Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
MethodsX ; 9: 101811, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36065218

RESUMO

Studying the biology of sleep requires accurate and efficient assessment of the sleep stages. However, analysis of sleep-wake cycles in mice and other laboratory animals remains a time-consuming and laborious process. In this study, we developed a Python script and a process for the streamlined analysis of sleep data that includes real-time processing of electroencephalogram (EEG) and electromyogram (EMG) signals that is compatible with commercial sleep-recording software that supports user datagram protocol (UDP) communication. The process consists of EEG/EMG data acquisition, automated threshold calculation for real-time determination of sleep stages, sleep staging and EEG power spectrum analysis. It also allows data storage in the format that facilitates further analysis of the sleep pattern in mice. The described method is aimed at increasing efficiency of sleep stage scoring and analysis in mice thus facilitating sleep research. • A process of EEG/EMG recording and streamline analysis of sleep-wake cycle in real time in mice. • The compatibility with commercial sleep-recording software that can generate a UDP stream. • The capability of further analysis of recorded data by an open-source software.

2.
Sci Rep ; 12(1): 12278, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35853986

RESUMO

Non-invasive stimulation technologies are emerging as potential treatment options for a range of neurodegenerative disorders. Experimental evidence suggests that stimuli-evoked changes in slow brain rhythms may mitigate or even prevent neuropathological and behavioral impairments. Slow wave activity is prevalent during sleep and can be triggered non-invasively by sensory stimulation targeting the visual system or directly via activation of neurons locally using optogenetics. Here, we developed new tools for delivering visual stimulation using light-emitting diodes in freely moving mice while awake and during sleep. We compared these tools to traditional optogenetic approaches used for local stimulation of neurons in the cerebral cortex. We then used these tools to compare the effects of low-frequency visual versus optogenetic stimulations on the slow wave activity and sleep pattern in mice. Visual stimulation effectively enhanced slow wave activity without disrupting the sleep pattern. Optogenetic stimulation of cortical GABAergic neurons increased NREM sleep. These results suggest that visual stimulation can be effective at boosting slow wave activity without having adverse effects on sleep and thus holds great potential as a non-invasive stimulation treatment strategy.


Assuntos
Sono de Ondas Lentas , Animais , Eletroencefalografia , Camundongos , Optogenética , Estimulação Luminosa , Sono/fisiologia , Sono de Ondas Lentas/fisiologia , Vigília/fisiologia
3.
Nat Methods ; 19(7): 871-880, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35681062

RESUMO

Regulation of receptor tyrosine kinase (RTK) activity is necessary for studying cell signaling pathways in health and disease. We developed a generalized approach for engineering RTKs optically controlled with far-red light. We targeted the bacterial phytochrome DrBphP to the cell surface and allowed its light-induced conformational changes to be transmitted across the plasma membrane via transmembrane helices to intracellular RTK domains. Systematic optimization of these constructs has resulted in optically regulated epidermal growth factor receptor, HER2, TrkA, TrkB, FGFR1, IR1, cKIT and cMet, named eDrRTKs. eDrRTKs induced downstream signaling in mammalian cells in tens of seconds. The ability to activate eDrRTKs with far-red light enabled spectral multiplexing with fluorescent probes operating in a shorter spectral range, allowing for all-optical assays. We validated eDrTrkB performance in mice and found that minimally invasive stimulation in the neocortex with penetrating via skull far-red light-induced neural activity, early immediate gene expression and affected sleep patterns.


Assuntos
Fitocromo , Animais , Membrana Celular/metabolismo , Luz , Mamíferos , Camundongos , Transdução de Sinais
4.
Curr Biol ; 30(12): 2379-2385.e4, 2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32413301

RESUMO

The ability to rapidly arouse from sleep is important for survival. However, increased arousals in patients with sleep apnea and other disorders prevent restful sleep and contribute to cognitive, metabolic, and physiologic dysfunction [1, 2]. Little is currently known about which neural systems mediate these brief arousals, hindering the development of treatments that restore normal sleep. The basal forebrain (BF) receives inputs from many nuclei of the ascending arousal system, including the brainstem parabrachial neurons, which promote arousal in response to elevated blood carbon dioxide levels, as seen in sleep apnea [3]. Optical inhibition of the terminals of parabrachial neurons in the BF impairs cortical arousals to hypercarbia [4], but which BF cell types mediate cortical arousals in response to hypercarbia or other sensory stimuli is unknown. Here, we tested the role of BF parvalbumin (PV) neurons in arousal using optogenetic techniques in mice. Optical stimulation of BF-PV neurons produced rapid transitions to wakefulness from non-rapid eye movement (NREM) sleep but did not affect REM-wakefulness transitions. Unlike previous studies of BF glutamatergic and cholinergic neurons, arousals induced by stimulation of BF-PV neurons were brief and only slightly increased total wake time, reminiscent of clinical findings in sleep apnea [5, 6]. Bilateral optical inhibition of BF-PV neurons increased the latency to arousal produced by exposure to hypercarbia or auditory stimuli. Thus, BF-PV neurons are an important component of the brain circuitry that generates brief arousals from sleep in response to stimuli, which may indicate physiological dysfunction or danger to the organism.


Assuntos
Estimulação Acústica , Nível de Alerta/fisiologia , Carboidratos/administração & dosagem , Neurônios/fisiologia , Ração Animal/análise , Animais , Prosencéfalo Basal/fisiologia , Dieta , Camundongos , Parvalbuminas/metabolismo , Sono/fisiologia , Vigília/fisiologia
5.
Sci Rep ; 9(1): 3607, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30837664

RESUMO

The thalamic reticular nucleus (TRN) is implicated in schizophrenia pathology. However, it remains unclear whether alterations of TRN activity can account for abnormal electroencephalographic activity observed in patients, namely reduced spindles (10-15 Hz) during sleep and increased delta (0.5-4 Hz) and gamma-band activity (30-80 Hz) during wakefulness. Here, we utilized optogenetic and reverse-microdialysis approaches to modulate activity of the major subpopulation of TRN GABAergic neurons, which express the calcium-binding protein parvalbumin (PV), and are implicated in schizophrenia dysfunction. An automated algorithm with enhanced efficiency and reproducibility compared to manual detection was used for sleep spindle assessment. A novel, low power, waxing-and-waning optogenetic stimulation paradigm preferentially induced spindles that were indistinguishable from spontaneously occurring sleep spindles without altering the behavioral state, when compared to a single pulse laser stimulation used by us and others. Direct optogenetic inhibition of TRN-PV neurons was ineffective in blocking spindles but increased both wakefulness and cortical delta/gamma activity, as well as impaired the 40 Hz auditory steady-state response. For the first time we demonstrate that spindle density is markedly reduced by (i) optogenetic stimulation of a major GABA/PV inhibitory input to TRN arising from basal forebrain parvalbumin neurons (BF-PV) and; (ii) localized pharmacological inhibition of low-threshold calcium channels, implicated as a genetic risk factor for schizophrenia. Together with clinical findings, our results support impaired TRN-PV neuron activity as a potential cause of schizophrenia-linked abnormalities in cortical delta, gamma, and spindle activity. Modulation of the BF-PV input to TRN may improve these neural abnormalities.


Assuntos
Neurônios GABAérgicos/fisiologia , Parvalbuminas/metabolismo , Esquizofrenia/fisiopatologia , Sono/fisiologia , Núcleos Talâmicos/fisiologia , Vigília/fisiologia , Animais , Fenômenos Eletrofisiológicos , Camundongos , Optogenética
6.
Sleep ; 42(2)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30476300

RESUMO

Study Objectives: Sleep spindles are abnormal in several neuropsychiatric conditions and have been implicated in associated cognitive symptoms. Accordingly, there is growing interest in elucidating the pathophysiology behind spindle abnormalities using rodent models of such disorders. However, whether sleep spindles can reliably be detected in mouse electroencephalography (EEG) is controversial necessitating careful validation of spindle detection and analysis techniques. Methods: Manual spindle detection procedures were developed and optimized to generate an algorithm for automated detection of events from mouse cortical EEG. Accuracy and external validity of this algorithm were then assayed via comparison to sigma band (10-15 Hz) power analysis, a proxy for sleep spindles, and pharmacological manipulations. Results: We found manual spindle identification in raw mouse EEG unreliable, leading to low agreement between human scorers as determined by F1-score (0.26 ± 0.07). Thus, we concluded it is not possible to reliably score mouse spindles manually using unprocessed EEG data. Manual scoring from processed EEG data (filtered, cubed root-mean-squared), enabled reliable detection between human scorers, and between human scorers and algorithm (F1-score > 0.95). Algorithmically detected spindles correlated with changes in sigma-power and were altered by the following conditions: sleep-wake state changes, transitions between NREM and REM sleep, and application of the hypnotic drug zolpidem (10 mg/kg, intraperitoneal). Conclusions: Here we describe and validate an automated paradigm for rapid and reliable detection of spindles from mouse EEG recordings. This technique provides a powerful tool to facilitate investigations of the mechanisms of spindle generation, as well as spindle alterations evident in mouse models of neuropsychiatric disorders.


Assuntos
Ondas Encefálicas/fisiologia , Eletroencefalografia/métodos , Sono REM/fisiologia , Sono de Ondas Lentas/fisiologia , Algoritmos , Animais , Bioensaio , Coleta de Dados , Feminino , Humanos , Hipnóticos e Sedativos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Registros , Zolpidem/farmacologia
7.
Neuron ; 96(5): 1153-1167.e5, 2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-29103805

RESUMO

The precise neural circuitry that mediates arousal during sleep apnea is not known. We previously found that glutamatergic neurons in the external lateral parabrachial nucleus (PBel) play a critical role in arousal to elevated CO2 or hypoxia. Because many of the PBel neurons that respond to CO2 express calcitonin gene-related peptide (CGRP), we hypothesized that CGRP may provide a molecular identifier of the CO2 arousal circuit. Here, we report that selective chemogenetic and optogenetic activation of PBelCGRP neurons caused wakefulness, whereas optogenetic inhibition of PBelCGRP neurons prevented arousal to CO2, but not to an acoustic tone or shaking. Optogenetic inhibition of PBelCGRP terminals identified a network of forebrain sites under the control of a PBelCGRP switch that is necessary to arouse animals from hypercapnia. Our findings define a novel cellular target for interventions that may prevent sleep fragmentation and the attendant cardiovascular and cognitive consequences seen in obstructive sleep apnea. VIDEO ABSTRACT.


Assuntos
Nível de Alerta/genética , Hipercapnia/genética , Hipercapnia/fisiopatologia , Sono/genética , Estimulação Acústica , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Dióxido de Carbono/metabolismo , Dióxido de Carbono/farmacologia , Eletroencefalografia , Eletromiografia , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/fisiopatologia , Neurônios , Optogenética , Técnicas de Patch-Clamp , Prosencéfalo/fisiopatologia , Respiração , Síndromes da Apneia do Sono/fisiopatologia
9.
Curr Opin Neurobiol ; 44: 159-166, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28538168

RESUMO

The diverse cell-types of the basal forebrain control sleep-wake states, cortical activity and reward processing. Large, slow-firing, cholinergic neurons suppress cortical delta activity and promote cortical plasticity in response to reinforcers. Large, fast-firing, cortically-projecting GABAergic neurons promote wakefulness and fast cortical activity. In particular, parvalbumin/GABAergic neurons promote neocortical gamma band activity. Conversely, excitation of slower-firing somatostatin/GABAergic neurons promotes sleep through inhibition of cortically-projecting neurons. Activation of glutamatergic neurons promotes wakefulness, likely by exciting other cortically-projecting neurons. Similarly, cholinergic neurons indirectly promote wakefulness by excitation of wake-promoting, cortically-projecting GABAergic neurons and/or inhibition of sleep-promoting somatostatin/GABAergic neurons. Both glia and neurons increase the levels of adenosine during prolonged wakefulness. Adenosine presynaptically inhibits glutamatergic inputs to wake-promoting cholinergic and GABAergic/parvalbumin neurons, promoting sleep.


Assuntos
Prosencéfalo Basal/citologia , Prosencéfalo Basal/fisiologia , Neurônios/fisiologia , Animais , Humanos , Neurônios/citologia , Sono/fisiologia , Vigília/fisiologia
10.
Proc Natl Acad Sci U S A ; 112(11): 3535-40, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25733878

RESUMO

Cortical gamma band oscillations (GBO, 30-80 Hz, typically ∼40 Hz) are involved in higher cognitive functions such as feature binding, attention, and working memory. GBO abnormalities are a feature of several neuropsychiatric disorders associated with dysfunction of cortical fast-spiking interneurons containing the calcium-binding protein parvalbumin (PV). GBO vary according to the state of arousal, are modulated by attention, and are correlated with conscious awareness. However, the subcortical cell types underlying the state-dependent control of GBO are not well understood. Here we tested the role of one cell type in the wakefulness-promoting basal forebrain (BF) region, cortically projecting GABAergic neurons containing PV, whose virally transduced fibers we found apposed cortical PV interneurons involved in generating GBO. Optogenetic stimulation of BF PV neurons in mice preferentially increased cortical GBO power by entraining a cortical oscillator with a resonant frequency of ∼40 Hz, as revealed by analysis of both rhythmic and nonrhythmic BF PV stimulation. Selective saporin lesions of BF cholinergic neurons did not alter the enhancement of cortical GBO power induced by BF PV stimulation. Importantly, bilateral optogenetic inhibition of BF PV neurons decreased the power of the 40-Hz auditory steady-state response, a read-out of the ability of the cortex to generate GBO used in clinical studies. Our results are surprising and novel in indicating that this presumptively inhibitory BF PV input controls cortical GBO, likely by synchronizing the activity of cortical PV interneurons. BF PV neurons may represent a previously unidentified therapeutic target to treat disorders involving abnormal GBO, such as schizophrenia.


Assuntos
Prosencéfalo Basal/fisiologia , Ritmo Gama/fisiologia , Neurônios/fisiologia , Parvalbuminas/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Channelrhodopsins , Neurônios Colinérgicos/fisiologia , Potenciais Evocados Auditivos/fisiologia , Proteínas Luminescentes/metabolismo , Camundongos , Optogenética , Reprodutibilidade dos Testes , Transdução Genética
11.
PLoS One ; 4(7): e6346, 2009 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-19623260

RESUMO

Ten years ago the sleep disorder narcolepsy was linked to the neuropeptide hypocretin (HCRT), also known as orexin. This disorder is characterized by excessive day time sleepiness, inappropriate triggering of rapid-eye movement (REM) sleep and cataplexy, which is a sudden loss of muscle tone during waking. It is still not known how HCRT regulates REM sleep or muscle tone since HCRT neurons are localized only in the lateral hypothalamus while REM sleep and muscle atonia are generated from the brainstem. To identify a potential neuronal circuit, the neurotoxin hypocretin-2-saporin (HCRT2-SAP) was used to lesion neurons in the ventral lateral periaquaductal gray (vlPAG). The first experiment utilized hypocretin knock-out (HCRT-ko) mice with the expectation that deletion of both HCRT and its target neurons would exacerbate narcoleptic symptoms. Indeed, HCRT-ko mice (n = 8) given the neurotoxin HCRT2-SAP (16.5 ng/23nl/sec each side) in the vlPAG had levels of REM sleep and sleep fragmentation that were considerably higher compared to HCRT-ko given saline (+39%; n = 7) or wildtype mice (+177%; n = 9). However, cataplexy attacks did not increase, nor were levels of wake or non-REM sleep changed. Experiment 2 determined the effects in mice where HCRT was present but the downstream target neurons in the vlPAG were deleted by the neurotoxin. This experiment utilized an FVB-transgenic strain of mice where eGFP identifies GABA neurons. We verified this and also determined that eGFP neurons were immunopositive for the HCRT-2 receptor. vlPAG lesions in these mice increased REM sleep (+79% versus saline controls) and it was significantly correlated (r = 0.89) with loss of eGFP neurons. These results identify the vlPAG as one site that loses its inhibitory control over REM sleep, but does not cause cataplexy, as a result of hypocretin deficiency.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neuropeptídeos/farmacologia , Neuropeptídeos/fisiologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Proteínas Inativadoras de Ribossomos Tipo 1/farmacologia , Sono REM/efeitos dos fármacos , Toxinas Biológicas/farmacologia , Animais , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/genética , Orexinas , Substância Cinzenta Periaquedutal/patologia , Substância Cinzenta Periaquedutal/fisiopatologia , Saporinas
12.
J Neurosci ; 29(5): 1580-5, 2009 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-19193905

RESUMO

Narcolepsy is a human sleep disorder resulting from the loss of neurons containing the neuropeptide orexin, also known as hypocretin. Cataplexy, which is a sudden loss of muscle tone during waking, is an important diagnostic symptom of narcolepsy. In humans and canines with narcolepsy, cataplexy is considered to be a separate and distinct behavioral state. However, in the mouse model of the disease this issue is not resolved. The present study monitored the activity of forty four neurons in the rostral pons in hypocretin knock-out mice. Majority of the neurons were active during wake and REM sleep, while four neurons were selectively active during REM sleep. All of these neurons were less active during cataplexy compared with REM sleep. Thus, although cataplexy and REM sleep share many common features, including the muscle atonia, cataplexy is a distinct state in mice.


Assuntos
Cataplexia/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neurônios/fisiologia , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Ponte/fisiologia , Sono/genética , Animais , Cataplexia/genética , Eletroencefalografia/métodos , Camundongos , Camundongos Knockout , Orexinas , Ponte/citologia , Sono/fisiologia , Sono REM/genética , Sono REM/fisiologia
13.
Eur J Neurosci ; 28(7): 1382-93, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18973565

RESUMO

Gene transfer has proven to be an effective neurobiological tool in a number of neurodegenerative diseases, but it is not known if it can correct a sleep disorder. Narcolepsy is a neurodegenerative sleep disorder linked to the loss of neurons containing the neuropeptide orexin, also known as hypocretin. Here, a replication-defective herpes simplex virus-1 amplicon-based vector was constructed to transfer the gene for mouse prepro-orexin into mice with a genetic deletion of the orexin gene. After in vitro tests confirmed successful gene transfer into cells, the gene vector was delivered to the lateral hypothalamus of orexin knockout (KO) mice where the orexin peptide was robustly expressed in the somata and processes of numerous neurons, and the peptide product was detected in the cerebrospinal fluid. During the 4-day life-span of the vector the incidence of cataplexy declined by 60%, and the levels of rapid eye movement sleep during the second half of the night were similar to levels in wild-type mice, indicating that narcoleptic sleep-wake behavior in orexin KO mice can be improved by targeted gene transfer.


Assuntos
Técnicas de Transferência de Genes , Região Hipotalâmica Lateral/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Narcolepsia/genética , Narcolepsia/metabolismo , Neuropeptídeos/genética , Sono/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Genes Reporter/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/cirurgia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Narcolepsia/terapia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Orexinas , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Resultado do Tratamento
14.
Brain Res ; 1205: 47-54, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18343358

RESUMO

Ablation of the SCN, an established circadian clock, does not abolish food entrainment, suggesting that the food-entrainable oscillator (FEO) must lie outside the SCN. Typically, animals show anticipatory locomotor activity and rise in core body temperature under the influence of the FEO. Signals from the FEO would, therefore, converge onto arousal neurons so that the animal might forage for food. In the present study, we investigate whether the neuropeptide orexin, which has been linked to arousal, might transduce the arousal signal. Orexin-knockout (orexin-KO) and wildtype (WT) mice (both C57BL/6J derived) were implanted with MiniMitter transmitters that recorded core body temperature and activity (12 h LD cycle). After a week of ad-libitum feeding, the mice were given access to food for 4 h (ZT 4-8) for nine days followed by 2-days of fasting. When orexin-KO mice were placed in a restricted feeding schedule, both core body temperature and activity entrained to the feeding schedule. In these mice gross locomotor activity was severely blunted during the nine day period of restricted feeding (-79.4+/-6.3%) from the WT, but they showed an increase in core body temperature in anticipation to the meal time similar to the WT mice. There was no difference in the amount of food intake between the genotypes. We conclude that orexin is not required for entrainment of activity and temperature to a restricted feeding schedule, but is required for the robust expression of gross locomotor activity in anticipation of the scheduled feeding.


Assuntos
Restrição Calórica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Atividade Motora/fisiologia , Neuropeptídeos/genética , Neuropeptídeos/fisiologia , Periodicidade , Temperatura , Animais , Nível de Alerta/fisiologia , Jejum/fisiologia , Genótipo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orexinas , Transdução de Sinais/fisiologia
15.
J Neurosci ; 25(40): 9195-204, 2005 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-16207879

RESUMO

The thalamic intergeniculate leaflet (IGL) is involved in mediating effects of both photic and nonphotic stimuli on mammalian circadian rhythms. IGL neurons containing neuropeptide Y (NPY) have been implicated in mediating nonphotic effects, but little is known about those involved in photic entrainment. We used juxtacellular recording/labeling in rats to characterize both photic responses and neurochemical phenotypes of neurons in the lateral geniculate area, focusing on the IGL and ventral lateral geniculate (VLG). Single neurons were recorded to characterize photic responsiveness and were labeled with Neurobiotin (Nb); tissue was stained for Nb, NPY, and in some cases for orexin A. Three classes of neurons were identified in the IGL/VLG. Type I neurons lacked NPY and showed sustained activations during retinal illumination and moderate firing rates in darkness. Type II neurons contained large amounts of NPY throughout the soma and showed varied responses to illumination: suppression, complex responses, or no response. Type III neurons had patches of NPY both on the external soma surface and within the soma, apparently representing internalization of NPY. Type III neurons resembled type I cells in their sustained activation by illumination but were virtually silent during the intervening dark period. These neurons appear to receive NPY input, presumably from other IGL cells, which may suppress their activity during darkness. These results demonstrate the presence of several classes of neurons in the IGL defined by their functional and anatomical features and reinforce the role of the IGL/VLG complex in integrating photic and nonphotic inputs to the circadian system.


Assuntos
Potenciais de Ação/fisiologia , Ritmo Circadiano/fisiologia , Corpos Geniculados/citologia , Neurônios/citologia , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Imuno-Histoquímica/métodos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Microscopia Confocal/métodos , Neurônios/classificação , Neurônios/fisiologia , Neuropeptídeo Y/metabolismo , Neuropeptídeos/metabolismo , Orexinas , Estimulação Luminosa/métodos , Ratos , Ratos Sprague-Dawley
16.
J Neurosci Res ; 75(1): 133-42, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14689456

RESUMO

Rapid-eye-movement (REM) sleep is normally preceded by non-REM sleep; however, every non-REM sleep episode is not followed by REM sleep. It has been proposed that, for the regulation of REM sleep, the brain areas modulating waking and non-REM sleep are likely to communicate with neurons promoting REM sleep. The former has been reported earlier, and in this study the latter has been investigated. Under surgical anaesthesia, cats were prepared for electrophysiological recording of sleep-wakefulness and electrical stimulation of caudal brainstem as well as preopticoanterior hypothalamic hypnogenic areas. Insulated microwires of 25-32 microm were used to record 52 single neuronal activities from the brainstem along with bipolar electroencephalogram, electromyogram, electrooculogram, and pontogeniculooccipital waves in freely moving, normally behaving cats. The neurons were classified into five groups based on changes in firing rates associated with different sleep-waking states compared with quiet wakefulness. Thereafter, the responses of these neurons to 1-Hz stimulation of the two non-REM sleep-promoting areas were studied. At the end of experiment, the stimulating and recording sites were histologically identified. It was observed that, among the affected neurons, the caudal brainstem non-REM sleep-promoting area excited more REM-on neurons, whereas the preopticoanterior hypothalamus hypnogenic area inhibited more awake-active neurons. Thus, the results suggest that, at the single neuronal level, the caudal brainstem non-REM sleep-modulating area, rather than the preopticoanterior hypothalamic hypnogenic area in the brain, plays a modulatory role in triggering REM sleep initiation at a certain depth of sleep.


Assuntos
Tronco Encefálico/citologia , Neurônios/fisiologia , Sono REM/fisiologia , Vigília/fisiologia , Potenciais de Ação/fisiologia , Animais , Comportamento Animal , Tronco Encefálico/fisiologia , Gatos , Estimulação Elétrica , Eletroencefalografia , Eletromiografia , Eletroculografia , Feminino , Lateralidade Funcional , Hipotálamo/anatomia & histologia , Hipotálamo/fisiologia , Hipotálamo/efeitos da radiação , Masculino , Inibição Neural , Neurônios/classificação , Neurônios/efeitos da radiação , Sono/fisiologia , Técnicas Estereotáxicas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA