Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.806
Filter
1.
Headache ; 64(7): 825-837, 2024.
Article in English | MEDLINE | ID: mdl-38837259

ABSTRACT

OBJECTIVE: In this pilot prospective cohort study, we aimed to evaluate, using high-density electroencephalography (HD-EEG), the longitudinal changes in functional connectivity (FC) in patients with chronic migraine (CM) treated with onabotulinumtoxinA (OBTA). BACKGROUND: OBTA is a treatment for CM. Several studies have shown the modulatory action of OBTA on the central nervous system; however, research on migraine is limited. METHODS: This study was conducted at the Neurology Unit of "Policlinico Tor Vergata," Rome, Italy, and included 12 adult patients with CM treated with OBTA and 15 healthy controls (HC). Patients underwent clinical scales at enrollment (T0) and 3 months (T1) from the start of treatment. HD-EEG was recorded using a 64-channel system in patients with CM at T0 and T1. A source reconstruction method was used to identify brain activity. FC in δ-θ-α-ß-low-γ bands was analyzed using the weighted phase-lag index. FC changes between HCs and CM at T0 and T1 were assessed using cross-validation methods to estimate the results' reliability. RESULTS: Compared to HCs at T0, patients with CM showed hyperconnected networks in δ (p = 0.046, area under the receiver operating characteristic curve [AUC: 0.76-0.98], Cohen's κ [0.65-0.93]) and ß (p = 0.031, AUC [0.68-0.95], Cohen's κ [0.51-0.84]), mainly involving orbitofrontal, occipital, temporal pole and orbitofrontal, superior temporal, occipital, cingulate areas, and hypoconnected networks in α band (p = 0.029, AUC [0.80-0.99], Cohen's κ [0.42-0.77]), predominantly involving cingulate, temporal pole, and precuneus. Patients with CM at T1, compared to T0, showed hypoconnected networks in δ band (p = 0.032, AUC [0.73-0.99], Cohen's κ [0.53-0.90]) and hyperconnected networks in α band (p = 0.048, AUC [0.58-0.93], Cohen's κ [0.37-0.78]), involving the sensorimotor, orbitofrontal, cingulate, and temporal cortex. CONCLUSION: These preliminary results showed that patients with CM presented disrupted EEG-FC compared to controls restored by a single session of OBTA treatment, suggesting a primary central modulatory action of OBTA.


Subject(s)
Botulinum Toxins, Type A , Electroencephalography , Migraine Disorders , Humans , Botulinum Toxins, Type A/pharmacology , Botulinum Toxins, Type A/administration & dosage , Pilot Projects , Migraine Disorders/drug therapy , Migraine Disorders/physiopathology , Female , Male , Adult , Electroencephalography/drug effects , Middle Aged , Chronic Disease , Prospective Studies , Neuromuscular Agents/pharmacology , Neuromuscular Agents/administration & dosage , Brain/drug effects , Brain/physiopathology , Brain/diagnostic imaging
2.
Clin Neurophysiol ; 163: 152-159, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749380

ABSTRACT

OBJECTIVE: Brivaracetam (BRV) is a recent antiseizure medication (ASM) approved as an add-on therapy for people with focal epilepsy. BRV has a good efficacy and safety profile compared to other ASMs. However, its specific effects on resting-state EEG activity and connectivity are unknown. The aim of this study is to evaluate quantitative EEG changes induced by BRV therapy in a population of adult people with drug-resistant epilepsy (PwE) compared to healthy controls (HC). METHODS: We performed a longitudinal, retrospective, pharmaco-EEG study on a population of 23 PwE and a group of 25 HC. Clinical outcome was dichotomized into drug-responders (i.e., >50% reduction in seizures' frequency; RES) and non-responders (N-RES) after two years of BRV. EEG parameters were compared between PwE and HC at baseline (pre-BRV) and after three months of BRV therapy (post-BRV). We investigated BRV-related variations in EEG connectivity using the phase locking value (PLV). RESULTS: BRV therapy did not induce modifications in power spectrum density across different frequency bands. PwE presented lower PLV connectivity values compared to HC in all frequency bands. RES exhibited lower theta PLV connectivity compared to HC before initiating BRV and experienced an increase after BRV, eliminating the significant difference from HC. CONCLUSIONS: This study shows that BRV does not alter the EEG power spectrum in PwE, supporting its favourable neuropsychiatric side-effect profile, and induces the disappearance of EEG connectivity differences between PwE and HC. SIGNIFICANCE: The integration of EEG quantitative analysis in epilepsy can provide insights into the efficacy, mechanism of action, and side effects of ASMs.


Subject(s)
Anticonvulsants , Drug Resistant Epilepsy , Electroencephalography , Pyrrolidinones , Humans , Male , Female , Adult , Electroencephalography/drug effects , Electroencephalography/methods , Drug Resistant Epilepsy/drug therapy , Drug Resistant Epilepsy/physiopathology , Pyrrolidinones/therapeutic use , Pyrrolidinones/adverse effects , Anticonvulsants/therapeutic use , Anticonvulsants/pharmacology , Anticonvulsants/adverse effects , Middle Aged , Retrospective Studies , Longitudinal Studies , Young Adult
3.
Toxicol Appl Pharmacol ; 488: 116970, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38777098

ABSTRACT

Soman produces excitotoxic effects by inhibiting acetylcholinesterase in the cholinergic synapses and neuromuscular junctions, resulting in soman-induced sustained status epilepticus (SSE). Our previous work showed delayed intramuscular (i.m.) treatment with A1 adenosine receptor agonist N-bicyclo-[2.2.1]-hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA) alone suppressed soman-induced SSE and prevented neuropathology. Using this same rat soman seizure model, we tested if delayed therapy with ENBA (60 mg/kg, i.m.) would terminate seizure, protect neuropathology, and aid in survival when given in conjunction with current standard medical countermeasures (MCMs): atropine sulfate, 2-PAM, and midazolam (MDZ). Either 15- or 30-min following soman-induced SSE onset, male rats received atropine and 2-PAM plus either MDZ or MDZ + ENBA. Electroencephalographic (EEG) activity, physiologic parameters, and motor function were recorded. Either 2- or 14-days following exposure surviving rats were euthanized and perfused for histology. All animals treated with MDZ + ENBA at both time points had 100% EEG seizure termination and reduced total neuropathology compared to animals treated with MDZ (2-day, p = 0.015 for 15-min, p = 0.002 for 30-min; 14-day, p < 0.001 for 15-min, p = 0.006 for 30-min), showing ENBA enhanced MDZ's anticonvulsant and neuroprotectant efficacy. However, combined MDZ + ENBA treatment, when compared to MDZ treatment groups, had a reduction in the 14-day survival rate regardless of treatment time, indicating possible enhancement of MDZ's neuronal inhibitory effects by ENBA. Based on our findings, ENBA shows promise as an anticonvulsant and neuroprotectant in a combined treatment regimen following soman exposure; when given as an adjunct to standard MCMs, the dose of ENBA needs to be adjusted.


Subject(s)
Adenosine A1 Receptor Agonists , Rats, Sprague-Dawley , Seizures , Soman , Animals , Soman/toxicity , Male , Adenosine A1 Receptor Agonists/pharmacology , Rats , Injections, Intramuscular , Seizures/chemically induced , Seizures/drug therapy , Seizures/prevention & control , Neuroprotective Agents/pharmacology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Anticonvulsants/administration & dosage , Electroencephalography/drug effects , Adenosine/analogs & derivatives , Adenosine/administration & dosage , Adenosine/pharmacology , Atropine/pharmacology , Atropine/administration & dosage , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Midazolam/pharmacology , Midazolam/therapeutic use
4.
Anesthesiology ; 141(2): 353-364, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38718376

ABSTRACT

BACKGROUND: Unlike expired sevoflurane concentration, propofol lacks a biomarker for its brain effect site concentration, leading to dosing imprecision particularly in infants. Electroencephalography monitoring can serve as a biomarker for propofol effect site concentration, yet proprietary electroencephalography indices are not validated in infants. The authors evaluated spectral edge frequency (SEF95) as a propofol anesthesia biomarker in infants. It was hypothesized that the SEF95 targets will vary for different clinical stimuli and an inverse relationship existed between SEF95 and propofol plasma concentration. METHODS: This prospective study enrolled infants (3 to 12 months) to determine the SEF95 ranges for three clinical endpoints of anesthesia (consciousness-pacifier placement, pain-electrical nerve stimulation, and intubation-laryngoscopy) and correlation between SEF95 and propofol plasma concentration at steady state. Dixon's up-down method was used to determine target SEF95 for each clinical endpoint. Centered isotonic regression determined the dose-response function of SEF95 where 50% and 90% of infants (ED50 and ED90) did not respond to the clinical endpoint. Linear mixed-effect model determined the association of propofol plasma concentration and SEF95. RESULTS: Of 49 enrolled infants, 44 evaluable (90%) showed distinct SEF95 for endpoints: pacifier (ED50, 21.4 Hz; ED90, 19.3 Hz), electrical stimulation (ED50, 12.6 Hz; ED90, 10.4 Hz), and laryngoscopy (ED50, 8.5 Hz; ED90, 5.2 Hz). From propofol 0.5 to 6 µg/ml, a 1-Hz SEF95 increase was linearly correlated to a 0.24 (95% CI, 0.19 to 0.29; P < 0.001) µg/ml decrease in plasma propofol concentration (marginal R2 = 0.55). CONCLUSIONS: SEF95 can be a biomarker for propofol anesthesia depth in infants, potentially improving dosing accuracy and utilization of propofol anesthesia in this population.


Subject(s)
Anesthetics, Intravenous , Electroencephalography , Propofol , Humans , Propofol/blood , Propofol/administration & dosage , Infant , Prospective Studies , Electroencephalography/drug effects , Electroencephalography/methods , Anesthetics, Intravenous/blood , Anesthetics, Intravenous/administration & dosage , Female , Male , Biomarkers/blood , Dose-Response Relationship, Drug , Endpoint Determination
5.
BMC Anesthesiol ; 24(1): 167, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702608

ABSTRACT

The exact mechanisms and the neural circuits involved in anesthesia induced unconsciousness are still not fully understood. To elucidate them valid animal models are necessary. Since the most commonly used species in neuroscience are mice, we established a murine model for commonly used anesthetics/sedatives and evaluated the epidural electroencephalographic (EEG) patterns during slow anesthesia induction and emergence. Forty-four mice underwent surgery in which we inserted a central venous catheter and implanted nine intracranial electrodes above the prefrontal, motor, sensory, and visual cortex. After at least one week of recovery, mice were anesthetized either by inhalational sevoflurane or intravenous propofol, ketamine, or dexmedetomidine. We evaluated the loss and return of righting reflex (LORR/RORR) and recorded the electrocorticogram. For spectral analysis we focused on the prefrontal and visual cortex. In addition to analyzing the power spectral density at specific time points we evaluated the changes in the spectral power distribution longitudinally. The median time to LORR after start anesthesia ranged from 1080 [1st quartile: 960; 3rd quartile: 1080]s under sevoflurane anesthesia to 1541 [1455; 1890]s with ketamine. Around LORR sevoflurane as well as propofol induced a decrease in the theta/alpha band and an increase in the beta/gamma band. Dexmedetomidine infusion resulted in a shift towards lower frequencies with an increase in the delta range. Ketamine induced stronger activity in the higher frequencies. Our results showed substance-specific changes in EEG patterns during slow anesthesia induction. These patterns were partially identical to previous observations in humans, but also included significant differences, especially in the low frequencies. Our study emphasizes strengths and limitations of murine models in neuroscience and provides an important basis for future studies investigating complex neurophysiological mechanisms.


Subject(s)
Anesthetics, Inhalation , Dexmedetomidine , Electroencephalography , Ketamine , Propofol , Sevoflurane , Animals , Mice , Ketamine/pharmacology , Ketamine/administration & dosage , Sevoflurane/pharmacology , Sevoflurane/administration & dosage , Dexmedetomidine/pharmacology , Electroencephalography/drug effects , Electroencephalography/methods , Propofol/pharmacology , Propofol/administration & dosage , Male , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/administration & dosage , Reflex, Righting/drug effects , Reflex, Righting/physiology , Mice, Inbred C57BL , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage , Anesthetics, Intravenous/pharmacology , Anesthetics, Intravenous/administration & dosage , Anesthesia/methods
6.
Physiol Meas ; 45(5)2024 May 21.
Article in English | MEDLINE | ID: mdl-38697205

ABSTRACT

Objectives.The purpose of this study is to investigate the age dependence of bilateral frontal electroencephalogram (EEG) coupling characteristics, and find potential age-independent depth of anesthesia monitoring indicators for the elderlies.Approach.We recorded bilateral forehead EEG data from 41 patients (ranged in 19-82 years old), and separated into three age groups: 18-40 years (n= 12); 40-65 years (n= 14), >65 years (n= 15). All these patients underwent desflurane maintained general anesthesia (GA). We analyzed the age-related EEG spectra, phase amplitude coupling (PAC), coherence and phase lag index (PLI) of EEG data in the states of awake, GA, and recovery.Main results.The frontal alpha power shows age dependence in the state of GA maintained by desflurane. Modulation index in slow oscillation-alpha and delta-alpha bands showed age dependence and state dependence in varying degrees, the PAC pattern also became less pronounced with increasing age. In the awake state, the coherence in delta, theta and alpha frequency bands were all significantly higher in the >65 years age group than in the 18-40 years age group (p< 0.05 for three frequency bands). The coherence in alpha-band was significantly enhanced in all age groups in GA (p< 0.01) and then decreased in recovery state. Notably, the PLI in the alpha band was able to significantly distinguish the three states of awake, GA and recovery (p< 0.01) and the results of PLI in delta and theta frequency bands had similar changes to those of coherence.Significance.We found the EEG coupling and synchronization between bilateral forehead are age-dependent. The PAC, coherence and PLI portray this age-dependence. The PLI and coherence based on bilateral frontal EEG functional connectivity measures and PAC based on frontal single-channel are closely associated with anesthesia-induced unconsciousness.


Subject(s)
Desflurane , Electroencephalography , Humans , Desflurane/pharmacology , Adult , Middle Aged , Aged , Electroencephalography/drug effects , Young Adult , Male , Female , Aged, 80 and over , Adolescent , Aging/physiology , Aging/drug effects , Frontal Lobe/drug effects , Frontal Lobe/physiology , Isoflurane/analogs & derivatives , Isoflurane/pharmacology , Anesthetics, Inhalation/pharmacology , Anesthesia, General
7.
PeerJ ; 12: e17342, 2024.
Article in English | MEDLINE | ID: mdl-38737745

ABSTRACT

Background: N-Ethylmaleimide (NEM), an agonist of the potassium chloride cotransporters 2 (KCC2) receptor, has been correlated with neurosuppressive outcomes, including decreased pain perception and the prevention of epileptic seizures. Nevertheless, its relationship with sleep-inducing effects remains unreported. Objective: The present study aimed to investigate the potential enhancement of NEM on the sleep-inducing properties of alprazolam (Alp). Methods: The test of the righting reflex was used to identify the appropriate concentrations of Alp and NEM for inducing sleep-promoting effects in mice. Total sleep duration and sleep quality were evaluated through EEG/EMG analysis. The neural mechanism underlying the sleep-promoting effect was examined through c-fos immunoreactivity in the brain using immunofluorescence. Furthermore, potential CNS-side effects of the combination Alp and NEM were assessed using LABORAS automated home-cage behavioral phenotyping. Results: Combination administration of Alp (1.84 mg/kg) and NEM (1.0 mg/kg) significantly decreased sleep latency and increased sleep duration in comparison to administering 1.84 mg/kg Alp alone. This effect was characterized by a notable increase in REM duration. The findings from c-fos immunoreactivity indicated that NEM significantly suppressed neuron activation in brain regions associated with wakefulness. Additionally, combination administration of Alp and NEM showed no effects on mouse neural behaviors during automated home cage monitoring. Conclusions: This study is the first to propose and demonstrate a combination therapy involving Alp and NEM that not only enhances the hypnotic effect but also mitigates potential CNS side effects, suggesting its potential application in treating insomnia.


Subject(s)
Alprazolam , Drug Synergism , Sleep , Animals , Alprazolam/pharmacology , Alprazolam/administration & dosage , Mice , Male , Sleep/drug effects , Electroencephalography/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Brain/drug effects , Brain/metabolism , Reflex, Righting/drug effects , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage
8.
J Clin Anesth ; 95: 111459, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38599161

ABSTRACT

STUDY OBJECTIVE: Processed electroencephalography (pEEG) may help clinicians optimize depth of general anesthesia. Avoiding excessive depth of anesthesia may reduce intraoperative hypotension and the need for vasopressors. We tested the hypothesis that pEEG-guided - compared to non-pEEG-guided - general anesthesia reduces the amount of norepinephrine needed to keep intraoperative mean arterial pressure above 65 mmHg in patients having vascular surgery. DESIGN: Randomized controlled clinical trial. SETTING: University Medical Center Hamburg-Eppendorf, Hamburg, Germany. PATIENTS: 110 patients having vascular surgery. INTERVENTIONS: pEEG-guided general anesthesia. MEASUREMENTS: Our primary endpoint was the average norepinephrine infusion rate from the beginning of induction of anesthesia until the end of surgery. MAIN RESULT: 96 patients were analyzed. The mean ± standard deviation average norepinephrine infusion rate was 0.08 ± 0.04 µg kg-1 min-1 in patients assigned to pEEG-guided and 0.12 ± 0.09 µg kg-1 min-1 in patients assigned to non-pEEG-guided general anesthesia (mean difference 0.04 µg kg-1 min-1, 95% confidence interval 0.01 to 0.07 µg kg-1 min-1, p = 0.004). Patients assigned to pEEG-guided versus non-pEEG-guided general anesthesia, had a median time-weighted minimum alveolar concentration of 0.7 (0.6, 0.8) versus 0.8 (0.7, 0.8) (p = 0.006) and a median percentage of time Patient State Index was <25 of 12 (1, 41) % versus 23 (3, 49) % (p = 0.279). CONCLUSION: pEEG-guided - compared to non-pEEG-guided - general anesthesia reduced the amount of norepinephrine needed to keep mean arterial pressure above 65 mmHg by about a third in patients having vascular surgery. Whether reduced intraoperative norepinephrine requirements resulting from pEEG-guided general anesthesia translate into improved patient-centered outcomes remains to be determined in larger trials.


Subject(s)
Anesthesia, General , Electroencephalography , Norepinephrine , Vascular Surgical Procedures , Vasoconstrictor Agents , Humans , Anesthesia, General/methods , Norepinephrine/administration & dosage , Male , Female , Middle Aged , Aged , Electroencephalography/drug effects , Vascular Surgical Procedures/adverse effects , Vasoconstrictor Agents/administration & dosage , Hypotension/prevention & control , Arterial Pressure/drug effects , Monitoring, Intraoperative/methods
9.
Psychopharmacology (Berl) ; 241(7): 1417-1426, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38467891

ABSTRACT

Ibogaine is a potent atypical psychedelic that has gained considerable attention due to its antiaddictive and antidepressant properties in preclinical and clinical studies. Previous research from our group showed that ibogaine suppresses sleep and produces an altered wakefulness state, which resembles natural REM sleep. However, after systemic administration, ibogaine is rapidly metabolized to noribogaine, which also shows antiaddictive effects but with a distinct pharmacological profile, making this drug a promising therapeutic candidate. Therefore, we still ignore whether the sleep/wake alterations depend on ibogaine or its principal metabolite noribogaine. To answer this question, we conducted polysomnographic recordings in rats following the administration of pure noribogaine. Our results show that noribogaine promotes wakefulness while reducing slow-wave sleep and blocking REM sleep, similar to our previous results reported for ibogaine administration. Thus, we shed new evidence on the mechanisms by which iboga alkaloids work in the brain.


Subject(s)
Ibogaine , Polysomnography , Sleep, REM , Wakefulness , Animals , Sleep, REM/drug effects , Wakefulness/drug effects , Wakefulness/physiology , Male , Rats , Ibogaine/analogs & derivatives , Ibogaine/pharmacology , Ibogaine/administration & dosage , Rats, Sprague-Dawley , Sleep, Slow-Wave/drug effects , Sleep, Slow-Wave/physiology , Hallucinogens/pharmacology , Hallucinogens/administration & dosage , Electroencephalography/drug effects
10.
Anaesth Crit Care Pain Med ; 43(3): 101377, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38494158

ABSTRACT

BACKGROUND: Remimazolam is a safe and effective new benzodiazepine sedative that has unique advantages in anesthesia induction and maintenance. The differences in the electroencephalogram bispectral index (BIS) during general anesthesia between propofol and remimazolam deserve further exploration. METHODS: Single-center randomized crossover study. Patients who required multiple hysteroscopic surgery were randomly assigned to use remimazolam (0.27 mg/kg for induction and 1 mg/kg/h for maintenance) first and then propofol (2.0 mg/kg for induction and 6 mg/kg/h for maintenance) during hysteroscopic surgery again 3 months later, or in the opposite order. Both drugs were used at the latest ED95 for unconsciousness. The BIS values (primary endpoint), intraoperative conditions, and incidence of adverse reactions (secondary endpoints) were compared at each time point. BIS values were analyzed with a mixed model of repeated measurements (MMRM). RESULTS: Seventeen patients completed the study. The lowest BIS value in the remimazolam regimen was significantly higher than that in the propofol regimen (p = 0.001). The MMRM analysis of the BIS values revealed significant differences between the regimens at each time point (p < 0.001). The intraoperative diastolic blood pressure and heart rate changes were smaller, the recovery was faster, and there were fewer adverse reactions and less injection pain, but a greater incidence of intraoperative body movement and hiccups, in the remimazolam regimen. CONCLUSION: The trial indicated that remimazolam maintained a higher BIS level than propofol. The correlation between the BIS and the depth of anesthesia induced by remimazolam needs to be further studied. TRIAL REGISTRATION: This trial is registered at ClinicalTrials.gov: ChiCTR2200064551.


Subject(s)
Anesthesia, General , Benzodiazepines , Cross-Over Studies , Electroencephalography , Hypnotics and Sedatives , Propofol , Humans , Female , Propofol/administration & dosage , Propofol/adverse effects , Adult , Anesthesia, General/methods , Benzodiazepines/administration & dosage , Benzodiazepines/adverse effects , Middle Aged , Electroencephalography/drug effects , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Consciousness Monitors , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/adverse effects , Hysteroscopy/adverse effects , Hysteroscopy/methods
12.
J Anesth ; 38(3): 371-376, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38376589

ABSTRACT

PURPOSE: The effect-site concentration (Ce) at loss of response (Ce-LOR) to propofol closely correlates both with Ce as electroencephalographic alpha power becomes highest (Ce-alpha) and with Ce at onset of burst suppression (BS) (Ce-OBS), when no opioids are administered. Co-administration of opioids dose-dependently decreases Ce-LOR. We investigated the influence of remifentanil on the relationship between these three Ces. METHODS: After receiving approval from our local ethical committee, with written informed consent, we enrolled 90 participants (ASA-PS I or II) who were scheduled for elective surgery. Participants were randomly assigned to three groups: constant remifentanil Ce 0 ng/ml (Remi_0); 1 ng/mL (Remi_1); and 2 ng/mL (Remi_2). We recorded both raw EEG and EEG-derived parameters on a computer. After reaching remifentanil equilibrium, we administered propofol using a target-controlled infusion pump such that propofol Ce increased to about 0.3 µg/mL/min. After determining Ce-LOR, we administered 0.6 mg/kg of rocuronium and started mask ventilation. The study protocol ended after observation of BS. RESULTS: Three participants were excluded. Ce-LOR in each group (Remi_0, Remi_1, Remi_2) was 2.00 ± 0.58 µg/mL, 1.43 ± 0.49 µg/mL, and 1.37 ± 0.42 µg/mL. Ce-alpha was 2.91 ± 0.63 µg/mL, 2.30 ± 0.41 µg/mL, and 2.12 ± 0.39 µg/mL. Ce-OBS was 3.80 ± 0.69 µg/mL, 3.25 ± 0.68 µg/mL, and 2.90 ± 0.57 µg/mL. In three other instances, Ce was decreased by remifentanil. Generalized linear model analysis revealed that remifentanil had no influence on the relationship between the three Ces. CONCLUSION: During propofol anesthesia, even low concentrations of remifentanil shifted concentration-related electroencephalographic changes.


Subject(s)
Anesthetics, Intravenous , Electroencephalography , Propofol , Remifentanil , Humans , Remifentanil/administration & dosage , Remifentanil/pharmacology , Propofol/administration & dosage , Propofol/pharmacology , Electroencephalography/drug effects , Electroencephalography/methods , Male , Female , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/pharmacology , Prospective Studies , Adult , Middle Aged , Dose-Response Relationship, Drug , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Piperidines/pharmacology , Piperidines/administration & dosage
13.
Korean J Anesthesiol ; 77(3): 289-305, 2024 06.
Article in English | MEDLINE | ID: mdl-38228393

ABSTRACT

Intraoperative electroencephalography (EEG) monitoring under pediatric anesthesia has begun to attract increasing interest, driven by the availability of pediatric-specific EEG monitors and the realization that traditional dosing methods based on patient movement or changes in hemodynamic response often lead to imprecise dosing, especially in younger infants who may experience adverse events (e.g., hypotension) due to excess anesthesia. EEG directly measures the effects of anesthetics on the brain, which is the target end-organ responsible for inducing loss of consciousness. Over the past ten years, research on anesthesia and computational neuroscience has improved our understanding of intraoperative pediatric EEG monitoring and expanded the utility of EEG in clinical practice. We now have better insights into neurodevelopmental changes in the developing pediatric brain, functional connectivity, the use of non-proprietary EEG parameters to guide anesthetic dosing, epileptiform EEG changes during induction, EEG changes from spinal/regional anesthesia, EEG discontinuity, and the use of EEG to improve clinical outcomes. This review article summarizes the recent literature on EEG monitoring in perioperative pediatric anesthesia, highlighting several of the topics mentioned above.


Subject(s)
Electroencephalography , Humans , Electroencephalography/methods , Electroencephalography/drug effects , Child , Monitoring, Intraoperative/methods , Infant , Intraoperative Neurophysiological Monitoring/methods , Anesthesia/methods , Anesthetics/administration & dosage , Anesthetics/adverse effects , Child, Preschool
14.
Neuropsychopharmacology ; 49(7): 1120-1128, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38287172

ABSTRACT

Neural complexity correlates with one's level of consciousness. During coma, anesthesia, and sleep, complexity is reduced. During altered states, including after lysergic acid diethylamide (LSD), complexity is increased. In the present analysis, we examined whether low doses of LSD (13 and 26 µg) were sufficient to increase neural complexity in the absence of altered states of consciousness. In addition, neural complexity was assessed after doses of two other drugs that significantly altered consciousness and mood: delta-9-tetrahydrocannabinol (THC; 7.5 and 15 mg) and methamphetamine (MA; 10 and 20 mg). In three separate studies (N = 73; 21, LSD; 23, THC; 29, MA), healthy volunteers received placebo or drug in a within-subjects design over three laboratory visits. During anticipated peak drug effects, resting state electroencephalography (EEG) recorded Limpel-Ziv complexity and spectral power. LSD, but not THC or MA, dose-dependently increased neural complexity. LSD also reduced delta and theta power. THC reduced, and MA increased, alpha power, primarily in frontal regions. Neural complexity was not associated with any subjective drug effect; however, LSD-induced reductions in delta and theta were associated with elation, and THC-induced reductions in alpha were associated with altered states. These data inform relationships between neural complexity, spectral power, and subjective states, demonstrating that increased neural complexity is not necessary or sufficient for altered states of consciousness. Future studies should address whether greater complexity after low doses of LSD is related to cognitive, behavioral, or therapeutic outcomes, and further examine the role of alpha desynchronization in mediating altered states of consciousness.


Subject(s)
Dose-Response Relationship, Drug , Dronabinol , Electroencephalography , Lysergic Acid Diethylamide , Methamphetamine , Humans , Methamphetamine/administration & dosage , Methamphetamine/pharmacology , Dronabinol/pharmacology , Dronabinol/administration & dosage , Male , Adult , Lysergic Acid Diethylamide/pharmacology , Lysergic Acid Diethylamide/administration & dosage , Female , Young Adult , Electroencephalography/drug effects , Hallucinogens/administration & dosage , Hallucinogens/pharmacology , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/administration & dosage
15.
Cogn Affect Behav Neurosci ; 24(2): 269-278, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38168850

ABSTRACT

Translation of drug targets from preclinical studies to clinical trials has been aided by cross-species behavioral tasks, but evidence for brain-based engagement during task performance is still required. Cross-species progressive ratio breakpoint tasks (PRBTs) measure motivation-related behavior and are pharmacologically and clinically sensitive. We recently advanced elevated parietal alpha power as a cross-species electroencephalographic (EEG) biomarker of PRBT engagement. Given that amphetamine increases breakpoint in mice, we tested its effects on breakpoint and parietal alpha power in both humans and mice. Twenty-three healthy participants performed the PRBT with EEG after amphetamine or placebo in a double-blind design. C57BL/6J mice were trained on PRBT with EEG (n = 24) and were treated with amphetamine or vehicle. A second cohort of mice was trained on PRBT without EEG (n = 40) and was treated with amphetamine or vehicle. In humans, amphetamine increased breakpoint. In mice, during concomitant EEG, 1 mg/kg of amphetamine significantly decreased breakpoint. In cohort 2, however, 0.3 mg/kg of amphetamine increased breakpoint consistent with human findings. Increased alpha power was observed in both species as they reached breakpoint, replicating previous findings. Amphetamine did not affect alpha power in either species. Amphetamine increased effort in humans and mice. Consistent with previous reports, elevated parietal alpha power was observed in humans and mice as they performed the PRBT. Amphetamine did not affect this EEG biomarker of effort. Hence, these findings support the pharmacological predictive validity of the PRBT to measure effort in humans and mice and suggest that this EEG biomarker is not directly reflective of amphetamine-induced changes in effort.


Subject(s)
Amphetamine , Central Nervous System Stimulants , Electroencephalography , Mice, Inbred C57BL , Motivation , Amphetamine/pharmacology , Humans , Animals , Male , Electroencephalography/drug effects , Adult , Young Adult , Double-Blind Method , Motivation/drug effects , Motivation/physiology , Female , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/administration & dosage , Mice , Alpha Rhythm/drug effects , Alpha Rhythm/physiology
16.
Anaesthesia ; 77 Suppl 1: 78-91, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35001380

ABSTRACT

Status epilepticus causes prolonged or repetitive seizures that, if left untreated, can lead to neuronal injury, severe disability, coma and death in paediatric and adult populations. While convulsive status epilepticus can be diagnosed using clinical features alone, non-convulsive status epilepticus requires confirmation by electroencephalogram. Early seizure control remains key in preventing the complications of status epilepticus. This is especially true for convulsive status epilepticus, which has stronger evidence supporting the benefit of treatment on outcomes. When status epilepticus becomes refractory, often due to gamma-aminobutyric acid and N-methyl-D-aspartate receptor modulation, anaesthetic drugs are needed to suppress seizure activity, of which there is limited evidence regarding the selection, dose or duration of their use. Seizure monitoring with electroencephalogram is often needed when patients do not return to baseline or during anaesthetic wean; however, it is resource-intensive, costly, only available in highly specialised centres and has not been shown to improve functional outcomes. Thus, the treatment goals and aggressiveness of therapy remain under debate, especially for non-convulsive status epilepticus, where prolonged therapeutic coma can lead to severe complications. This review presents an evidence-based, clinically-oriented and comprehensive review of status epilepticus and its definitions, aetiologies, treatments, outcomes and prognosis at different stages of the patient's journey.


Subject(s)
Anesthetics/therapeutic use , Anticonvulsants/therapeutic use , Disease Management , Evidence-Based Medicine/methods , Status Epilepticus/drug therapy , Status Epilepticus/surgery , Anesthetics/pharmacology , Anticonvulsants/pharmacology , Electroencephalography/drug effects , Electroencephalography/methods , Humans
17.
Front Neural Circuits ; 16: 1065374, 2022.
Article in English | MEDLINE | ID: mdl-36589861

ABSTRACT

Background: Volatile anesthetics including sevoflurane and isoflurane enhance oscillations of cortical electroencephalogram (EEG), partly by their modulations on glutamate-mediated excitatory synaptic transmission. Expression of NMDA receptors is increased during neonatal development. However, how the development of NMDA receptors influences EEG under volatile anesthesia remains unclear. Methods: Expressions of NMDA receptor subtypes (NR1, NR2A, and NR2B) during neonatal development were measured by Western blotting. MAC (minimal alveolar concentration) of isoflurane and sevoflurane that inducing loss of righting reflex (LORR) and no response to tail-clamp (immobility) were measured to verify the effect of NR1 expression on anesthetic potency during neonatal development. Cortical electroencephalogram recording was used to examine the influence of NR1 expression on the power density of EEG. Results: The expressions of GluNR1, GluNR2A and GluNR2B receptors were gradually increased during neonatal development in cortex, hippocampus and thalamus of rats. Knockdown of NR1 enhanced the sedative potency of volatile anesthetics but not on immobility potency in postnatal day 14 (P14)-P17 rats. For cortical EEG, along with the increased concentration of volatile anesthetics, cortical slow-delta oscillations of P5 rats were inhibited, theta and alpha oscillations were not changed significantly; while these oscillations were enhanced until high anesthetic concentrations in P21 rats. Knockdown of NR1 in forebrain suppressed the enhancement of cortical EEG oscillations in P21 rats. Conclusion: The development of NMDA receptors may contribute to the enhancement of cortical EEG oscillations under volatile anesthetics.


Subject(s)
Anesthetics, Inhalation , Cerebral Cortex , Electroencephalography , Receptors, N-Methyl-D-Aspartate , Animals , Rats , Anesthetics, Inhalation/pharmacology , Electroencephalography/drug effects , Isoflurane/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Sevoflurane/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/embryology
18.
J Neurophysiol ; 126(5): 1660-1669, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34644166

ABSTRACT

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.


Subject(s)
Anesthesia , Anesthetics, General/pharmacology , Auditory Perception/drug effects , Ketamine/pharmacology , Superior Olivary Complex/drug effects , Xylazine/pharmacology , Animals , Electroencephalography/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Gerbillinae , Male
19.
PLoS One ; 16(10): e0258647, 2021.
Article in English | MEDLINE | ID: mdl-34673803

ABSTRACT

BACKGROUND: Bispectral index (BIS) monitoring is a widely used non-invasive method to monitor the depth of anesthesia. However, in the event of surgeries requiring a frontal approach, placement of the electrode may be impossible at the designated area to achieve a proper BIS measurement. METHODS: We developed an investigational interface device to connect needle-electrodes to BIS sensors. The safety and clinical performance were investigated in patients who underwent surgery. Direct BIS values from a disposable BIS electrode and indirect values via the interface device were simultaneously recorded from the same areas of electrode placement in a single patient. The agreement between the direct and indirect BIS values was statistically analyzed. RESULTS: The interface device with a silver electrode demonstrated sufficient electric conduction to transmit electroencephalogram signals. The overall BIS curves were similar to those of direct BIS monitoring. Direct and indirect BIS values from 18 patients were statistically analyzed using a linear mixed model and a significant concordance was confirmed (indirect BIS = 7.0405 + 0.8286 * direct BIS, p<0.0001). Most observed data (2582/2787 data points, 92.64%) had BIS unit differences of 10 or less. CONCLUSIONS: The interface device provides an opportunity for intraoperative BIS monitoring of patients, whose clinical situation does not permit the placement of conventional adhesive sensors at the standard location.


Subject(s)
Anesthesia, General/methods , Biosensing Techniques/methods , Electrodes , Electroencephalography/drug effects , Electroencephalography/instrumentation , Monitoring, Intraoperative/methods , Neurosurgical Procedures/methods , Adult , Aged , Female , Humans , Male , Middle Aged
20.
CNS Drugs ; 35(11): 1207-1215, 2021 11.
Article in English | MEDLINE | ID: mdl-34687005

ABSTRACT

BACKGROUND: Cannabidiol has been shown to be effective in seizure reduction in patients with Dravet syndrome, Lennox-Gastaut syndrome, and tuberous sclerosis. However, very little is known about its potential to reduce interictal epileptiform activity and improve sleep architecture. OBJECTIVE: The objective of this prospective study was to evaluate the influence of cannabidiol therapy on the frequency of interictal epileptiform discharges (IEDs) and sleep microstructure in a cohort of children with drug-resistant epilepsy. METHODS: Children with drug-resistant epilepsy were prospectively followed from November 2019 to January 2021 during an open-label trial of cannabidiol at a dose of 20 mg/kg/day (to a maximum of 50 mg/kg/day) and stable concomitant medication. Electroencephalograms were recorded at baseline (T0) and after 3 months (T1). Two independent raters, blinded to clinical outcome, evaluated 5-min segments of sleep stage 2 or low-noise awake state. IEDs were visually identified and rates per minute calculated. Sleep microstructure was considered improved if sleep structures were seen at T1 that were not present at T0. IED rates at T0 and T1 were compared and correlated with seizure outcome, cannabidiol dose, initial IED rate, and disease duration. RESULTS: In total, 35 children (mean ± standard deviation age 10.1 ± 0.86) were included. The IED rate at T1 was significantly lower than at T0 (19.6 ± 19.5 vs. 36.8 ± 27.2, respectively; p < 0.0001). We found a moderate correlation between IED reduction and percentage of seizure reduction compared with baseline (Pearson's r = 0.39; p = 0.02), a moderate negative correlation between IED reduction and IED rate at T0 (r = - 0.34; p = 0.04), and a trend towards a moderate negative correlation between IED reduction and disease duration (r = - 0.32; p = 0.06). Sleep was recorded in 23 patients. Sleep microstructure was initially abnormal in 56.5% of sleep recordings and improved in 84.6% of those cases. CONCLUSION: Our results strongly suggest the utility of cannabidiol in reducing IEDs and improving sleep microstructure in children with drug-resistant epilepsy. Larger controlled studies are needed to evaluate the clinical relevance of this effect in different epilepsy types. TRIAL REGISTRATION: DRKS00013177; 25 June 2019.


Subject(s)
Anticonvulsants/administration & dosage , Cannabidiol/administration & dosage , Drug Resistant Epilepsy/drug therapy , Drug Resistant Epilepsy/physiopathology , Electroencephalography/drug effects , Sleep/drug effects , Administration, Oral , Child , Cohort Studies , Drug Resistant Epilepsy/diagnosis , Electroencephalography/methods , Female , Humans , Male , Prospective Studies , Sleep/physiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL