Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Obes (Lond) ; 41(8): 1256-1262, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28392556

RESUMO

BACKGROUND: Identifying whether components of total energy expenditure (EE) are affected by orexin receptor (OXR1 and OXR2) stimulation or antagonism with dual orexin receptor antagonists (DORAs) has relevance for obesity treatment. Orexin receptor stimulation reduces weight gain by increasing total EE and EE during spontaneous physical activity (SPA). OBJECTIVE: The purpose of this study was to determine if a DORA (TCS-1102) in the ventrolateral preoptic area (VLPO) reduced orexin-A-induced arousal, SPA, total EE and EE during sleep, rest, wake and SPA and whether the DORA alone reduced total EE and its components. We hypothesized that: (1) a DORA would reduce orexin-A induced increases in arousal, SPA, components of total EE, reductions in sleep and the EE during sleep and (2) the DORA alone would reduce baseline (non-stimulated) SPA and total EE. SUBJECTS/METHODS: Sleep, wakefulness, SPA and EE were determined after microinjection of the DORA (TCS-1102) and orexin-A in the VLPO of male Sprague-Dawley rats with a unilateral cannula targeted towards the VLPO. Individual components of total EE were determined based on time-stamped data. RESULTS: The DORA reduced orexin-A-induced increases in arousal, SPA, total EE and EE during SPA, wake, rest and sleep 1 h post injection (P<0.05). Orexin-A significantly reduced sleep and significantly increased EE during sleep 1 h post injection (P<0.05). Furthermore, the DORA alone significantly reduced total EE, EE during sleep (NREM and REM) and resting EE 2 h post injection (P<0.05). CONCLUSIONS: These data suggest that orexin-A reduces weight gain by stimulating total EE through increases in EE during SPA, rest and sleep. Residual effects of the DORA alone include decreases in total EE and EE during sleep and rest, which may promote weight gain.


Assuntos
Metabolismo Energético/fisiologia , Orexinas/metabolismo , Área Pré-Óptica/metabolismo , Animais , Metabolismo Energético/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Antagonistas dos Receptores de Orexina/farmacologia , Orexinas/antagonistas & inibidores , Área Pré-Óptica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Sono/fisiologia , Vigília/efeitos dos fármacos , Vigília/fisiologia , Aumento de Peso/efeitos dos fármacos
2.
Neuroscience ; 301: 323-37, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26073698

RESUMO

Neuropeptide B and W (NPB and NPW) are cognate peptide ligands for NPBWR1 (GPR7), a G protein-coupled receptor. In rodents, they have been implicated in the regulation of energy homeostasis, neuroendocrine/autonomic responses, and social interactions. Although localization of these peptides and their receptors in adult rodent brain has been well documented, their expression in mouse brain during development is unknown. Here we demonstrate the transient expression of NPW mRNA in the dorsomedial hypothalamus (DMH) of postnatal mouse brain and its co-localization with neuropeptide Y (NPY) mRNA. Neurons expressing both NPW and NPY mRNAs begin to emerge in the DMH at about postnatal day 0 (P-0) through P-3. Their expression is highest around P-14, declines after P-21, and by P-28 only a faint expression of NPW and NPY mRNA remains. In P-18 brains, we detected NPW neurons in the region spanning the subincertal nucleus (SubI), the lateral hypothalamic (LH) perifornical (PF) areas, and the DMH, where the highest expression of NPW mRNA was observed. The majority of these postnatal hypothalamic NPW neurons co-express NPY mRNA. A cross of NPW-iCre knock-in mice with a Cre-dependent tdTomato reporter line revealed that more than half of the reporter-positive neurons in the adult DMH, which mature from the transiently NPW-expressing neurons, are sensitive to peripherally administrated leptin. These data suggest that the DMH neurons that transiently co-express NPW and NPY in the peri-weaning period might play a role in regulating energy homeostasis during postnatal development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Homeostase/fisiologia , Hipotálamo/metabolismo , Neuropeptídeos/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Homeostase/efeitos dos fármacos , Homeostase/genética , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/crescimento & desenvolvimento , Leptina/farmacologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/farmacologia , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo , Fatores de Transcrição STAT/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
3.
Neuroscience ; 284: 217-224, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25286384

RESUMO

Orexins are bioactive peptides, which have been shown to play a pivotal role in vigilance state transitions: the loss of orexin-producing neurons (orexin neurons) leads to narcolepsy with cataplexy in the human. However, the effect of the need for sleep (i.e., sleep pressure) on orexin neurons remains largely unknown. Here, we found that immunostaining intensities of the α1 subunit of the GABAA receptor and neuroligin 2, which is involved in inhibitory synapse specialization, on orexin neurons of mouse brain were significantly increased by 6-h sleep deprivation. In contrast, we noted that immunostaining intensities of the α2, γ2, and ß2/3 subunits of the GABAA receptor and Huntingtin-associated protein 1, which is involved in GABAAR trafficking, were not changed by 6-h sleep deprivation. Using a slice patch recording, orexin neurons demonstrated increased sensitivity to a GABAA receptor agonist together with synaptic plasticity changes after sleep deprivation when compared with an ad lib sleep condition. In summary, the GABAergic input property of orexin neurons responds rapidly to sleep deprivation. This molecular response of orexin neurons may thus play a role in the changes that accompany the need for sleep following prolonged wakefulness, in particular the decreased probability of a transition to wakefulness once recovery sleep has begun.


Assuntos
Encéfalo/patologia , Regulação da Expressão Gênica/fisiologia , Neurônios/fisiologia , Orexinas/metabolismo , Receptores de GABA-A/metabolismo , Privação do Sono/patologia , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Relação Dose-Resposta a Droga , GABAérgicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Orexinas/genética , Técnicas de Patch-Clamp , Privação do Sono/fisiopatologia , Vigília/fisiologia
4.
Neuroscience ; 156(4): 819-29, 2008 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-18809470

RESUMO

Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide that has been implicated in energy homeostasis. Pharmacological studies with MCH and its receptor antagonists have suggested additional behavioral roles for the neuropeptide in the control of mood and vigilance states. These suggestions have been supported by a report of modified sleep in the MCH-1 receptor knockout mouse. Here we found that MCH knockout (MCH(-)(/)(-)) mice slept less during both the light and dark phases under baseline conditions. In response to fasting, MCH(-)(/)(-) mice exhibited marked hyperactivity, accelerated weight loss and an exaggerated decrease in rapid eye movement (REM) sleep. Following a 6-h period of sleep deprivation, however, the sleep rebound in MCH(-)(/)(-) mice was normal. Thus MCH(-)(/)(-) mice adapt poorly to fasting, and their loss of bodyweight under this condition is associated with behavioral hyperactivity and abnormal expression of REM sleep. These results support a role for MCH in vigilance state regulation in response to changes in energy homeostasis and may relate to a recent report of initial clinical trials with a novel MCH-1 receptor antagonist. When combined with caloric restriction, the treatment of healthy, obese subjects with this compound resulted in some subjects experiencing vivid dreams and sleep disturbances.


Assuntos
Jejum/fisiologia , Hipercinese/genética , Hormônios Hipotalâmicos/deficiência , Melaninas/deficiência , Hormônios Hipofisários/deficiência , Sono REM/fisiologia , Análise de Variância , Animais , Comportamento Animal , Composição Corporal/genética , Eletroencefalografia/métodos , Eletromiografia/métodos , Hormônios Hipotalâmicos/genética , Melaninas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/genética , Hormônios Hipofisários/genética , Sono REM/genética , Análise Espectral , Vigília/genética
5.
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
6.
Neuroscience ; 110(3): 431-43, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11906784

RESUMO

During rapid eye movement (REM) sleep, activity of non-retinal origin is propagated into central visual-system pathways in a manner similar, in pattern and intensity, to central visual-system activity that is exogenously generated in waking. It has been hypothesized that REM sleep, which is more abundantly represented early in life than later, functions to provide adjunct 'afferent' input for shaping synaptic connectivity during brain maturation. Here we present data that support this proposal. Recent studies have described a developmentally regulated form of in vitro long-term potentiation (LTP) in the visual cortex that is experience- and age-dependent. In immature rats, suppression of retinal activation of the visual system by removal of visual experience (dark rearing) extends the age when the developmentally regulated form of LTP can be produced. This study tests whether suppression of REM-state activation of the visual system also lengthens the developmental period in which this specific form of LTP can be elicited. Young rats were deprived of REM sleep by the multiple-small-platforms-over-water method during the typically latest week for induction of such LTP in slices of visual cortex. After this week, we could still induce LTP in slices from nearly all the REM-sleep-deprived rats (8/9) but not from age-matched rats that had not lost REM sleep (0/5). The control rats had been housed on large platforms that allow the animals to obtain REM sleep. Only body weights and the concentration of thyrotrophin-releasing hormone in the hypothalamus distinguished home-caged, normal-sleeping controls from both groups of platform animals. On all measures, stress levels were not dissimilar in the two platforms groups. After 7 days of behavioral suppression of REM sleep in immature rats, and consequent reduction of the intense, extra-retinal activity endogenously generated during this sleep state, we found that the period was extended in which developmentally regulated synaptic plasticity (LTP) could be elicited in slices of visual neocortex. These studies support the role of REM sleep and its associated neuronal activity in brain maturation.


Assuntos
Envelhecimento/fisiologia , Diferenciação Celular/fisiologia , Potenciação de Longa Duração/fisiologia , Vias Neurais/crescimento & desenvolvimento , Neurônios/fisiologia , Privação do Sono/fisiopatologia , Transmissão Sináptica/fisiologia , Córtex Visual/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Comportamento Animal/fisiologia , Peso Corporal/fisiologia , Hormônio Liberador da Corticotropina/metabolismo , Estimulação Elétrica , Feminino , Vias Neurais/citologia , Vias Neurais/fisiologia , Neurônios/citologia , Gravidez , Ratos , Ratos Long-Evans , Somatostatina/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Córtex Visual/citologia , Córtex Visual/fisiologia
7.
Neuron ; 30(2): 345-54, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11394998

RESUMO

Orexins (hypocretins) are a pair of neuropeptides implicated in energy homeostasis and arousal. Recent reports suggest that loss of orexin-containing neurons occurs in human patients with narcolepsy. We generated transgenic mice in which orexin-containing neurons are ablated by orexinergic-specific expression of a truncated Machado-Joseph disease gene product (ataxin-3) with an expanded polyglutamine stretch. These mice showed a phenotype strikingly similar to human narcolepsy, including behavioral arrests, premature entry into rapid eye movement (REM) sleep, poorly consolidated sleep patterns, and a late-onset obesity, despite eating less than nontransgenic littermates. These results provide evidence that orexin-containing neurons play important roles in regulating vigilance states and energy homeostasis. Orexin/ataxin-3 mice provide a valuable model for studying the pathophysiology and treatment of narcolepsy.


Assuntos
Proteínas de Transporte/metabolismo , Transtornos da Alimentação e da Ingestão de Alimentos/genética , Hipotálamo/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular , Narcolepsia/genética , Proteínas do Tecido Nervoso/genética , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Neurotransmissores/metabolismo , Obesidade/genética , Fases do Sono/genética , Animais , Ataxina-3 , Transtornos da Alimentação e da Ingestão de Alimentos/fisiopatologia , Feminino , Humanos , Hipotálamo/patologia , Doença de Machado-Joseph/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Narcolepsia/fisiopatologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Proteínas Nucleares , Obesidade/fisiopatologia , Orexinas , Peptídeos/genética , Proteínas Repressoras , Deleção de Sequência , Fases do Sono/fisiologia , Sono REM/genética , Fatores de Transcrição
8.
Toxicol Appl Pharmacol ; 173(1): 1-6, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11350209

RESUMO

The specific hydrolytic activity of PON1 paraoxonase/arylesterase enzymes in liver and blood provides a natural barrier against the entry of organophosphate toxins into the central and peripheral nervous systems. Inherited differences in PON1 enzyme concentrations may determine levels of susceptibility to organophosphate injury in humans. To test whether boosting serum levels of PON1 enzymes by gene therapy might provide increased protection, we compared the degree of inactivation of whole brain acetylcholinesterase of mice exposed to chlorpyrifos 4 days after intravenous injection of recombinant adenoviruses containing PON1-LQ or PON1-LR genes or no PON1 gene. Both recombinant viruses containing PON1 genes boosted serum arylesterase concentrations by approximately 60% and significantly prevented the inactivation of brain acetylcholinesterase. Some mice were completely protected. These findings indicate that boosting serum levels of PON1 enzymes by a gene delivery vector raises the threshold for organophosphate toxicity by hydrolytic destruction before the chemical can enter the brain.


Assuntos
Hidrolases de Éster Carboxílico/genética , Esterases/genética , Terapia Genética , Inseticidas/intoxicação , Compostos Organofosforados , Acetilcolinesterase/metabolismo , Adenoviridae/genética , Animais , Arildialquilfosfatase , Encéfalo/enzimologia , Hidrolases de Éster Carboxílico/sangue , Carcinoma Hepatocelular , Linhagem Celular , Esterases/sangue , Humanos , Neoplasias Hepáticas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Proteínas Recombinantes , Células Tumorais Cultivadas
9.
Annu Rev Neurosci ; 24: 429-58, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11283317

RESUMO

Orexin-A and orexin-B are neuropeptides originally identified as endogenous ligands for two orphan G-protein-coupled receptors. Orexin neuropeptides (also known as hypocretins) are produced by a small group of neurons in the lateral hypothalamic and perifornical areas, a region classically implicated in the control of mammalian feeding behavior. Orexin neurons project throughout the central nervous system (CNS) to nuclei known to be important in the control of feeding, sleep-wakefulness, neuroendocrine homeostasis, and autonomic regulation. orexin mRNA expression is upregulated by fasting and insulin-induced hypoglycemia. C-fos expression in orexin neurons, an indicator of neuronal activation, is positively correlated with wakefulness and negatively correlated with rapid eye movement (REM) and non-REM sleep states. Intracerebroventricular administration of orexins has been shown to significantly increase food consumption, wakefulness, and locomotor activity in rodent models. Conversely, an orexin receptor antagonist inhibits food consumption. Targeted disruption of the orexin gene in mice produces a syndrome remarkably similar to human and canine narcolepsy, a sleep disorder characterized by excessive daytime sleepiness, cataplexy, and other pathological manifestations of the intrusion of REM sleep-related features into wakefulness. Furthermore, orexin knockout mice are hypophagic compared with weight and age-matched littermates, suggesting a role in modulating energy metabolism. These findings suggest that the orexin neuropeptide system plays a significant role in feeding and sleep-wakefulness regulation, possibly by coordinating the complex behavioral and physiologic responses of these complementary homeostatic functions.


Assuntos
Proteínas de Transporte/fisiologia , Ingestão de Alimentos/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Neuropeptídeos/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Comportamento Alimentar , Homeostase , Humanos , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Neurotransmissores/fisiologia , Orexinas , Transdução de Sinais
10.
Mol Cell Endocrinol ; 162(1-2): 9-16, 2000 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-10854693

RESUMO

Estrogens have numerous reproductive and nonreproductive functions in brain. The actions of estrogens are mediated by estrogen receptors (ERs), and estrogens are believed to down-regulate their own receptors in many tissues. Assuming this to be true, if estrogens are removed there should be an upregulation of ERs. We have developed a mouse model in which estrogen synthesis is completely eliminated by homologous recombination to delete the gene encoding aromatase cytochrome P450 (P450(arom)). The P450(arom) enzyme catalyzes the synthesis of estrogens from androgens in the brain. The localization and density of ERs was studied in the brains of aromatase knockout (ArKO) and wild type male mice by using immunohistochemistry with a peptide antibody to ERalpha (ER-21) and computer imaging. In the wild-type animals a high density of ERalpha was found in a small number of hypothalamic cells; in the medial preoptic area, periventricular, arcuate, and ventromedial nuclei. A low and medium density of ERalpha was observed in cells of the lateral preoptic area, supraoptic, bed nucleus of the stria terminalis, and in central, medial and anterior cortical amygdaloid nuclei. The number of cells containing ERalpha-immunoreactivity was significantly increased (244%) in the medial preoptic area of the ArKO mice. In neither wild type nor ArKO animals was immunoreactivity observed in the cerebral cortex or striatum. There was intense ER-immunostaining in the nucleus of neurons in both wild type and ArKO mice. These data indicate that in the absence of estrogens there is as much as a 2-fold increase in the number of cells with ERalpha-immunoreactivity in certain hypothalamic and limbic regions. Thus, estrogens can down-regulate ERalpha in brain.


Assuntos
Aromatase/genética , Aromatase/metabolismo , Prosencéfalo/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Aromatase/deficiência , Núcleo Celular/metabolismo , Receptor alfa de Estrogênio , Estrogênios/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Prosencéfalo/anatomia & histologia , Distribuição Tecidual , Regulação para Cima
11.
Toxicol Appl Pharmacol ; 165(1): 99-105, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10814558

RESUMO

Pyridostigmine bromide (PB), a reversible inhibitor of acetylcholinesterase (AChE), is used for the treatment of myasthenia gravis. PB has also been provided to military personnel for preexposure protection against potential soman release. The entry of PB into the brain is typically minimal, but recently published data in mice suggest that a brief forced swim stress increases the permeability of the blood-brain barrier to PB. From these results, PB administered under stressful conditions was proposed to induce long-lasting central cholinergic deficits, potentially explaining the neurological and neuropsychological symptoms presented by some Gulf War veterans. In undertaking to replicate these results in the Long-Evans rat, no evidence of a stress-potentiated central effect of PB, administered at doses up 5.0 mg/kg ip, was found. Three stress protocols were used: restraint, forced swim, or a combined restraint/forced swim. Wistar rats were also tested in some of the protocols to ensure that the results were generalizable across rat strains, and plasma corticosterone levels were measured to test the effectiveness of the stressors employed. In contrast to the previously reported findings in the mouse, stress significantly reduced the entry of PB into rat brain, as measured by reduced inhibition of AChE activity: a 12.5% reduction in whole brain AChE activity after treatment with 5.0 mg/kg PB under control conditions declined to 9% after stress exposure. It is apparent, therefore, that the interaction between stress and PB requires further study, and previous data should be reassessed before they are used as a basis for interpreting symptoms presented by veterans.


Assuntos
Barreira Hematoencefálica , Corticosterona/metabolismo , Brometo de Piridostigmina/farmacocinética , Estresse Fisiológico/metabolismo , Acetilcolinesterase/metabolismo , Anestésicos Inalatórios/farmacologia , Animais , Anti-Inflamatórios/sangue , Anti-Inflamatórios/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/metabolismo , Sistema Nervoso Central/metabolismo , Inibidores da Colinesterase/farmacocinética , Corticosterona/sangue , Corticosterona/fisiologia , Halotano/farmacologia , Masculino , Camundongos , Permeabilidade , Fisostigmina/farmacologia , Ratos , Ratos Long-Evans , Ratos Wistar , Estresse Fisiológico/sangue
12.
Semin Clin Neuropsychiatry ; 5(1): 6-19, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10704534

RESUMO

This is an exciting period for basic sleep research, because we are now beginning to understand some of the mechanisms controlling the changes in consciousness associated with sleep and wakefulness. Witness the recent discoveries of a probable genetic involvement in narcolepsy, the identification of hypothalamic structures promoting sleep, and the mounting evidence that adenosine is an endogenous sleep factor. We review these and other recent developments that help us understand the neuroanatomical and neurophysiological basis of some sleep disorders. For a detailed discussion of specific sleep disorders and clinical issues, the reader is referred to other sources. Overviews are also available covering rapid eye movement (REM) and non-REM sleep physiology, the role of humoral factors in sleep, and the relationship between the immune system and sleep. In fact, where appropriate, here we draw directly on material from our earlier summaries of work in the field. We begin with a brief review of the organization of sleep and wakefulness to provide the background for the subsequent discussions of the anatomy and neurophysiology of the neural control of different vigilance states and associated sleep disorders. For example, a brief description of the neural mechanisms of REM sleep will be followed by an outline of selected sleep disorders related to REM sleep. In summary, we make no attempt here to include all sleep disorders and only review a few selected examples, those in which there is an understanding based on knowledge of central nervous system physiology. Unfortunately we are not now able to include sleep apnea, because the discovery of sleep apnea not only was a defining moment for clinical sleep research, but to this day remains the principal presenting complaint at some sleep disorder clinics.


Assuntos
Mapeamento Encefálico , Eletroencefalografia , Fases do Sono/fisiologia , Transtornos do Sono-Vigília/fisiopatologia , Encéfalo/fisiopatologia , Ritmo Circadiano/fisiologia , Humanos , Vias Neurais/fisiopatologia , Neurônios/fisiologia , Transtornos do Sono-Vigília/diagnóstico , Transtornos do Sono-Vigília/etiologia , Sono REM/fisiologia , Vigília/fisiologia
13.
Neuroscience ; 94(1): 21-31, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10613493

RESUMO

Neurochemical and functional abnormalities of the striatum have been reported in schizophrenic brains, but the cellular substrates of these changes are not known. We hypothesized that schizophrenia may involve an abnormality in one of the key modulators of striatal output, the cholinergic interneuron. We measured the densities of cholinergic neurons in the striatum in schizophrenic and control brains in a blind analysis, using as a marker of this cell population immunoreactivity for choline acetyltransferase, the synthetic enzyme of acetylcholine. As an independent marker, we used immunoreactivity for calretinin, a protein which is co-localized with choline acetyltransferase in virtually all of the cholinergic interneurons of the striatum. A significant decrease in choline acetyltransferase-positive and calretinin-positive cell densities was found in the schizophrenic cases compared with controls in the striatum as a whole [for the choline acetyltransferase-positive cells: controls: 3.21 +/- 0.48 cells/mm2 (mean +/- S.D.), schizophrenics: 2.43 +/- 0.68 cells(mm2; P < 0.02]. The decrease was patchy in nature and most prominent in the ventral striatum (for the choline acetyltransferase-positive cells: controls: 3.47 +/- 0.59 cells/mm2, schizophrenics: 2.52 +/- 0.64 cells/ mm2; P < 0.005) which included the ventral caudate nucleus and nucleus accumbens region. Three of the schizophrenic cases with the lowest densities of cholinergic neurons had not been treated with neuroleptics for periods from more than a month to more than 20 years. A decrease in the number or function of the cholinergic interneurons of the striatum may disrupt activity in the ventral striatal-pallidal-thalamic-prefrontal cortex pathway and thereby contribute to abnormalities in function of the prefrontal cortex in schizophrenia.


Assuntos
Fibras Colinérgicas/patologia , Corpo Estriado/patologia , Interneurônios/patologia , Esquizofrenia/metabolismo , Esquizofrenia/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Química Encefálica , Calbindina 2 , Contagem de Células , Colina O-Acetiltransferase/análise , Fibras Colinérgicas/química , Fibras Colinérgicas/enzimologia , Corpo Estriado/metabolismo , Humanos , Interneurônios/enzimologia , Interneurônios/ultraestrutura , Pessoa de Meia-Idade , Proteína G de Ligação ao Cálcio S100/análise
14.
Cell ; 98(4): 437-51, 1999 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-10481909

RESUMO

Neurons containing the neuropeptide orexin (hypocretin) are located exclusively in the lateral hypothalamus and send axons to numerous regions throughout the central nervous system, including the major nuclei implicated in sleep regulation. Here, we report that, by behavioral and electroencephalographic criteria, orexin knockout mice exhibit a phenotype strikingly similar to human narcolepsy patients, as well as canarc-1 mutant dogs, the only known monogenic model of narcolepsy. Moreover, modafinil, an anti-narcoleptic drug with ill-defined mechanisms of action, activates orexin-containing neurons. We propose that orexin regulates sleep/wakefulness states, and that orexin knockout mice are a model of human narcolepsy, a disorder characterized primarily by rapid eye movement (REM) sleep dysregulation.


Assuntos
Proteínas de Transporte/metabolismo , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular , Narcolepsia/genética , Neuropeptídeos/deficiência , Neuropeptídeos/metabolismo , Precursores de Proteínas/deficiência , Idade de Início , Animais , Compostos Benzidrílicos/farmacologia , Compostos Benzidrílicos/uso terapêutico , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Doenças do Cão/genética , Cães , Eletroencefalografia , Eletromiografia , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modafinila , Narcolepsia/tratamento farmacológico , Narcolepsia/metabolismo , Narcolepsia/fisiopatologia , Narcolepsia/veterinária , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neuropeptídeos/genética , Neuropeptídeos/fisiologia , Receptores de Orexina , Orexinas , Fenótipo , Postura , Precursores de Proteínas/genética , Receptores Acoplados a Proteínas G , Receptores de Neuropeptídeos/deficiência , Receptores de Neuropeptídeos/genética , Sono/fisiologia , Sono REM/fisiologia , Especificidade da Espécie , Comportamento Estereotipado
15.
J Sleep Res ; 8(3): 197-203, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10476006

RESUMO

Leptin (ob protein) is an adipose tissue derived circulating hormone that acts at specific receptors in the hypothalamus to reduce food intake. The protein is also critically involved in energy balance and metabolic status. Here the effect of leptin on sleep architecture in rats was evaluated because food consumption and metabolic status are known to influence sleep. Sprague-Dawley rats were chronically implanted with electrodes for EEG and EMG recording and diurnal sleep parameters were quantified over 9-h periods following leptin administration. Murine recombinant leptin (rMuLep) was administered systemically to rats that either had undergone 18 h of prior food deprivation or had received food ad libitum. In the normally fed rats, leptin significantly decreased the duration of rapid eye movement sleep (REMS) by about 30% and increased the duration of slow wave sleep (SWS) by about 13%, the latter effect reflecting enhanced power in the delta frequency band. These results are consistent with studies that have linked changes in metabolic rate with effects on sleep. Leptin administration has previously been shown to alter neuroendocrine parameters that could have mediated these changes in sleep architecture. Unexpectedly, prior food deprivation negated the effect of leptin on both REMS and SWS, a result that emphasizes the significance of the apparent coupling between sleep parameters and energy status.


Assuntos
Ritmo Delta , Privação de Alimentos , Leptina/sangue , Sono REM/fisiologia , Animais , Eletrodos Implantados , Eletroencefalografia , Eletromiografia , Ingestão de Energia/fisiologia , Comportamento Alimentar/fisiologia , Hipotálamo/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
16.
Brain Res ; 773(1-2): 223-6, 1997 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-9409726

RESUMO

This study examined whether damage to dopamine (DA) nerve terminals via inhibition of energy metabolism in the striatum would result in the retrograde loss of cell bodies in the substantia nigra. Infusion of 2 micromol malonate into the left striatum of rats resulted in a 67% loss of striatal DA and a 40% loss of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra. No change in the number of Nissl-positive-TH-negative neurons was observed. These findings demonstrate the retrograde destruction of DA cell bodies in the substantia nigra resulting from energy impairment at their terminal projection site.


Assuntos
Corpo Estriado/metabolismo , Dopamina/metabolismo , Metabolismo Energético/fisiologia , Malonatos/toxicidade , Substância Negra/patologia , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Metabolismo Energético/efeitos dos fármacos , Lateralidade Funcional , Infusões Parenterais , Masculino , Malonatos/administração & dosagem , Terminações Nervosas/efeitos dos fármacos , Terminações Nervosas/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Substância Negra/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/análise
17.
Neurodegeneration ; 5(4): 299-312, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9117541

RESUMO

The neurotoxin MPTP has been used to create an animal model of Parkinson's disease in the mouse, in part, because it causes a significant loss of dopaminergic neurons in the substantia nigra (nucleus A9). The purpose of the present study was to determine whether MPTP also causes degeneration of midbrain dopaminergic neurons in nuclei A8 and A10 in the mouse, as occurs in humans with Parkinson's disease. Two commonly used strains of mice were used: FVB/N and C57BL/6. MPTP was administered in cumulative doses of 50-300 mg/kg. Seven days later, dopamine concentrations were measured in the striatum using high performance liquid chromatography, and midbrain dopaminergic neurons were identified using an antibody against tyrosine hydroxylase. The cell locations were mapped with a computer imaging system. In the FVB/N strain, there was a dose-dependent decrease in striatal dopamine concentrations. Although the highest dose (300 mg/kg) caused an 86% reduction in striatal dopamine concentrations, there was only a moderate and non-significant loss of midbrain dopaminergic neurons. In the C57BL/6 strain, however, a high dose of MPTP (240 mg/kg) caused a significant reduction in both striatal dopamine concentrations (95%), and midbrain dopaminergic cells; 69% loss of nucleus A8 cells, 75% loss of nucleus A9 cells, and in nucleus A10 subnuclei there was 42% loss of ventral tegmental area cells, 55% loss of interfascicular nucleus cells, and no loss of cells in the central linear nucleus. These data (1) provide further evidence for differential susceptibility to MPTP toxicity among different mouse strains, (2) indicate that a significant depletion of striatal dopamine is not necessarily due to degeneration of midbrain dopaminergic neurons, (3) provide the precise locations of midbrain dopaminergic cells that are vulnerable to MPTP, which will aid future studies that seek to determine the mechanism/s by which-MPTP selectively destroys only certain midbrain dopaminergic neurons, and (4) indicate that MPTP produces midbrain dopaminergic neuronal degeneration in the same nuclei in the C57BL16 mouse that degenerate in humans with Parkinson's disease.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Dopaminérgicos/farmacologia , Dopamina/metabolismo , Degeneração Neural , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Imuno-Histoquímica/métodos , Masculino , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Coloração e Rotulagem , Tirosina 3-Mono-Oxigenase/metabolismo
18.
Neurodegeneration ; 5(4): 313-8, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9117542

RESUMO

The calcium-binding protein calbindin-D28k (CB) is located in midbrain dopaminergic (DA) neurons that are less vulnerable to degeneration in Parkinson's disease and in an animal model of the disorder, the MPTP-treated monkey. The present study sought to determine whether CB-containing DA neurons are also less vulnerable to degeneration in the MPTP-treated mouse. Double-labelling immunocytochemical staining and computer imaging techniques were employed to map and quantify the tyrosine hydroxylase-, CB- and CB-containing tyrosine hydroxylase neurons in portions of nucleus A9 and nucleus A10 (ventral tegmental area and central linear nucleus) following MPTP treatment in the C57BL/6 mouse. A cumulative dose of 140 mg/kg MPTP produced a significantly greater loss of DA neurons that lack CB in both nucleus A9 (71 +/- 4%) and the ventral tegmental area (70 +/- 4%), compared to the loss of DA neurons that contain CB (44 +/- 6% and 25 +/- 14%, respectively). In the central linear nucleus there was no loss of CB-containing DA neurons. These data demonstrate that the presence of CB in midbrain DA neurons identifies a population of cells in the mouse that are less vulnerable to MPTP-induced degeneration. The mouse, therefore, can serve as a useful model in which to investigate the putative neuroprotective effects of CB in an animal model of Parkinson's disease.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Dopaminérgicos/farmacologia , Dopamina/metabolismo , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Degeneração Neural , Proteína G de Ligação ao Cálcio S100/metabolismo , Animais , Calbindina 1 , Calbindinas , Imunofluorescência , Masculino , Mesencéfalo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
19.
Neuroscience ; 75(2): 523-33, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8931015

RESUMO

The calcium-binding proteins Calbindin-D28k and calretinin are co-localized with dopamine in some of the midbrain dopaminergic neurons in the rat and monkey; the present study sought to examine the pattern of co-localization in the mouse. Double immunofluorescence staining procedures were used for tyrosine hydroxylase (a dopaminergic cell marker) and Calbindin-D28k or calretinin. Midbrain dopaminergic neurons were examined at four rostrocaudal levels, and the percentage of cells that contained both tyrosine hydroxylase and either of the two calcium-binding proteins was determined in nucleus A8 (retrorubral field), nucleus A9 (substantia nigra pars compacta, pars reticulata and pars lateralis) and nucleus A10 (nucleus paranigralis, ventral tegmental area, interfascicular nucleus, central linear nucleus). The two calcium-binding proteins were distributed similarly in midbrain dopaminergic neurons in the several nuclear groups that comprise nuclei A8, A9 and A10. The calcium-binding proteins were found in the majority (50-100%) of nucleus A10 neurons, whereas in nuclei A8 and A9 (except for the substantia nigra pars lateralis) less than 40% of the cells contained either calcium-binding protein. The pattern of co-localization in the mouse is similar to that reported for the rat and monkey. The calcium-binding proteins mark the population of midbrain dopaminergic neurons that are less vulnerable to degeneration in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease.


Assuntos
Dopamina/metabolismo , Mesencéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo , Animais , Calbindina 1 , Calbindina 2 , Calbindinas , Técnica Direta de Fluorescência para Anticorpo , Imuno-Histoquímica , Masculino , Mesencéfalo/enzimologia , Mesencéfalo/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Neurônios/enzimologia , Neurônios/ultraestrutura , Tirosina 3-Mono-Oxigenase/metabolismo
20.
J Comp Neurol ; 369(3): 361-71, 1996 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-8743418

RESUMO

The dopaminergic (DA) neurons in the midbrain play a role in cognition, affect and movement. The purpose of the present study was to map and quantify the number of DA neurons in the midbrain, within the nuclei that constitute cell groups A8, A9 and A10, in the mouse. Two strains of mice were used; the C57BL/6 strain was chosen because it is commonly used in neurobiological studies, and the FVB/N strain was chosen because it is used frequently in transgenic studies. DA neurons were identified, in every fifth 20-microns-thick coronal section, using an antibody against tyrosine hydroxylase. Cell locations were entered into a computer imaging system. The FVB/N strain has 42% more midbrain DA neurons than the C57BL/6 strain; on one side of the brain there were 15,135 +/- 356 neurons (mean +/- S.E.M.) in the FVB/N strain, and 10,645 +/- 315 neurons in the C57BL/6 strain. In both strains, approximately 11% of the neurons were located in nucleus A8 (the DA neurons in the retrorubral field), 38% in nucleus A9 (the DA neurons in the substantia nigra pars compacta, pars reticulata, and pars lateralis), and 51% in nucleus A10 (the DA neurons in midline regions such as the ventral tegmental area, central linear nucleus, and interfascicular nucleus). The number of midbrain DA cells, and their distribution within the three nuclear groups, is discussed with respect to findings in other species.


Assuntos
Mapeamento Encefálico , Dopamina/fisiologia , Mesencéfalo/citologia , Camundongos Endogâmicos C57BL/anatomia & histologia , Neurônios/química , Animais , Contagem de Células , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Transgênicos , Neostriado/citologia , Neurônios/enzimologia , Tirosina 3-Mono-Oxigenase/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...