Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Brain Res Bull ; 191: 40-47, 2022 12.
Article in English | MEDLINE | ID: mdl-36243208

ABSTRACT

Although several previous studies have suggested a relationship between sleep and the stress response, the mechanism underlying this relationship remains largely unknown. Here, we show that fibroblast growth factor 21 (FGF21), a lipid metabolism-related hormone, may play a role in this relationship. In this study, we examined differences in the stress response between FGF21 knockout (KO) mice and wild-type (WT) mice after social defeat stress (SDS). When the amount of non-rapid eye movement (NREM) sleep, rapid eye movement (REM) sleep and wakefulness were averaged over the dark period after SDS, only KO mice showed significant differences in NREM sleep and wakefulness. In the social interaction test, KO mice seemed to be more prone to social avoidance. Our real-time (RT) -PCR results revealed that the mRNA expression of the stress- and sleep-related gene gamma-aminobutyric acid A receptor subunit alpha 2 was significantly lower in WT mice than in KO mice. Moreover, KO mice showed lower plasma levels of ketone bodies, which also affect sleep/wake regulation, than WT mice. These results suggested that FGF21 might influence sleep/wake regulation by inducing production of an anti-stress agent and/or ketone bodies, which may result in resilience to social stress.


Subject(s)
Sleep , Wakefulness , Animals , Mice , Electroencephalography , Ketone Bodies , Mice, Inbred C57BL , Mice, Knockout , Sleep/physiology , Wakefulness/physiology , Stress, Physiological
2.
Brain Res Bull ; 188: 39-46, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35868501

ABSTRACT

AIMS: This study aimed to evaluate the involvement of the orexin system in predictable chronic mild stress (PCMS) and the effects of suvorexant, a dual orexin receptor antagonist, on nociceptive behavior in PCMS. MATERIALS AND METHODS: Male C57BL/6 J mice were separated into various PCMS groups: a control group with sawdust on the floor of the rearing cage (C), a group with mesh wire on the floor (M), and a group with water just below the mesh wire (W). Activation of lateral hypothalamic orexin neurons was assessed using immunofluorescence. In another experiment, half of the mice in each group were administered an intraperitoneal injection of suvorexant (10 mg/kg), and the remaining mice were injected with the same amount of vehicle (normal saline). Thermal hyperalgesia was examined using tail immersion and hot plate tests, while mechanical hyperalgesia was investigated using the tail pinch test after 21 days of PCMS. KEY FINDINGS: Animals subjected to PCMS showed an increased percentage of activated orexin neurons in the lateral hypothalamic region after 21 days. Mice raised in the PCMS environment showed increased pain sensitivity in several pain tests; however, the symptoms were significantly reduced by suvorexant administration. SIGNIFICANCE: The findings revealed that PCMS activates hypothalamic orexin neuronal activity, and the use of suvorexant can help attenuate PCMS-induced thermal and mechanical hyperalgesia.


Subject(s)
Hyperalgesia , Orexin Receptor Antagonists , Animals , Azepines , Hyperalgesia/drug therapy , Male , Mice , Mice, Inbred C57BL , Orexin Receptor Antagonists/pharmacology , Orexin Receptors , Orexins/pharmacology , Pain , Pharmaceutical Preparations , Triazoles
3.
Int J Mol Sci ; 22(19)2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34639172

ABSTRACT

Circadian phase shifts in peripheral clocks induced by changes in feeding rhythm often result in insulin resistance. However, whether the hypothalamic control system for energy metabolism is involved in the feeding rhythm-related development of insulin resistance is unknown. Here, we show the physiological significance and mechanism of the involvement of the agouti-related protein (AgRP) in evening feeding-associated alterations in insulin sensitivity. Evening feeding during the active dark period increased hypothalamic AgRP expression and skeletal muscle insulin resistance in mice. Inhibiting AgRP expression by administering an antisense oligo or a glucocorticoid receptor antagonist mitigated these effects. AgRP-producing neuron-specific glucocorticoid receptor-knockout (AgRP-GR-KO) mice had normal skeletal muscle insulin sensitivity even under evening feeding schedules. Hepatic vagotomy enhanced AgRP expression in the hypothalamus even during ad-lib feeding in wild-type mice but not in AgRP-GR-KO mice. The findings of this study indicate that feeding in the late active period may affect hypothalamic AgRP expression via glucocorticoids and induce skeletal muscle insulin resistance.


Subject(s)
Agouti-Related Protein/metabolism , Energy Metabolism , Feeding Behavior , Glucocorticoids/pharmacology , Hypothalamus/pathology , Insulin Resistance , Muscle, Skeletal/pathology , Agouti-Related Protein/genetics , Animals , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Receptors, Glucocorticoid/metabolism
4.
Sci Rep ; 11(1): 14231, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244555

ABSTRACT

Even though it has been well documented that stress can lead to the development of sleep disorders and the intensification of pain, their relationships have not been fully understood. The present study was aimed at investigating the effects of predictable chronic mild stress (PCMS) on sleep-wake states and pain threshold, using the PCMS rearing conditions of mesh wire (MW) and water (W) for 21 days. Exposure to PCMS decreased the amount of non-rapid eye movement (NREM) sleep during the dark phase. Moreover, the chronicity of PCMS decreased slow-wave activity (SWA) during NREM sleep in the MW and W groups in both the light and dark phases. Mechanical and aversively hot thermal hyperalgesia were more intensified in the PCMS groups than the control. Higher plasma corticosterone levels were seen in mice subjected to PCMS, whereas TNF-α expression was found higher in the hypothalamus in the W and the trigeminal ganglion in the MW group. The W group had higher expression levels of IL-6 in the thalamus as well. The PCMS paradigm decreased SWA and may have intensified mechanical and thermal hyperalgesia. The current study also suggests that rearing under PCMS may cause impaired sleep quality and heightened pain sensation to painful mechanical and aversively hot thermal stimuli.


Subject(s)
Facial Pain/physiopathology , Locomotion/physiology , Sleep Wake Disorders/physiopathology , Animals , Corticosterone/blood , Electroencephalography , Facial Pain/blood , Male , Mice , Mice, Inbred C57BL , Pain/blood , Pain/physiopathology , Pain Threshold , Real-Time Polymerase Chain Reaction , Sleep Deprivation/blood , Sleep Deprivation/physiopathology , Sleep Wake Disorders/blood , Sleep, REM/physiology , Wakefulness/physiology
5.
Behav Brain Res ; 403: 113143, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33516739

ABSTRACT

Mast cells (MCs) exist intracranially and have been reported to affect higher brain functions in rodents. However, the role of MCs in the regulation of emotionality and social behavior is unclear. In the present study, using male mice, we examined the relationship between MCs and social behavior and investigated the underlying mechanisms. Wild-type male mice intraventricularly injected with a degranulator of MCs exhibited a marked increase in a three-chamber sociability test. In addition, removal of MCs in Mast cell-specific Toxin Receptor-mediated Conditional cell Knock out (Mas-TRECK) male mice showed reduced social preference levels in a three-chamber sociability test without other behavioral changes, such as anxiety-like and depression-like behavior. Mas-TRECK male mice also had reduced serotonin content and serotonin receptor expression and increased oxytocin receptor expression in the brain. These results suggested that MCs may contribute to the regulation of social behavior in male mice. This effect may be partially mediated by serotonin derived from MCs in the brain.


Subject(s)
Behavior, Animal/physiology , Brain , Mast Cells/physiology , Receptors, Serotonin/metabolism , Serotonin/metabolism , Social Behavior , Animals , Brain/cytology , Brain/drug effects , Brain/physiology , Male , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , p-Methoxy-N-methylphenethylamine/pharmacology
6.
Physiol Behav ; 214: 112760, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31785270

ABSTRACT

Peroxisome proliferator-activated receptor alpha (PPARα) is a transcription factor that belongs to the nuclear receptor family and plays an important role in regulating gene expression associated with lipid metabolism. PPARα promotes hepatic fatty acid oxidation and ketogenesis in response to fasting. Because energy metabolism is known to affect sleep regulation, manipulations that change PPARα are likely to affect sleep and other physiological phenotypes. In this study, we examined the role of PPARα in sleep/wake regulation using PPARα knockout (KO) mice. Sleep, body temperature (BT), locomotor activity, arterial pressure (AP) and heart rate (HR) were recorded in KO mice and wild-type (WT) controls under ad libitum-fed conditions and 24-hour food deprivation (FD). KO and WT mice were identical in basal sleep amount, BT, mean AP and HR, although KO mice showed enhanced sleepiness (enhanced EEG slow-wave activity). In response to FD, KO mice showed a large drop in wakefulness and locomotor activity at the end of the dark phase, whereas WT mice did not. Similarly, AP and HR, which were suppressed by FD, decreased more in KO than in WT mice. Compared to WT mice, KO mice showed a reduced concentration of plasma ketone bodies and decreased mRNA expression of the ketogenic enzyme gene Hmgcs2 in the liver and brain under FD conditions. These results suggest that PPARα and/or lipid metabolism is involved in the maintenance of wakefulness and locomotor activity during fasting in mice.


Subject(s)
Fasting/physiology , PPAR alpha/deficiency , Sleep/physiology , Animals , Blood Glucose/metabolism , Fatty Acids, Nonesterified/blood , Heart/physiopathology , Ketone Bodies/blood , Male , Mice , Mice, Knockout , PPAR alpha/genetics , Photoperiod , Triglycerides/blood , Wakefulness/physiology
7.
Sci Rep ; 9(1): 16344, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31704978

ABSTRACT

Idiopathic restless legs syndrome (RLS) has a genetic basis wherein BTBD9 is associated with a higher risk of RLS. Hemodialysis patients also exhibit higher rates of RLS compared with the healthy population. However, little is known about the relationship of BTBD9 and end-stage renal disease to RLS pathophysiology. Here we evaluated sleep and leg muscle activity of Btbd9 mutant (MT) mice after administration of serum from patients with either idiopathic or RLS due to end-stage renal disease (renal RLS) and investigated the efficacy of treatment with the dopamine agonist rotigotine. At baseline, the amount of rapid eye movement (REM) sleep was decreased and leg muscle activity during non-REM (NREM) sleep was increased in MT mice compared to wild-type (WT) mice. Wake-promoting effects of rotigotine were attenuated by injection of serum from RLS patients in both WT and MT mice. Leg muscle activity during NREM sleep was increased only in MT mice injected with serum from RLS patients of ideiopatic and renal RLS. Subsequent treatment with rotigotine ameliorated this altered leg muscle activity. Together these results support previous reports showing a relationship between the Btbd9/dopamine system and RLS, and elucidate in part the pathophysiology of RLS.


Subject(s)
Muscles/physiology , Musculoskeletal Physiological Phenomena , Nerve Tissue Proteins/physiology , Restless Legs Syndrome/blood , Serum/chemistry , Sleep, REM/physiology , Tetrahydronaphthalenes/pharmacology , Thiophenes/pharmacology , Animals , Case-Control Studies , Disease Models, Animal , Dopamine Agonists/pharmacology , Humans , Kidney Failure, Chronic/therapy , Male , Mice , Mice, Knockout , Muscles/drug effects , Renal Dialysis/adverse effects , Restless Legs Syndrome/etiology , Restless Legs Syndrome/pathology , Sleep, REM/drug effects
8.
J Physiol Sci ; 69(6): 1019-1028, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31664642

ABSTRACT

We aimed to identify the neurotransmitters and brain regions involved in exercise efficiency in mice during continuous complicated exercises. Male C57BL/6J mice practiced treadmill running with intermittent obstacles on a treadmill for 8 days. Oxygen uptake (VO2) during treadmill running was measured as exercise efficiency. After obstacle exercise training, the VO2 measured during treadmill running with obstacles decreased significantly. Obstacle exercise-induced c-Fos expressions and dopamine turnover (DOPAC/dopamine) in the septum after obstacle exercise training were significantly higher than that before training. The dopamine turnover was correlated with exercise efficiency on the 3rd day after exercise training. Furthermore, the training effect on exercise efficiency was significantly decreased by injection of dopamine receptor antagonists into the septum and was associated with decreased c-Fos expressions in the septum and hippocampus of the mice. These results suggest that dopaminergic function in the septum is involved in exercise efficiency during continuous complicated exercises.


Subject(s)
Dopamine/pharmacology , Oxygen Consumption/physiology , Physical Conditioning, Animal/physiology , Septum of Brain/drug effects , Animals , Benzazepines/pharmacology , Biomarkers , Dopamine Antagonists/pharmacology , Gene Expression Regulation/drug effects , Genes, fos/physiology , Male , Mice , Mice, Inbred C57BL , Receptors, Dopamine D1/antagonists & inhibitors , Running , Serotonin/metabolism , Sulpiride/pharmacology
9.
Biochem Biophys Res Commun ; 514(1): 166-172, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31029425

ABSTRACT

Orexin is known as an important neuropeptide in the regulation of energy metabolism. However, the role of orexin in exercise-induced leptin sensitivity in the hypothalamus has been unclear. In this study, we determined the effect of transient treadmill exercise on leptin sensitivity in the mediobasal hypothalamus (MBH) of mice and examined the role of orexin in post-exercise leptin sensitivity. Treadmill running for 45 min increased the orexin neuron activity in mice. Intraperitoneal injection of a submaximal dose of leptin after exercise stimulated the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in MBH of mice post-exercise compared with that in non-exercised mice, although intracerebroventricular (icv) injection of leptin did not enhance STAT3 phosphorylation, even after exercise. Icv injection of an orexin receptor antagonist, SB334867 reduced STAT3 phosphorylation, which was enhanced by icv injection of orexin but not by direct injection of orexin into MBH. Exercise increased the phosphorylation of extracellular signal-regulated kinases (ERKs) in the MBH of mice, while ERK phosphorylation was reduced by SB334867. Leptin injection after exercise increased the leptin level in MBH, whereas icv injection of SB334867 suppressed the increase in the leptin level in MBH of mice. These results indicate that the activation of orexin neurons by exercise may contribute to the enhancement of leptin sensitivity in MBH. This effect may be mediated by increased transportation of circulating leptin into MBH, with the involvement of ERK phosphorylation.


Subject(s)
Hypothalamus/physiology , Leptin/pharmacology , Orexins/metabolism , Animals , Benzoxazoles/pharmacology , Exercise Test , Hypothalamus/drug effects , Male , Mice, Inbred C57BL , Naphthyridines/pharmacology , Neurons/drug effects , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/metabolism , Orexins/pharmacology , Phosphorylation , Physical Conditioning, Animal , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Urea/analogs & derivatives , Urea/pharmacology
10.
Life Sci ; 219: 219-230, 2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30653972

ABSTRACT

AIMS: Psychosocial stress is a form of mental stress associated with human relationships that underlies the pathogenesis of mental disorders such as depression. Previous studies have suggested that intake of energy-dense foods, also known as "palatable foods," can relieve psychosocial stress. However, it remains unclear whether the volume of palatable food affects abnormal behavior induced by psychosocial stress. In the present study, we aimed to determine whether levels of high-fat food intake significantly influence psychosocial stress using the social-defeat stress (SDS) paradigm. MAIN METHODS: Mice subjected to SDS ate either a high-fat or normal chow diet for 10 days. Behavioral tests were conducted following the completion of the SDS paradigm. The hypothalamus, liver, and blood were examined post-mortem. KEY FINDINGS: Mice with sufficient intake of high-fat chow immediately following exposure to SDS did not exhibit social avoidance behavior, suggesting that a high-fat diet may improve social behavior. However, inadequate intake of high-fat food, which did not alter cholesterol metabolism or hypothalamic-pituitary-adrenal axis activity, was not associated with such benefits, instead increased anxiety-like behavior. SIGNIFICANCE: The results of the present study demonstrate that eating a high-fat diet may attenuate stress, but that this benefit disappears with insufficient intake of high-fat foods. The benefits of a high-fat diet under SDS may be related to cholesterol metabolism in the liver.


Subject(s)
Avoidance Learning , Diet, High-Fat , Social Behavior , Stress, Psychological/psychology , Animals , Brain/metabolism , Cholesterol/blood , Corticosterone/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Stress, Psychological/diet therapy
11.
Behav Brain Res ; 359: 664-670, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30278189

ABSTRACT

Peroxisome proliferator-activated receptor alpha (PPARα) is a member of the nuclear receptor superfamily and regulates fatty acid oxidation. Although PPARα is expressed not only in the peripheral tissues but also in the brain, its role in higher brain function is unclear. In this study, we investigated the role of PPARα in the control of behavior, including memory/learning and mood change, using PPARα knockout (KO) mice. A significant difference between wild-type (WT) and KO mice was seen in the passive avoidance test, demonstrating that KO mice showed enhanced fear leaning. In the amygdala of KO mice, the levels of dopamine and its metabolites were increased, and the mRNA expression of dopamine degrading enzyme was decreased. When dopamine D1 receptor antagonist was administered, the enhanced fear learning observed in KO mice was attenuated. These results suggest that PPARα is involved in the regulation of emotional memory via the dopamine pathway in the amygdala.


Subject(s)
Fear/physiology , Learning/physiology , PPAR alpha/deficiency , Amygdala/drug effects , Amygdala/metabolism , Animals , Anxiety/metabolism , Benzazepines/pharmacology , Depression/metabolism , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Fear/psychology , Male , Mice, 129 Strain , Mice, Knockout , PPAR alpha/genetics , RNA, Messenger/metabolism , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/metabolism
12.
Sci Rep ; 7(1): 13640, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29057915

ABSTRACT

We have developed a chronic mild stress (MS) mouse model by simply rearing mice on a wire net for 3 weeks and investigated the effects of MS on glucose homeostasis and sleep. MS mice showed impaired glucose tolerance and disturbed sleep. One-week treatment with a histamine H1 receptor antagonist (H1RA) ameliorated the glucose intolerance and improved sleep quality in MS mice. MS mice showed an increased number of mast cells in both adipose tissue and the brain. Inhibition of mast cell function ameliorated the impairment in both glucose tolerance and sleep. Together, these findings indicate that mast cells may represent an important pathophysiological mediator in sleep and energy homeostasis.


Subject(s)
Glucose Intolerance/metabolism , Glucose/metabolism , Mast Cells/metabolism , Sleep Wake Disorders/metabolism , Stress, Psychological/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Chronic Disease , Disease Models, Animal , Glucose Intolerance/drug therapy , Glucose Intolerance/pathology , Glucose Tolerance Test , Histamine H1 Antagonists/pharmacology , Homeostasis/drug effects , Homeostasis/physiology , Male , Mast Cells/drug effects , Mast Cells/pathology , Mice, Inbred ICR , Mice, Transgenic , Random Allocation , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/pathology , Stress, Psychological/drug therapy , Stress, Psychological/pathology
13.
Life Sci ; 160: 18-26, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27436545

ABSTRACT

AIMS: Sleep and feeding behaviors closely interact to maintain energy homeostasis. While it is known that sleep disorders can lead to various metabolic issues such as insulin resistance, the mechanism for this effect is poorly understood. We thus investigated whether different feeding rhythms during the active period affect sleep-wake regulation. MAIN METHODS: For 2weeks, mice were randomly assigned to 1 of 3 feeding schedules as follows: free access to lab chow during the active period (ZT12-24, Ad-lib group), free access to lab chow during the first half of the active period (ZT12-18; Morning group), or free access to lab chow during the second half of the active period (ZT18-24, Evening group). Food intake, body weight, body temperature, locomotor activity, and sleep were evaluated. The hypothalamus and cerebral cortex were examined post-mortem. KEY FINDINGS: No alterations in food intake or body weight were observed among the 3 groups. The Evening group showed lower slow-wave activity (SWA) than the other 2 groups, in addition to higher expression of orexin mRNA in the hypothalamus and higher concentrations of dopamine and its metabolites in the cerebral cortex. AMPK phosphorylation was increased in the hypothalamus of mice in the Evening group; however, AMPK inhibition had no effect on SWA. SIGNIFICANCE: We concluded that late feeding reduces SWA in NREM sleep via a mechanism that involves orexin-mediated arousal in the hypothalamus and elevated monoamines in the cerebral cortex. These data have important implications for the relationship between sleep-wake disturbances and metabolic disorders.


Subject(s)
Feeding Behavior/physiology , Adenylate Kinase/metabolism , Animals , Biogenic Monoamines/metabolism , Body Temperature , Body Weight , Energy Metabolism , Gene Expression , Homeostasis , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Activity , Phosphorylation , Sleep/physiology , Sleep Deprivation
14.
Physiol Behav ; 151: 264-71, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26232088

ABSTRACT

It is well-established that exercise can influence psychological conditions, cognitive function, and energy metabolism in peripheral tissues including the skeletal muscle. However, it is not clear whether exercise can influence social interaction with others and alleviate defeat stress. This study investigated the effect of voluntary wheel running on impaired social interaction induced by chronic social defeat stress (SDS) using the resident-intruder social defeat model. Mice were divided into three groups: control, stress alone, and stress+exercise. SDS was performed by exposing C57BL/6 mice to retired ICR mice for 2.5 min. The C57BL/6 mice were continuously defeated by these resident (aggressor) mice and, following 5 days of SDS, experienced 2 days of rest with no SDS. Mice in the stress+exercise group were allowed to voluntarily run on a wheel for 2h after every SDS exposure. Two weeks later, compared to the control group, the stress group showed a higher ratio of time spent in the corner zone of a social interaction paradigm even though SDS did not elicit depressive- and anxiety-like behaviors. We also observed that voluntary exercise, which did not affect muscle weight and gene expression, decreased social avoidance behavior of stressed mice without clear changes in brain monoamine levels. Interestingly, food intake in the stress+exercise group was the greatest among the three groups. To test the effect of the exercise-induced increase in food intake on social behavior, we set up a pair-fed group where food intake was restricted. We then compared these mice to mice in the stress alone group. We found that the ratio of time spent in the corner zone of the social interaction test was not different between ad libitum- and pair-fed groups, although pair-fed mice spent more time in the corner zone when an aggressor mouse was present than when it was absent. In addition, pair-feeding did not show exercise-induced reductions of adrenal gland weight and enhanced the loss of body fat. Our findings indicate that voluntary exercise reduces social avoidance behavior induced by SDS. Further, we determined that SDS and exercise-induced increases in food intake partially influence energy metabolism and social avoidance behavior.


Subject(s)
Eating , Motor Activity , Running/psychology , Social Behavior , Stress, Psychological/physiopathology , Animals , Anxiety/physiopathology , Brain/metabolism , Depression/physiopathology , Disease Models, Animal , Dominance-Subordination , Dopamine/metabolism , Eating/physiology , Gene Expression/physiology , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Motor Activity/physiology , Running/physiology , Serotonin/metabolism , Volition
15.
J Med Invest ; 61(1-2): 79-83, 2014.
Article in English | MEDLINE | ID: mdl-24705753

ABSTRACT

It has been reported that negative emotional changes and conditions affect the visual faculties of humans at the neural level. On the other hand, the effects of emotion on color perception in particular, which are based on evoked potentials, are unknown. In the present study, we investigated whether different anxiety levels affect the color information processing for each of 3 wavelengths by using flash visual evoked potentials (FVEPs) and State-Trait Anxiety Inventory. In results, significant positive correlations were observed between FVEP amplitudes and state or trait anxiety scores in the long (sensed as red) and middle (sensed as green) wavelengths. On the other hand, short-wavelength-evoked FVEPs were not correlated with anxiety level. Our results suggest that negative emotional conditions may affect color sense processing in humans.


Subject(s)
Anxiety/physiopathology , Color Perception/physiology , Color , Evoked Potentials, Visual/physiology , Adult , Anxiety/psychology , Emotions/physiology , Humans , Intelligence Tests , Male , Severity of Illness Index
16.
Neuropharmacology ; 79: 399-404, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24361452

ABSTRACT

A link has been established between energy metabolism and sleep homeostasis. The ketone bodies acetoacetate and ß-hydroxybutyrate, generated from the breakdown of fatty acids, are major metabolic fuels for the brain under conditions of low glucose availability. Ketogenesis is modulated by the activity of peroxisome proliferator-activated receptor alpha (PPARα), and treatment with a PPAR activator has been shown to induce a marked increase in plasma acetoacetate and decreased ß-hydroxybutyrate in mice, accompanied by increased slow-wave activity during non-rapid eye movement (NREM) sleep. The present study investigated the role of ketone bodies in sleep regulation. Six-hour sleep deprivation increased plasma ketone bodies and their ratio (acetoacetate/ß-hydroxybutyrate) in 10-week-old male mice. Moreover, sleep deprivation increased mRNA expression of ketogenic genes such as PPARα and 3-hydroxy-3-methylglutarate-CoA synthase 2 in the brain and decreased ketolytic enzymes such as succinyl-CoA: 3-oxoacid CoA transferase. In addition, central injection of acetoacetate, but not ß-hydroxybutyrate, markedly increased slow-wave activity during NREM sleep and suppressed glutamate release. Central metabolism of ketone bodies, especially acetoacetate, appears to play a role in the regulation of sleep homeostasis.


Subject(s)
Homeostasis/physiology , Ketone Bodies/metabolism , Sleep Deprivation/physiopathology , Sleep/physiology , 3-Hydroxybutyric Acid/metabolism , Acetoacetates/metabolism , Animals , Brain/physiopathology , Food Deprivation/physiology , Gene Expression/physiology , Glutamic Acid/metabolism , Liver/physiopathology , Male , Mice , Mice, Inbred ICR , PPAR alpha/metabolism , RNA, Messenger/metabolism , Sleep Stages/physiology
17.
PLoS One ; 8(10): e78434, 2013.
Article in English | MEDLINE | ID: mdl-24205232

ABSTRACT

Mast cell activation and degranulation can result in the release of various chemical mediators, such as histamine and cytokines, which significantly affect sleep. Mast cells also exist in the central nervous system (CNS). Since up to 50% of histamine contents in the brain are from brain mast cells, mediators from brain mast cells may significantly influence sleep and other behaviors. In this study, we examined potential involvement of brain mast cells in sleep/wake regulations, focusing especially on the histaminergic system, using mast cell deficient (W/W(v)) mice. No significant difference was found in the basal amount of sleep/wake between W/W(v) mice and their wild-type littermates (WT), although W/W(v) mice showed increased EEG delta power and attenuated rebound response after sleep deprivation. Intracerebroventricular injection of compound 48/80, a histamine releaser from mast cells, significantly increased histamine levels in the ventricular region and enhanced wakefulness in WT mice, while it had no effect in W/W(v) mice. Injection of H1 antagonists (triprolidine and mepyramine) significantly increased the amounts of slow-wave sleep in WT mice, but not in W/W(v) mice. Most strikingly, the food-seeking behavior observed in WT mice during food deprivation was completely abolished in W/W(v) mice. W/W(v) mice also exhibited higher anxiety and depression levels compared to WT mice. Our findings suggest that histamine released from brain mast cells is wake-promoting, and emphasizes the physiological and pharmacological importance of brain mast cells in the regulation of sleep and fundamental neurobehavior.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , Brain/physiology , Histamine Release/physiology , Histamine/metabolism , Mast Cells/physiology , Wakefulness/physiology , Animals , Electroencephalography/methods , Male , Mast Cells/metabolism , Mice , Sleep/physiology , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology
18.
PLoS One ; 8(5): e64263, 2013.
Article in English | MEDLINE | ID: mdl-23741310

ABSTRACT

Nutritional state in the gestation period influences fetal growth and development. We hypothesized that undernutrition during gestation would affect offspring sleep architecture and/or homeostasis. Pregnant female mice were assigned to either control (fed ad libitum; AD) or 50% dietary restriction (DR) groups from gestation day 12 to parturition. After parturition, dams were fed AD chow. After weaning, the pups were also fed AD into adulthood. At adulthood (aged 8-9 weeks), we carried out sleep recordings. Although offspring mice displayed a significantly reduced body weight at birth, their weights recovered three days after birth. Enhancement of electroencephalogram (EEG) slow wave activity (SWA) during non-rapid eye movement (NREM) sleep was observed in the DR mice over a 24-hour period without changing the diurnal pattern or amounts of wake, NREM, or rapid eye movement (REM) sleep. In addition, DR mice also displayed an enhancement of EEG-SWA rebound after a 6-hour sleep deprivation and a higher threshold for waking in the face of external stimuli. DR adult offspring mice exhibited small but significant increases in the expression of hypothalamic peroxisome proliferator-activated receptor α (Pparα) and brain-specific carnitine palmitoyltransferase 1 (Cpt1c) mRNA, two genes involved in lipid metabolism. Undernutrition during pregnancy may influence sleep homeostasis, with offspring exhibiting greater sleep pressure.


Subject(s)
Caloric Restriction , Diet , Hypothalamus/physiology , Prenatal Exposure Delayed Effects/physiopathology , Sleep/physiology , Animals , Body Weight , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Electroencephalography , Female , Gene Expression , Homeostasis , Mice , PPAR alpha/genetics , PPAR alpha/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Sleep, REM , Wakefulness
19.
Genes Cells ; 18(6): 502-18, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23621645

ABSTRACT

Identifying the stages of sleep, or sleep staging, is an unavoidable step in sleep research and typically requires visual inspection of electroencephalography (EEG) and electromyography (EMG) data. Currently, scoring is slow, biased and prone to error by humans and thus is the most important bottleneck for large-scale sleep research in animals. We have developed an unsupervised, fully automated sleep staging method for mice that allows less subjective and high-throughput evaluation of sleep. Fully Automated Sleep sTaging method via EEG/EMG Recordings (FASTER) is based on nonparametric density estimation clustering of comprehensive EEG/EMG power spectra. FASTER can accurately identify sleep patterns in mice that have been perturbed by drugs or by genetic modification of a clock gene. The overall accuracy is over 90% in every group. 24-h data are staged by a laptop computer in 10 min, which is faster than an experienced human rater. Dramatically improving the sleep staging process in both quality and throughput FASTER will open the door to quantitative and comprehensive animal sleep research.


Subject(s)
Electroencephalography , Electromyography , Electronic Data Processing/methods , Sleep Stages/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Neuropharmacology ; 72: 58-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23624141

ABSTRACT

The retinoic acid (RA, a vitamin A metabolite) receptor (RAR) is a transcription factor. Vitamin A/RA administration improves the Alzheimer's disease (AD)- and age-related attenuation of memory/learning in mouse models. Recently, a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) was identified as a key molecule in RA-mediated anti-AD mechanisms. We investigated the effect of chronic administration of the RAR agonist Am80 (tamibarotene) on ADAM10 expression in senescence-accelerated mice (SAMP8). Moreover, we estimated changes in the expression of the amyloid precursor protein (APP), amyloid beta (Aß), and hairy/enhancer of split (Hes), which are mediated by ADAM10. Spatial working memory and the levels of a hippocampal proliferation marker (Ki67) were also assessed in these mice. ADAM10 mRNA and protein expression was significantly reduced in the hippocampus of 13-month-old SAMP8 mice; their expression improved significantly after Am80 administration. Further, after Am80 administration, the expression levels of Hes5 and Ki67 were restored and the deterioration of working memory was suppressed, whereas APP and Aß levels remained unchanged. Our results suggest that Am80 administration effectively improves dementia by activating the hippocampal ADAM10-Notch-Hes5 proliferative pathway.


Subject(s)
ADAM Proteins/metabolism , Aging/genetics , Amyloid Precursor Protein Secretases/metabolism , Benzoates/pharmacology , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Membrane Proteins/metabolism , Tetrahydronaphthalenes/pharmacology , ADAM Proteins/genetics , ADAM10 Protein , Amyloid Precursor Protein Secretases/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzoates/chemistry , Cell Proliferation/drug effects , Gene Expression Regulation/genetics , Ki-67 Antigen/metabolism , Maze Learning/drug effects , Membrane Proteins/genetics , Memory, Short-Term/drug effects , Mice , Mice, Mutant Strains , RNA, Messenger/metabolism , Receptors, Retinoic Acid/agonists , Repressor Proteins/metabolism , Tetrahydronaphthalenes/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL