sevoflurane increased the DV of (11)C-flumazenil significantly (P < 0.05) in all brain areas studied except the pons and the white matter. In the propofol group the increases were significant (P < 0.05) in the caudatus, putamen, cerebellum, thalamus and the frontal, temporal, and parietal cortices. Furthermore, the DV increases in the frontal, occipital, parietal, and temporal cortical areas and in the putamen were statistically significantly larger in the sevoflurane than in the propofol group. Our findings support the involvement of GABA(A) receptors in the mechanism of action of both anesthetics in humans.


Assuntos
Anestésicos Inalatórios/farmacologia , Anestésicos Intravenosos/farmacologia , Flumazenil/metabolismo , Moduladores GABAérgicos/metabolismo , Éteres Metílicos/farmacologia , Propofol/farmacologia , Receptores de GABA-A/metabolismo , Adulto , Anestesia , Gasometria , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Dióxido de Carbono/metabolismo , Eletrocardiografia/efeitos dos fármacos , Eletroencefalografia/efeitos dos fármacos , Flumazenil/farmacocinética , Moduladores GABAérgicos/farmacocinética , Hemodinâmica/efeitos dos fármacos , Humanos , Masculino , Modelos Biológicos , Oxigênio/sangue , Tomografia por Emissão de Pósitrons , Receptores de GABA-A/efeitos dos fármacos , Reflexo/efeitos dos fármacos , Sevoflurano , Distribuição Tecidual
16.
J Nucl Med Technol ; 32(2): 79-82, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15175404

RESUMO

OBJECTIVES: The increasing use of PET for assessing cerebral blood flow, oxygen metabolism, and blood volume in critically ill patients has created a need for reliable technical solutions for delivering (15)O-tracer gases to mechanically ventilated subjects. Our objective was to create such a solution. METHODS: We designed a ventilator add-on unit that enables complex functional brain studies using labeled oxygen and carbon monoxide gases as tracers. The unit manages both steady-state and bolus inhalations, and the latter can be manually initiated using a remote trigger. All parts conducting breathing gases can be sterilized. The unit can be operated during both spontaneous pressure support breathing and volume-controlled ventilation. It supports the standard safety features and alarms of the ventilator and includes an overflow valve in the bolus reservoir. RESULTS: The count rate curves obtained using the new unit were similar to those from the standard bag-inhalation method. CONCLUSION: The unit we describe offers an economical and easily operated solution for providing uninterrupted ventilator treatment while performing PET brain studies, and the provided treatment meets intensive care criteria.


Assuntos
Monóxido de Carbono , Radioisótopos de Oxigênio , Tomografia por Emissão de Pósitrons/métodos , Respiração Artificial/instrumentação , Encéfalo/diagnóstico por imagem , Desenho de Equipamento , Humanos , Reprodutibilidade dos Testes , Fatores de Tempo
17.
Anesthesiology ; 100(5): 1065-71, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15114201

RESUMO

BACKGROUND: The authors have recently shown with positron emission tomography that subanesthetic doses of racemic ketamine increase cerebral blood flow but do not affect oxygen consumption significantly. In this study, the authors wanted to assess the effects of racemic ketamine on regional glucose metabolic rate (rGMR) in similar conditions to establish whether ketamine truly induces disturbed coupling between cerebral blood flow and metabolism. METHODS: 18F-labeled fluorodeoxyglucose was used as a positron emission tomography tracer to quantify rGMR on 12 brain regions of interest of nine healthy male volunteers at baseline and during a 300-ng/ml ketamine target concentration level. In addition, voxel-based analysis was performed for the relative changes in rGMR using statistical parametric mapping. RESULTS: The mean +/- SD measured ketamine serum concentration was 326.4+/-86.3 ng/ml. The mean arterial pressure was slightly increased (maximally by 16.4%) during ketamine infusion (P < 0.001). Ketamine increased absolute rGMR significantly in most regions of interest studied. The greatest increases were detected in the thalamus (14.6+/-15.9%; P = 0.029) and in the frontal (13.6+/-13.1%; P = 0.011) and parietal cortices (13.1+/-11.2%; P = 0.007). Absolute rGMR was not decreased anywhere in the brain. The voxel-based analysis revealed relative rGMR increases in the frontal, temporal, and parietal cortices. CONCLUSIONS: Global increases in rGMR seem to parallel ketamine-induced increases in cerebral blood flow detected in the authors' earlier study. Therefore, ketamine-induced disturbance of coupling between cerebral blood flow and metabolism is highly unlikely. The previously observed decrease in oxygen extraction fraction may be due to nonoxidative glucose metabolism during ketamine-induced increase in glutamate release.


Assuntos
Anestésicos Dissociativos/administração & dosagem , Encéfalo/efeitos dos fármacos , Glucose/metabolismo , Ketamina/administração & dosagem , Adulto , Análise de Variância , Encéfalo/metabolismo , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Fluordesoxiglucose F18/metabolismo , Humanos , Masculino , Tomografia Computadorizada de Emissão/métodos
18.
Anesthesiology ; 99(3): 603-13, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12960544

RESUMO

BACKGROUND: Anesthetic agents, especially volatile anesthetics and nitrous oxide (N2O), are suspected to perturb cerebral homeostasis and vascular reactivity. The authors quantified the effects of sevoflurane and propofol as sole anesthetics and in combination with N2O on regional cerebral blood flow (rCBF), metabolic rate of oxygen (rCMRO2), and blood volume (rCBV) in the living human brain using positron emission tomography. METHODS: 15O-labeled water, oxygen, and carbon monoxide were used as positron emission tomography tracers to determine rCBF, rCMRO2 and rCBV, respectively, in eight healthy male subjects during the awake state (baseline) and at four different anesthetic regimens: (1) sevoflurane alone, (2) sevoflurane plus 70% N2O (S+N), (3) propofol alone, and (4) propofol plus 70% N2O (P+N). Sevoflurane and propofol were titrated to keep a constant hypnotic depth (Bispectral Index 40) throughout anesthesia. End-tidal carbon dioxide was strictly kept at preinduction level. RESULTS: The mean +/- SD end-tidal concentration of sevoflurane was 1.5 +/- 0.3% during sevoflurane alone and 1.2 +/- 0.3% during S+N (P < 0.001). The measured propofol concentration was 3.7 +/- 0.7 microg/ml during propofol alone and 3.5 +/- 0.7 microg/ml during P+N (not significant). Sevoflurane alone decreased rCBF in some (to 73-80% of baseline, P < 0.01), and propofol in all brain structures (to 53-70%, P < 0.001). Only propofol reduced also rCBV (in the cortex and cerebellum to 83-86% of baseline, P < 0.05). Both sevoflurane and propofol similarly reduced rCMRO2 in all brain areas to 56-70% and 50-68% of baseline, respectively (P < 0.05). The adjunct N2O counteracted some of the rCMRO2 and rCBF reductions caused by drugs alone, and especially during S+N, a widespread reduction (P < 0.05 for all cortex and cerebellum vs. awake) in the oxygen extraction fraction was seen. Adding of N2O did not alter the rCBV effects of sevoflurane and propofol alone. CONCLUSIONS: Propofol reduced rCBF and rCMRO2 comparably. Sevoflurane reduced rCBF less than propofol but rCMRO2 to an extent similar to propofol. These reductions in flow and metabolism were partly attenuated by adjunct N2O. S+N especially reduced the oxygen extraction fraction, suggesting disturbed flow-activity coupling in humans at a moderate depth of anesthesia.


Assuntos
Anestesia Geral , Anestésicos Inalatórios , Anestésicos Intravenosos , Volume Sanguíneo/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Éteres Metílicos , Óxido Nitroso , Propofol , Adulto , Algoritmos , Encéfalo/diagnóstico por imagem , Química Encefálica/efeitos dos fármacos , Mapeamento Encefálico , Dióxido de Carbono/sangue , Relação Dose-Resposta a Droga , Interações Medicamentosas , Eletroencefalografia/efeitos dos fármacos , Humanos , Cinética , Masculino , Consumo de Oxigênio/efeitos dos fármacos , Sevoflurano , Tomografia Computadorizada de Emissão
19.
Anesthesiology ; 99(3): 614-23, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12960545

RESUMO

BACKGROUND: Animal experiments have demonstrated neuroprotection by ketamine. However, because of its propensity to increase cerebral blood flow, metabolism, and intracranial pressure, its use in neurosurgery or trauma patients has been questioned. METHODS: 15O-labeled water, oxygen, and carbon monoxide were used as positron emission tomography tracers to determine quantitative regional cerebral blood flow (rCBF), metabolic rate of oxygen (rCMRO2), and blood volume (rCBV), respectively, on selected regions of interest of nine healthy male volunteers at baseline and during three escalating concentrations of ketamine (targeted to 30, 100, and 300 ng/ml). In addition, voxel-based analysis for relative changes in rCBF and rCMRO2 was performed using statistical parametric mapping. RESULTS: The mean +/- SD measured ketamine serum concentrations were 37 +/- 8, 132 +/- 19, and 411 +/- 71 ng/ml. Mean arterial pressure was slightly elevated (maximally by 15.3%, P < 0.001) during ketamine infusion. Ketamine increased rCBF in a concentration-dependent manner. In the region-of-interest analysis, the greatest absolute changes were detected at the highest ketamine concentration level in the anterior cingulate (38.2% increase from baseline, P < 0.001), thalamus (28.5%, P < 0.001), putamen (26.8%, P < 0.001), and frontal cortex (25.4%, P < 0.001). Voxel-based analysis revealed marked relative rCBF increases in the anterior cingulate, frontal cortex, and insula. Although absolute rCMRO2 was not changed in the region-of-interest analysis, subtle relative increases in the frontal, parietal, and occipital cortices and decreases predominantly in the cerebellum were detected in the voxel-based analysis. rCBV increased only in the frontal cortex (4%, P = 0.022). CONCLUSIONS: Subanesthetic doses of ketamine induced a global increase in rCBF but no changes in rCMRO2. Consequently, the regional oxygen extraction fraction was decreased. Disturbed coupling of cerebral blood flow and metabolism is, however, considered unlikely because ketamine has been previously shown to increase cerebral glucose metabolism. Only a minor increase in rCBV was detected. Interestingly, the most profound changes in rCBF were observed in structures related to pain processing.


Assuntos
Anestésicos Dissociativos/farmacologia , Volume Sanguíneo/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ketamina/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Adulto , Afeto/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hemodinâmica/efeitos dos fármacos , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Mecânica Respiratória/efeitos dos fármacos , Tomografia Computadorizada de Emissão
20.
Anesthesiology ; 96(6): 1358-70, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12170048

RESUMO

BACKGROUND: The authors report a positron emission tomography (PET) study on humans with parallel exploration of the dose-dependent effects of an intravenous (propofol) and a volatile (sevoflurane) anesthetic agent on regional cerebral blood flow (rCBF) using quantitative and relative (Statistical Parametric Mapping [SPM]) analysis. METHODS: Using H(2)(15)O, rCBF was assessed in 16 healthy (American Society of Anesthesiologists [ASA] physical status I) volunteers awake and at three escalating drug concentrations: 1, 1.5, and 2 MAC/EC(50), or specifically, at either 2, 3, and 4% end-tidal sevoflurane (n = 8), or 6, 9, and 12 microg/ml plasma concentration of propofol (n = 8). Rocuronium was used for muscle relaxation. RESULTS: Both drugs decreased the bispectral index and blood pressure dose-dependently. Comparison between adjacent levels showed that sevoflurane initially (0 vs. 1 MAC) reduced absolute rCBF by 36-53% in all areas, then (1 vs. 1.5 MAC) increased rCBF in the frontal cortex, thalamus, and cerebellum (7-16%), and finally (1.5 vs. 2 MAC) caused a dual effect with a 23% frontal reduction and a 38% cerebellar increase. In the propofol group, flow was also initially reduced by 62-70%, with minor further effects. In the SPM analysis of the "awake to 1 MAC/EC(50)" step, both anesthetic agents reduced relative rCBF in the cuneus, precuneus, posterior limbic system, and the thalamus or midbrain; additionally, propofol reduced relative rCBF in the parietal and frontal cortices. CONCLUSIONS: Both anesthetic agents caused a global reduction of rCBF (propofol > sevoflurane) at the 1 MAC/EC(50) level. The effect was maintained at higher propofol concentrations, whereas 2 MAC sevoflurane caused noticeable flow redistribution. Despite the marked global changes, SPM analysis enabled detailed localization of regions with the greatest relative decreases.


Assuntos
Anestésicos Inalatórios/farmacologia , Anestésicos Intravenosos/farmacologia , Circulação Cerebrovascular/efeitos dos fármacos , Éteres Metílicos/farmacologia , Propofol/farmacologia , Tomografia Computadorizada de Emissão , Adulto , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eletroencefalografia/efeitos dos fármacos , Humanos , Masculino , Sevoflurano
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA