Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Anesthesiology ; 135(4): 633-648, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34270686

RESUMO

BACKGROUND: Parabrachial nucleus excitation reduces cortical delta oscillation (0.5 to 4 Hz) power and recovery time associated with anesthetics that enhance γ-aminobutyric acid type A receptor action. The effects of parabrachial nucleus excitation on anesthetics with other molecular targets, such as dexmedetomidine and ketamine, remain unknown. The hypothesis was that parabrachial nucleus excitation would cause arousal during dexmedetomidine and ketamine anesthesia. METHODS: Designer Receptors Exclusively Activated by Designer Drugs were used to excite calcium/calmodulin-dependent protein kinase 2α-positive neurons in the parabrachial nucleus region of adult male rats without anesthesia (nine rats), with dexmedetomidine (low dose: 0.3 µg · kg-1 · min-1 for 45 min, eight rats; high dose: 4.5 µg · kg-1 · min-1 for 10 min, seven rats), or with ketamine (low dose: 2 mg · kg-1 · min-1 for 30 min, seven rats; high dose: 4 mg · kg-1 · min-1 for 15 min, eight rats). For control experiments (same rats and treatments), the Designer Receptors Exclusively Activated by Designer Drugs were not excited. The electroencephalogram and anesthesia recovery times were recorded and analyzed. RESULTS: Parabrachial nucleus excitation reduced delta power in the prefrontal electroencephalogram with low-dose dexmedetomidine for the 150-min analyzed period, excepting two brief periods (peak median bootstrapped difference [clozapine-N-oxide - saline] during dexmedetomidine infusion = -6.06 [99% CI = -12.36 to -1.48] dB, P = 0.007). However, parabrachial nucleus excitation was less effective at reducing delta power with high-dose dexmedetomidine and low- and high-dose ketamine (peak median bootstrapped differences during high-dose [dexmedetomidine, ketamine] infusions = [-1.93, -0.87] dB, 99% CI = [-4.16 to -0.56, -1.62 to -0.18] dB, P = [0.006, 0.019]; low-dose ketamine had no statistically significant decreases during the infusion). Recovery time differences with parabrachial nucleus excitation were not statistically significant for dexmedetomidine (median difference for [low, high] dose = [1.63, 11.01] min, 95% CI = [-20.06 to 14.14, -20.84 to 23.67] min, P = [0.945, 0.297]) nor low-dose ketamine (median difference = 12.82 [95% CI: -3.20 to 39.58] min, P = 0.109) but were significantly longer for high-dose ketamine (median difference = 11.38 [95% CI: 1.81 to 24.67] min, P = 0.016). CONCLUSIONS: These results suggest that the effectiveness of parabrachial nucleus excitation to change the neurophysiologic and behavioral effects of anesthesia depends on the anesthetic's molecular target.


Assuntos
Ritmo Delta/efeitos dos fármacos , Dexmedetomidina/farmacologia , Ácido Glutâmico , Ketamina/farmacologia , Neurônios/efeitos dos fármacos , Núcleos Parabraquiais/efeitos dos fármacos , Anestesia/métodos , Anestésicos Dissociativos/farmacologia , Animais , Proteínas de Ligação ao Cálcio/fisiologia , Ritmo Delta/fisiologia , Ácido Glutâmico/fisiologia , Hipnóticos e Sedativos/farmacologia , Masculino , Neurônios/fisiologia , Núcleos Parabraquiais/fisiologia , Ratos , Ratos Sprague-Dawley
2.
Schizophr Res ; 208: 293-299, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30738699

RESUMO

Decreased brain activity in the frontal region, as indicated by increased slow wave EEG power measured by electrodes place on the skull over this area, in association with negative symptoms has previously been shown to distinguish ultra-high risk (UHR) individuals who later transitioned to psychosis (UHR-P) from those who did not transition (UHR-NP). The aims of the current study were to: 1) replicate these results and 2) investigate whether similar association between increased frontal slow wave activity and functioning shows any value in the prediction of transition to psychosis in UHR individuals. The brain activity, recorded using EEG, of 44 UHR individuals and 38 healthy controls was included in the analyses. Symptom severity was assessed in UHR participants and functioning was measured in both groups. The power in the theta frequency band in the frontal region of UHR individuals was higher than in controls. However, there was no difference between the UHR-P and the UHR-NP groups, and no change in slow frequency power following transition to psychosis. The correlation between delta frequency power and negative symptoms previously observed was not present in our UHR cohort, and there was no association between frontal delta or theta and functioning in either group. Increased delta power was rather correlated with depressive symptoms in the UHR group. Future research will be needed to better understand when, in the course of the illness, does the slow wave activity in the frontal area becomes impaired.


Assuntos
Eletroencefalografia , Lobo Frontal/fisiopatologia , Transtornos Psicóticos/fisiopatologia , Adolescente , Adulto , Austrália , Estudos de Coortes , Correlação de Dados , Ritmo Delta/efeitos dos fármacos , Depressão/diagnóstico , Depressão/fisiopatologia , Depressão/psicologia , Progressão da Doença , Eletroencefalografia/efeitos dos fármacos , Função Executiva/efeitos dos fármacos , Função Executiva/fisiologia , Ácidos Graxos Ômega-3/uso terapêutico , Feminino , Lobo Frontal/efeitos dos fármacos , Humanos , Masculino , Prognóstico , Escalas de Graduação Psiquiátrica/estatística & dados numéricos , Psicometria , Transtornos Psicóticos/diagnóstico , Transtornos Psicóticos/tratamento farmacológico , Transtornos Psicóticos/psicologia , Valores de Referência , Risco , Fatores de Risco , Ritmo Teta/efeitos dos fármacos , Adulto Jovem
3.
Neuroscience ; 404: 541-556, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30738854

RESUMO

Aging is associated with sleep-wake disruption, dampening of circadian amplitudes, and a reduced homeostatic sleep response. Aging is also associated with a decline in hypothalamic cell proliferation. We hypothesized that the aging-related decline in cell-proliferation contributes to the dysfunction of preoptic-hypothalamic sleep-wake and circadian systems and consequent sleep-wake disruption. We determined if cytosine-ß-D-arabinofuranoside (AraC), an antimitotic agent known to suppress hypothalamic cell proliferation and neurogenesis, causes sleep-wake instability in young mice. The sleep-wake profiles were compared during baseline, during 4 weeks of artificial cerebrospinal fluid (aCSF) + 5-bromo-2'-deoxyuridine (BrdU) or AraC+BrdU infusion into the lateral ventricle, and 8 weeks after treatments. The sleep-wake architecture after AraC treatment was further compared with sleep-wake profiles in aged mice. Compared to aCSF+BrdU, 4 weeks of AraC+BrdU infusion significantly decreased (-96%) the number of BrdU+ cells around the third ventricular wall and adjacent preoptic-hypothalamic area and produced a) sleep disruption during the light phase with decreases in non-rapid eye movement (nonREM) (-9%) and REM sleep (-21%) amounts, and increased numbers of shorter (<2 min; 142 versus 98 episodes/12 h) and decreased numbers of longer (>5 min; 19 versus 26 episodes/12 h) nonREM sleep episodes; and b) wake disruption during the dark phase, with increased numbers of shorter (138 versus 91 episodes/12 h) and decreased numbers of longer active waking (17 versus 24 episodes/12 h) episodes. AraC-treated mice also exhibited lower delta activity within nonREM recovery sleep. The sleep-wake architecture of AraC-treated mice was similar to that observed in aged mice. These findings are consistent with a hypothesis that a decrease in hypothalamic cell proliferation/neurogenesis is detrimental to sleep-wake and circadian systems and may underlie sleep-wake disturbance in aging.


Assuntos
Envelhecimento/fisiologia , Proliferação de Células/fisiologia , Hipotálamo/fisiologia , Neurogênese/fisiologia , Sono/fisiologia , Vigília/fisiologia , Fatores Etários , Envelhecimento/efeitos dos fármacos , Animais , Antimitóticos/administração & dosagem , Antimitóticos/toxicidade , Proliferação de Células/efeitos dos fármacos , Ritmo Delta/efeitos dos fármacos , Ritmo Delta/fisiologia , Hipotálamo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/efeitos dos fármacos , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos
4.
BMJ Case Rep ; 20172017 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-28756378

RESUMO

We report on a 17-year-old girl with absence status epilepticus who developed recurrent motionless confusional state and continuous generalised 3-4 Hz rhythmic delta waves on electroencephalogram (EEG). The patient had no history of absence, myoclonus or generalised convulsion. Her seizure was resistant to a combination of antiepileptic drugs including carbamazepine. Ictal positron emission tomography using [18F]fluorodeoxyglucose ([18F]FDG-PET) revealed hypermetabolism of the bilateral thalamus and cerebellum and hypometabolism of the frontal, parietal and posterior cingulate cortices. We diagnosed her seizure as absence status and obtained remission by changing medication. The findings of ictal metabolic alteration in previous studies and in our case confirm the pathogenic importance of the thalamus in absence status and that associated cortical deactivation and cerebellar activation may be related to the generation or maintenance of epileptic EEG discharges.


Assuntos
Córtex Cerebral/fisiopatologia , Ritmo Delta/efeitos dos fármacos , Eletroencefalografia , Tomografia por Emissão de Pósitrons , Estado Epiléptico/diagnóstico por imagem , Tálamo/fisiopatologia , Adolescente , Anticonvulsivantes/uso terapêutico , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/efeitos dos fármacos , Clonazepam/uso terapêutico , Ritmo Delta/fisiologia , Feminino , Fluordesoxiglucose F18 , Humanos , Lamotrigina , Compostos Radiofarmacêuticos , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/fisiopatologia , Tálamo/diagnóstico por imagem , Tálamo/efeitos dos fármacos , Resultado do Tratamento , Triazinas/uso terapêutico
5.
Front Neural Circuits ; 11: 36, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28725184

RESUMO

Although general anesthetics are routinely administered to surgical patients to induce loss of consciousness, the mechanisms underlying anesthetic-induced unconsciousness are not fully understood. In rats, we characterized changes in the extradural EEG and intracranial local field potentials (LFPs) within the prefrontal cortex (PFC), parietal cortex (PC), and central thalamus (CT) in response to progressively higher doses of the inhaled anesthetic sevoflurane. During induction with a low dose of sevoflurane, beta/low gamma (12-40 Hz) power increased in the frontal EEG and PFC, PC and CT LFPs, and PFC-CT and PFC-PFC LFP beta/low gamma coherence increased. Loss of movement (LOM) coincided with an abrupt decrease in beta/low gamma PFC-CT LFP coherence. Following LOM, cortically coherent slow-delta (0.1-4 Hz) oscillations were observed in the frontal EEG and PFC, PC and CT LFPs. At higher doses of sevoflurane sufficient to induce loss of the righting reflex, coherent slow-delta oscillations were dominant in the frontal EEG and PFC, PC and CT LFPs. Dynamics similar to those observed during induction were observed as animals emerged from sevoflurane anesthesia. We conclude that the rat is a useful animal model for sevoflurane-induced EEG oscillations in humans, and that coherent slow-delta oscillations are a correlate of sevoflurane-induced behavioral arrest and loss of righting in rats.


Assuntos
Anestésicos Inalatórios/farmacologia , Ritmo Delta/efeitos dos fármacos , Éteres Metílicos/farmacologia , Lobo Parietal/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Animais , Ritmo beta/efeitos dos fármacos , Sincronização Cortical/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eletrodos Implantados , Ritmo Gama/efeitos dos fármacos , Masculino , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Lobo Parietal/fisiologia , Córtex Pré-Frontal/fisiologia , Ratos Sprague-Dawley , Reflexo de Endireitamento/efeitos dos fármacos , Reflexo de Endireitamento/fisiologia , Sevoflurano , Tálamo/fisiologia
6.
J Neurosci Methods ; 235: 83-91, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-25004203

RESUMO

BACKGROUND: The advent of optogenetics has given neuroscientists the opportunity to excite or inhibit neuronal population activity with high temporal resolution and cellular selectivity. Thus, when combined with recordings of neuronal ensemble activity in freely moving animals optogenetics can provide an unprecedented snapshot of the contribution of neuronal assemblies to (patho)physiological conditions in vivo. Still, the combination of optogenetic and silicone probe (or tetrode) recordings does not allow investigation of the role played by voltage- and transmitter-gated channels of the opsin-transfected neurons and/or other adjacent neurons in controlling neuronal activity. NEW METHOD AND RESULTS: We demonstrate that optogenetics and silicone probe recordings can be combined with intracerebral reverse microdialysis for the long-term delivery of neuroactive drugs around the optic fiber and silicone probe. In particular, we show the effect of antagonists of T-type Ca(2+) channels, hyperpolarization-activated cyclic nucleotide-gated channels and metabotropic glutamate receptors on silicone probe-recorded activity of the local opsin-transfected neurons in the ventrobasal thalamus, and demonstrate the changes that the block of these thalamic channels/receptors brings about in the network dynamics of distant somatotopic cortical neuronal ensembles. COMPARISON WITH EXISTING METHODS: This is the first demonstration of successfully combining optogenetics and neuronal ensemble recordings with reverse microdialysis. This combination of techniques overcomes some of the disadvantages that are associated with the use of intracerebral injection of a drug-containing solution at the site of laser activation. CONCLUSIONS: The combination of reverse microdialysis, silicone probe recordings and optogenetics can unravel the short and long-term effects of specific transmitter- and voltage-gated channels on laser-modulated firing at the site of optogenetic stimulation and the actions that these manipulations exert on distant neuronal populations.


Assuntos
Equipamentos e Provisões Elétricas , Microdiálise/métodos , Neurônios/fisiologia , Optogenética/métodos , Tálamo/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Ritmo Delta/efeitos dos fármacos , Ritmo Delta/fisiologia , Eletroencefalografia/instrumentação , Eletroencefalografia/métodos , Eletromiografia , Masculino , Microdiálise/instrumentação , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Vias Neurais/cirurgia , Neurônios/efeitos dos fármacos , Procedimentos Neurocirúrgicos , Optogenética/instrumentação , Ratos Wistar , Receptores de Glutamato Metabotrópico/metabolismo , Processamento de Sinais Assistido por Computador , Silicones , Tálamo/efeitos dos fármacos , Tálamo/cirurgia
7.
Acta Physiol Hung ; 99(3): 332-43, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22982721

RESUMO

Orexin A and orexin B are neuropeptides produced by a group of neurons located in the lateral hypothalamus which send widespread projections virtually to the whole neuraxis. Several studies indicated that orexins play a crucial role in the sleep-wake regulation and in the pathomechanism of the sleep disorder narcolepsy. As no data are available related to the EEG effects of orexin A in healthy, freely moving rats, the aim of the present experiments was to analyze EEG power changes in the generally used frequency bands after intracerebroventricular orexin A administration.Orexin A administration (0.84 and 2.8 nM/rat) differently affected fronto-occipital EEG waves in the different frequency bands recorded for 24 hours. Delta (1-4 Hz) and alpha (10-16 Hz) power decreased, while theta (4-10 Hz) and beta (16-48 Hz) power increased. Decrease of the delta power was followed by a rebound in case of the higher orexin A dose. This complex picture might be explained by the activation of several systems by the orexin A administration. Among these systems, cortical and thalamic circuits as well as the role of the neurons containing corticotrophin-releasing factor might be of significant importance.


Assuntos
Ritmo Circadiano/efeitos dos fármacos , Eletroencefalografia/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neuropeptídeos/farmacologia , Neuropeptídeos/fisiologia , Neurotransmissores/farmacologia , Ritmo alfa/efeitos dos fármacos , Animais , Ritmo beta/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Ritmo Delta/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Injeções Intraventriculares , Masculino , Atividade Motora/efeitos dos fármacos , Neurotransmissores/fisiologia , Orexinas , Ratos , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Tálamo/fisiologia , Ritmo Teta/efeitos dos fármacos
8.
CNS Neurosci Ther ; 18(8): 623-30, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22632633

RESUMO

AIMS: Safranal (2,6,6-trimethyl-1,3-cyclohexadiene-1-carboxaldehyde, C(10) H(14) O) is an active ingredient in the saffron, which is used in traditional medicine. It has been reported to have sedative and anti-epileptic effects, but its hypnotic effects remain uncertain. The aim of this study was to evaluate effects of safranal on sleep-wake cycle. METHODS: We established hypnotic-model mice treated with a low dose of pentobarbital 20 mg/kg, and administered different doses of safranal, vehicle, or diazepam. The change of sleep-wake cycle was assessed by sleep recording and c-Fos expression in the brain was analyzed by immunohistochemistry. RESULTS: Safranal increased the duration of non-rapid eye movement (NREM) sleep, shortened NREM sleep latency, and enhanced the delta power activity of NREM sleep. Immunohistochemical evaluation revealed that safranal increased c-Fos expression in the ventrolateral preoptic nucleus (VLPO), one of the putative sleep centers, and decreased it in the arousal histaminergic tuberomammillary nuclei (TMN). CONCLUSION: These findings indicate that safranal enhances NREM sleep in pentobarbital-treated mice. The hypnotic effects of safranal may be related to the activation of the sleep-promoting neurons in the VLPO and the simultaneous inhibition of the wakefulness-promoting neurons in the TMN, suggesting that safranal may be a hypnotic substance.


Assuntos
Cicloexenos/farmacologia , Hipnóticos e Sedativos/farmacologia , Pentobarbital/farmacologia , Sono/efeitos dos fármacos , Terpenos/farmacologia , Animais , Nível de Alerta/efeitos dos fármacos , Ritmo Delta/efeitos dos fármacos , Eletroencefalografia , Eletromiografia , Região Hipotalâmica Lateral/efeitos dos fármacos , Região Hipotalâmica Lateral/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Polissonografia , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fases do Sono/efeitos dos fármacos
9.
Schizophr Res ; 138(2-3): 206-11, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22520856

RESUMO

It has recently been shown that treatment with long-chain omega-3 polyunsaturated fatty acids (PUFAs) could decrease the rate of transition to psychosis, and improve psychiatric symptoms and global functioning in people at ultra-high risk (UHR) for psychosis. Previous studies have suggested that resting state brain activity measured with electroencephalography (EEG) may represent an objective biomarker of changes in neural function associated with supplementation with omega-3 PUFAs. It has also been proposed that although resting state EEG cannot, by itself, predict transition to psychosis in UHR individuals, the combination of resting state EEG with negative symptoms may be a valid predictor of transition. The present study investigated whether treatment with omega-3 PUFAs influenced resting state EEG in UHR participants, and whether or not the association of the participants' resting state EEG with their levels of negative symptoms was dependent on their transition status. The brain activity of 73 UHR participants was recorded in the context of a randomized, placebo-controlled trial of the effects of supplementation with omega-3 PUFAs. The UHR participants who subsequently transitioned to psychosis (UHR+) did not differ from those who did not transition (UHR-) in terms of resting state EEG power in any frequency band. However, negative symptom scores were associated with increased delta activity in the frontal region of the UHR+ participants, but not in the UHR- participants. Treatment with omega-3 PUFAs did not induce changes in resting state EEG in either group. The results suggest that decreased frontal delta activity, in combination with high levels of negative symptoms, may be a risk factor for subsequent transition to psychosis in UHR individuals.


Assuntos
Ritmo Delta/efeitos dos fármacos , Ácidos Graxos Ômega-3/uso terapêutico , Transtornos Psicóticos/prevenção & controle , Transtornos Psicóticos/fisiopatologia , Adolescente , Progressão da Doença , Eletroencefalografia , Feminino , Humanos , Masculino , Fatores de Risco , Adulto Jovem
10.
J Neurophysiol ; 107(11): 3181-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22423006

RESUMO

Work on schizophrenia demonstrates the involvement of the hippocampus in the disease and points specifically to hyperactivity of CA1. Many symptoms of schizophrenia can be mimicked by N-methyl-d-aspartate receptor (NMDAR) antagonist; notably, delta frequency oscillations in the awake state are enhanced in schizophrenia, an abnormality that can be mimicked by NMDAR antagonist action in the thalamus. Given that CA1 receives input from the nucleus reuniens of the thalamus, we sought to determine whether an NMDAR antagonist in the thalamus can affect hippocampal processes. We found that a systemic NMDAR antagonist (ketamine; 50 mg/kg) increased the firing rate of cells in the reuniens and CA1 in awake rats. Furthermore, ketamine increased the power of delta oscillations in both structures. The thalamic origin of the change in hippocampal properties was demonstrated in three ways: 1) oscillations in the two structures were coherent; 2) the hippocampal changes induced by systematic ketamine were reduced by thalamic injection of muscimol; and 3) the hippocampal changes could be induced by local injection of ketamine into the thalamus. Lower doses of ketamine (20 mg/kg) did not evoke delta oscillations but did increase hippocampal gamma power, an effect not dependent on the thalamus. There are thus at least two mechanisms for ketamine action on the hippocampus: a low-dose mechanism that affects gamma through a nonthalamic mechanism and a high-dose mechanism that increases CA1 activity and delta oscillations as a result of input from the thalamus. Both mechanisms may be important in producing symptoms of schizophrenia.


Assuntos
Ritmo Delta/fisiologia , Hipocampo/fisiologia , Ketamina/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/fisiologia , Tálamo/fisiologia , Animais , Ritmo Delta/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Ketamina/toxicidade , Masculino , Ratos , Ratos Long-Evans , Esquizofrenia/induzido quimicamente , Tálamo/efeitos dos fármacos
11.
Behav Neurol ; 25(1): 3-11, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22207418

RESUMO

BACKGROUND: Visual delta event-related (ERO) and evoked oscillations (EO) of Alzheimer patients (AD) are different than healthy. In the present study, the analysis is extented to include auditory ERO and EO in AD. The rationale is to reveal whether the auditory ERO delta responses are also reduced, and whether this is a general phenomenon in Alzheimer patients upon applying stimuli with cognitive load. METHODS: Thirty-four mild AD subjects [17 de-novo and 17 medicated (cholinergic)] and seventeen healthy controls were included. Auditory oddball paradigm and sensory auditory stimuli were applied to the subjects. Oscillatory responses were analyzed by measuring maximum amplitudes in delta frequency range (0.5-3.5 Hz). RESULTS: Auditory delta ERO (0.5-3.5 Hz) responses of healthy controls were higher than either de-novo AD or medicated AD group, without a difference between two AD subgroups. Furthermore, the auditory EO after presentation of tone bursts yielded no group difference. CONCLUSION: Our findings imply that delta ERO is highly unstable in AD patients in comparison to age-matched healthy controls only during the cognitive paradigm. Our results favor the hypothesis that neural delta networks are activated during cognitive tasks and that the reduced delta response is a general phenomenon in AD, due to cognitive impairment.


Assuntos
Doença de Alzheimer/fisiopatologia , Ritmo Delta/fisiologia , Potenciais Evocados Auditivos/fisiologia , Estimulação Acústica/métodos , Estimulação Acústica/psicologia , Idoso , Doença de Alzheimer/tratamento farmacológico , Inibidores da Colinesterase/farmacologia , Inibidores da Colinesterase/uso terapêutico , Ritmo Delta/efeitos dos fármacos , Potenciais Evocados Auditivos/efeitos dos fármacos , Feminino , Humanos , Masculino , Testes Neuropsicológicos/estatística & dados numéricos , Desempenho Psicomotor/efeitos dos fármacos , Desempenho Psicomotor/fisiologia
12.
Nat Neurosci ; 13(1): 9-17, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19966841

RESUMO

The slow (<1 Hz) rhythm, the most important electroencephalogram (EEG) signature of non-rapid eye movement (NREM) sleep, is generally viewed as originating exclusively from neocortical networks. Here we argue that the full manifestation of this fundamental sleep oscillation in a corticothalamic module requires the dynamic interaction of three cardinal oscillators: one predominantly synaptically based cortical oscillator and two intrinsic, conditional thalamic oscillators. The functional implications of this hypothesis are discussed in relation to other EEG features of NREM sleep, with respect to coordinating activities in local and distant neuronal assemblies and in the context of facilitating cellular and network plasticity during slow-wave sleep.


Assuntos
Córtex Cerebral/fisiologia , Ritmo Delta , Fases do Sono/fisiologia , Tálamo/fisiologia , Potenciais de Ação/efeitos dos fármacos , Anestésicos/farmacologia , Animais , Relógios Biológicos , Mapeamento Encefálico , Gatos , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Ritmo Delta/efeitos dos fármacos , Humanos , Técnicas In Vitro , Modelos Biológicos , Rede Nervosa/fisiologia , Neurônios/fisiologia , Tálamo/citologia , Tálamo/efeitos dos fármacos
13.
Neuropharmacology ; 57(4): 369-74, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19615388

RESUMO

AMP-activated protein kinase (AMPK) is an energy-sensing molecular signal involved in glucose and lipid metabolism. The known interaction of sleep with energy metabolism led us to investigate the role of central AMPK in sleep homeostasis. Sleep deprivation (SD) for 6 h increased p-AMPK protein in the hypothalamus and also increased the mRNA level of Ca(2+)/calmodulin (CaM)-dependent protein kinase kinase beta (CaMKK2), an activator of AMPK, and carnitine palmitoyltransferase 1 (CPT1), a downstream signaling factor of AMPK. Central injection of compound C (CC), an inhibitor of AMPK, suppressed EEG delta power during NREM sleep, while 5-aminoimidazole-4-carboxamide riboside (AICAR), an activator of AMPK, enhanced EEG delta power. The treatment of both CC and AICAR attenuated rebound responses of delta power in NREM sleep after SD. These results indicate that central AMPK is involved in the regulation of sleep depth and sleep homeostasis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Encéfalo/fisiologia , Homeostase/fisiologia , Sono/fisiologia , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Fármacos do Sistema Nervoso Central/farmacologia , Ritmo Delta/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Hipotálamo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , RNA Mensageiro/metabolismo , Ribonucleotídeos/farmacologia , Sono/efeitos dos fármacos , Privação do Sono/fisiopatologia
14.
Biol Psychiatry ; 66(2): 177-84, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19058790

RESUMO

BACKGROUND: K-complexes (KCs) are evoked delta frequency electroencephalogram (EEG) responses during sleep that occur when large numbers of healthy cortical cells burst fire in a synchronized manner. The KC amplitude and incidence are sensitive measures of normal healthy brain aging. Given the known neurodegenerative consequences of alcohol abuse it was hypothesized that alcoholism would be associated with further KC amplitude and incidence reductions. METHODS: Eighty-four subjects (42 alcoholics) screened for medical, psychiatric, and sleep problems participated. The protocol involved the presentation of auditory stimuli during stage 2 sleep throughout a night in the laboratory. The KCs were identified and averaged, to enable measurement of the P2, N550, and P900 peaks. RESULTS: Compared with control subjects, alcoholic men and women had lower KC incidence (p < .001) and P2 (p < .001), N550 (p < .05), and P900 (p < .05) amplitudes. There was a significant diagnosis x site interaction (p < .001), indicating the group difference was largest at frontal sites. Longer sobriety correlated with increased N550 amplitude (p < .01). CONCLUSIONS: The KC incidence and amplitude were negatively impacted in alcoholic men and women with exacerbation of the normal aging effects, particularly over frontal scalp regions. The observed relationship between improvements in KC measures and increased time of abstinence suggests that these measures might provide a useful marker of brain recovery with continued abstinence from alcohol.


Assuntos
Alcoolismo/fisiopatologia , Ritmo Delta/efeitos dos fármacos , Sono/efeitos dos fármacos , Estimulação Acústica , Adulto , Eletroencefalografia , Potenciais Evocados Auditivos/fisiologia , Feminino , Lobo Frontal/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Escalas de Graduação Psiquiátrica , Temperança
15.
Endocrinology ; 149(10): 5262-71, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18787029

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors belonging to the nuclear receptor family. PPARs play a critical role in lipid and glucose metabolism. We examined whether chronic treatment with bezafibrate, a PPAR agonist, would alter sleep and body temperature (BT). Mice fed with a control diet were monitored for BT, electroencephalogram (EEG), and electromyogram for 48 h under light-dark conditions. After obtaining the baseline recording, the mice were provided with bezafibrate-supplemented food for 2 wk, after which the same recordings were performed. Two-week feeding of bezafibrate decreased BT, especially during the latter half of the dark period. BT rhythm and sleep/wake rhythm were phase advanced about 2-3 h by bezafibrate treatment. Bezafibrate treatment also increased the EEG delta-power in nonrapid eye movement sleep compared with the control diet attenuating its daily amplitude. Furthermore, bezafibrate-treated mice showed no rebound of EEG delta-power in nonrapid eye movement sleep after 6 h sleep deprivation, whereas values in control mice largely increased relative to baseline. DNA microarray, and real-time RT-PCR analysis showed that bezafibrate treatment increased levels of Neuropeptide Y mRNA in the hypothalamus at both Zeitgeber time (ZT) 10 and ZT22, and decreased proopiomelanocortin-alpha mRNA in the hypothalamus at ZT10. These findings demonstrate that PPARs participate in the control of both BT and sleep regulation, which accompanied changes in gene expression in the hypothalamus. Activation of PPARs may enhance deep sleep and improve resistance to sleep loss.


Assuntos
Bezafibrato/farmacologia , Temperatura Corporal/efeitos dos fármacos , Ritmo Delta/efeitos dos fármacos , Hipolipemiantes/farmacologia , Fases do Sono/efeitos dos fármacos , Animais , Regulação da Temperatura Corporal/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos ICR , Neuropeptídeo Y/genética , Neuropeptídeo Y/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Pró-Opiomelanocortina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vigília/efeitos dos fármacos
16.
Biol Pharm Bull ; 30(2): 363-6, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17268081

RESUMO

In the present study, we studied the effect of valerian extract preparation (BIM) containing valerian extract, golden root (Rhodiola rosea L.) extract and L-theanine (gamma-glutamylethylamide) on the sleep-wake cycle using sleep-disturbed model rats in comparison with that of valerian extract. A significant shortening in sleep latency was observed with valerian extract and the BIM at a dose of 1000 mg/kg. On the other hand, valerian extract and the BIM caused no significant effects on total times of wakefulness, non-rapid eye movement (non-REM) sleep and REM sleep. Valerian extract and the BIM at a dose of 1000 mg/kg also had no significant effect on delta activity. In conclusion, it became clear that the BIM could be useful as a herbal medicine having a sleep-inducing effect without causing an alteration of the sleep-wakefulness cycle.


Assuntos
Glutamatos/farmacologia , Hipnóticos e Sedativos/farmacologia , Rhodiola/química , Distúrbios do Início e da Manutenção do Sono/tratamento farmacológico , Valeriana/química , Animais , Ritmo Delta/efeitos dos fármacos , Eletromiografia , Masculino , Extratos Vegetais/farmacologia , Raízes de Plantas/química , Ratos , Ratos Wistar , Sono/efeitos dos fármacos , Distúrbios do Início e da Manutenção do Sono/fisiopatologia , Vigília/efeitos dos fármacos
17.
Acta Med Okayama ; 59(3): 89-92, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16049561

RESUMO

The present study was performed to investigate the effects of valerian extract on the sleep-wake cycle using sleep-disturbed model rats. A significant shortening in sleep latency was observed with valerian extract at doses of 1000 and 3000 mg/kg. On the other hand, valerian extract had no significant effects on total times of wakefulness, non-rapid eye movement (non-REM) sleep, or REM sleep, even at a dose of 3000 mg/kg. Valerian extract at doses of 1000 and 3000 mg/kg showed a significant increase in the delta activity during non-REM sleep. In conclusion, valerian extract may be useful as an herbal medicine having not only sleep-inducing effects but also sleep quality-enhancement effects.


Assuntos
Fitoterapia , Extratos Vegetais/farmacologia , Distúrbios do Início e da Manutenção do Sono/tratamento farmacológico , Sono REM/efeitos dos fármacos , Valeriana , Vigília/efeitos dos fármacos , Animais , Ritmo Delta/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar
18.
Homeopathy ; 94(2): 86-91, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15892488

RESUMO

Histamine is a central neurotransmitter, it increases arousal via H1 receptors. This study examines the effect of ultra-diluted histamine on arousal through changes in the sleep pattern of Wistar rats. The spectral density in delta (0.5-2.5 Hz) band, one of the three major spectral components of the sleep-electroencephalogram, was analyzed against time. Rats were randomized to receive histamine 30c (histamine 30c, 0.05 ml every 20 min during the first 2 h orally), histamine intraperitoneal pre-treatment/histamine 30c (histamine 6mg/kg i.p., followed by histamine 30c) or solvent control. The mean delta band spectral density was lower in the histamine 30c and histamine pretreatment/histamine 30c groups than the control group. Significant differences between histamine 30c and baseline during the first 2 h imply an immediate effect. These results also suggest a dynamic process in which the system spontaneously evolves between two locally stationary states according to a power law. From the time perspective, the system approaches, asymptotically, an equifinal state.


Assuntos
Nível de Alerta/efeitos dos fármacos , Ritmo Delta/efeitos dos fármacos , Histamina/farmacologia , Fases do Sono/efeitos dos fármacos , Administração Oral , Análise de Variância , Animais , Relação Dose-Resposta a Droga , Histamina/administração & dosagem , Masculino , Distribuição Aleatória , Ratos , Ratos Wistar , Fatores de Tempo
19.
Vet Hum Toxicol ; 37(6): 521-7, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8588286

RESUMO

A minimally-invasive method of quantitative electroencephalography (qEEG) that requires no anesthetics and parallels techniques of naturalistic stimulation was developed and validated for regulatory testing of drugs and chemicals in rats. Male and female Fischer 344 rats were utilized in a randomized-block design to measure qEEG target parameters associated with a range of cholinesterase inhibition. For this study, physostigmine was administered ip at doses of 0.05, 0.2 or 1.0 mg/kg, resulting in average cholinesterase inhibition in plasma (28, 38 and 70%), erythrocytes (19, 24 and 36%), and brain (2, 10 and 31%) which correlated well with increased total power and amplitude changes. Additional treatment-related effects consisted of decreased relative alpha and beta, increased relative delta, and a left-shift in the spectral-edge frequency. In a second study, male and female Sprague-Dawley rats were utilized in a treatment-by-subjects design to determine qEEG target parameter changes due to the M2 autoreceptor agonist oxotremorine. Repeated incremental doses (0.05, 0.1, 0.2 mg/kg; ip) of oxotremorine resulted in increased beta contribution, a right-shift in the spectral-edge frequency and decreased alpha contribution. These qEEG results with physostigmine and oxotremorine correlate well with receptor-specific and general muscarinic effects, making it a reliable contribution to analysis of agonist and antagonist effects of cholinergic compounds.


Assuntos
Encéfalo/efeitos dos fármacos , Inibidores da Colinesterase/toxicidade , Agonistas Muscarínicos/toxicidade , Oxotremorina/toxicidade , Fisostigmina/toxicidade , Ritmo alfa/efeitos dos fármacos , Animais , Ritmo beta/efeitos dos fármacos , Encéfalo/enzimologia , Inibidores da Colinesterase/administração & dosagem , Inibidores da Colinesterase/metabolismo , Colinesterases/sangue , Simulação por Computador , Ritmo Delta/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Eletroencefalografia , Eritrócitos/efeitos dos fármacos , Eritrócitos/enzimologia , Feminino , Injeções Intraperitoneais , Masculino , Agonistas Muscarínicos/administração & dosagem , Agonistas Muscarínicos/metabolismo , Oxotremorina/administração & dosagem , Oxotremorina/metabolismo , Fisostigmina/administração & dosagem , Fisostigmina/metabolismo , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley
20.
Anesthesiology ; 78(3): 498-509, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8457051

RESUMO

BACKGROUND: The N1 and P3 waves of the auditory event-related potential provide information on consciousness and cortical function. The N1 wave is reduced during states of low vigilance. The P3 wave occurs only for stimuli that somehow capture the subject's attention. There are two types of P3:P3a and P3b. The P3a predominates frontally and probably occurs when the subject simply notices the stimulus. The P3b predominates parietally and indicates conscious awareness of the evoking stimulus. The N1 and P3 were recorded in 12 patients during cardiac surgery under sufentanil anesthesia to search for unintentional awareness. The study was limited to the period before cardiopulmonary bypass. METHODS: After premedication with diazepam, morphine, and scopolamine, sufentanil was used for induction (mean dose, 7.9 micrograms/kg) and maintenance (4 micrograms/kg) of anesthesia. No other anesthetics were administered. Recordings were obtained before induction, during induction after loss of consciousness, after tracheal intubation before incision, and before cardiopulmonary bypass. RESULTS: The N1 was attenuated significantly by sufentanil but was not abolished. The P3b occurred only during preinduction. There was no P3 during induction. There was a P3a during postintubation and precardiopulmonary bypass. CONCLUSIONS: The attenuation of N1 from induction onward reflects a decrease in the level of arousal caused by sufentanil. A P3a during postintubation and precardiopulmonary bypass indicates that pitch discrimination at the cortical level occurs but does not prove that conscious awareness has occurred. Whether or not the P3a reflects the regaining of consciousness is not known.


Assuntos
Anestesia Intravenosa , Atenção/efeitos dos fármacos , Conscientização/efeitos dos fármacos , Procedimentos Cirúrgicos Cardíacos , Estado de Consciência/efeitos dos fármacos , Potenciais Evocados Auditivos/efeitos dos fármacos , Sufentanil/farmacologia , Estimulação Acústica , Atenção/fisiologia , Conscientização/fisiologia , Ponte Cardiopulmonar , Estado de Consciência/fisiologia , Ritmo Delta/efeitos dos fármacos , Eletroencefalografia/efeitos dos fármacos , Potenciais Evocados Auditivos/fisiologia , Feminino , Humanos , Intubação Intratraqueal , Masculino , Memória/efeitos dos fármacos , Memória/fisiologia , Pessoa de Meia-Idade , Tempo de Reação , Sufentanil/administração & dosagem , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA