Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Physiol (Oxf) ; 198(3): 287-94, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19694625

ABSTRACT

AIM: The effect of orexin on wakefulness has been suggested to be largely mediated by activation of histaminergic neurones in the tuberomammillary nucleus (TMN) via orexin receptor-2 (OX(2)R). However, orexin receptors in other regions of the brain might also play important roles in maintenance of wakefulness. To dissect the role of the histaminergic system as a downstream mediator of the orexin system in the regulation of sleep/wake states without compensation by the orexin receptor-1 (OX(1)R) mediated pathways, we analysed the phenotype of Histamine-1 receptor (H(1)R) and OX(1)R double-deficient (H(1)R(-/-);OX(1)R(-/-)) mice. These mice lack OX(1)R-mediated pathways in addition to deficiency of H(1)R, which is thought to be the most important system in downstream of OX(2)R. METHODS: We used H(1)R deficient (H(1)R(-/-)) mice, H(1)R(-/-);OX(1)R(-/-) mice, OX(1)R and OX(2)R double-deficient (OX(1)R(-/-);OX(2)R(-/-)) mice, and wild type controls. Rapid eye movement (REM) sleep, non-REM (NREM) sleep and awake states were determined by polygraphic electroencephalographic/electromyographic recording. RESULTS: No abnormality in sleep/wake states was observed in H(1)R(-/-) mice, consistent with previous studies. H(1)R(-/-);OX(1)R(-/-) mice also showed a sleep/wake phenotype comparable to that of wild type mice, while OX(1)R(-/-); OX(2)R(-/-) mice showed severe fragmentation of sleep/wake states. CONCLUSION: Our observations showed that regulation of the sleep/wake states is completely achieved by OX(2)R-expressing neurones without involving H(1)R-mediated pathways. The maintenance of basal physiological sleep/wake states is fully achieved without both H(1) and OX(1) receptors. Downstream pathways of OX(2)R other than the histaminergic system might play an important role in the maintenance of sleep/wake states.


Subject(s)
Antigens, Surface/metabolism , Receptors, Cell Surface/metabolism , Receptors, Histamine H1/metabolism , Sleep/physiology , Wakefulness/physiology , Animals , Brain/physiology , Electroencephalography , Electromyography , Male , Mice , Mice, Knockout , Neurons/physiology , Orexin Receptors , Receptors, Cell Surface/deficiency , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine H1/deficiency , Receptors, Neuropeptide/deficiency , Receptors, Neuropeptide/metabolism , Sleep, REM/physiology
2.
Neuroscience ; 156(4): 819-29, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18809470

ABSTRACT

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.


Subject(s)
Fasting/physiology , Hyperkinesis/genetics , Hypothalamic Hormones/deficiency , Melanins/deficiency , Pituitary Hormones/deficiency , Sleep, REM/physiology , Analysis of Variance , Animals , Behavior, Animal , Body Composition/genetics , Electroencephalography/methods , Electromyography/methods , Hypothalamic Hormones/genetics , Melanins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Pituitary Hormones/genetics , Sleep, REM/genetics , Spectrum Analysis , Wakefulness/genetics
3.
Neuroscience ; 130(4): 983-95, 2005.
Article in English | MEDLINE | ID: mdl-15652995

ABSTRACT

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.


Subject(s)
Benzhydryl Compounds/pharmacology , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Neuropeptides/genetics , Wakefulness/drug effects , Animals , Attention/drug effects , Attention/physiology , Brain/metabolism , Dose-Response Relationship, Drug , Electroencephalography/drug effects , Genotype , Immunohistochemistry , Male , Mice , Mice, Knockout , Modafinil , Narcolepsy/genetics , Narcolepsy/physiopathology , Neurons/drug effects , Neurons/metabolism , Orexins , Proto-Oncogene Proteins c-fos/metabolism , Sleep, REM/drug effects , Sleep, REM/physiology , Wakefulness/physiology
4.
J Neurosci ; 21(19): RC168, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567079

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Dynorphins/metabolism , Intracellular Signaling Peptides and Proteins , Neurons/metabolism , Neuropeptides/metabolism , Protein Precursors/metabolism , Animals , Ataxin-3 , Carrier Proteins/genetics , Dynorphins/genetics , Fornix, Brain/cytology , Fornix, Brain/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neuropeptides/deficiency , Neuropeptides/genetics , Nuclear Proteins , Orexins , Protein Precursors/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins , Transcription Factors
5.
Neuron ; 30(2): 345-54, 2001 May.
Article in English | MEDLINE | ID: mdl-11394998

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Feeding and Eating Disorders/genetics , Hypothalamus/physiopathology , Intracellular Signaling Peptides and Proteins , Narcolepsy/genetics , Nerve Tissue Proteins/genetics , Neurons/physiology , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Obesity/genetics , Sleep Stages/genetics , Animals , Ataxin-3 , Feeding and Eating Disorders/physiopathology , Female , Humans , Hypothalamus/pathology , Machado-Joseph Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Narcolepsy/physiopathology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/pathology , Nuclear Proteins , Obesity/physiopathology , Orexins , Peptides/genetics , Repressor Proteins , Sequence Deletion , Sleep Stages/physiology , Sleep, REM/genetics , Transcription Factors
6.
Annu Rev Neurosci ; 24: 429-58, 2001.
Article in English | MEDLINE | ID: mdl-11283317

ABSTRACT

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.


Subject(s)
Carrier Proteins/physiology , Eating/physiology , Intracellular Signaling Peptides and Proteins , Neuropeptides/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Feeding Behavior , Homeostasis , Humans , Mice , Mice, Knockout , Neurons/physiology , Neurotransmitter Agents/physiology , Orexins , Signal Transduction
7.
Cell ; 98(4): 437-51, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10481909

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Disease Models, Animal , Intracellular Signaling Peptides and Proteins , Narcolepsy/genetics , Neuropeptides/deficiency , Neuropeptides/metabolism , Protein Precursors/deficiency , Age of Onset , Animals , Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/therapeutic use , Carrier Proteins/genetics , Carrier Proteins/physiology , Dog Diseases/genetics , Dogs , Electroencephalography , Electromyography , Humans , Hypothalamus/drug effects , Hypothalamus/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Modafinil , Narcolepsy/drug therapy , Narcolepsy/metabolism , Narcolepsy/physiopathology , Narcolepsy/veterinary , Neurons/drug effects , Neurons/pathology , Neuropeptides/genetics , Neuropeptides/physiology , Orexin Receptors , Orexins , Phenotype , Posture , Protein Precursors/genetics , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/deficiency , Receptors, Neuropeptide/genetics , Sleep/physiology , Sleep, REM/physiology , Species Specificity , Stereotyped Behavior
SELECTION OF CITATIONS
SEARCH DETAIL
...