Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Comp Neurol ; 532(5): e25622, 2024 May.
Article in English | MEDLINE | ID: mdl-38712635

ABSTRACT

Histamine H1 receptor (H1R) in the central nervous system plays an important role in various functions, including learning and memory, aggression, feeding behaviors, and wakefulness, as evidenced by studies utilizing H1R knockout mice and pharmacological interventions. Although previous studies have reported the widespread distribution of H1R in the brains of rats, guinea pigs, monkeys, and humans, the detailed distribution in the mouse brain remains unclear. This study provides a comprehensive description of the distribution of H1R mRNA in the mouse brain using two recently developed techniques: RNAscope and in situ hybridization chain reaction, both of which offer enhanced sensitivity and resolution compared to traditional methodologies such as radioisotope labeling, which were used in previous studies. The H1R mRNA expression was observed throughout the entire brain, including key regions implicated in sleep-wake regulatory functions, such as the pedunculopontine tegmental nucleus and dorsal raphe. Additionally, strong H1R mRNA signals were identified in the paraventricular hypothalamus and ventromedial hypothalamus, which may explain the potential mechanisms underlying histamine-mediated feeding regulation. Notably, we identified strong H1R mRNA expression in previously unreported cerebral regions, such as the dorsal endopiriform nucleus, bed nucleus of the accessory olfactory tract, and postsubiculum. These findings significantly contribute to our understanding of the multifaceted roles of H1R in diverse brain functions.


Subject(s)
Brain Mapping , Brain , RNA, Messenger , Receptors, Histamine H1 , Animals , Male , Mice , Brain/metabolism , Brain Mapping/methods , In Situ Hybridization , Mice, Inbred C57BL , Receptors, Histamine H1/metabolism , RNA, Messenger/metabolism
2.
Pharmacol Ther ; 249: 108505, 2023 09.
Article in English | MEDLINE | ID: mdl-37541595

ABSTRACT

Benzodiazepine, a classical medication utilized in the treatment of insomnia, operates by augmenting the activity of the GABAA receptor. This underscores the significance of GABAergic neurotransmission in both the initiation and maintenance of sleep. Nevertheless, an increasing body of evidence substantiates the notion that GABA-mediated neurotransmission also assumes a vital role in promoting wakefulness in specific neuronal circuits. Despite the longstanding belief in the pivotal function of GABA in regulating the sleep-wake cycle, there exists a dearth of comprehensive documentation regarding the specific regions within the central nervous system where GABAergic neurons are crucial for these functions. In this review, we delve into the involvement of GABAergic neurons in the regulation of sleep-wake cycles, with particular focus on those located in the preoptic area (POA) and ventral tegmental area (VTA). Recent research, including our own, has further underscored the importance of GABAergic neurotransmission in these areas for the regulation of sleep-wake cycles.


Subject(s)
Sleep , Wakefulness , Humans , GABAergic Neurons , Central Nervous System , Receptors, GABA-A , gamma-Aminobutyric Acid
3.
Front Pharmacol ; 14: 1138666, 2023.
Article in English | MEDLINE | ID: mdl-37153764

ABSTRACT

Background: Insomnia is associated with psychiatric illnesses such as bipolar disorder or schizophrenia. Treating insomnia improves psychotic symptoms severity, quality of life, and functional outcomes. Patients with psychiatric disorders are often dissatisfied with the available therapeutic options for their insomnia. In contrast, positive allosteric modulation of adenosine A2A receptors (A2ARs) leads to slow-wave sleep without cardiovascular side effects in contrast to A2AR agonists. Methods: We investigated the hypnotic effects of A2AR positive allosteric modulators (PAMs) in mice with mania-like behavior produced by ablating GABAergic neurons in the ventral medial midbrain/pons area and in a mouse model of schizophrenia by knocking out of microtubule-associated protein 6. We also compared the properties of sleep induced by A2AR PAMs in mice with mania-like behavior with those induced by DORA-22, a dual orexin receptor antagonist that improves sleep in pre-clinical models, and the benzodiazepine diazepam. Results: A2AR PAMs suppress insomnia associated with mania- or schizophrenia-like behaviors in mice. A2AR PAM-mediated suppression of insomnia in mice with mania-like behavior was similar to that mediated by DORA-22, and, unlike diazepam, did not result in abnormal sleep. Conclusion: A2AR allosteric modulation may represent a new therapeutic avenue for sleep disruption associated with bipolar disorder or psychosis.

5.
iScience ; 23(6): 101240, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32563157

ABSTRACT

Individuals with the neuropsychiatric disorder mania exhibit hyperactivity, elevated mood, and a decreased need for sleep. The brain areas and neuronal populations involved in mania-like behaviors, however, have not been elucidated. In this study, we found that ablating the ventral medial midbrain/pons (VMP) GABAergic neurons induced mania-like behaviors in mice, including hyperactivity, anti-depressive behaviors, reduced anxiety, increased risk-taking behaviors, distractibility, and an extremely shortened sleep time. Strikingly, these mice also showed no rebound sleep after sleep deprivation, suggesting abnormal sleep homeostatic regulation. Dopamine D2 receptor deficiency largely abolished the sleep reduction induced by ablating the VMP GABAergic neurons without affecting the hyperactivity and anti-depressive behaviors. Our data demonstrate that VMP GABAergic neurons are involved in the expression of mania-like behaviors, which can be segregated to the short-sleep and other phenotypes on the basis of the dopamine D2 receptors.

6.
Front Neurosci ; 13: 740, 2019.
Article in English | MEDLINE | ID: mdl-31379490

ABSTRACT

Roughly one-third of the human lifetime is spent in sleep, yet the reason for sleep remains unclear. Understanding the physiologic function of sleep is crucial toward establishing optimal health. Several proposed concepts address different aspects of sleep physiology, including humoral and circuit-based theories of sleep-wake regulation, the homeostatic two-process model of sleep regulation, the theory of sleep as a state of adaptive inactivity, and observations that arousal state and sleep homeostasis can be dissociated in pathologic disorders. Currently, there is no model that places the regulation of arousal and sleep homeostasis in a unified conceptual framework. Adenosine is well known as a somnogenic substance that affects normal sleep-wake patterns through several mechanisms in various brain locations via A1 or A2A receptors (A1Rs or A2ARs). Many cells and processes appear to play a role in modulating the extracellular concentration of adenosine at neuronal A1R or A2AR sites. Emerging evidence suggests that A1Rs and A2ARs have different roles in the regulation of sleep. In this review, we propose a model in which A2ARs allow the brain to sleep, i.e., these receptors provide sleep gating, whereas A1Rs modulate the function of sleep, i.e., these receptors are essential for the expression and resolution of sleep need. In this model, sleep is considered a brain state established in the absence of arousing inputs.

7.
Sci Rep ; 9(1): 1931, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760783

ABSTRACT

Lipocalin-type prostaglandin (PG) D synthase (L-PGDS) is responsible for the production of PGD2 in adipocytes and is selectively induced by a high-fat diet (HFD) in adipose tissue. In this study, we investigated the effects of HFD on obesity and insulin resistance in two distinct types of adipose-specific L-PGDS gene knockout (KO) mice: fatty acid binding protein 4 (fabp4, aP2)-Cre/L-PGDS flox/flox and adiponectin (AdipoQ)-Cre/L-PGDS flox/flox mice. The L-PGDS gene was deleted in adipocytes in the premature stage of the former strain and after maturation of the latter strain. The L-PGDS expression and PGD2 production levels decreased in white adipose tissue (WAT) under HFD conditions only in the aP2-Cre/L-PGDS flox/flox mice, but were unchanged in the AdipoQ-Cre/L-PGDS flox/flox mice. When fed an HFD, aP2-Cre/L-PGDS flox/flox mice significantly reduced body weight gain, adipocyte size, and serum cholesterol and triglyceride levels. In WAT of the HFD-fed aP2-Cre/L-PGDS flox/flox mice, the expression levels of the adipogenic, lipogenic, and M1 macrophage marker genes were decreased, whereas those of the lipolytic and M2 macrophage marker genes were enhanced or unchanged. Insulin sensitivity was improved in the HFD-fed aP2-Cre/L-PGDS flox/flox mice. These results indicate that PGD2 produced by L-PGDS in premature adipocytes is involved in the regulation of body weight gain and insulin resistance under nutrient-dense conditions.


Subject(s)
Adipocytes/metabolism , Insulin Resistance , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Obesity/metabolism , Prostaglandin D2/biosynthesis , Adipocytes/pathology , Animals , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Intramolecular Oxidoreductases/genetics , Lipocalins/genetics , Mice , Mice, Transgenic , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Prostaglandin D2/genetics
8.
Neurochem Int ; 124: 256-263, 2019 03.
Article in English | MEDLINE | ID: mdl-30690114

ABSTRACT

Sleep and wakefulness are controlled by a wide range of neuronal populations in the mammalian brain. Activation of adenosine A2A receptor (A2AR)-expressing neurons in the nucleus accumbens (NAc) core promotes slow-wave sleep (SWS). The neuronal mechanism by which activation of NAc A2AR neurons induces SWS, however, is unknown. We hypothesized that the ability of NAc activation to induce sleep is mediated by the classic somnogen adenosine, which can be formed by various processes in all types of cells. Here, to investigate whether astrocytes are involved in the ability of the NAc to regulate SWS, we ablated glial fibrillary acidic protein (GFAP)-positive cells in the NAc core of mice by virus-mediated expression of diphtheria toxin (DT) receptors and intraperitoneal administration of DT. Analysis of electroencephalogram and electromyogram recordings of DT-treated wild-type mice revealed that SWS was remarkably increased at 1 week after DT treatment, whereas sleep-wake behavior was unchanged in DT-treated A2AR knockout mice. Cell ablation was associated with an increased number of GFAP-positive cells and activation of microglia in the NAc. In-vivo microdialysis revealed significantly increased levels of extracellular adenosine in the NAc at 1 week after DT treatment. Our findings suggest that elevated adenosine levels in the NAc core promote SWS by acting on A2ARs and provide the first evidence that adenosine is an endogenous candidate for activating NAc A2AR neurons that have the ability to induce SWS.


Subject(s)
Adenosine/metabolism , Astrocytes/metabolism , Extracellular Fluid/metabolism , Neurons/metabolism , Nucleus Accumbens/metabolism , Sleep, Slow-Wave/physiology , Ablation Techniques/methods , Animals , Mice , Mice, Knockout , Mice, Transgenic , Nucleus Accumbens/surgery , Receptor, Adenosine A2A/metabolism
9.
Front Cell Neurosci ; 12: 357, 2018.
Article in English | MEDLINE | ID: mdl-30364224

ABSTRACT

Injection of nanomolar amounts of prostaglandin D2 (PGD2) into the rat brain has dose and time-dependent somnogenic effects, and the PGD2-induced sleep is indistinguishable from physiologic sleep. Sleep-inducing PGD2 is produced in the brain by lipocalin-type PGD2 synthase (LPGDS). Three potential intracranial sources of LPGDS have been identified: oligodendrocytes, choroid plexus, and leptomeninges. We aimed at the identification of the site of synthesis of somnogenic PGD2 and therefore, generated a transgenic mouse line with the LPGDS gene amenable to conditional deletion using Cre recombinase (flox-LPGDS mouse). To identify the cell type responsible for producing somnogenic PGD2, we engineered animals lacking LPGDS expression specifically in oligodendrocytes (OD-LPGDS KO), choroid plexus (CP-LPGDS KO), or leptomeninges (LM-LPGDS KO). We measured prostaglandins and LPGDS concentrations together with PGD synthase activity in the brain of these mice. While the LPGDS amount and PGD synthase activity were drastically reduced in the OD- and LM-LPGDS KO mice, they were unchanged in the CP-LPGDS KO mice compared with control animals. We then recorded electroencephalograms, electromyograms, and locomotor activity to measure sleep in 10-week-old mice with specific knockdown of LPGDS in each of the three targets. Using selenium tetrachloride, a specific PGDS inhibitor, we demonstrated that sleep is inhibited in OD-LPGDS and CP-LPGDS KO mice, but not in the LM-LPGDS KO mice. We concluded that somnogenic PGD2 is produced primarily by the leptomeninges, and not by oligodendrocytes or choroid plexus.

10.
J Neurosci ; 38(47): 10080-10092, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30282729

ABSTRACT

Sleep-wake behavior is controlled by a wide range of neuronal populations in the mammalian brain. Although the ventral midbrain/pons (VMP) area is suggested to participate in sleep-wake regulation, the neuronal mechanisms have remained unclear. Here, we found that nonspecific cell ablation or selective ablation of GABAergic neurons by expressing diphtheria toxin fragment A in the VMP in male mice induced a large increase in wakefulness that lasted at least 4 weeks. In contrast, selective ablation of dopaminergic neurons in the VMP had little effect on wakefulness. Chemogenetic inhibition of VMP GABAergic neurons also markedly increased wakefulness. The wake-promoting effect of the VMP GABAergic neuron ablation or inhibition was attenuated to varying degrees by the administration of dopamine D1 or D2/3 receptor antagonists and abolished by the administration of both antagonists together. In contrast, chemogenetic activation of VMP GABAergic neurons very strongly increased slow-wave sleep and reduced wakefulness. These findings suggest that VMP GABAergic neurons regulate dopaminergic actions in the sleep-wake behavior of mice.SIGNIFICANCE STATEMENT Current understanding of the neuronal mechanisms and populations that regulate sleep-wake behavior is incomplete. Here, we identified a GABAergic ventral midbrain/pons area that is necessary for controlling the daily amount of sleep and wakefulness in mice. We also found that these inhibitory neurons control wakefulness by suppressing dopaminergic systems. Surprisingly, activation of these neurons strongly induced slow-wave sleep while suppressing wakefulness. Our study reveals a new brain mechanism critical for sleep-wake regulation.


Subject(s)
GABAergic Neurons/physiology , Mesencephalon/physiology , Pons/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Dopamine Antagonists/pharmacology , Electroencephalography/methods , GABAergic Neurons/drug effects , Male , Mesencephalon/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pons/drug effects , Sleep/drug effects , Wakefulness/drug effects
11.
Nat Commun ; 8(1): 734, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963505

ABSTRACT

Sleep control is ascribed to a two-process model, a widely accepted concept that posits homoeostatic drive and a circadian process as the major sleep-regulating factors. Cognitive and emotional factors also influence sleep-wake behaviour; however, the precise circuit mechanisms underlying their effects on sleep control are unknown. Previous studies suggest that adenosine has a role affecting behavioural arousal in the nucleus accumbens (NAc), a brain area critical for reinforcement and reward. Here, we show that chemogenetic or optogenetic activation of excitatory adenosine A2A receptor-expressing indirect pathway neurons in the core region of the NAc strongly induces slow-wave sleep. Chemogenetic inhibition of the NAc indirect pathway neurons prevents the sleep induction, but does not affect the homoeostatic sleep rebound. In addition, motivational stimuli inhibit the activity of ventral pallidum-projecting NAc indirect pathway neurons and suppress sleep. Our findings reveal a prominent contribution of this indirect pathway to sleep control associated with motivation.In addition to circadian and homoeostatic drives, motivational levels influence sleep-wake cycles. Here the authors demonstrate that adenosine receptor-expressing neurons in the nucleus accumbens core that project to the ventral pallidum are inhibited by motivational stimuli and are causally involved in the control of slow-wave sleep.


Subject(s)
Nucleus Accumbens/physiology , Sleep/physiology , Animals , Circadian Rhythm , Female , Male , Mice , Mice, Inbred C57BL , Motivation , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2A/physiology
12.
Neurosci Res ; 118: 66-73, 2017 May.
Article in English | MEDLINE | ID: mdl-28434991

ABSTRACT

The mesolimbic dopamine pathway between the ventral tegmental area (VTA) and the nucleus accumbens (NAc) plays a central role in motivational behaviors. Recent findings indicate that the VTA and NAc are also involved in sleep/wake regulation - the topic of this review. First, we present an overview of the growing evidence from rodent studies revealing a wake-regulatory role of VTA dopamine neurons. We also discuss brain areas and their neurotransmitters or neuromodulators that may regulate the activity of wake-promoting VTA dopamine neurons. This is followed by a summary of current knowledge of the role of the NAc in regulating slow-wave sleep and a discussion of where and how this control of sleep physiology might be regulated by upstream neurotransmitters and neuromodulators, including dopamine and the classic somnogen adenosine.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/physiology , Nucleus Accumbens/physiology , Sleep/physiology , Ventral Tegmental Area/physiology , Wakefulness/physiology , Adenosine/metabolism , Animals , Neurotransmitter Agents/physiology
13.
Brain Struct Funct ; 222(6): 2907-2915, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28124114

ABSTRACT

A growing body of evidence suggests that dopamine plays a role in sleep-wake regulation, but the dopamine-producing brain areas that control sleep-wake states are unclear. In this study, we chemogenetically activated dopamine neurons in the ventral midbrain of mice to examine the role of these neurons in sleep-wake regulation. We found that activation of dopamine neurons in the ventral tegmental area (VTA), but not in the substantia nigra, strongly induced wakefulness, although both cell populations expressed the neuronal activity marker c-Fos after chemogenetic stimulation. Analysis of the pattern of behavioral states revealed that VTA activation increased the duration of wakefulness and decreased the number of wakefulness episodes, indicating that wakefulness was consolidated by VTA activation. The increased wakefulness evoked by VTA activation was completely abolished by pretreatment with the dopamine D2/D3 receptor antagonist raclopride, but not by the D1 receptor antagonist SCH23390. These findings indicate that the activation of VTA dopamine neurons promotes wakefulness via D2/D3 receptors.


Subject(s)
Dopaminergic Neurons/metabolism , Receptors, Dopamine D2/metabolism , Ventral Tegmental Area/metabolism , Wakefulness , Animals , Behavior, Animal/drug effects , Dopamine D2 Receptor Antagonists/pharmacology , Dopamine Plasma Membrane Transport Proteins/deficiency , Dopamine Plasma Membrane Transport Proteins/genetics , Dopaminergic Neurons/drug effects , Electroencephalography , Electromyography , Genotype , Mice, Knockout , Phenotype , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Dopamine D3/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Time Factors , Ventral Tegmental Area/drug effects , Wakefulness/drug effects
14.
J Vis Exp ; (107): e53678, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26863349

ABSTRACT

Recording of the epidural electroencephalogram (EEG) and electromyogram (EMG) in small animals, like mice and rats, has been pivotal to study the homeodynamics and circuitry of sleep-wake regulation. In many laboratories, a cable-based sleep recording system is used to monitor the EEG and EMG in freely behaving mice in combination with computer software for automatic scoring of the vigilance states on the basis of power spectrum analysis of EEG data. A description of this system is detailed herein. Steel screws are implanted over the frontal cortical area and the parietal area of 1 hemisphere for monitoring EEG signals. In addition, EMG activity is monitored by the bilateral placement of wires in both neck muscles. Non-rapid eye movement (Non-REM; NREM) sleep is characterized by large, slow brain waves with delta activity below 4 Hz in the EEG, whereas a shift from low-frequency delta activity to a rapid low-voltage EEG in the theta range between 6 and 10 Hz can be observed at the transition from NREM to REM sleep. By contrast, wakefulness is identified by low- to moderate-voltage brain waves in the EEG trace and significant EMG activity.


Subject(s)
Electroencephalography/methods , Electromyography/methods , Sleep/physiology , Algorithms , Animals , Brain/physiology , Electrodes, Implanted , Fourier Analysis , Male , Mice , Mice, Inbred C57BL , Rats , Sleep, REM/physiology , Wakefulness/physiology
15.
Brain Behav Immun ; 47: 172-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25532785

ABSTRACT

When living organisms become sick as a result of a bacterial infection, a suite of brain-mediated responses occur, including fever, anorexia and sleepiness. Systemic administration of lipopolysaccharide (LPS), a common constituent of bacterial cell walls, increases body temperature and non-rapid eye movement (NREM) sleep in animals and induces the production of pro-inflammatory prostaglandins (PGs). PGE2 is the principal mediator of fever, and both PGE2 and PGD2 regulate sleep-wake behavior. The extent to which PGE2 and PGD2 are involved in the effect of LPS on NREM sleep remains to be clarified. Therefore, we examined LPS-induced changes in body temperature and NREM sleep in mice with nervous system-specific knockouts (KO) for the PGE2 receptors type EP3 or EP4, in mice with total body KO of microsomal PGE synthase-1 or the PGD2 receptor type DP, and in mice treated with the cyclooxygenase (COX) inhibitor meloxicam. We observed that LPS-induced NREM sleep was slightly attenuated in mice lacking EP4 receptors in the nervous system, but was not affected in any of the other KO mice or in mice pretreated with the COX inhibitor. These results suggest that the effect of LPS on NREM sleep is partially dependent on PGs and is likely mediated mainly by other pro-inflammatory substances. In addition, our data show that the main effect of LPS on body temperature is hypothermia in the absence of nervous system EP3 receptors or in the presence of a COX inhibitor.


Subject(s)
Dinoprostone/metabolism , Lipopolysaccharides/pharmacology , Prostaglandin D2/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sleep/drug effects , Animals , Body Temperature/drug effects , Body Temperature/genetics , Cyclooxygenase Inhibitors/pharmacology , Meloxicam , Mice , Mice, Knockout , Receptors, Prostaglandin E, EP3 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Sleep/genetics , Thiazines/pharmacology , Thiazoles/pharmacology
16.
J Neurosci ; 33(23): 9734-42, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23739970

ABSTRACT

Narcolepsy is characterized by excessive sleepiness and cataplexy, sudden episodes of muscle weakness during waking that are thought to be an intrusion of rapid eye movement sleep muscle atonia into wakefulness. One of the most striking aspects of cataplexy is that it is often triggered by strong, generally positive emotions, but little is known about the neural pathways through which positive emotions trigger muscle atonia. We hypothesized that the amygdala is functionally important for cataplexy because the amygdala has a role in processing emotional stimuli and it contains neurons that are active during cataplexy. Using anterograde and retrograde tracing in mice, we found that GABAergic neurons in the central nucleus of the amygdala heavily innervate neurons that maintain waking muscle tone such as those in the ventrolateral periaqueductal gray, lateral pontine tegmentum, locus ceruleus, and dorsal raphe. We then found that bilateral, excitotoxic lesions of the amygdala markedly reduced cataplexy in orexin knock-out mice, a model of narcolepsy. These lesions did not alter basic sleep-wake behavior but substantially reduced the triggering of cataplexy. Lesions also reduced the cataplexy events triggered by conditions associated with high arousal and positive emotions (i.e., wheel running and chocolate). These observations demonstrate that the amygdala is a functionally important part of the circuitry underlying cataplexy and suggest that increased amygdala activity in response to emotional stimuli could directly trigger cataplexy by inhibiting brainstem regions that suppress muscle atonia.


Subject(s)
Amygdala/metabolism , Amygdala/pathology , Cataplexy/metabolism , Cataplexy/prevention & control , Intracellular Signaling Peptides and Proteins/deficiency , Neuropeptides/deficiency , Animals , Cataplexy/pathology , Electroencephalography/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orexins
17.
J Neurosci ; 33(23): 9743-51, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23739971

ABSTRACT

Narcolepsy is characterized by chronic sleepiness and cataplexy, episodes of profound muscle weakness that are often triggered by strong, positive emotions. Narcolepsy with cataplexy is caused by a loss of orexin (also known as hypocretin) signaling, but almost nothing is known about the neural mechanisms through which positive emotions trigger cataplexy. Using orexin knock-out mice as a model of narcolepsy, we found that palatable foods, especially chocolate, markedly increased cataplexy and activated neurons in the medial prefrontal cortex (mPFC). Reversible suppression of mPFC activity using an engineered chloride channel substantially reduced cataplexy induced by chocolate but did not affect spontaneous cataplexy. In addition, neurons in the mPFC innervated parts of the amygdala and lateral hypothalamus that contain neurons active during cataplexy and that innervate brainstem regions known to regulate motor tone. These observations indicate that the mPFC is a critical site through which positive emotions trigger cataplexy.


Subject(s)
Cacao , Cataplexy/metabolism , Cataplexy/physiopathology , Prefrontal Cortex/physiology , Animals , Cataplexy/genetics , Electroencephalography/methods , Feeding Behavior/physiology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/deficiency , Neuropeptides/genetics , Orexins
18.
Mol Nutr Food Res ; 56(8): 1315-23, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22715053

ABSTRACT

SCOPE: We found that rubiscolin-6, a δ opioid agonist peptide derived from d-ribulose-1,5-bisphosphate carboxylase/oxygenase (Rubisco), a major protein of green leaves, stimulates food intake after oral administration in mice. We therefore investigated its mechanism. METHODS AND RESULTS: Orexigenic activity after oral administration of rubiscolin-6 was blocked by central administration of naltrindole, an antagonist for δ opioid receptor, suggesting that orally administered rubiscolin-6 stimulates food intake via central δ opioid receptor activation. The orexigenic activity of rubiscolin-6 was inhibited by celecoxib, a cyclooxygenase (COX)-2 inhibitor. The hypothalamic mRNA expression of COX-2 and lipocallin-type (L) prostaglandin D synthase (PGDS) was elevated in response to rubiscolin-6 administration. Rubiscolin-6 stimulated food intake in wild-type and hematopoietic (H)-PGDS knockout (KO), but not L-PGDS KO mice. Interestingly, rubiscolin-6 stimulated food intake in L-PGDS(flox) /Nescre mice, which were deficient in L-PGDS in the brain parenchyma, but not leptomeninges. The orexigenic effect of rubiscolin-6 was abolished by genetic deletion of DP(1) receptor for PGD(2) , and by MK0524 or BIBO3304, an antagonist of DP(1) receptor or of Y(1) receptor for neuropeptide Y, respectively. CONCLUSION: Orally administered rubiscolin-6 may stimulate food intake through COX-2 and leptomeningeal L-PGDS, followed by DP(1) and Y(1) receptors, downstream of the central δ opioid receptor.


Subject(s)
Eating/drug effects , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Peptide Fragments/pharmacology , Ribulose-Bisphosphate Carboxylase/pharmacology , Administration, Oral , Animals , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Intramolecular Oxidoreductases/genetics , Lipocalins/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Neuropeptide Y/metabolism , Peptide Fragments/administration & dosage , Receptors, Opioid, delta/agonists , Ribulose-Bisphosphate Carboxylase/administration & dosage
19.
Mol Pain ; 5: 58, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19785772

ABSTRACT

BACKGROUND: The role of the diffusible messenger nitric oxide (NO) in the regulation of pain transmission is still a debate of matter, pro-nociceptive and/or anti-nociceptive. S-Nitrosylation, the reversible post-translational modification of selective cysteine residues in proteins, has emerged as an important mechanism by which NO acts as a signaling molecule. The occurrence of S-nitrosylation in the spinal cord and its targets that may modulate pain transmission remain unclarified. The "biotin-switch" method and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry were employed for identifying S-nitrosylated proteins. RESULTS: Here we show that actin was a major protein S-nitrosylated in the spinal cord by the NO donor, S-nitroso-N-acetyl-DL-penicillamine (SNAP). Interestingly, actin was S-nitrosylated, more in the S2 fraction than in the P2 fraction of the spinal homogenate. Treatment of PC12 cells with SNAP caused rapid S-nitrosylation of actin and inhibited dopamine release from the cells. Just like cytochalasin B, which depolymerizes actin, SNAP decreased the amount of filamentous actin cytoskeleton just beneath the membrane. The inhibition of dopamine release was not attenuated by inhibitors of soluble guanylyl cyclase and cGMP-dependent protein kinase. CONCLUSION: The present study demonstrates that actin is a major S-nitrosylated protein in the spinal cord and suggests that NO directly regulates neurotransmitter release by S-nitrosylation in addition to the well-known phosphorylation by cGMP-dependent protein kinase.


Subject(s)
Actins/metabolism , Neurotransmitter Agents/metabolism , Nitric Oxide/pharmacology , S-Nitroso-N-Acetylpenicillamine/metabolism , Amino Acid Sequence , Animals , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Dopamine/metabolism , Mice , Molecular Sequence Data , PC12 Cells , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Proteins/chemistry , Rats , Signal Transduction/drug effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spinal Cord/drug effects , Spinal Cord/enzymology
20.
Proc Natl Acad Sci U S A ; 105(50): 19992-7, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19066225

ABSTRACT

Adenosine has been proposed to promote sleep through A(1) receptors (A(1)R's) and/or A(2A) receptors in the brain. We previously reported that A(2A) receptors mediate the sleep-promoting effect of prostaglandin D(2), an endogenous sleep-inducing substance, and that activation of these receptors induces sleep and blockade of them by caffeine results in wakefulness. On the other hand, A(1)R has been suggested to increase sleep by inhibition of the cholinergic region of the basal forebrain. However, the role and target sites of A(1)R in sleep-wake regulation remained controversial. In this study, immunohistochemistry revealed that A(1)R was expressed in histaminergic neurons of the rat tuberomammillary nucleus (TMN). In vivo microdialysis showed that the histamine release in the frontal cortex was decreased by microinjection into the TMN of N(6)-cyclopentyladenosine (CPA), an A(1)R agonist, adenosine or coformycin, an inhibitor of adenosine deaminase, which catabolizes adenosine to inosine. Bilateral injection of CPA into the rat TMN significantly increased the amount and the delta power density of non-rapid eye movement (non-REM; NREM) sleep but did not affect REM sleep. CPA-promoted sleep was observed in WT mice but not in KO mice for A(1)R or histamine H(1) receptor, indicating that the NREM sleep promoted by A(1)R-specific agonist depended on the histaminergic system. Furthermore, the bilateral injection of adenosine or coformycin into the rat TMN increased NREM sleep, which was completely abolished by coadministration of 1,3-dimethyl-8-cyclopenthylxanthine, a selective A(1)R antagonist. These results indicate that endogenous adenosine in the TMN suppresses the histaminergic system via A(1)R to promote NREM sleep.


Subject(s)
Adenosine/administration & dosage , Hypothalamic Area, Lateral/drug effects , Receptor, Adenosine A1/physiology , Receptors, Histamine H1/physiology , Sleep Stages/drug effects , Adenosine A1 Receptor Agonists , Adenosine Deaminase Inhibitors , Animals , Coformycin/pharmacology , Enzyme Inhibitors/pharmacology , Histamine/metabolism , Hypothalamic Area, Lateral/physiology , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A1/genetics , Sleep Stages/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...