Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mol Psychiatry ; 28(1): 28-33, 2023 01.
Article in English | MEDLINE | ID: mdl-35858991

ABSTRACT

Many psychiatric and neurological disorders present deficits in both the social and cognitive domain. In this perspectives article, we provide an overview and the potential of the existence of an extensive neurobiological substrate underlying the close relationship between these two domains. By mapping the rodent brain regions involved in the social and/or cognitive domain, we show that the vast majority of brain regions involved in the cognitive domain are also involved in the social domain. The identified neuroanatomical overlap has an evolutionary basis, as complex social behavior requires cognitive skills, and aligns with the reported functional interactions of processes underlying cognitive and social performance. Based on the neuroanatomical mapping, recent (pre-)clinical findings, and the evolutionary perspective, we emphasize that the social domain requires more focus as an important treatment target and/or biomarker, especially considering the presently limited treatment strategies for these disorders.


Subject(s)
Brain Diseases , Brain , Humans , Social Behavior , Cognition , Brain Mapping
2.
Neuropsychobiology ; 80(5): 374-392, 2021.
Article in English | MEDLINE | ID: mdl-33588406

ABSTRACT

BACKGROUND/AIMS: Olfactory dysfunction can provide valuable insight into early pathophysiological processes of brain disorders. Olfactory processing of chemosensory and odour sensitivity relies on segregating salient odours from background odours cues. Odour-evoked fast oscillations in the olfactory bulb (OB) are hypothesized to be an important index of odour quality coding. The present preclinical work aimed at better understanding connectivity associated with odour coding and behavioural odour discrimination. METHODS: Network oscillations and functional connectivity (FC) were measured in C57BL/6 mice performing the olfactory associative odour learning (OL) test, using multichannel local field potential recordings in key olfactory networks. Cholinergic modulation of odour processing was investigated using the muscarinic antagonist scopolamine. RESULTS: At the behavioural level, olfactory memory, which refers to the acquisition and recollection of a reference odour by reduced exploration time, was observed in animals that correctly learned the task. Significant decrease in mean investigation and retrieval time of the associated odour-food reward was observed between trials. At the network level, the associated odour during sniffing behaviour was associated with enhanced coherence in the ß and γ frequency oscillations across the olfactory pathway, with marked changes observed between the OB and anterior piriform cortex (PC). The enhanced phase-amplitude cross-frequency coupling in the OB and the weak coupling index in the hippocampal CA1 suggests a role of the OB network in olfaction encoding and processing. Scopolamine impaired behavioural and FC underlying recall and retrieval of the associated odour. CONCLUSION: The results suggest that the acquisition and formation of odour reference memory rely primarily on FC at the OB-PC network and confirm the role of muscarinic receptors in olfactory retrieval processing.


Subject(s)
Odorants , Olfactory Bulb , Animals , Cholinergic Agents , Mice , Mice, Inbred C57BL , Olfactory Pathways
3.
Clin Neurophysiol ; 131(1): 285-307, 2020 01.
Article in English | MEDLINE | ID: mdl-31501011

ABSTRACT

In 1999, the International Federation of Clinical Neurophysiology (IFCN) published "IFCN Guidelines for topographic and frequency analysis of EEGs and EPs" (Nuwer et al., 1999). Here a Workgroup of IFCN experts presents unanimous recommendations on the following procedures relevant for the topographic and frequency analysis of resting state EEGs (rsEEGs) in clinical research defined as neurophysiological experimental studies carried out in neurological and psychiatric patients: (1) recording of rsEEGs (environmental conditions and instructions to participants; montage of the EEG electrodes; recording settings); (2) digital storage of rsEEG and control data; (3) computerized visualization of rsEEGs and control data (identification of artifacts and neuropathological rsEEG waveforms); (4) extraction of "synchronization" features based on frequency analysis (band-pass filtering and computation of rsEEG amplitude/power density spectrum); (5) extraction of "connectivity" features based on frequency analysis (linear and nonlinear measures); (6) extraction of "topographic" features (topographic mapping; cortical source mapping; estimation of scalp current density and dura surface potential; cortical connectivity mapping), and (7) statistical analysis and neurophysiological interpretation of those rsEEG features. As core outcomes, the IFCN Workgroup endorsed the use of the most promising "synchronization" and "connectivity" features for clinical research, carefully considering the limitations discussed in this paper. The Workgroup also encourages more experimental (i.e. simulation studies) and clinical research within international initiatives (i.e., shared software platforms and databases) facing the open controversies about electrode montages and linear vs. nonlinear and electrode vs. source levels of those analyses.


Subject(s)
Electroencephalography/methods , Mental Disorders/physiopathology , Nervous System Diseases/physiopathology , Rest/physiology , Artifacts , Biomedical Research , Brain Mapping/methods , Brain Waves/physiology , Databases as Topic , Electrodes , Electroencephalography/instrumentation , Electroencephalography/standards , Electroencephalography Phase Synchronization/physiology , Environment , Humans , Information Storage and Retrieval/methods , Neurophysiology , Scalp , Simulation Training , Software , Wakefulness/physiology
4.
Sci Rep ; 9(1): 16363, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31705038

ABSTRACT

Multiple animal models have been created to gain insight into Alzheimer's disease (AD) pathology. Among the most commonly used models are transgenic mice overexpressing human amyloid precursor protein (APP) with mutations linked to familial AD, resulting in the formation of amyloid ß plaques, one of the pathological hallmarks observed in AD patients. However, recent evidence suggests that the overexpression of APP by itself can confound some of the reported observations. Therefore, we investigated in the present study the AppNL-G-Fmodel, an App knock-in (App-KI) mouse model that develops amyloidosis in the absence of APP-overexpression. Our findings at the behavioral, electrophysiological, and histopathological level confirmed an age-dependent increase in Aß1-42 levels and plaque deposition in these mice in accordance with previous reports. This had apparently no consequences on cognitive performance in a visual discrimination (VD) task, which was largely unaffected in AppNL-G-F mice at the ages tested. Additionally, we investigated neurophysiological functioning of several brain areas by phase-amplitude coupling (PAC) analysis, a measure associated with adequate cognitive functioning, during the VD task (starting at 4.5 months) and the exploration of home environment (at 5 and 8 months of age). While we did not detect age-dependent changes in PAC during home environment exploration for both the wild-type and the AppNL-G-F mice, we did observe subtle changes in PAC in the wild-type mice that were not present in the AppNL-G-F mice.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Brain Waves/physiology , Cognition/physiology , Disease Models, Animal , Neurons/pathology , Plaque, Amyloid/pathology , Animals , Behavior, Animal , Discrimination Learning , Gene Knock-In Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neurons/metabolism , Plaque, Amyloid/metabolism , Visual Perception
5.
Neuropsychobiology ; 72(3-4): 151-64, 2015.
Article in English | MEDLINE | ID: mdl-26901596

ABSTRACT

The contemporary value of animal pharmaco-electroencephalography (p-EEG)-based applications are strongly interlinked with progress in recording and neuroscience analysis methodology. While p-EEG in humans and animals has been shown to be closely related in terms of underlying neuronal substrates, both translational and back-translational approaches are being used to address extrapolation issues and optimize the translational validity of preclinical animal p-EEG paradigms and data. Present applications build further on animal p-EEG and pharmaco-sleep EEG findings, but also on stimulation protocols, more specifically pharmaco-event-related potentials. Pharmaceutical research into novel treatments for neurological and psychiatric diseases has employed an increasing number of pharmacological as well as transgenic models to assess the potential therapeutic involvement of different neurochemical systems and novel drug targets as well as underlying neuronal connectivity and synaptic function. Consequently, p-EEG studies, now also readily applied in modeled animals, continue to have an important role in drug discovery and development, with progressively more emphasis on its potential as a central readout for target engagement and as a (translational) functional marker of neuronal circuit processes underlying normal and pathological brain functioning. In a similar vein as was done for human p-EEG studies, the contribution of animal p-EEG studies can further benefit by adherence to guidelines for methodological standardization, which are presently under construction by the International Pharmaco-EEG Society (IPEG).


Subject(s)
Central Nervous System Agents/pharmacology , Electroencephalography , Mental Disorders/physiopathology , Nervous System Diseases/physiopathology , Translational Research, Biomedical , Animals , Brain Waves/drug effects , Central Nervous System Agents/therapeutic use , Disease Models, Animal , Humans , Mental Disorders/drug therapy , Nervous System Diseases/drug therapy , Translational Research, Biomedical/instrumentation , Translational Research, Biomedical/methods
6.
Neuropsychobiology ; 72(3-4): 139-50, 2015.
Article in English | MEDLINE | ID: mdl-26901675

ABSTRACT

Current research on the effects of pharmacological agents on human neurophysiology finds its roots in animal research, which is also reflected in contemporary animal pharmaco-electroencephalography (p-EEG) applications. The contributions, present value and translational appreciation of animal p-EEG-based applications are strongly interlinked with progress in recording and neuroscience analysis methodology. After the pioneering years in the late 19th and early 20th century, animal p-EEG research flourished in the pharmaceutical industry in the early 1980s. However, around the turn of the millennium the emergence of structurally and functionally revealing imaging techniques and the increasing application of molecular biology caused a temporary reduction in the use of EEG as a window into the brain for the prediction of drug efficacy. Today, animal p-EEG is applied again for its biomarker potential - extensive databases of p-EEG and polysomnography studies in rats and mice hold EEG signatures of a broad collection of psychoactive reference and test compounds. A multitude of functional EEG measures has been investigated, ranging from simple spectral power and sleep-wake parameters to advanced neuronal connectivity and plasticity parameters. Compared to clinical p-EEG studies, where the level of vigilance can be well controlled, changes in sleep-waking behaviour are generally a prominent confounding variable in animal p-EEG studies and need to be dealt with. Contributions of rodent pharmaco-sleep EEG research are outlined to illustrate the value and limitations of such preclinical p-EEG data for pharmacodynamic and chronopharmacological drug profiling. Contemporary applications of p-EEG and pharmaco-sleep EEG recordings in animals provide a common and relatively inexpensive window into the functional brain early in the preclinical and clinical development of psychoactive drugs in comparison to other brain imaging techniques. They provide information on the impact of drugs on arousal and sleep architecture, assessing their neuropharmacological characteristics in vivo, including central exposure and information on kinetics. In view of the clear disadvantages as well as advantages of animal p-EEG as compared to clinical p-EEG, general statements about the usefulness of EEG as a biomarker to demonstrate the translatability of p-EEG effects should be made with caution, however, because they depend on the particular EEG or sleep parameter that is being studied. The contribution of animal p-EEG studies to the translational characterisation of centrally active drugs can be furthered by adherence to guidelines for methodological standardisation, which are presently under construction by the International Pharmaco-EEG Society (IPEG).


Subject(s)
Brain Waves/drug effects , Central Nervous System Agents/pharmacology , Electroencephalography/history , Translational Research, Biomedical/history , Animals , Brain/drug effects , Brain/physiology , Electroencephalography/methods , History, 19th Century , History, 20th Century , Humans , Models, Animal , Sleep/drug effects , Sleep/physiology
7.
Neuropsychobiology ; 67(3): 127-67, 2013.
Article in English | MEDLINE | ID: mdl-23548759

ABSTRACT

The International Pharmaco-EEG Society (IPEG) presents guidelines summarising the requirements for the recording and computerised evaluation of pharmaco-sleep data in man. Over the past years, technical and data-processing methods have advanced steadily, thus enhancing data quality and expanding the palette of sleep assessment tools that can be used to investigate the activity of drugs on the central nervous system (CNS), determine the time course of effects and pharmacodynamic properties of novel therapeutics, hence enabling the study of the pharmacokinetic/pharmacodynamic relationship, and evaluate the CNS penetration or toxicity of compounds. However, despite the presence of robust guidelines on the scoring of polysomnography -recordings, a review of the literature reveals inconsistent -aspects in the operating procedures from one study to another. While this fact does not invalidate results, the lack of standardisation constitutes a regrettable shortcoming, especially in the context of drug development programmes. The present guidelines are intended to assist investigators, who are using pharmaco-sleep measures in clinical research, in an effort to provide clear and concise recommendations and thereby to standardise methodology and facilitate comparability of data across laboratories.


Subject(s)
Electroencephalography/standards , Pharmacology, Clinical/standards , Polysomnography/standards , Practice Guidelines as Topic/standards , Sleep/drug effects , Societies, Medical/standards , Humans , Pharmacology, Clinical/methods
8.
Neuropsychobiology ; 66(4): 201-20, 2012.
Article in English | MEDLINE | ID: mdl-23075830

ABSTRACT

The International Pharmaco-EEG Society (IPEG) presents updated guidelines summarising the requirements for the recording and computerised evaluation of pharmaco-EEG data in man. Since the publication of the first pharmaco-EEG guidelines in 1982, technical and data processing methods have advanced steadily, thus enhancing data quality and expanding the palette of tools available to investigate the action of drugs on the central nervous system (CNS), determine the pharmacokinetic and pharmacodynamic properties of novel therapeutics and evaluate the CNS penetration or toxicity of compounds. However, a review of the literature reveals inconsistent operating procedures from one study to another. While this fact does not invalidate results per se, the lack of standardisation constitutes a regrettable shortcoming, especially in the context of drug development programmes. Moreover, this shortcoming hampers reliable comparisons between outcomes of studies from different laboratories and hence also prevents pooling of data which is a requirement for sufficiently powering the validation of novel analytical algorithms and EEG-based biomarkers. The present updated guidelines reflect the consensus of a global panel of EEG experts and are intended to assist investigators using pharmaco-EEG in clinical research, by providing clear and concise recommendations and thereby enabling standardisation of methodology and facilitating comparability of data across laboratories.


Subject(s)
Electroencephalography/standards , Societies, Scientific/standards , Consensus , Drug Evaluation/standards , Electroencephalography/methods , Humans
9.
Eur J Pharmacol ; 680(1-3): 63-8, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-22314225

ABSTRACT

Abnormalities in the regulation of the hypothalamic stress hormone corticotropin-releasing factor (CRF) are thought to play a critical role in mood disorders. Consequently, CRF receptor antagonists have been proposed as potential novel therapeutic agents of these conditions. Sleep disturbance is common in depressed patients and changed sleep-wake architecture is considered as potential predictor or surrogate marker of response to treatment. The aim of our study was to characterise the effects of oral administration of the corticotropin-releasing factor CRF(1) receptor antagonist R278995/CRA0450 (3 and 10mg/kg) on sleep-wake organization and electroencephalographic (EEG) components in Sprague-Dawley rats, and to determine whether the changes observed in the sleep-EEG pattern resemble those seen with antidepressants. At 3mg/kg, R278995/CRA0450 produced minor changes in sleep behaviour, while an overall reduction in power spectra was observed during deep slow wave sleep. At 10mg/kg, R278995/CRA0450 consistently reduced rapid eye movement (REM) sleep (-75.4%) and increased the REM sleep onset latency (+67%, 92.1±4.9min for vehicle vs. 153.8±24min for R278995/CRA0450), in the absence of systematic changes in spectral EEG pattern, which are characteristic anti-depressant-like effects. These findings in rats indicate that the corticotropin-releasing factor CRF(1) receptor antagonist R278995/CRA0450 is centrally active under standard conditions as it inhibits REM sleep and promotes wakefulness. The characteristic changes found in the sleep EEG model further support the hypothesis that R278995/CRA0450 could exert a non-sedative, antidepressant-like action.


Subject(s)
Benzenesulfonates/pharmacology , Quinolines/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Sleep, REM/drug effects , Administration, Oral , Animals , Antidepressive Agents/pharmacology , Electroencephalography/methods , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/metabolism , Sleep/drug effects , Wakefulness/drug effects
10.
J Biopharm Stat ; 20(4): 768-86, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20496205

ABSTRACT

This paper proposes a modified model averaging approach for linear discriminant analysis. This approach is used in combination with a doubly hierarchical supervised learning analysis and applied to preclinical pharmaco-electroencephalographical data for classification of psychotropic drugs. Classification of a test dataset was highly improved with this method.


Subject(s)
Artificial Intelligence , Drug Evaluation, Preclinical/methods , Electroencephalography/drug effects , Models, Statistical , Psychotropic Drugs/pharmacology , Algorithms , Animals , Discriminant Analysis , Likelihood Functions , Linear Models , Psychotropic Drugs/classification , Rats , Regression Analysis , Selection Bias , Sleep Stages/drug effects , Wakefulness/drug effects
11.
Eur J Pharmacol ; 603(1-3): 62-72, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19046965

ABSTRACT

Compiling pharmacological evidence implicates metabotropic glutamate mGlu(2) receptors in the regulation of emotional states and suggests positive modulators as a novel therapeutic approach of Anxiety/Depression and Schizophrenia. Here, we investigated subcutaneous effects of the metabotropic glutamate mGlu(2/3) agonist (LY354740) on sleep-wake architecture in rat. To confirm the specific effects on rapid eye movement (REM) sleep were mediated via metabotropic glutamate mGlu(2) receptors, we characterized the sleep-wake cycles in metabotropic glutamate mGlu(2) receptor deficient mice (mGlu(2)R(-/-)) and their arousal response to LY354740. We furthermore examined effects on sleep behavior in rats of the positive allosteric modulator, biphenyl-indanone A (BINA) alone and in combination with LY354740 at sub-effective doses. LY354740 (1, 3 and 10 mg/kg) dose-dependently suppressed REM sleep and prolonged its onset latency. Metabotropic glutamate mGlu(2)R(-/-) and their wild type (WT) littermates exhibited similar spontaneous sleep-wake phenotype, while LY354740 (10 mg/kg) significantly affected REM sleep variables in WT but not in the mutant. In rats, BINA (1, 3, 10, 20, 40 mg/kg) dose-dependently suppressed REM sleep, lengthened its onset latency and slightly enhanced passive waking. Additionally, combined treatment elicited a synergistic action on REM sleep variables. Our findings show common changes of REM sleep variables following modulation of metabotropic glutamate mGlu(2) receptor and support an active role of this receptor in the regulation of REM sleep. The synergistic action of BINA on LY354740's effects on sleep pattern implies that positive modulators would tune the endogenous glutamate tone suggesting potential benefit in the treatment of psychiatric disorders, in which REM sleep overdrive is manifested.


Subject(s)
Receptors, Metabotropic Glutamate/metabolism , Sleep/drug effects , Wakefulness/drug effects , Allosteric Regulation , Animals , Biphenyl Compounds/administration & dosage , Biphenyl Compounds/pharmacology , Bridged Bicyclo Compounds/administration & dosage , Bridged Bicyclo Compounds/chemical synthesis , Bridged Bicyclo Compounds/pharmacology , Drug Combinations , Glutamic Acid/metabolism , Indans/administration & dosage , Indans/pharmacology , Mice , Rats , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/deficiency , Sleep, REM/drug effects , Substrate Specificity
12.
J Biopharm Stat ; 18(6): 1120-35, 2008.
Article in English | MEDLINE | ID: mdl-18991112

ABSTRACT

This paper proposes a general and simple procedure that can be applied to establish classification rules for application to multiple-class longitudinal data. The procedure is applied to preclinical pharmaco-electroencephalogram (EEG) studies aiming at characterizing psychotropic drug effects on the basis of spectral EEG analysis. It is a flexible hierarchical supervised learning tool that takes into account the specific nature of the multiple drug classes, as well as the longitudinal aspect of the data. Several variations of this procedure are applied to the EEG data, generally producing comparable results, in particular similar association between the sleeping stages and the psychotropic drug classes.


Subject(s)
Discriminant Analysis , Drug Evaluation, Preclinical/statistics & numerical data , Electroencephalography/statistics & numerical data , Models, Statistical , Psychotropic Drugs/pharmacology , Sleep/drug effects , Wakefulness/drug effects , Animals , Dose-Response Relationship, Drug , Linear Models , Longitudinal Studies , Models, Animal , Psychotropic Drugs/classification , Rats , Reproducibility of Results , Time Factors
13.
Eur J Pharmacol ; 579(1-3): 177-88, 2008 Jan 28.
Article in English | MEDLINE | ID: mdl-18062961

ABSTRACT

Melanin-concentrating hormone (MCH) is a hypothalamic peptide that centrally regulates food intake, energy balance and emotion. Interestingly, MCH and melanin-concentrating hormone MCH(1) receptors are distributed in brain areas known to regulate vigilance states. Effects of subcutaneous administration of two selective melanin-concentrating hormone MCH(1) receptor antagonists, labeled A and B were examined over a broad dose range (1, 3, 10, 20, 40 mg/kg) on rat sleep-wake architecture. Both compounds have a nanomolar antagonist activity at recombinant human melanin-concentrating hormone MCH(1) receptor (IC(50)=44.1+/-6.1 nM and 26.6+/-5.4 nM, respectively) and potently inhibited the MCH-induced mobilization of [Ca(2+)] (IC(50) 29.1+/-8.1 nM and 10.5+/-4.1 nM, respectively). The selectivity of both compounds was further confirmed on a panel of receptors, transporters and channels. In vivo, both compounds dose-dependently decreased deep sleep primarily by decreasing the mean duration of episodes during the first 4 h post-administration. In parallel, REM sleep and intermediate stage sleep were decreased while active and passive waking increased. Deep sleep and REM sleep onset latencies were significantly prolonged at higher doses. No homeostatic rebound of deep sleep was observed, while a tendency for recovery of REM sleep was found during subsequent dark phase. Together, the results support a role of the melanin-concentrating hormone MCH(1) receptor in the regulation of deep slow-wave sleep-REM sleep cycle. Therapeutic application of melanin-concentrating hormone MCH(1) receptor-inhibiting agents should take into account the significant decreases in deep sleep without recovery as these may interfere with sleep dependent memory consolidation.


Subject(s)
Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/metabolism , Sleep Stages/physiology , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Injections, Subcutaneous , Male , Rats , Rats, Sprague-Dawley , Sleep Stages/drug effects , Sleep, REM/drug effects , Sleep, REM/physiology
14.
Eur J Pharm Sci ; 30(1): 45-55, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17134886

ABSTRACT

The limitations of blood sampling in pharmacokinetic (PK)/pharmacodynamic (PD) studies in behavioral animal models could in part be overcome by a mixed effects modeling approach. This analysis characterizes and evaluates the population PK of fluvoxamine in rat plasma using nonlinear mixed effects modeling. The model is assessed for its utility in animal behavioral PK/PD studies. In six studies with a different experimental setup, study site and/or sampling design, rats received an intravenous infusion of 1, 3.7 or 7.3mg/kg fluvoxamine. A population three-compartment PK model adequately described the fluvoxamine plasma concentrations. Body weight was included as a covariate and mean population PK parameters for CL, V(1), V(2), Q(2), V(3) and Q(3) were 25.1 ml/min, 256 ml, 721 ml, 30.3 ml/min, 136 ml and 1.0 ml/min, respectively. Inter-individual variability was identified on CL (39.5%), V(1) (43.5%), V(2) (50.1%) and Q(2) (25.7%). A predictive check and bootstrap analysis confirmed the predictive ability, model stability and precision of the parameter estimates. Body weight was identified as a significant covariate of the inter-compartmental clearance Q(2). The pharmacokinetics was independent of factors such as dose, surgery (for instrumentation) and study site. The utility of the model in animal behavioral studies was demonstrated in a PK/PD analysis of the effects on REM sleep in which a sparse PK sampling design was used. By using the pertinent information from the population PK model, individual PK profiles and the PK/PD correlation could be adequately described.


Subject(s)
Behavior, Animal/drug effects , Fluvoxamine , Models, Biological , Selective Serotonin Reuptake Inhibitors , Animals , Fluvoxamine/pharmacokinetics , Fluvoxamine/pharmacology , Infusions, Intravenous , Male , Rats , Rats, Wistar , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , Selective Serotonin Reuptake Inhibitors/pharmacology , Sleep, REM/drug effects
15.
Neuropharmacology ; 47(2): 163-74, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15223295

ABSTRACT

A pharmacological dissociation of the relation between electroencephalographic (EEG) activity and behavior has been described for the benzodiazepines. While a decrease in high frequency EEG activity is associated with a decrease in arousal in drug-free conditions, sedative benzodiazepines increase beta activity. Non-benzodiazepine GABA(A) receptor modulators can increase beta activity as well. To further study the relationship between rat behavior and EEG under GABA(A) receptor modulation, EEG effects of diazepam (2.5 mg/kg) and zolpidem (2.5 mg/kg) were studied during different behaviors. Both drugs modulate the GABA(A) receptor, albeit that zolpidem shows alpha(1) subunit selectivity while diazepam is non-selective. A detailed analysis of rat open field behavior was made with a distinction of 25 behavioral elements. The EEG was segmented according to each behavioral element and a corresponding power spectrum calculated. Both diazepam and zolpidem increased EEG beta frequencies, characteristic for the benzodiazepines. However, the beta and gamma increase was specific for active behavior and not for inactivity. Interestingly, diazepam and zolpidem seemed to amplify, rather than dissociate, the relation between behavior and the EEG. It is hypothesized that the large increase in beta-3/gamma activity caused by diazepam and zolpidem is a compensatory mechanism that allows for behavioral activation, despite pharmacologically induced sedation.


Subject(s)
Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Beta Rhythm/drug effects , Diazepam/pharmacology , Electroencephalography/drug effects , Hypnotics and Sedatives/pharmacology , Pyridines/pharmacology , Animals , Cerebral Cortex/drug effects , Electrodes , Hippocampus/drug effects , Male , Rats , Rats, Long-Evans , Solvents , Zolpidem
16.
Physiol Behav ; 78(2): 241-6, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12576121

ABSTRACT

Chronic neuropathic pain patients often report sleep disturbances such as reduced amount of sleep and excessive daytime tiredness. The aim of this study was to evaluate possible abnormalities in sleep patterns in a widely used animal model of neuropathic pain. Adult male Sprague-Dawley rats were chronically implanted with electrodes for electroencephalogram (EEG) and electromyogram (EMG) registrations to allow continuous 24-h polygraphic recording. Subsequently, a chronic constriction injury (CCI) was inflicted on eight rats in accordance with the CCI model of neuropathic pain and a sham operation was performed on another eight rats. The polygraphic recordings were repeated 13, 27, 55, and 146 days after surgery. Although the CCI animals developed significant mechanical and cold allodynia and heat hyperalgesia, there were no significant differences between the CCI rats and the sham-operated control animals in the spontaneous EEG/EMG in homecage-like conditions. It is concluded that in the chronic phase, this neuropathic pain model does not produce clear sleep disturbances. Such an absence of general suffering from sleep disturbances is advantageous to the CCI model as it makes use of the model more acceptable ethically. Nonetheless, this outcome appears to be in contrast with the clinical situation in neuropathic pain and therefore could also be seen as a disadvantage for the face validity of the CCI model.


Subject(s)
Electroencephalography , Nervous System Diseases/complications , Pain/etiology , Pain/physiopathology , Sleep , Animals , Chronic Disease , Cold Temperature , Electromyography , Hot Temperature , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Hyperesthesia/etiology , Hyperesthesia/physiopathology , Ligation , Male , Nervous System Diseases/etiology , Rats , Rats, Sprague-Dawley , Sciatic Nerve
SELECTION OF CITATIONS
SEARCH DETAIL
...