Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 559
Filter
Add more filters

Publication year range
1.
Nature ; 586(7827): 87-94, 2020 10.
Article in English | MEDLINE | ID: mdl-32939091

ABSTRACT

Advanced imaging methods now allow cell-type-specific recording of neural activity across the mammalian brain, potentially enabling the exploration of how brain-wide dynamical patterns give rise to complex behavioural states1-12. Dissociation is an altered behavioural state in which the integrity of experience is disrupted, resulting in reproducible cognitive phenomena including the dissociation of stimulus detection from stimulus-related affective responses. Dissociation can occur as a result of trauma, epilepsy or dissociative drug use13,14, but despite its substantial basic and clinical importance, the underlying neurophysiology of this state is unknown. Here we establish such a dissociation-like state in mice, induced by precisely-dosed administration of ketamine or phencyclidine. Large-scale imaging of neural activity revealed that these dissociative agents elicited a 1-3-Hz rhythm in layer 5 neurons of the retrosplenial cortex. Electrophysiological recording with four simultaneously deployed high-density probes revealed rhythmic coupling of the retrosplenial cortex with anatomically connected components of thalamus circuitry, but uncoupling from most other brain regions was observed-including a notable inverse correlation with frontally projecting thalamic nuclei. In testing for causal significance, we found that rhythmic optogenetic activation of retrosplenial cortex layer 5 neurons recapitulated dissociation-like behavioural effects. Local retrosplenial hyperpolarization-activated cyclic-nucleotide-gated potassium channel 1 (HCN1) pacemakers were required for systemic ketamine to induce this rhythm and to elicit dissociation-like behavioural effects. In a patient with focal epilepsy, simultaneous intracranial stereoencephalography recordings from across the brain revealed a similarly localized rhythm in the homologous deep posteromedial cortex that was temporally correlated with pre-seizure self-reported dissociation, and local brief electrical stimulation of this region elicited dissociative experiences. These results identify the molecular, cellular and physiological properties of a conserved deep posteromedial cortical rhythm that underlies states of dissociation.


Subject(s)
Brain Waves/physiology , Cerebral Cortex/physiology , Dissociative Disorders/physiopathology , Action Potentials/drug effects , Animals , Behavior/drug effects , Brain Waves/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/drug effects , Dissociative Disorders/diagnostic imaging , Electrophysiology , Female , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ketamine/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Optogenetics , Self Report , Thalamus/cytology , Thalamus/diagnostic imaging , Thalamus/drug effects , Thalamus/physiology
2.
Neuroimage ; 292: 120606, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38604538

ABSTRACT

Radon is a naturally occurring gas that contributes significantly to radiation in the environment and is the second leading cause of lung cancer globally. Previous studies have shown that other environmental toxins have deleterious effects on brain development, though radon has not been studied as thoroughly in this context. This study examined the impact of home radon exposure on the neural oscillatory activity serving attention reorientation in youths. Fifty-six participants (ages 6-14 years) completed a classic Posner cuing task during magnetoencephalography (MEG), and home radon levels were measured for each participant. Time-frequency spectrograms indicated stronger theta (3-7 Hz, 300-800 ms), alpha (9-13 Hz, 400-900 ms), and beta responses (14-24 Hz, 400-900 ms) during the task relative to baseline. Source reconstruction of each significant oscillatory response was performed, and validity maps were computed by subtracting the task conditions (invalidly cued - validly cued). These validity maps were examined for associations with radon exposure, age, and their interaction in a linear regression design. Children with greater radon exposure showed aberrant oscillatory activity across distributed regions critical for attentional processing and attention reorientation (e.g., dorsolateral prefrontal cortex, and anterior cingulate cortex). Generally, youths with greater radon exposure exhibited a reverse neural validity effect in almost all regions and showed greater overall power relative to peers with lesser radon exposure. We also detected an interactive effect between radon exposure and age where youths with greater radon exposure exhibited divergent developmental trajectories in neural substrates implicated in attentional processing (e.g., bilateral prefrontal cortices, superior temporal gyri, and inferior parietal lobules). These data suggest aberrant, but potentially compensatory neural processing as a function of increasing home radon exposure in areas critical for attention and higher order cognition.


Subject(s)
Attention , Magnetoencephalography , Radon , Humans , Adolescent , Child , Male , Female , Radon/toxicity , Radon/adverse effects , Attention/radiation effects , Attention/physiology , Environmental Exposure/adverse effects , Brain/radiation effects , Brain Waves/radiation effects , Brain Waves/physiology , Brain Waves/drug effects , Orientation/physiology
3.
Neuroimage ; 249: 118891, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35007718

ABSTRACT

Recent neuroimaging studies have demonstrated that spontaneous brain activity exhibits rich spatiotemporal structure that can be characterized as the exploration of a repertoire of spatially distributed patterns that recur over time. The repertoire of brain states may reflect the capacity for consciousness, since general anesthetics suppress and psychedelic drugs enhance such dynamics. However, the modulation of brain activity repertoire across varying states of consciousness has not yet been studied in a systematic and unified framework. As a unique drug that has both psychedelic and anesthetic properties depending on the dose, ketamine offers an opportunity to examine brain reconfiguration dynamics along a continuum of consciousness. Here we investigated the dynamic organization of cortical activity during wakefulness and during altered states of consciousness induced by different doses of ketamine. Through k-means clustering analysis of the envelope data of source-localized electroencephalographic (EEG) signals, we identified a set of recurring states that represent frequency-specific spatial coactivation patterns. We quantified the effect of ketamine on individual brain states in terms of fractional occupancy and transition probabilities and found that ketamine anesthesia tends to shift the configuration toward brain states with low spatial variability. Furthermore, by assessing the temporal dynamics of the occurrence and transitions of brain states, we showed that subanesthetic ketamine is associated with a richer repertoire, while anesthetic ketamine induces dynamic changes in brain state organization, with the repertoire richness evolving from a reduced level to one comparable to that of normal wakefulness before recovery of consciousness. These results provide a novel description of ketamine's modulation of the dynamic configuration of cortical activity and advance understanding of the neurophysiological mechanism of ketamine in terms of the spatial, temporal, and spectral structures of underlying whole-brain dynamics.


Subject(s)
Anesthetics, Dissociative/pharmacology , Brain Waves/drug effects , Cerebral Cortex/drug effects , Consciousness/drug effects , Electroencephalography/methods , Ketamine/pharmacology , Wakefulness/drug effects , Adult , Anesthesia, General , Anesthetics, Dissociative/administration & dosage , Humans , Ketamine/administration & dosage
4.
Nutr Neurosci ; 25(1): 159-168, 2022 Jan.
Article in English | MEDLINE | ID: mdl-32116139

ABSTRACT

Objectives: Removing artificial food coloring (AFC) is a common dietary intervention for children with Attention-Deficit/Hyperactivity Disorder (ADHD), but has not been tested in young adults. This pilot study examined the effects of AFC on ADHD symptoms and electroencephalography (EEG) in college students with and without ADHD.Methods: At baseline, control and ADHD participants completed the Adult ADHD Self-Report Scale (ASRS), simple and complex attention measures, and resting-state EEG recordings. ADHD participants (n = 18) and a subset of controls (extended control group or EC, n = 11) avoided AFC in their diet for 2 weeks and then were randomized to a double-blind, placebo-controlled crossover challenge. Subjects received either 225 mg AFC disguised in chocolate cookies or placebo chocolate cookies for 3 days each week, with testing on the third day each week. Baseline comparisons were made using Student's t-test or Wilcoxon rank sum tests and challenge period analyses were run using General Linear Modeling.Results: The ADHD group had significantly greater scores on the ASRS (p < 0.001), confirming a symptom differential between groups; however, there were no differences in attentional measures or EEG at baseline. The AFC challenge resulted in an increase in posterior mean gamma power (p = 0.05), a decrease in posterior relative alpha power (p = 0.04), and a marginal increase in inattentive symptoms (p = 0.08) in the ADHD group. There were no effects of AFC in the EC group.Discussion: This study indicates that AFC exposure may affect brainwave activity and ADHD symptoms in college students with ADHD. Larger studies are needed to confirm these findings.


Subject(s)
Attention Deficit Disorder with Hyperactivity/chemically induced , Attention Deficit Disorder with Hyperactivity/physiopathology , Electroencephalography , Food Coloring Agents/adverse effects , Pilot Projects , Brain Waves/drug effects , Brain Waves/physiology , Cross-Over Studies , Double-Blind Method , Humans , Students , Young Adult
5.
Neuroimage ; 245: 118659, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34767940

ABSTRACT

Studying changes in cortical oscillations can help elucidate the mechanistic link between receptor physiology and the clinical effects of anaesthetic drugs. Propofol, a GABA-ergic drug produces divergent effects on visual cortical activity: increasing induced gamma-band responses (GBR) while decreasing evoked responses. Dexmedetomidine, an α2- adrenergic agonist, differs from GABA-ergic sedatives both mechanistically and clinically as it allows easy arousability from deep sedation with less cognitive side-effects. Here we use magnetoencephalography (MEG) to characterize and compare the effects of GABA-ergic (propofol) and non-GABA-ergic (dexmedetomidine) sedation, on visual and motor cortical oscillations. Sixteen male participants received target-controlled infusions of propofol and dexmedetomidine, producing mild-sedation, in a placebo-controlled, cross-over study. MEG data was collected during a combined visuomotor task. The key findings were that propofol significantly enhanced visual stimulus induced GBR (44% increase in amplitude) while dexmedetomidine decreased it (40%). Propofol also decreased the amplitudes of the Mv100 (visual M100) (27%) and Mv150 (52%) visual evoked fields (VEF), whilst dexmedetomidine had no effect on these. During the motor task, neither drug had any significant effect on movement related gamma synchrony (MRGS), movement related beta de-synchronisation (MRBD) or Mm100 (movement-related M100) movement-related evoked fields (MEF), although dexmedetomidine slowed the Mm300. Dexmedetomidine increased (92%) post-movement beta synchronisation/rebound (PMBR) power while propofol reduced it (70%, statistically non- significant). Overall, dexmedetomidine and propofol, at equi-sedative doses, produce contrasting effects on visual induced GBR, VEF, PMBR and MEF. These findings provide a mechanistic link between the known receptor physiology of these sedative drugs with their known clinical effects and may be used to explore mechanisms of other anaesthetic drugs on human consciousness.


Subject(s)
Brain Waves/drug effects , Dexmedetomidine/pharmacology , Hypnotics and Sedatives/pharmacology , Magnetoencephalography/methods , Motor Cortex/drug effects , Propofol/pharmacology , Adult , Conscious Sedation , Consciousness/drug effects , Cross-Over Studies , Humans , Male , Movement/physiology , Wakefulness , Young Adult
6.
Neuroimage ; 224: 117415, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33011419

ABSTRACT

The ability of different groups of cortical neurons to engage in causal interactions that are at once differentiated and integrated results in complex dynamic patterns. Complexity is low during periods of unconsciousness (deep sleep, anesthesia, unresponsive wakefulness syndrome) in which the brain tends to generate a stereotypical pattern consisting of alternating active and silent periods of neural activity-slow oscillations- and is high during wakefulness. But how is cortical complexity built up? Is it a continuum? An open question is whether cortical complexity can vary within the same brain state. Here we recorded with 32-channel multielectrode arrays from the cortical surface of the mouse and used both spontaneous dynamics (wave propagation entropy and functional complexity) and a perturbational approach (a variation of the perturbation complexity index) to measure complexity at different anesthesia levels. Variations in anesthesia level within the bistable regime of slow oscillations (0.1-1.5 Hz) resulted in a modulation of the slow oscillation frequency. Both perturbational and spontaneous complexity increased with decreasing anesthesia levels, in correlation with the decrease in coherence of the underlying network. Changes in complexity level are related to, but not dependent on, changes in excitability. We conclude that cortical complexity can vary within a single brain state dominated by slow oscillations, building up to the higher complexity associated with consciousness.


Subject(s)
Anesthetics, General/pharmacology , Brain Waves/drug effects , Cerebral Cortex/drug effects , Anesthesia, General , Animals , Brain Waves/physiology , Cerebral Cortex/physiology , Electric Stimulation , Electroencephalography , Hypnotics and Sedatives/pharmacology , Isoflurane/pharmacology , Ketamine/pharmacology , Medetomidine/pharmacology , Mice
7.
Neuroimage ; 245: 118769, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34861394

ABSTRACT

The effects of hippocampal neuronal afterdischarges (nAD) on hemodynamic parameters, such as blood-oxygen-level-dependent (BOLD) signals) and local cerebral blood volume (CBV) changes, as well as neuronal activity and metabolic parameters in the dentate gyrus, was investigated in rats by combining in vivo electrophysiology with functional magnetic resonance imaging (fMRI) or 1H-nuclear magnetic resonance spectroscopy (1H-NMRS). Brief electrical high-frequency pulse-burst stimulation of the right perforant pathway triggered nAD, a seizure-like activity, in the right dentate gyrus with a high incidence, a phenomenon that in turn caused a sustained decrease in BOLD signals for more than 30 min. The decrease was associated with a reduction in CBV but not with signs of hypoxic metabolism. nAD also triggered transient changes mainly in the low gamma frequency band that recovered within 20 min, so that the longer-lasting altered hemodynamics reflected a switch in blood supply rather than transient changes in ongoing neuronal activity. Even in the presence of reduced baseline BOLD signals, neurovascular coupling mechanisms remained intact, making long-lasting vasospasm unlikely. Subsequently generated nAD did not further alter the baseline BOLD signals. Similarly, nAD did not alter baseline BOLD signals when acetaminophen was previously administered, because acetaminophen alone had already caused a similar decrease in baseline BOLD signals as observed after the first nAD. Thus, at least two different blood supply states exist for the hippocampus, one low and one high, with both states allowing similar neuronal activity. Both acetaminophen and nAD switch from the high to the low blood supply state. As a result, the hemodynamic response function to an identical stimulus differed after nAD or acetaminophen, although the triggered neuronal activity was similar.


Subject(s)
Brain Waves/physiology , Electrocorticography , Hippocampus/physiology , Magnetic Resonance Imaging , Neuroimaging , Neurovascular Coupling/physiology , Proton Magnetic Resonance Spectroscopy , Seizures/physiopathology , Animals , Brain Waves/drug effects , Disease Models, Animal , Hippocampus/drug effects , Male , Neurovascular Coupling/drug effects , Rats , Rats, Wistar , Seizures/metabolism
8.
J Neurophysiol ; 126(4): 1265-1275, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34469699

ABSTRACT

The electroencephalogram (EEG) provides an objective, neural correlate of consciousness. Opioid receptors modulate mammalian neuronal excitability, and this fact was used to characterize how opioids administered to mice alter EEG power and states of consciousness. The present study tested the hypothesis that antinociceptive doses of fentanyl, morphine, or buprenorphine differentially alter the EEG and states of sleep and wakefulness in adult, male C57BL/6J mice. Mice were anesthetized and implanted with telemeters that enabled wireless recordings of cortical EEG and electromyogram (EMG). After surgical recovery, EEG and EMG were used to objectively score states of consciousness as wakefulness, rapid eye movement (REM) sleep, or non-REM (NREM) sleep. Measures of EEG power (dB) were quantified as δ (0.5-4 Hz), θ (4-8 Hz), α (8-13 Hz), σ (12-15 Hz), ß (13-30 Hz), and γ (30-60 Hz). Compared with saline (control), fentanyl and morphine decreased NREM sleep, morphine eliminated REM sleep, and buprenorphine eliminated NREM sleep and REM sleep. Opioids significantly and differentially disrupted the temporal organization of sleep/wake states, altered specific EEG frequency bands, and caused dissociated states of consciousness. The results are discussed relative to the fact that opioids, pain, and sleep modulate interacting states of consciousness.NEW & NOTEWORTHY This study discovered that antinociceptive doses of fentanyl, morphine, and buprenorphine significantly and differentially disrupt EEG-defined states of consciousness in C57BL/6J mice. These data are noteworthy because: 1) buprenorphine is commonly used in medication-assisted therapy for opioid addiction, and 2) there is evidence that disordered sleep can promote addiction relapse. The results contribute to community phenotyping efforts by making publicly available all descriptive and inferential statistics from this study (Supplemental Tables S1-S8).


Subject(s)
Analgesics, Opioid/pharmacology , Analgesics/pharmacology , Brain Waves/drug effects , Buprenorphine/pharmacology , Consciousness/drug effects , Dissociative Disorders/chemically induced , Electrocorticography/drug effects , Fentanyl/pharmacology , Morphine/pharmacology , Sleep Stages/drug effects , Wakefulness/drug effects , Analgesics/administration & dosage , Analgesics, Opioid/administration & dosage , Animals , Buprenorphine/administration & dosage , Disease Models, Animal , Electroencephalography , Electromyography , Fentanyl/administration & dosage , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage
9.
Hum Brain Mapp ; 42(16): 5446-5457, 2021 11.
Article in English | MEDLINE | ID: mdl-34464488

ABSTRACT

People with HIV (PWH) use cannabis at a higher rate than the general population, but the influence on neural activity is not well characterized. Cannabis use among PWH may have a beneficial effect, as neuroinflammation is known to be a critical problem in PWH and cannabis use has been associated with a reduction in proinflammatory markers. Thus, it is important to understand the net impact of cannabis use on brain and cognitive function in PWH. In this study, we collected magnetoencephalographic (MEG) brain imaging data on 81 participants split across four demographically matched groups (i.e., PWH using cannabis, controls using cannabis, non-using PWH, and non-using controls). Participants completed a visuospatial processing task during MEG. Time-frequency resolved voxel time series were extracted to identify the dynamics of oscillatory and pre-stimulus baseline neural activity. Our results indicated strong theta (4-8 Hz), alpha (10-16 Hz), and gamma (62-72 Hz) visual oscillations in parietal-occipital brain regions across all participants. PWH exhibited significant behavioral deficits in visuospatial processing, as well as reduced theta oscillations and elevated pre-stimulus gamma activity in visual cortices, all of which replicate prior work. Strikingly, chronic cannabis use was associated with a significant reduction in pre-stimulus gamma activity in the visual cortices, such that PWH no longer statistically differed from controls. These results provide initial evidence that cannabis use may normalize some neural aberrations in PWH. This study fills an important gap in understanding the impact of cannabis use on brain and cognitive function in PWH.


Subject(s)
Brain Waves , Cannabinoid Receptor Modulators/pharmacology , Cognitive Dysfunction , HIV Infections/complications , Medical Marijuana/pharmacology , Visual Cortex , Visual Perception , Adult , Brain Waves/drug effects , Brain Waves/physiology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Female , Humans , Magnetoencephalography , Male , Middle Aged , Visual Cortex/drug effects , Visual Cortex/physiology , Visual Perception/drug effects , Visual Perception/physiology
10.
Cerebrovasc Dis ; 50(2): 216-221, 2021.
Article in English | MEDLINE | ID: mdl-33465768

ABSTRACT

OBJECTIVE: The frequency of seizures after stroke is high, with a severe impact on the quality of life. However, little is known about their prevention. Therefore, we investigated whether early administration of diazepam prevents the development of seizures in acute stroke patients. METHODS: We performed a substudy of the EGASIS trial, a multicenter double-blind, randomized trial in which acute stroke patients were treated with diazepam or placebo for 3 days. Follow-up was after 2 weeks and 3 months. The occurrence of seizures was registered prospectively as one of the prespecified secondary outcomes. RESULTS: 784 EGASIS patients were eligible for this substudy (389 treated with diazepam [49.6%] and 395 treated with placebo [50.4%]). Seizures were reported in 19 patients (2.4% of the total patient group). Seizures occurred less frequently in patients treated with diazepam (1.5 vs. 3.3% in the placebo group); however, this difference was only statistically significant in patients with a cortical anterior circulation infarction (0.9% in the diazepam group vs. 4.6% in the placebo group, incidence rate ratio 0.20, 95% CI: 0.05-0.78, p = 0.02, NNT = 27). CONCLUSION: We found that a 3-day treatment with diazepam after acute cortical anterior circulation stroke prevents the occurrence of seizures in the first 3 months following stroke.


Subject(s)
Anticonvulsants/therapeutic use , Brain Waves/drug effects , Brain/drug effects , Diazepam/therapeutic use , Seizures/prevention & control , Stroke/drug therapy , Adult , Aged , Aged, 80 and over , Anticonvulsants/adverse effects , Brain/physiopathology , Diazepam/adverse effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Netherlands , Prospective Studies , Seizures/diagnosis , Seizures/etiology , Seizures/physiopathology , Stroke/complications , Stroke/diagnosis , Stroke/physiopathology , Time Factors , Treatment Outcome , Young Adult
11.
Br J Anaesth ; 126(2): 477-485, 2021 02.
Article in English | MEDLINE | ID: mdl-33160604

ABSTRACT

BACKGROUND: Desflurane and sevoflurane are commonly used during inhalational anaesthesia, but few studies have investigated their effects on deep cerebral neuronal activity. In addition, the association between subthalamic nucleus (STN) neurophysiology and general anaesthesia induced by volatile anaesthetics are not yet identified. This study aimed to identify differences in neurophysiological characteristics of the STN during comparable minimal alveolar concentration (MAC) desflurane and sevoflurane anaesthesia for deep brain stimulation (DBS) in patients with Parkinson's disease. METHODS: Twelve patients with similar Parkinson's disease severity received desflurane (n=6) or sevoflurane (n=6) during DBS surgery. We obtained STN spike firing using microelectrode recording at 0.5-0.6 MAC and compared firing rate, power spectral density, and coherence. RESULTS: Neuronal firing rate was lower with desflurane (47.4 [26.7] Hz) than with sevoflurane (63.9 [36.5] Hz) anaesthesia (P<0.001). Sevoflurane entrained greater gamma oscillation power than desflurane (62.9% [0.9%] vs 57.0% [1.5%], respectively; P=0.002). There was greater coherence in the theta band of the desflurane group compared with the sevoflurane group (13% vs 6%, respectively). Anaesthetic choice did not differentially influence STN mapping accuracy or the clinical outcome of DBS electrode implantation. CONCLUSIONS: Desflurane and sevoflurane produced distinct neurophysiological profiles in humans that may be associated with their analgesic and hypnotic actions.


Subject(s)
Anesthetics, Inhalation/administration & dosage , Brain Waves/drug effects , Desflurane/administration & dosage , Parkinson Disease/therapy , Sevoflurane/administration & dosage , Subthalamic Nucleus/drug effects , Adult , Aged , Deep Brain Stimulation , Electroencephalography , Female , Humans , Intraoperative Neurophysiological Monitoring , Male , Middle Aged , Parkinson Disease/diagnosis , Parkinson Disease/physiopathology , Subthalamic Nucleus/physiopathology , Treatment Outcome
12.
Br J Anaesth ; 126(2): 445-457, 2021 02.
Article in English | MEDLINE | ID: mdl-33461725

ABSTRACT

Electroencephalographic (EEG) activity is used to monitor the neurophysiology of the brain, which is a target organ of general anaesthesia. Besides its use in evaluating hypnotic states, neurophysiologic reactions to noxious stimulation can also be observed in the EEG. Recognising and understanding these responses could help optimise intraoperative analgesic management. This review describes three types of changes in the EEG induced by noxious stimulation when the patient is under general anaesthesia: (1) beta arousal, (2) (paradoxical) delta arousal, and (3) alpha dropout. Beta arousal is an increase in EEG power in the beta-frequency band (12-25 Hz) in response to noxious stimulation, especially at lower doses of anaesthesia drugs in the absence of opioids. It is usually indicative of a cortical depolarisation and increased cortical activity. At higher concentrations of anaesthetic drug, and with insufficient opioids, delta arousal (increased power in the delta band [0.5-4 Hz]) and alpha dropout (decreased alpha power [8-12 Hz]) are associated with noxious stimuli. The mechanisms of delta arousal are not well understood, but the midbrain reticular formation seems to play a role. Alpha dropout may indicate a return of thalamocortical communication, from an idling mode to an operational mode. Each of these EEG changes reflect an incomplete modulation of pain signals and can be mitigated by administration of opioid or the use of regional anaesthesia techniques. Future studies should evaluate whether titrating analgesic drugs in response to these EEG signals reduces postoperative pain and influences other postoperative outcomes, including the potential development of chronic pain.


Subject(s)
Analgesics/administration & dosage , Anesthesia, General , Anesthetics, General/administration & dosage , Brain Waves/drug effects , Brain/drug effects , Electroencephalography , Intraoperative Neurophysiological Monitoring , Nociception/drug effects , Pain Threshold/drug effects , Pain, Postoperative/prevention & control , Brain/physiopathology , Dose-Response Relationship, Drug , Humans , Pain, Postoperative/physiopathology , Physical Stimulation , Predictive Value of Tests , Time Factors , Treatment Outcome
13.
Br J Anaesth ; 127(3): 435-446, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33972091

ABSTRACT

BACKGROUND: We recently showed that a neurosteroid analogue, (3ß,5ß,17ß)-3-hydroxyandrostane-17-carbonitrile (3ß-OH), induced hypnosis in rats. The aim of the present study was to evaluate the hypnotic and anaesthetic potential of 3ß-OH further using electroencephalography. METHODS: We used behavioural assessment and cortical electroencephalogram (EEG) spectral power analysis to examine hypnotic and anaesthetic effects of 3ß-OH (30 and 60 mg kg-1) administered intraperitoneally or intravenously to young adult male and female rats. RESULTS: We found dose-dependent sex differences in 3ß-OH-induced hypnosis and EEG changes. Both male and female rats responded similarly to i.p. 3ß-OH 30 mg kg-1. However, at the higher dose (60 mg kg-1, i.p.), female rats had two-fold longer duration of spontaneous immobility than male rats (203.4 [61.6] min vs 101.3 [32.1] min), and their EEG was suppressed in the low-frequency range (2-6 Hz), in contrast to male rats. Although a sex-dependent hypnotic effect was not confirmed after 30 mg kg-1 i.v., female rats appeared more sensitive to 3ß-OH with relatively small changes within delta (1-4 Hz) and alpha (8-13 Hz) bands. Finally, 3ß-OH had a rapid onset of action and potent hypnotic/anaesthetic effect after 60 mg kg-1 i.v. in rats of both sexes; however, all female rats and only half of the male rats reached burst suppression, an EEG pattern usually associated with profound inhibition of thalamocortical networks. CONCLUSIONS: Based on its behavioural effects and EEG signature, 3ß-OH is a potent hypnotic in rats, with female rats being more sensitive than male rats.


Subject(s)
Androstanols/pharmacology , Brain Waves/drug effects , Cerebral Cortex/drug effects , Electrocorticography , Immobility Response, Tonic/drug effects , Neurosteroids/pharmacology , Nitriles/pharmacology , Androstanols/administration & dosage , Animals , Cerebral Cortex/physiopathology , Dose-Response Relationship, Drug , Female , Injections, Intraperitoneal , Injections, Intravenous , Male , Neurosteroids/administration & dosage , Nitriles/administration & dosage , Rats, Sprague-Dawley , Sex Factors , Time Factors
14.
Cereb Cortex ; 30(6): 3451-3466, 2020 05 18.
Article in English | MEDLINE | ID: mdl-31989160

ABSTRACT

Sleep slow waves are known to participate in memory consolidation, yet slow waves occurring under anesthesia present no positive effects on memory. Here, we shed light onto this paradox, based on a combination of extracellular recordings in vivo, in vitro, and computational models. We find two types of slow waves, based on analyzing the temporal patterns of successive slow-wave events. The first type is consistently observed in natural slow-wave sleep, while the second is shown to be ubiquitous under anesthesia. Network models of spiking neurons predict that the two slow wave types emerge due to a different gain on inhibitory versus excitatory cells and that different levels of spike-frequency adaptation in excitatory cells can account for dynamical distinctions between the two types. This prediction was tested in vitro by varying adaptation strength using an agonist of acetylcholine receptors, which demonstrated a neuromodulatory switch between the two types of slow waves. Finally, we show that the first type of slow-wave dynamics is more sensitive to external stimuli, which can explain how slow waves in sleep and anesthesia differentially affect memory consolidation, as well as provide a link between slow-wave dynamics and memory diseases.


Subject(s)
Cerebral Cortex/physiology , Neurons/physiology , Receptors, Cholinergic/physiology , Sleep, Slow-Wave/physiology , Anesthesia, General , Anesthetics, Dissociative/pharmacology , Anesthetics, Intravenous/pharmacology , Animals , Brain Waves/drug effects , Brain Waves/physiology , Cats , Cerebral Cortex/drug effects , Cholinergic Agonists/pharmacology , Computer Simulation , Entorhinal Cortex/drug effects , Entorhinal Cortex/physiology , Humans , In Vitro Techniques , Ketamine/pharmacology , Macaca , Memory Consolidation , Mice , Motor Cortex/drug effects , Motor Cortex/physiology , Neural Inhibition , Neurons/drug effects , Parietal Lobe/drug effects , Parietal Lobe/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Primary Visual Cortex/drug effects , Primary Visual Cortex/physiology , Rats , Receptors, Cholinergic/drug effects , Sleep, Slow-Wave/drug effects , Sufentanil/pharmacology , Temporal Lobe/drug effects , Temporal Lobe/physiology
15.
Anesth Analg ; 132(5): 1254-1264, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33857967

ABSTRACT

General anesthesia is characterized by loss of consciousness, amnesia, analgesia, and immobility. Important molecular targets of general anesthetics have been identified, but the neural circuits underlying the discrete end points of general anesthesia remain incompletely understood. General anesthesia and natural sleep share the common feature of reversible unconsciousness, and recent developments in neuroscience have enabled elegant studies that investigate the brain nuclei and neural circuits underlying this important end point. A common approach to measure cortical activity across the brain is electroencephalogram (EEG), which can reflect local neuronal activity as well as connectivity among brain regions. The EEG oscillations observed during general anesthesia depend greatly on the anesthetic agent as well as dosing, and only some resemble those observed during sleep. For example, the EEG oscillations during dexmedetomidine sedation are similar to those of stage 2 nonrapid eye movement (NREM) sleep, but high doses of propofol and ether anesthetics produce burst suppression, a pattern that is never observed during natural sleep. Sleep is primarily driven by withdrawal of subcortical excitation to the cortex, but anesthetics can directly act at both subcortical and cortical targets. While some anesthetics appear to activate specific sleep-active regions to induce unconsciousness, not all sleep-active regions play a significant role in anesthesia. Anesthetics also inhibit cortical neurons, and it is likely that each class of anesthetic drugs produces a distinct combination of subcortical and cortical effects that lead to unconsciousness. Conversely, arousal circuits that promote wakefulness are involved in anesthetic emergence and activating them can induce emergence and accelerate recovery of consciousness. Modern neuroscience techniques that enable the manipulation of specific neural circuits have led to new insights into the neural circuitry underlying general anesthesia and sleep. In the coming years, we will continue to better understand the mechanisms that generate these distinct states of reversible unconsciousness.


Subject(s)
Anesthesia, General , Anesthetics, General/adverse effects , Brain Waves/drug effects , Brain/drug effects , Consciousness/drug effects , Sleep , Anesthesia Recovery Period , Anesthesia, General/adverse effects , Animals , Brain/physiology , Brain Mapping , Electroencephalography , Humans , Neural Pathways/drug effects , Neural Pathways/physiology , Terminology as Topic
16.
Anesth Analg ; 133(5): 1269-1279, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34081045

ABSTRACT

BACKGROUND: Depth-of-anesthesia monitoring is often utilized for patients receiving xenon anesthesia. Processed electroencephalogram (EEG) depth-of-anesthesia monitoring relies to a significant extent on frequency domain analysis of the frontal EEG, and there is evidence that the spectral features observed under anesthesia vary significantly between anesthetic agents. The spectral features of the EEG during xenon anesthesia for a surgical procedure have not previously been described. METHODS: Twenty-four participants scheduled for general anesthesia for lithotripsy were randomized to receive either xenon anesthesia or sevoflurane anesthesia. Frontal EEG recordings were obtained from each participant via the Brain Anesthesia Response Monitor (BARM). Twenty-two EEG recordings were suitable for analysis: 11 in participants who received sevoflurane and 11 in participants who received xenon. Spectrograms for the duration of the anesthetic episode were produced for each participant. Group-level spectral analysis was calculated for two 30-second EEG epochs: one recorded at awake baseline and the other during maintenance anesthesia. A linear mixed-effects model was utilized to compare the changes in 5 frequency bands from baseline to maintenance between the 2 groups. RESULTS: The spectrograms of sevoflurane participants illustrate an increase in frontal delta (0.5-4 Hz), theta (4-8 Hz), and alpha (8-13 Hz) band power during maintenance anesthesia. In contrast, spectrograms of the xenon participants did not illustrate an increase in alpha power. The results of the linear mixed-effects model indicate that both agents were associated with a significant increase in delta power from baseline to maintenance. There was no significant difference in the magnitude of this increase observed between the agents. In contrast, sevoflurane anesthesia was associated with significantly greater absolute power in the theta, alpha, and beta (13-30 Hz) bands when compared to xenon. In terms of relative power, xenon was associated with a significant increase in delta power compared to sevoflurane, while sevoflurane was associated with greater increases in relative theta, alpha, and beta power. CONCLUSIONS: Both xenon anesthesia and sevoflurane anesthesia were associated with significant increases in delta power. Sevoflurane anesthesia was also associated with increases in theta, alpha, and beta power, while xenon anesthesia was associated with greater consolidation of power in the delta band. Xenon anesthesia and sevoflurane anesthesia are associated with distinct spectral features. These findings suggest that appropriate depth-of-anesthesia monitoring may require the development of agent-specific spectral measures of unconsciousness.


Subject(s)
Anesthesia, General , Anesthetics, Inhalation/administration & dosage , Brain Waves/drug effects , Brain/drug effects , Electroencephalography , Intraoperative Neurophysiological Monitoring , Sevoflurane/administration & dosage , Xenon/administration & dosage , Aged , Anesthesia, General/adverse effects , Anesthetics, Inhalation/adverse effects , Brain/physiology , Consciousness/drug effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Prospective Studies , Sevoflurane/adverse effects , Time Factors , Treatment Outcome , Victoria , Xenon/adverse effects
17.
Anesth Analg ; 132(4): e50-e55, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33560660

ABSTRACT

Many general anesthetics potentiate gamma-aminobutyric acid (GABA) A receptors but their neuroanatomic sites of action are less clear. GABAergic neurons in the rostromedial tegmental nucleus (RMTg) send inhibitory projections to multiple arousal-promoting nuclei, but the role of these neurons in modulating consciousness is unknown. In this study, designer receptors exclusively activated by designer drugs (DREADDs) were targeted to RMTg GABAergic neurons of Vgat-ires-Cre mice. DREADDs expression was found in the RMTg and other brainstem regions. Activation of these neurons decreased movement and exploratory behavior, impaired motor coordination, induced electroencephalogram (EEG) oscillations resembling nonrapid eye movement (NREM) sleep without loss of righting and reduced the dose requirement for sevoflurane-induced unconsciousness. These results suggest that GABAergic neurons in the RMTg and other brainstem regions promote sedation and facilitate sevoflurane-induced unconsciousness.


Subject(s)
Anesthetics, Inhalation/pharmacology , Behavior, Animal/drug effects , Brain Stem/drug effects , Consciousness/drug effects , GABAergic Neurons/drug effects , Receptors, G-Protein-Coupled/metabolism , Sevoflurane/pharmacology , Sleep/drug effects , Animals , Brain Stem/metabolism , Brain Waves/drug effects , Exploratory Behavior/drug effects , Female , GABAergic Neurons/metabolism , Male , Mice, Transgenic , Motor Activity/drug effects
18.
Proc Natl Acad Sci U S A ; 115(40): E9469-E9478, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30228120

ABSTRACT

The suprachiasmatic nucleus (SCN), the master circadian clock in mammals, sends major output signals to the subparaventricular zone (SPZ) and further to the paraventricular nucleus (PVN), the neural mechanism of which is largely unknown. In this study, the intracellular calcium levels were measured continuously in cultured hypothalamic slices containing the PVN, SPZ, and SCN. We detected ultradian calcium rhythms in both the SPZ-PVN and SCN regions with periods of 0.5-4.0 hours, the frequency of which depended on the local circadian rhythm in the SPZ-PVN region. The ultradian rhythms were synchronous in the entire SPZ-PVN region and a part of the SCN. Because the ultradian rhythms were not detected in the SCN-only slice, the origin of ultradian rhythm is the SPZ-PVN region. In association with an ultradian bout, a rapid increase of intracellular calcium in a millisecond order was detected, the frequency of which determined the amplitude of an ultradian bout. The synchronous ultradian rhythms were desynchronized and depressed by a sodium channel blocker tetrodotoxin, suggesting that a tetrodotoxin-sensitive network is involved in synchrony of the ultradian bouts. In contrast, the ultradian rhythm is abolished by glutamate receptor blockers, indicating the critical role of glutamatergic mechanism in ultradian rhythm generation, while a GABAA receptor blocker increased the frequency of ultradian rhythm and modified the circadian rhythm in the SCN. A GABAergic network may refine the circadian output signals. The present study provides a clue to unraveling the loci and network mechanisms of the ultradian rhythm.


Subject(s)
Brain Waves/physiology , Calcium Signaling/physiology , Circadian Clocks/physiology , GABAergic Neurons/metabolism , Paraventricular Hypothalamic Nucleus/physiology , Animals , Brain Waves/drug effects , Calcium Signaling/drug effects , Circadian Clocks/drug effects , GABA-A Receptor Antagonists/pharmacology , GABAergic Neurons/cytology , Mice , Paraventricular Hypothalamic Nucleus/cytology , Tetrodotoxin/pharmacology
19.
Int J Neurosci ; 131(6): 580-590, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32228330

ABSTRACT

INTRODUCTION: There is evidence to suggest that melatonin diminishes non-rapid eye movement sleep (NREMS) latency in patients with Alzheimer´s disease (AD). However, melatonin's effects on cortical activity during NREMS in AD have not been studied. The objective of this research was to analyze the effects of melatonin on cortical activity during the stages of NREMS in 8 mild-to-moderate AD patients that received 5-mg of fast-release melatonin. METHODS: During a single-blind, placebo-controlled crossover study, polysomnographic recordings were obtained from C3-A1, C4-A2, F7-T3, F8-T4, F3-F4 and O1-O2. Also, the relative power (RP) and EEG coherences of the delta, theta, alpha1, alpha2, beta1, beta2 and gamma bands were calculated during NREMS-1, NREMS-2 and NREMS-3. These sleep latencies and all EEG data were then compared between the placebo and melatonin conditions. RESULTS: During NREMS-2, a significant RP increase was observed in the theta band of the left-central hemisphere. During NREMS-3, significant RP decreases in the beta bands were recorded in the right-central hemisphere, compared to the placebo group. After melatonin administration, significant decreases of EEG coherences in the beta2, beta1 and gamma bands were observed in the right hemisphere during NREMS-3. DISCUSSION: We conclude that short NREMS onset related to melatonin intake in AD patients is associated with a significant RP increase in the theta band and a decrease in RP and EEG coherences in the beta and gamma bands during NREMS-3. These results suggest that the GABAergic pathways are preserved in mild-to-moderate AD.


Subject(s)
Alzheimer Disease/complications , Brain Waves/drug effects , Central Nervous System Depressants/pharmacology , Electroencephalography Phase Synchronization/drug effects , Melatonin/pharmacology , Sleep Stages/drug effects , Sleep Wake Disorders/drug therapy , Aged , Alzheimer Disease/physiopathology , Brain Waves/physiology , Central Nervous System Depressants/administration & dosage , Cross-Over Studies , Humans , Male , Melatonin/administration & dosage , Middle Aged , Pilot Projects , Polysomnography , Severity of Illness Index , Single-Blind Method , Sleep Stages/physiology , Sleep Wake Disorders/etiology , Sleep Wake Disorders/physiopathology
20.
J Stroke Cerebrovasc Dis ; 30(9): 105651, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33581988

ABSTRACT

Chronic kidney disease and seizures often co-exist. When seizures are provoked in patients with kidney disease, their treatment poses a particular challenge. Seizures may be provoked in the context of uremia, and toxic substances associated with uremic encephalopathy. In that case, the mainstay of therapy is to treat the uremia before consideration for anticonvulsant therapy. Treatment of seizures in the setting of chronic kidney disease requires special attention to selection of anticonvulsant medications and knowledge of the altered pharmacokinetics of these medications, which may require special titration schedule in that setting. The purpose of this review is to summarize the current knowledge about inter-relation of seizures and kidney disease. The review will also help practitioners who treat patients with renal failure and coexisting seizures in choosing the best treatment options.


Subject(s)
Anticonvulsants/therapeutic use , Brain Waves/drug effects , Brain/drug effects , Epilepsy/drug therapy , Kidney Diseases/therapy , Renal Dialysis , Uremia/therapy , Anticonvulsants/adverse effects , Anticonvulsants/pharmacokinetics , Brain/physiopathology , Electroencephalography , Epilepsy/diagnosis , Epilepsy/etiology , Epilepsy/physiopathology , Humans , Kidney Diseases/complications , Kidney Diseases/diagnosis , Kidney Diseases/physiopathology , Male , Middle Aged , Predictive Value of Tests , Renal Dialysis/adverse effects , Risk Factors , Treatment Outcome , Uremia/complications , Uremia/diagnosis , Uremia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL