Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Physiol ; 594(4): 1069-85, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26613645

RESUMO

KEY POINTS: The basal forebrain is an important component of the ascending arousal system and may be a key site through which the orexin neurons promote arousal. It has long been known that orexin-A and -B excite basal forebrain cholinergic neurons, but orexin-producing neurons also make the inhibitory peptide dynorphin. Using whole-cell recordings in brain slices, we found that dynorphin-A directly inhibits basal forebrain cholinergic neurons via κ-opioid receptors, and decreases afferent excitatory synaptic input to these neurons. While the effects of dynorphin-A and orexin-A desensitize over multiple applications, co-application of dynorphin-A and orexin-A produces a sustained response that reverses depending on the membrane potential of basal forebrain cholinergic neurons. At -40 mV the net effect of the co-application is inhibition by dynorphin-A, whereas at -70 mV the excitatory response to orexin-A prevails. ABSTRACT: The basal forebrain (BF) is an essential component of the ascending arousal systems and may be a key site through which the orexin (also known as hypocretin) neurons drive arousal and promote the maintenance of normal wakefulness. All orexin neurons also make dynorphin, and nearly all brain regions innervated by the orexin neurons express kappa opiate receptors, the main receptor for dynorphin. This is remarkable because orexin excites target neurons including BF neurons, but dynorphin has inhibitory effects. We identified the sources of dynorphin input to the magnocellular preoptic nucleus and substantia innominata (MCPO/SI) in mice and determined the effects of dynorphin-A on MCPO/SI cholinergic neurons using patch-clamp recordings in brain slices. We found that the orexin neurons are the main source of dynorphin input to the MCPO/SI region, and dynorphin-A inhibits MCPO/SI cholinergic neurons through κ-opioid receptors by (1) activation of a G protein-coupled inwardly rectifying potassium current, (2) inhibition of a voltage-gated Ca(2+) current and (3) presynaptic depression of the glutamatergic input to these neurons. The responses both to dynorphin-A and to orexin-A desensitize, but co-application of dynorphin-A and orexin-A produces a sustained response. In addition, the polarity of the response to the co-application depends on the membrane potential of BF neurons; at -40 mV the net effect of the co-application is inhibition by dynorphin-A, whereas at -70 mV the excitatory response to orexin-A prevails. This suggests that depending on their state of activation, BF cholinergic neurons can be excited or inhibited by signals from the orexin neurons.


Assuntos
Neurônios Colinérgicos/metabolismo , Dinorfinas/metabolismo , Área Pré-Óptica/metabolismo , Substância Inominada/metabolismo , Sinapses/metabolismo , Animais , Canais de Cálcio/metabolismo , Neurônios Colinérgicos/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Orexinas/metabolismo , Área Pré-Óptica/citologia , Área Pré-Óptica/fisiologia , Receptores Opioides/metabolismo , Substância Inominada/citologia , Substância Inominada/fisiologia , Sinapses/fisiologia , Potenciais Sinápticos
2.
J Physiol ; 592(7): 1601-17, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24344163

RESUMO

Considerable electrophysiological and pharmacological evidence has long suggested an important role for acetylcholine in the regulation of rapid-eye-movement (REM) sleep. For example, injection of the cholinergic agonist carbachol into the dorsomedial pons produces an REM sleep-like state with muscle atonia and cortical activation, both of which are cardinal features of REM sleep. Located within this region of the pons is the sublaterodorsal nucleus (SLD), a structure thought to be both necessary and sufficient for generating REM sleep muscle atonia. Subsets of glutamatergic SLD neurons potently contribute to motor inhibition during REM sleep through descending projections to motor-related glycinergic/GABAergic neurons in the spinal cord and ventromedial medulla. Prior electrophysiological and pharmacological studies examining the effects of acetylcholine on SLD neurons have, however, produced conflicting results. In the present study, we sought to clarify how acetylcholine influences the activity of spinally projecting SLD (SLDsp) neurons. We used retrograde tracing in combination with patch-clamp recordings and recorded pre- and postsynaptic effects of carbachol on SLDsp neurons. Carbachol acted presynaptically by increasing the frequency of glutamatergic miniature excitatory postsynaptic currents. We also found that carbachol directly excited SLDsp neurons by activating an Na(+)-Ca(2+) exchanger. Both pre- and postsynaptic effects were mediated by co-activation of M1 and M3 muscarinic receptors. These observations suggest that acetylcholine produces synergistic, excitatory pre- and postsynaptic responses on SLDsp neurons that, in turn, probably serve to promote muscle atonia during REM sleep.


Assuntos
Carbacol/farmacologia , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , Agonistas Muscarínicos/farmacologia , Ponte/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Acetilcolina/metabolismo , Animais , Neurônios Colinérgicos/metabolismo , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Ácido Glutâmico/metabolismo , Técnicas In Vitro , Masculino , Camundongos Endogâmicos C57BL , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Neurônios Motores/metabolismo , Inibição Neural/efeitos dos fármacos , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Ponte/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M3/agonistas , Receptor Muscarínico M3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sono REM/efeitos dos fármacos , Trocador de Sódio e Cálcio/efeitos dos fármacos , Trocador de Sódio e Cálcio/metabolismo , Medula Espinal/metabolismo , Fatores de Tempo
3.
Neurobiol Aging ; 33(7): 1318-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21257235

RESUMO

A recent publication suggested that hypocretin (Hcrt, orexin) may mediate the neuropathological process leading to Alzheimer's disease (AD) and that antagonism of hypocretin receptors decreases this process. Narcoleptics have an approximately 90% loss of Hcrt neurons and commensurate reductions in the levels of Hcrt in their cerebrospinal fluid beginning at disease onset, usually before the age of 30. If Hcrt mediates the disease process, narcoleptics should be protected against AD. We examined the postmortem neuropathology and clinical records of 12 sequentially encountered cases of human narcolepsy. We found that AD was present in 4 of these narcoleptics, a prevalence that is similar to that of the general population.


Assuntos
Doença de Alzheimer/complicações , Doença de Alzheimer/diagnóstico , Narcolepsia/complicações , Narcolepsia/diagnóstico , Idoso , Idoso de 80 Anos ou mais , Humanos , Masculino
4.
Acta Physiol (Oxf) ; 198(3): 223-35, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19723027

RESUMO

The orexin neurones play an essential role in driving arousal and in maintaining normal wakefulness. Lack of orexin neurotransmission produces a chronic state of hypoarousal characterized by excessive sleepiness, frequent transitions between wake and sleep, and episodes of cataplexy. A growing body of research now suggests that the basal forebrain (BF) may be a key site through which the orexin-producing neurones promote arousal. Here we review anatomical, pharmacological and electrophysiological studies on how the orexin neurones may promote arousal by exciting cortically projecting neurones of the BF. Orexin fibres synapse on BF cholinergic neurones and orexin-A is released in the BF during waking. Local application of orexins excites BF cholinergic neurones, induces cortical release of acetylcholine and promotes wakefulness. The orexin neurones also contain and probably co-release the inhibitory neuropeptide dynorphin. We found that orexin-A and dynorphin have specific effects on different classes of BF neurones that project to the cortex. Cholinergic neurones were directly excited by orexin-A, but did not respond to dynorphin. Non-cholinergic BF neurones that project to the cortex seem to comprise at least two populations with some directly excited by orexin-A that may represent wake-active, GABAergic neurones, whereas others did not respond to orexin-A but were inhibited by dynorphin and may be sleep-active, GABAergic neurones. This evidence suggests that the BF is a key site through which orexins activate the cortex and promote behavioural arousal. In addition, orexins and dynorphin may act synergistically in the BF to promote arousal and improve cognitive performance.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Neurotransmissores/metabolismo , Prosencéfalo/fisiologia , Animais , Nível de Alerta/fisiologia , Cognição/fisiologia , Sinergismo Farmacológico , Dinorfinas/metabolismo , Fenômenos Eletrofisiológicos , Ácido Glutâmico/metabolismo , Humanos , Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Microdiálise , Modelos Neurológicos , Orexinas , Hormônios Hipofisários/metabolismo , Transmissão Sináptica/fisiologia
5.
Neuroscience ; 161(4): 970-7, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19362119

RESUMO

Mice lacking orexin/hypocretin signaling have sudden episodes of atonia and paralysis during active wakefulness. These events strongly resemble cataplexy, episodes of sudden muscle weakness triggered by strong positive emotions in people with narcolepsy, but it remains unknown whether murine cataplexy is triggered by positive emotions. To determine whether positive emotions elicit murine cataplexy, we placed orexin knockout (KO) mice on a scheduled feeding protocol with regular or highly palatable food. Baseline sleep/wake behavior was recorded with ad libitum regular chow. Mice were then placed on a scheduled feeding protocol in which they received 60% of their normal amount of chow 3 h after dark onset for the next 10 days. Wild-type and KO mice rapidly entrained to scheduled feeding with regular chow, with more wake and locomotor activity prior to the feeding time. On day 10 of scheduled feeding, orexin KO mice had slightly more cataplexy during the food-anticipation period and more cataplexy in the second half of the dark period, when they may have been foraging for residual food. To test whether more palatable food increases cataplexy, mice were then switched to scheduled feeding with an isocaloric amount of Froot Loops, a food often used as a reward in behavioral studies. With this highly palatable food, orexin KO mice had much more cataplexy during the food-anticipation period and throughout the dark period. The increase in cataplexy with scheduled feeding, especially with highly palatable food, suggests that positive emotions may trigger cataplexy in mice, just as in people with narcolepsy. Establishing this connection helps validate orexin KO mice as an excellent model of human narcolepsy and provides an opportunity to better understand the mechanisms that trigger cataplexy.


Assuntos
Cataplexia/genética , Cataplexia/psicologia , Comportamento Alimentar/fisiologia , Alimentos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neuropeptídeos/genética , Análise de Variância , Animais , Eletroencefalografia , Eletromiografia , Emoções , Iluminação , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/genética , Orexinas , Estimulação Luminosa , Sono/genética , Fatores de Tempo , Vigília/genética
6.
J Neurophysiol ; 99(6): 3090-103, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18417630

RESUMO

Orexin-producing neurons are clearly essential for the regulation of wakefulness and sleep because loss of these cells produces narcolepsy. However, little is understood about how these neurons dynamically interact with other wake- and sleep-regulatory nuclei to control behavioral states. Using survival analysis of wake bouts in wild-type and orexin knockout mice, we found that orexins are necessary for the maintenance of long bouts of wakefulness, but orexin deficiency has little impact on wake bouts <1 min. Since orexin neurons often begin firing several seconds before the onset of waking, this suggests a surprisingly delayed onset (>1 min) of functional effects. This delay has important implications for understanding the control of wakefulness and sleep because increasing evidence suggests that different mechanisms are involved in the production of brief and sustained wake bouts. We incorporated these findings into a mathematical model of the mouse sleep/wake network. Orexins excite monoaminergic neurons and we hypothesize that orexins increase the monoaminergic inhibition of sleep-promoting neurons in the ventrolateral preoptic nucleus. We modeled orexin effects as a time-dependent increase in the strength of inhibition from wake- to sleep-promoting populations and the resulting simulated behavior accurately reflects the fragmented sleep/wake behavior of narcolepsy and leads to several predictions. By integrating neurophysiology of the sleep/wake network with emergent properties of behavioral data, this model provides a novel framework for investigating network dynamics and mechanisms associated with normal and pathologic sleep/wake behavior.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Neurológicos , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Comportamento Animal , Ritmo Circadiano , Simulação por Computador , Inibição Psicológica , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/deficiência , Orexinas , Sono/genética , Análise de Sobrevida , Fatores de Tempo , Vigília/genética
7.
Neurology ; 65(8): 1184-8, 2005 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-16247044

RESUMO

BACKGROUND: Narcolepsy with cataplexy is associated with a loss of orexin/hypocretin. It is speculated that an autoimmune process kills the orexin-producing neurons, but these cells may survive yet fail to produce orexin. OBJECTIVE: To examine whether other markers of the orexin neurons are lost in narcolepsy with cataplexy. METHODS: We used immunohistochemistry and in situ hybridization to examine the expression of orexin, neuronal activity-regulated pentraxin (NARP), and prodynorphin in hypothalami from five control and two narcoleptic individuals. RESULTS: In the control hypothalami, at least 80% of the orexin-producing neurons also contained prodynorphin mRNA and NARP. In the patients with narcolepsy, the number of cells producing these markers was reduced to about 5 to 10% of normal. CONCLUSIONS: Narcolepsy with cataplexy is likely caused by a loss of the orexin-producing neurons. In addition, loss of dynorphin and neuronal activity-regulated pentraxin may contribute to the symptoms of narcolepsy.


Assuntos
Hipotálamo/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Narcolepsia/etiologia , Narcolepsia/fisiopatologia , Doenças Neurodegenerativas/complicações , Doenças Neurodegenerativas/fisiopatologia , Neuropeptídeos/deficiência , Idoso , Autoanticorpos/imunologia , Doenças Autoimunes do Sistema Nervoso/complicações , Doenças Autoimunes do Sistema Nervoso/patologia , Doenças Autoimunes do Sistema Nervoso/fisiopatologia , Mapeamento Encefálico , Proteína C-Reativa/deficiência , Proteína C-Reativa/genética , Proteína C-Reativa/imunologia , Dinorfinas/deficiência , Dinorfinas/genética , Dinorfinas/imunologia , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Masculino , Pessoa de Meia-Idade , Narcolepsia/patologia , Degeneração Neural/genética , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Vias Neurais/metabolismo , Vias Neurais/patologia , Vias Neurais/fisiopatologia , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/patologia , Neuropeptídeos/genética , Neuropeptídeos/imunologia , Orexinas , RNA Mensageiro/metabolismo
8.
Neuroscience ; 132(3): 575-80, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15837119

RESUMO

Adenosine protects neurons during hypoxia by inhibiting excitatory synaptic transmission and preventing NMDA receptor activation. Using an adeno-associated viral (AAV) vector containing Cre recombinase, we have focally deleted adenosine A(1) receptors in specific hippocampal regions of adult mice. Recently, we found that deletion of A(1) receptors in the CA1 area blocks the postsynaptic responses to adenosine in CA1 pyramidal neurons, and deletion of A(1) receptors in CA3 neurons abolishes the presynaptic effects of adenosine on the Schaffer collateral input [J Neurosci 23 (2003) 5762]. In the current study, we used this technique to delete A(1) receptors focally from CA3 neurons to investigate whether presynaptic A(1) receptors protect synaptic transmission from hypoxia. We studied the effects of prolonged (1 h) hypoxia on the evoked field excitatory postsynaptic potentials (fEPSPs) in the CA1 region using in vitro slices. Focal deletion of the presynaptic A(1) receptors on the Schaffer collateral input slowed the depression of the fEPSPs in response to hypoxia and impaired the recovery of the fEPSPs after hypoxia. Delayed responses to hypoxia linearly correlated with impaired recovery. These findings provide direct evidence that the neuroprotective role of adenosine during hypoxia depends on the rapid inhibition of synaptic transmission by the activation of presynaptic A(1) receptors.


Assuntos
Hipóxia/metabolismo , Terminações Pré-Sinápticas/metabolismo , Receptor A1 de Adenosina/deficiência , Recuperação de Função Fisiológica/fisiologia , Transmissão Sináptica/fisiologia , Antagonistas do Receptor A1 de Adenosina , Animais , Dependovirus/fisiologia , Estimulação Elétrica/métodos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Hipocampo/efeitos da radiação , Hipóxia/fisiopatologia , Hibridização In Situ/métodos , Técnicas In Vitro , Integrases/fisiologia , Camundongos , Camundongos Knockout , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Inibição Neural/efeitos da radiação , Terminações Pré-Sinápticas/efeitos dos fármacos , Receptor A1 de Adenosina/genética , Transmissão Sináptica/efeitos dos fármacos , Teofilina/análogos & derivados , Teofilina/farmacologia , Fatores de Tempo
9.
Neuroscience ; 130(4): 983-95, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15652995

RESUMO

Narcolepsy-cataplexy, a disorder of excessive sleepiness and abnormalities of rapid eye movement (REM) sleep, results from deficiency of the hypothalamic orexin (hypocretin) neuropeptides. Modafinil, an atypical wakefulness-promoting agent with an unknown mechanism of action, is used to treat hypersomnolence in these patients. Fos protein immunohistochemistry has previously demonstrated that orexin neurons are activated after modafinil administration, and it has been hypothesized that the wakefulness-promoting properties of modafinil might therefore be mediated by the neuropeptide. Here we tested this hypothesis by immunohistochemical, electroencephalographic, and behavioral methods using modafinil at doses of 0, 10, 30 and 100 mg/kg i.p. in orexin-/- mice and their wild-type littermates. We found that modafinil produced similar patterns of neuronal activation, as indicated by Fos immunohistochemistry, in both genotypes. Surprisingly, modafinil more effectively increased wakefulness time in orexin-/- mice than in the wild-type mice. This may reflect compensatory facilitation of components of central arousal in the absence of orexin in the null mice. In contrast, the compound did not suppress direct transitions from wakefulness to REM sleep, a sign of narcolepsy-cataplexy in mice. Spectral analysis of the electroencephalogram in awake orexin-/- mice under baseline conditions revealed reduced power in the theta; band frequencies (8-9 Hz), an index of alertness or attention during wakefulness in the rodent. Modafinil administration only partly compensated for this attention deficit in the orexin null mice. We conclude that the presence of orexin is not required for the wakefulness-prolonging action of modafinil, but orexin may mediate some of the alerting effects of the compound.


Assuntos
Compostos Benzidrílicos/farmacologia , Encéfalo/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neuropeptídeos/genética , Vigília/efeitos dos fármacos , Animais , Atenção/efeitos dos fármacos , Atenção/fisiologia , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Eletroencefalografia/efeitos dos fármacos , Genótipo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Modafinila , Narcolepsia/genética , Narcolepsia/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Orexinas , Proteínas Proto-Oncogênicas c-fos/metabolismo , Sono REM/efeitos dos fármacos , Sono REM/fisiologia , Vigília/fisiologia
10.
Neurosci Lett ; 351(2): 115-9, 2003 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-14583395

RESUMO

Orexin increases blood pressure and orexin-immunoreactive (IR) axons robustly innervate the spinal cord. Seeking anatomical evidence for direct effects of orexin on sympathetic preganglionic neurons (SPN), we used immunohistochemistry to study the relationships between orexin-IR axons and SPN identified by immunoreactivity for choline acetyltransferase (ChAT) or for cholera toxin B retrogradely transported from the superior cervical ganglion (SCG). In the intermediolateral cell column (IML), varicose, orexin-positive axons closely apposed almost all SPN in segments T1 and T2, but appositions were rare in T4-L2. Orexin fibers also apposed ChAT-IR cell bodies in the intercalated nucleus and the central autonomic area from T1 to L2. Orexin-IR synapses were identified ultrastructurally on SPN projecting to the SCG. Since SPN involved in cardiovascular control cluster in the IML of mid- and lower thoracic cord, these findings suggest that orexin affects blood pressure by acting on supraspinal neurons rather than SPN.


Assuntos
Fibras Autônomas Pré-Ganglionares/metabolismo , Proteínas de Transporte/metabolismo , Vias Eferentes/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Neuropeptídeos/metabolismo , Medula Espinal/metabolismo , Acetilcolina/metabolismo , Fibras Adrenérgicas/metabolismo , Fibras Adrenérgicas/ultraestrutura , Animais , Fibras Autônomas Pré-Ganglionares/ultraestrutura , Barorreflexo/fisiologia , Pressão Sanguínea/fisiologia , Toxina da Cólera/metabolismo , Toxina da Cólera/farmacocinética , Colina O-Acetiltransferase/metabolismo , Vias Eferentes/ultraestrutura , Imuno-Histoquímica , Masculino , Microscopia Eletrônica , Orexinas , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Ratos , Ratos Sprague-Dawley , Medula Espinal/ultraestrutura , Gânglio Cervical Superior/metabolismo
11.
Neuroscience ; 119(4): 913-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12831851

RESUMO

The ventrolateral preoptic nucleus (VLPO) is a key regulator of behavioral state that promotes sleep by directly inhibiting brain regions that maintain wakefulness. Subarachnoid administration of adenosine (AD) or AD agonists promotes sleep and induces expression of Fos protein in VLPO neurons. Therefore, activation of VLPO neurons may contribute to the somnogenic actions of AD. To define the mechanism through which AD activates VLPO neurons, we prepared hypothalamic slices from 9 to 12-day-old rat pups and recorded from 43 neurons in the galaninergic VLPO cluster; nine neurons contained galanin mRNA by post hoc in situ hybridization. Bath application of AD (20 microM) to seven of these neurons had no direct effect but caused a significant decrease in the frequency of spontaneous miniature inhibitory postsynaptic currents in the presence of tetrodotoxin, indicating a presynaptic site of action. We conclude that AD-mediated disinhibition increases the excitability of VLPO neurons thus contributing to the somnogenic properties of AD.


Assuntos
Adenosina/metabolismo , Vias Aferentes/metabolismo , Área Pré-Óptica/metabolismo , Terminações Pré-Sinápticas/metabolismo , Transmissão Sináptica/fisiologia , Ácido gama-Aminobutírico/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Adenosina/farmacologia , Vias Aferentes/citologia , Vias Aferentes/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Feminino , Galanina/genética , Masculino , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Técnicas de Cultura de Órgãos , Área Pré-Óptica/citologia , Área Pré-Óptica/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Sono/fisiologia , Transmissão Sináptica/efeitos dos fármacos
12.
Science ; 294(5551): 2511-5, 2001 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-11752569

RESUMO

The circadian clock in the suprachiasmatic nucleus (SCN) is thought to drive daily rhythms of behavior by secreting factors that act locally within the hypothalamus. In a systematic screen, we identified transforming growth factor-alpha (TGF-alpha) as a likely SCN inhibitor of locomotion. TGF-alpha is expressed rhythmically in the SCN, and when infused into the third ventricle it reversibly inhibited locomotor activity and disrupted circadian sleep-wake cycles. These actions are mediated by epidermal growth factor (EGF) receptors on neurons in the hypothalamic subparaventricular zone. Mice with a hypomorphic EGF receptor mutation exhibited excessive daytime locomotor activity and failed to suppress activity when exposed to light. These results implicate EGF receptor signaling in the daily control of locomotor activity, and identify a neural circuit in the hypothalamus that likely mediates the regulation of behavior both by the SCN and the retina.


Assuntos
Ritmo Circadiano/fisiologia , Receptores ErbB/metabolismo , Hipotálamo/metabolismo , Atividade Motora , Sono/fisiologia , Núcleo Supraquiasmático/metabolismo , Animais , Relógios Biológicos/efeitos dos fármacos , Relógios Biológicos/fisiologia , Temperatura Corporal/efeitos dos fármacos , Ventrículos Cerebrais/metabolismo , Ritmo Circadiano/efeitos dos fármacos , Cricetinae , Escuridão , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/genética , Feminino , Ligantes , Luz , Masculino , Mesocricetus , Camundongos , Atividade Motora/efeitos dos fármacos , Vias Neurais/fisiologia , Neurônios/metabolismo , Mutação Puntual , Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Transdução de Sinais , Sono/efeitos dos fármacos , Fator de Crescimento Transformador alfa/administração & dosagem , Fator de Crescimento Transformador alfa/genética , Fator de Crescimento Transformador alfa/metabolismo , Fator de Crescimento Transformador alfa/farmacologia
13.
Nat Neurosci ; 4(12): 1165, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11713469

RESUMO

All known eukaryotic organisms exhibit physiological and behavioral rhythms termed circadian rhythms that cycle with a near-24-hour period; in mammals, light is the most potent stimulus for entraining endogenous rhythms to the daily light cycle. Photic information is transmitted via the retinohypothalamic tract (RHT) to the suprachiasmatic nucleus (SCN) in the hypothalamus, where circadian rhythms are generated, but the retinal photopigment that mediates circadian entrainment has remained elusive. Here we show that most retinal ganglion cells (RGCs) that project to the SCN express the photopigment melanopsin.


Assuntos
Ritmo Circadiano/fisiologia , Vias Neurais/metabolismo , Células Ganglionares da Retina/metabolismo , Opsinas de Bastonetes/genética , Estilbamidinas , Núcleo Supraquiasmático/metabolismo , Animais , Corantes Fluorescentes , Lateralidade Funcional/fisiologia , Transdução de Sinal Luminoso/fisiologia , Vias Neurais/citologia , Estimulação Luminosa , RNA Mensageiro/metabolismo , Ratos , Células Ganglionares da Retina/citologia , Núcleo Supraquiasmático/citologia
14.
Trends Neurosci ; 24(12): 726-31, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11718878

RESUMO

More than 70 years ago, von Economo predicted a wake-promoting area in the posterior hypothalamus and a sleep-promoting region in the preoptic area. Recent studies have dramatically confirmed these predictions. The ventrolateral preoptic nucleus contains GABAergic and galaninergic neurons that are active during sleep and are necessary for normal sleep. The posterior lateral hypothalamus contains orexin/hypocretin neurons that are crucial for maintaining normal wakefulness. A model is proposed in which wake- and sleep-promoting neurons inhibit each other, which results in stable wakefulness and sleep. Disruption of wake- or sleep-promoting pathways results in behavioral state instability.


Assuntos
Hipotálamo/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Humanos , Hipotálamo/citologia , Vias Neurais
15.
Neurology ; 57(10): 1896-9, 2001 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-11723285

RESUMO

The neuroexcitatory peptide hypocretin and its receptors are central to the pathophysiology of both human and animal models of the disease. In this study of American and Icelandic patients with narcolepsy, the authors found no significant association between narcolepsy and single-nucleotide polymorphisms in the genes for hypocretin or its two known receptors, hypocretin receptor-1 and hypocretin receptor-2.


Assuntos
Proteínas de Transporte/genética , Peptídeos e Proteínas de Sinalização Intracelular , Narcolepsia/genética , Neuropeptídeos/genética , Polimorfismo Genético , Precursores de Proteínas/genética , Adulto , Mapeamento Cromossômico , Comparação Transcultural , Éxons , Feminino , Predisposição Genética para Doença/genética , Testes Genéticos , Genótipo , Humanos , Islândia , Íntrons , Masculino , Pessoa de Meia-Idade , Narcolepsia/diagnóstico , Receptores de Orexina , Orexinas , Receptores Acoplados a Proteínas G , Receptores de Neuropeptídeos/genética , Estados Unidos
16.
Neuroscience ; 107(4): 653-63, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11720788

RESUMO

Considerable evidence indicates that adenosine may be an endogenous somnogen, yet the mechanism through which it promotes sleep is unknown. Adenosine may act via A1 receptors to promote sleep, but an A2a receptor antagonist can block the sleep induced by prostaglandin D(2). We previously reported that prostaglandin D(2) activates sleep-promoting neurons of the ventrolateral preoptic area, and we hypothesized that an A2a receptor agonist also should activate these neurons. Rats were instrumented for sleep recordings, and an injection cannula was placed in the subarachnoid space just anterior to the ventrolateral preoptic area. After an 8-10-day recovery period, the A2a receptor agonist CGS21680 (20 pmol/min) or saline was infused through the injection cannula, and the animals were killed 2 h later. The brains were stained using Fos immunohistochemistry, and the pattern of Fos expression was studied in the entire brain. CGS21680 increased non-rapid eye movement sleep and markedly increased the expression of Fos in the ventrolateral preoptic area and basal leptomeninges, but it reduced Fos expression in wake-active brain regions such as the tuberomammillary nucleus. CGS21680 also induced Fos in the shell and core of the nucleus accumbens and in the lateral subdivision of the central nucleus of the amygdala. To determine whether these effects may have been mediated through A1 receptors, an additional group of rats received subarachnoid infusion of the A1 receptor agonist N(6)-cyclopentyladenosine (2 pmol/min). In contrast to CGS21680, infusion of N(6)-cyclopentyladenosine into the subarachnoid space produced only a small decrease in rapid eye movement sleep, and the pattern of Fos expression induced by N(6)-cyclopentyladenosine was notable only for decreased Fos in regions near the infusion site. These findings suggest that an adenosine A2a receptor agonist may activate cells of the leptomeninges or nucleus accumbens that increase the activity of ventrolateral preoptic area neurons. These ventrolateral preoptic area neurons may then coordinate the inhibition of multiple wake-promoting regions, resulting in sleep.


Assuntos
Adenosina/análogos & derivados , Neurônios/metabolismo , Área Pré-Óptica/citologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , Agonistas do Receptor Purinérgico P1 , Sono/efeitos dos fármacos , Adenosina/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Química Encefálica/efeitos dos fármacos , Masculino , Neurônios/química , Fenetilaminas/farmacologia , Área Pré-Óptica/fisiologia , Proteínas Proto-Oncogênicas c-fos/análise , Ratos , Ratos Sprague-Dawley , Receptor A2A de Adenosina , Organismos Livres de Patógenos Específicos , Espaço Subaracnóideo , Vigília/efeitos dos fármacos
18.
Curr Biol ; 11(19): R769-71, 2001 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-11591333

RESUMO

Orexin-containing neurons regulate wakefulness, and loss of orexin produces narcolepsy. Recent studies of mice lacking orexin neurons have shown that these cells also play essential roles in the control of feeding and energy balance.


Assuntos
Proteínas de Transporte/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Narcolepsia/etiologia , Neurônios/fisiologia , Neuropeptídeos/fisiologia , Animais , Proteínas de Transporte/genética , Humanos , Neuropeptídeos/genética , Obesidade , Orexinas , Sono , Vigília
19.
J Neurosci ; 21(19): RC168, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11567079

RESUMO

Orexins (also called hypocretins) are peptide neurotransmitters expressed in neurons of the lateral hypothalamic area (LHA). Mice lacking the orexin peptides develop narcolepsy-like symptoms, whereas mice with a selective loss of the orexin neurons develop hypophagia and severe obesity in addition to the narcolepsy phenotype. These different phenotypes suggest that orexin neurons may contain neurotransmitters besides orexin that regulate feeding and energy balance. Dynorphin neurons are common in the LHA, and dynorphin has been shown to influence feeding; hence, we studied whether dynorphin and orexin are colocalized. In rats, double-label in situ hybridization revealed that nearly all (94%) neurons expressing prepro-orexin mRNA also expressed prodynorphin mRNA. The converse was also true: 96% of neurons in the LHA containing prodynorphin mRNA also expressed prepro-orexin mRNA. Double-label immunohistochemistry confirmed that orexin-A and dynorphin-A peptides were highly colocalized in the LHA. Wild-type mice and orexin knock-out mice showed abundant prodynorphin mRNA-expressing neurons in the LHA, but orexin/ataxin-3 mice with a selective loss of the orexin neurons completely lacked prodynorphin mRNA in this area, further confirming that within the LHA, dynorphin expression is restricted to the orexin neurons. These findings suggest that dynorphin-A may play an important role in the function of the orexin neurons.


Assuntos
Proteínas de Transporte/metabolismo , Dinorfinas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Precursores de Proteínas/metabolismo , Animais , Ataxina-3 , Proteínas de Transporte/genética , Dinorfinas/genética , Fórnice/citologia , Fórnice/metabolismo , Hipotálamo/citologia , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Proteínas Nucleares , Orexinas , Precursores de Proteínas/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras , Fatores de Transcrição
20.
Neurology ; 56(12): 1751-3, 2001 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-11425947

RESUMO

Idiopathic narcolepsy usually results from a loss of the hypothalamic neuropeptide orexin (hypocretin), but the cause of secondary narcolepsy resulting from focal brain lesions is unknown. The authors describe a young man who developed narcolepsy after a large hypothalamic stroke. His lesion included much of the hypothalamic region in which orexin is produced, and his CSF concentration of orexin was low. The authors hypothesize that a loss of orexin neurons or their relevant targets may be the specific neuropathology causing this and many other cases of secondary narcolepsy.


Assuntos
Proteínas de Transporte/líquido cefalorraquidiano , Diencéfalo/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular , Narcolepsia/líquido cefalorraquidiano , Narcolepsia/fisiopatologia , Neuropeptídeos/líquido cefalorraquidiano , Acidente Vascular Cerebral/líquido cefalorraquidiano , Adulto , Diencéfalo/patologia , Eletroencefalografia , Humanos , Imageamento por Ressonância Magnética , Masculino , Narcolepsia/patologia , Orexinas , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...