Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Stress ; 26(1): 21-28, 2023 01.
Article in English | MEDLINE | ID: mdl-36522611

ABSTRACT

Chronic sleep disorders (CSD) comprise a potential risk factor for metabolic and cardiovascular diseases, obesity and stroke. Thus, the identification of biomarkers for CSD is an important step in the early prevention of metabolic dysfunctions induced by sleep dysfunction. Diagnostic saliva samples can be easily and noninvasively collected. Thus, we aimed to identify whole microRNA (miRNA) profiles of saliva in control and psychophysiologically stressed CSD mouse models and compare them at Zeitgeber time (ZT) 0 (lights on) and ZT12 (lights off). The findings of two-way ANOVA revealed that the expression of 342 and 109 salivary miRNAs was affected by CSD and the time of day, respectively. Interactions were found in 122 miRNAs among which, we identified 197 (ZT0) and 62 (ZT12) upregulated, and 40 (ZT0) and seven (ZT12) downregulated miRNAs in CSD mice. We showed that miR-30c-5p, which is elevated in the plasma of patients with hypersomnia, was upregulated in the saliva of CSD mice collected at ZT0. The miRNAs, miR-10a-5p, miR-146b-5p, miR-150-5p, and miR-25-3p are upregulated in the serum of humans with poor sleep quality, and these were also upregulated in the saliva of CSD mice collected at ZT0. The miRNAs miR-30c, miR146b-5p, miR150, and miR-25-5p are associated with cardiovascular diseases, and we found that plasma concentrations of brain natriuretic peptides were significantly increased in CSD mice. The present findings showed that salivary miRNA profiles could serve as useful biomarkers for predicting CSD.


Subject(s)
Cardiovascular Diseases , MicroRNAs , Sleep Wake Disorders , Humans , Male , Mice , Animals , Stress, Psychological , MicroRNAs/genetics , Biomarkers , Disease Models, Animal , Sleep
2.
FASEB J ; 35(9): e21861, 2021 09.
Article in English | MEDLINE | ID: mdl-34416029

ABSTRACT

Duchenne muscular dystrophy (DMD) is an intractable genetic disease associated with progressive skeletal muscle weakness and degeneration. Recently, it was reported that intraperitoneal injections of ketone bodies partially ameliorated muscular dystrophy by increasing satellite cell (SC) proliferation. Here, we evaluated whether a ketogenic diet (KD) with medium-chain triglycerides (MCT-KD) could alter genetically mutated DMD in model rats. We found that the MCT-KD significantly increased muscle strength and fiber diameter in these rats. The MCT-KD significantly suppressed the key features of DMD, namely, muscle necrosis, inflammation, and subsequent fibrosis. Immunocytochemical analysis revealed that the MCT-KD promoted the proliferation of muscle SCs, suggesting enhanced muscle regeneration. The muscle strength of DMD model rats fed with MCT-KD was significantly improved even at the age of 9 months. Our findings suggested that the MCT-KD ameliorates muscular dystrophy by inhibiting myonecrosis and promoting the proliferation of muscle SCs. As far as we can ascertain, this is the first study to apply a functional diet as therapy for DMD in experimental animals. Further studies are needed to elucidate the underlying mechanisms of the MCT-KD-induced improvement of DMD.


Subject(s)
Diet, Ketogenic , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Muscular Dystrophy, Duchenne/diet therapy , Muscular Dystrophy, Duchenne/physiopathology , Triglycerides/chemistry , Triglycerides/pharmacology , Animals , Body Weight/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Disease Progression , Female , Fibrosis/diet therapy , Fibrosis/pathology , Inflammation/diet therapy , Inflammation/pathology , Ketones/blood , Ketosis , Male , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/pathology , Necrosis/diet therapy , Necrosis/pathology , Rats , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Triglycerides/therapeutic use
3.
Int J Mol Sci ; 23(22)2022 Nov 21.
Article in English | MEDLINE | ID: mdl-36430957

ABSTRACT

Identification of early biomarkers of stress is important for preventing mood and anxiety disorders. Saliva is an easy-to-collect and non-invasive diagnostic target. The aim of this study was to characterize the changes in salivary whole microRNAs (miRNAs) and metabolites in mice subjected to subchronic and mild social defeat stress (sCSDS). In this study, we identified seven upregulated and one downregulated miRNAs/PIWI-interacting RNA (piRNA) in the saliva of sCSDS mice. One of them, miR-208b-3p, which is reported as a reliable marker for myocardial infarction, was upregulated in the saliva of sCSDS mice. Histological analysis showed frequent myocardial interstitial fibrosis in the heart of such mice. In addition, gene ontology and pathway analyses suggested that the pathways related to energy metabolism, such as the oxidative phosphorylation and the pentose phosphate pathway, were significantly related to the miRNAs affected by sCSDS in saliva. In contrast, salivary metabolites were not significantly changed in the sCSDS mice, which is consistent with our previous metabolomic study on the plasma of sCSDS mice. Taken in the light of previous studies, the present study provides novel potential stress biomarkers for future diagnosis using saliva.


Subject(s)
MicroRNAs , Social Defeat , Mice , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mice, Inbred C57BL , Stress, Psychological/genetics , Stress, Psychological/metabolism , Metabolome , Disease Models, Animal , Biomarkers/metabolism
4.
Biochem Biophys Res Commun ; 529(2): 175-179, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703407

ABSTRACT

Sleep disturbances can contribute to cognitive decline and neuropsychiatric disorders. However, the underlying mechanisms of these processes are poorly understood. The present study evaluated the effects of a chronic sleep disorder (CSD) on long-term memory formation and anxiety-like behavior in our originally established mouse model of psychophysiological stress-induced CSD characterized by disrupted circadian rhythms of wheel-running activity and sleep-wake cycles. Model mice are continuously exposed to mild stress imposed by perpetually staying on a running-wheel to avoid water. The findings of novel object recognition (NORT) and open field (OFT) tests showed that CSD impaired recognition memory and elicited anxiety-like behavior, respectively. These results suggested that the CSD impaired cognitive function and emotional status. Thus, this CSD model could be useful for studying the underlying mechanisms of neurobehavioral difficulties caused by sleep disorders. We then examined the hippocampal mRNA expression of genes associated with learning and memory, and anxiety and depression. The CSD increased the mRNA expression of Crhr1, Ngf and Phlpp1, and suppressed that of Ace, Egr2 and Slc6a4. Based on the functions of these genes, we inferred that the increase in Crhr1 mRNA was associated with the pathogenesis of psychiatric conditions, whereas mRNA levels of the other five genes were directed towards symptom relief. Upregulating hippocampal Crhr1 expression might contribute in part to the activation of corticotropin-releasing hormone (CRH)-CRH receptor1 signaling that mediates CSD-evoked mental disorders.


Subject(s)
Anxiety/etiology , Cognitive Dysfunction/etiology , Memory, Long-Term , Sleep Wake Disorders/complications , Animals , Anxiety/physiopathology , Chronic Disease , Circadian Rhythm , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Male , Mice , Open Field Test , Sleep Wake Disorders/physiopathology , Stress, Psychological/complications , Stress, Psychological/physiopathology
5.
Biochem Biophys Res Commun ; 524(1): 129-134, 2020 03 26.
Article in English | MEDLINE | ID: mdl-31980181

ABSTRACT

Acute or chronic effects of consuming or skipping breakfast on cognitive performance in humans are controversial. To evaluate the effects of chronically skipping breakfast (SB) on hippocampus-dependent long-term memory formation, we examined hippocampal gene expression and applied the novel object recognition test (NORT) after two weeks of repeated fasting for six hours from lights off to mimic SB in mice. We also examined the effects of SB on circadian rhythms of locomotor activity, food intake, core body temperature (CBT) and sleep-wake cycles. Skipping breakfast slightly but significantly decreased total daily food intake without affecting body weight gain. Locomotor activity and CBT significantly decreased during the fasting period under SB. The degree of fasting-dependent CBT reduction gradually increased and then became stabilized after four days of SB. Electroencephalographic data revealed that repeated SB significantly decreased the duration of wakefulness and increased that of rapid eye movement (REM) and of non-REM (NREM) sleep during the period of SB. Furthermore, total daily amounts of wakefulness and NREM sleep were significantly decreased and increased, respectively, under SB, suggesting that SB disrupts sleep homeostasis. Skipping breakfast significantly suppressed mRNA expression of the memory-related genes, Camk2a, Fkbp5, Gadd45b, Gria1, Sirt1 and Tet1 in the hippocampus. Recognition memory assessed by NORT was impaired by SB in accordance with the gene expression profiles. These findings suggested that chronic SB causes dysregulated CBT, sleep-wake cycles and hippocampal gene expression, which results in impaired long-term memory formation.


Subject(s)
Body Temperature/physiology , Breakfast/physiology , Eating/physiology , Hippocampus/metabolism , Memory/physiology , Wakefulness/physiology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Circadian Rhythm/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fasting , Gene Expression Regulation , Homeostasis , Male , Memory, Long-Term/physiology , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sleep, REM/physiology , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
6.
Arch Biochem Biophys ; 677: 108160, 2019 11 30.
Article in English | MEDLINE | ID: mdl-31639326

ABSTRACT

Skeletal muscle mass is largely influenced by nutritional status and physical activity. Although feeding at specific times of the day (time-restricted feeding, TRF) modulates obesity and other metabolic functions, its effects on skeletal muscles remain unclear. We explored the effects of feeding mice only during the inactive (daytime feeding, DF) or active (nighttime feeding, NF) phases for one week. Daytime feeding did not abolish the nocturnal activity rhythm, although total daily activity was reduced in these mice. Temporal expression of the circadian clock genes, Per2 and Rev-erbα, became synchronized to the feeding cycle in the liver, but not in skeletal muscle. Skeletal muscle mass, grip strength, and cross-sectional area were significantly lower in DF, than in NF mice, although DF increased body weight gain and lipid accumulation. Expression of the atrophy-related ubiquitin ligases, Atrogin-1 and Murf1 and the autophagy-related genes, Lc3b and Bnip3, was induced during the active phase in the gastrocnemius muscles of DF, compared with those of NF mice. Plasma IGF-1 concentrations and Igf-1 expression in the livers and gastrocnemius muscles during the active phase were lower in DF, than in NF mice. Furthermore, exogenous IGF-1 injection significantly suppressed DF-induced reduction in gastrocnemius muscle mass, which might at least partly explain the association between decreased plasma IGF-1 concentrations and reductions in the skeletal muscle mass of DF mice. These findings suggest that feeding only during the inactive phase reduces skeletal muscle mass via a decrease in plasma IGF-1 concentrations during the active phase.


Subject(s)
Food Deprivation/physiology , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Animals , Circadian Clocks/genetics , Hand Strength , Insulin/metabolism , Male , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscular Atrophy/metabolism , Organ Size , RNA, Messenger/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction/physiology , Time Factors , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics
7.
Biol Pharm Bull ; 42(6): 1038-1043, 2019.
Article in English | MEDLINE | ID: mdl-31155580

ABSTRACT

Consuming food at uncommon times during the day might be associated with obesity in experimental animals and humans. We previously reported that mice become obese and their metabolism becomes disrupted when they consume food during the daytime (sleep phase feeding; SPF), but not during the nighttime (active phase feeding; APF). The goal of the present study was to clarify whether the molecular circadian clock is associated with the mechanisms that underly the metabolic disorders in mice brought about by SPF. We compared the effects of dominant negative Clock gene mutation on metabolic disruption and obesity brought about by SPF in mice. The consumption of food during SP increased body weight, adipose tissue mass and lipogenic gene expression in metabolic tissues, as well as hyperinsulinemia, hyperleptinemia and hepatic lipid accumulation in wild-type and Clock mutant mice, and there were no significant differences between genotypes except for the body weight increase which was attenuated by the Clock mutation. Temporal expression of Per2 was synchronized to feeding rhythms in the liver of both genotypes, although the expression of Dbp, a representative clock-controlled gene, was significantly damped in peripheral tissues of Clock mutant mice. These findings suggest that the molecular clock is not essentially associated with metabolic disruption caused by SPF. Desynchronized food consumption and central clock-dependent behaviour as well as rhythmic metabolic mechanisms might be associated with the metabolic disruption caused by SPF.


Subject(s)
CLOCK Proteins/genetics , Circadian Clocks , Feeding Behavior , Sleep , Adipose Tissue, White/metabolism , Animals , Body Weight , Lipid Metabolism , Liver/metabolism , Male , Mice, Inbred ICR , Mice, Knockout
8.
Biochem Biophys Res Commun ; 495(4): 2616-2621, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29288667

ABSTRACT

Sleep disturbances are associated with various metabolic diseases such as hypertension and diabetes. We had previously established a mouse model of a psychophysiological stress-induced chronic sleep disorder (CSD) characterized by disrupted circadian rhythms of wheel-running activity, core body temperature, and sleep-wake cycles. To evaluate the underlying mechanisms of metabolic disorders induced by CSD, we created mice with CSD for six weeks and fed them with a high-fat diet. Glucose intolerance with hyperglycemia resulted, although plasma insulin levels and body weight increases were identical between control and CSD mice. Gluconeogenesis and glycolysis were enhanced and suppressed, respectively, in the livers of CSD mice, because the mRNA expression of Pck1 was significantly increased, whereas that of Gck and Pklr were significantly decreased in the CSD mice. Adipose inflammation induced by the high-fat diet seemed suppressed by the CSD, because the mRNA expression levels of Adgre1, Ccl2, and Tnf were significantly downregulated in the adipose tissues of CSD mice. These findings suggest that CSD impair glucose tolerance by inducing gluconeogenesis and suppressing glycolysis. Hyperphasia with hypoleptinemia, hypercorticosteronemia, and increased plasma free fatty acids might be involved in the impaired glucose metabolism under a CSD. Further studies are needed to elucidate the endocrine and molecular mechanisms underlying the associations between sleep disorders and impaired glucose homeostasis that consequently causes diabetes.


Subject(s)
Glucose Intolerance/etiology , Glucose Intolerance/physiopathology , Sleep Wake Disorders/etiology , Sleep Wake Disorders/physiopathology , Stress, Psychological/complications , Stress, Psychological/physiopathology , Animals , Chronic Disease , Cytokines/metabolism , Male , Mice , Panniculitis/etiology , Panniculitis/physiopathology
9.
Int J Mol Sci ; 19(9)2018 Sep 18.
Article in English | MEDLINE | ID: mdl-30231537

ABSTRACT

Brain and muscle arnt-like protein 1 (BMAL1), is a transcription factor known to regulate circadian rhythm. BMAL1 was originally characterized by its high expression in the skeletal muscle. Since the skeletal muscle is the dominant organ system in energy metabolism, the possible functions of BMAL1 in the skeletal muscle include the control of metabolism. Here, we established that its involvement in the regulation of oxidative capacity in the skeletal muscle. Muscle-specific Bmal1 KO mice (MKO mice) displayed several physiological hallmarks for the increase of oxidative capacity. This included increased energy expenditure and oxygen consumption, high running endurance and resistance to obesity with improved metabolic profiles. Also, the phosphorylation status of AMP-activated protein kinase and its downstream signaling substrate acetyl-CoA carboxylase in the MKO mice were substantially higher than those in the Bmal1flox/flox mice. In addition, biochemical and histological studies confirmed the substantial activation of oxidative fibers in the skeletal muscle of the MKO mice. The mechanism includes the regulation of Cacna1s expression, followed by the activation of calcium-nuclear factor of activated T cells (NFAT) axis. We thus conclude that BMAL1 is a critical regulator of the muscular fatty acid level under nutrition overloading and that the mechanism involves the control of oxidative capacity.


Subject(s)
ARNTL Transcription Factors/genetics , Fats/metabolism , Gene Deletion , Muscle, Skeletal/metabolism , Obesity/genetics , Oxidative Stress , ARNTL Transcription Factors/metabolism , Animals , Diet, High-Fat/adverse effects , Insulin Resistance , Locomotion , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology
10.
Biochem Biophys Res Commun ; 483(1): 165-170, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28042033

ABSTRACT

The master clock in the suprachiasmatic nucleus synchronizes peripheral clocks via humoral and neural signals in mammals. Insulin is thought to be a critical Zeitgeber (synchronizer) for peripheral clocks because it induces transient clock gene expression in cultured cells. However, the extent to which fluctuations in feeding-dependent endogenous insulin affect the temporal expression of clock genes remains unclear. We therefore investigated the temporal expression profiles of clock genes in the peripheral tissues of mice fed for 8 h during either the daytime (DF) or the nighttime (NF) for one week to determine the involvement of feeding cycle-dependent endogenous insulin rhythms in the circadian regulation of peripheral clocks. The phase of circulating insulin fluctuations was reversed in DF compared with NF mice, although those of circulating corticosterone fluctuations and nocturnal locomotor activity were identical between these mice. The reversed feeding cycle affected the circadian phases of Per1 and Per2 gene expression in the liver and not in heart, lung, white adipose and skeletal muscle tissues. On the other hand, injected exogenous insulin significantly induced Akt phosphorylation in the heart and skeletal muscle as well as the liver, and significantly induced Per1 and Per2 gene expression in all examined tissues. These findings suggest that feeding cycles and feeding cycle-dependent endogenous insulin fluctuations are not dominant entrainment signals for peripheral clocks other than the liver, although exogenous insulin might reset peripheral oscillators in mammals.


Subject(s)
Circadian Clocks/genetics , Feeding Behavior/physiology , Insulin/blood , Animals , Corticosterone/blood , DNA-Binding Proteins/genetics , Gene Expression Regulation , Glucagon-Like Peptide 1/blood , Insulin/pharmacology , Liver/physiology , Male , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Transcription Factors/genetics
11.
Mol Genet Metab ; 121(2): 190-197, 2017 06.
Article in English | MEDLINE | ID: mdl-28410879

ABSTRACT

Real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis is a popular method for the measurement of mRNA expression level and is a critical tool for basic research. The identification of suitable reference genes that are stable and not affected by experimental conditions is a critical step in the accurate normalization of RT-PCR. On the other hand, the levels of numerous transcripts exhibit circadian oscillation in various peripheral tissues and it is thought to be regulated by feeding rhythms in addition to the molecular circadian clock. Here, we investigated the effects of feeding schedule on the temporal expression profiles of 13 common housekeeping genes in metabolic tissues of mice fed during either the sleep or the active phase. The expression of most of these genes fluctuated dependently on feeding rhythms in the liver and WAT, but not in skeletal muscle. Two-way analyses of variance (ANOVA) identified 18S ribosomal RNA (Rn18s) as the only gene that was stably expressed throughout the day independently of feeding schedules in the liver and WAT, although RefFinder software showed that peptidylprolyl isomerase A (Ppia) was the most stably expressed housekeeping gene. Both ANOVA and RefFinder software determined that Actb was the preferred reference gene for skeletal muscle. Furthermore, NormFinder proposed that the optimal pairs of reference genes were beta-2 microglobulin (B2m)-Ppia in the liver, Ppia-TATA box binding protein (Tbp) in WAT, and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide (Ywhaz)-glyceraldehyde-3-phosphate dehydrogenase (Gapdh) in skeletal muscle, and that their stability value was better than that of a single stable gene. The appropriate reference gene pairs for normalizing genes of interest in mouse circadian studies are B2m-Ppia in the liver, Ppia-Tbp in WAT, and Ywhaz-Gapdh in skeletal muscle.


Subject(s)
Circadian Clocks/genetics , Feeding Behavior , Gene Expression , Genes, Essential , Animals , Circadian Clocks/physiology , Gene Expression Profiling/methods , Liver/physiology , Mice , Muscle, Skeletal/physiology , RNA, Messenger , RNA, Ribosomal, 18S/genetics , Real-Time Polymerase Chain Reaction
12.
J Biol Chem ; 289(52): 36158-65, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25384980

ABSTRACT

The circadian clock regulates various behavioral and physiological rhythms in mammals. Circadian changes in olfactory functions such as neuronal firing in the olfactory bulb (OB) and olfactory sensitivity have recently been identified, although the underlying molecular mechanisms remain unknown. We analyzed the temporal profiles of glycan structures in the mouse OB using a high-density microarray that includes 96 lectins, because glycoconjugates play important roles in the nervous system such as neurite outgrowth and synaptogenesis. Sixteen lectin signals significantly fluctuated in the OB, and the intensity of all three that had high affinity for α1-2-fucose (α1-2Fuc) glycan in the microarray was higher during the nighttime. Histochemical analysis revealed that α1-2Fuc glycan is located in a diurnal manner in the lateral olfactory tract that comprises axon bundles of secondary olfactory neurons. The amount of α1-2Fuc glycan associated with the major target glycoprotein neural cell adhesion molecule (NCAM) varied in a diurnal fashion, although the mRNA and protein expression of Ncam1 did not. The mRNA and protein expression of Fut1, a α1-2-specific fucosyltransferase gene, was diurnal in the OB. Daily fluctuation of the α1-2Fuc glycan was obviously damped in homozygous Clock mutant mice with disrupted diurnal Fut1 expression, suggesting that the molecular clock governs rhythmic α1-2-fucosylation in secondary olfactory neurons. These findings suggest the possibility that the molecular clock is involved in the diurnal regulation of olfaction via α1-2-fucosylation in the olfactory system.


Subject(s)
CD56 Antigen/metabolism , CLOCK Proteins/genetics , Olfactory Receptor Neurons/metabolism , Animals , Circadian Rhythm , Fucose/metabolism , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Gene Expression , Gene Expression Regulation , Glycosylation , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Olfactory Nerve/cytology , Protein Processing, Post-Translational , Galactoside 2-alpha-L-fucosyltransferase
13.
Biochem Biophys Res Commun ; 458(2): 256-61, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25645021

ABSTRACT

The environmental light-dark (LD) cycle entrains the central circadian clock located in the suprachiasmatic nucleus (SCN) of mammals. The present study examined the effects of disrupted LD cycles on peripheral clocks in mice housed under a normal 12 h light-12 h dark cycle (LD 12:12) or an ultradian LD 3:3 cycle. Drinking behavior seemed to be free-running with a long period (26.03 h) under ultradian LD 3:3 cycles, in addition to light-induced direct suppression (masking effect). Core body temperature completely lost robust circadian rhythm and acquired a 6-h rhythm with a low amplitude under LD 3:3. Robust circadian expression of Per1, Per2, Clock and Bmal1 mRNAs was similarly flattened to intermediate levels in the liver, heart and white adipose tissue under LD 3:3. Robust circadian expression of Rev-erbα mRNA was completely damped in these tissues. Circadian expression of Dbp, a clock-controlled gene, was also disrupted in these tissues from mice housed under LD 3:3. The aberrant LD cycle seemed to induce the loss of circadian gene expression at the level of transcription, because rhythmic pre-mRNA expression of these genes was also abolished under LD 3:3. In addition to the direct effect of the aberrant LD cycle, abolished systemic time cues such as those of plasma corticosterone and body temperature might be involved in the disrupted expression of these circadian genes under LD 3:3. Our findings suggest that disrupted environmental LD cycles abolish the normal oscillation of peripheral clocks and induce internal desynchrony in mammals.


Subject(s)
Behavior, Animal/physiology , Body Temperature/physiology , Circadian Clocks/physiology , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Motor Activity/physiology , Photoperiod , Animals , Biological Clocks/physiology , Down-Regulation/physiology , Mice , Mice, Inbred ICR , Organ Specificity , Tissue Distribution
14.
J Nutr ; 145(2): 199-206, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25644338

ABSTRACT

BACKGROUND: Epidemiologic studies have shown that the consumption of whole grains can reduce the risk of type 2 diabetes mellitus, cardiovascular disease, and all-cause mortality. However, the underlying mechanisms remain a matter of debate. OBJECTIVE: We aimed to determine the effects of wheat bran-derived alkylresorcinols on diet-induced metabolic disorders in mice. METHODS: We fed C57BL/6J mice a normal refined diet or a high-fat, high-sucrose diet [29.1% fat, 20.7% protein, 34.0% carbohydrates containing 20.0% sucrose (w/w)] alone (FS) or containing 0.4% (wt:wt) alkylresorcinols (FS-AR) for 10 wk. RESULTS: The alkylresorcinols suppressed FS-induced increases in body weight by 31.0% as well as FS-induced hepatic triglyceride accumulation (means ± SEMs: 29.6 ± 3.18 and 19.8 ± 2.42 mg/g tissue in the FS and FS-AR groups, respectively), without affecting energy intake. We measured circadian changes in blood metabolic hormones and found that FS-induced hyperinsulinemia (5.1 and 2.1 µg/L at night in the FS and FS-AR groups, respectively) and hyperleptinemia (21.6 and 10.8 µg/L at night in the FS and FS-AR groups, respectively) were suppressed by alkylresorcinols. Glucose and insulin tolerance tests showed that alkylresorcinols significantly reduced fasting blood glucose concentrations (190 ± 3.62 and 160 ± 8.98 mg/dL in the FS and FS-AR groups, respectively) and suppressed glucose intolerance as well as insulin resistance induced by the FS diet. Furthermore, alkylresorcinols significantly increased insulin-stimulated hepatic serine/threonine protein kinase B phosphorylation compared to the FS diet (+81.3% and +57.4% for Ser473 and Thr308, respectively). On the other hand, pyruvate and starch tolerance tests suggested that alkylresorcinols did not affect gluconeogenesis and carbohydrate digestion, respectively. Alkylresorcinols significantly increased fecal cholesterol excretion by 39.6% and reduced blood cholesterol concentrations by 30.4%, while upregulating the expression of hepatic cholesterol synthetic genes such as sterol regulatory element binding protein 2 (Srebf2) and 3-hydroxy-3-methylglutaryl-Coenzyme A synthase 1 (Hmgcs1). CONCLUSIONS: These findings suggest that wheat alkylresorcinols increase glucose tolerance and insulin sensitivity by suppressing hepatic lipid accumulation and intestinal cholesterol absorption, which subsequently suppresses diet-induced obesity in mice.


Subject(s)
Diet, High-Fat/adverse effects , Glucose Intolerance/drug therapy , Obesity/drug therapy , Resorcinols/pharmacology , Sucrose/administration & dosage , Triticum/chemistry , Animals , Blood Glucose/metabolism , Cholesterol/blood , Dietary Carbohydrates/administration & dosage , Dietary Fiber/pharmacology , Energy Intake , Feces/chemistry , Hyperinsulinism/drug therapy , Insulin/blood , Insulin Resistance , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Triglycerides/blood
15.
Muscle Nerve ; 51(2): 276-81, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24910410

ABSTRACT

INTRODUCTION: Immobilization induced by experimental denervation leads to rapid and progressive alterations in structural and biochemical properties of skeletal muscle. Real-time reverse transcription-polymerase chain reaction (RT-PCR) is a popular method of elucidating the molecular mechanisms involved in muscle atrophy. Identification of suitable reference genes that are not affected by experimental conditions is a critical step in accurate normalization of real-time RT-PCR. METHODS: We investigated the impact of denervation-induced muscle atrophy for 2 weeks on the expression of common housekeeping genes. RESULTS: Denervation differentially affected the expression levels of these genes. RefFinder software identified TATA box binding protein (Tbp) as the most stable gene and showed that the stability of glyceraldehyde-3-phosphate dehydrogenase (Gapdh) and hypoxanthine guanine phosphoribosyl transferase (Hprt) genes was low, even though they are widely used for normalization. CONCLUSIONS: The appropriate reference gene for normalization of genes of interest in denervated muscle is Tbp.


Subject(s)
Gene Expression Regulation/physiology , Genes, Essential/genetics , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Sciatic Neuropathy/complications , TATA-Box Binding Protein/metabolism , Analysis of Variance , Animals , Denervation/adverse effects , Disease Models, Animal , Functional Laterality , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , RNA, Messenger/metabolism , TATA-Box Binding Protein/genetics , Time Factors
16.
Biochem Biophys Res Commun ; 450(1): 880-4, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24971530

ABSTRACT

Disordered circadian rhythms are associated with various psychiatric conditions and metabolic diseases. We recently established a mouse model of a psychophysiological stress-induced chronic sleep disorder (CSD) characterized by reduced amplitude of circadian wheel-running activity and sleep-wake cycles, sleep fragmentation and hyperphagia. Here, we evaluate day-night fluctuations in plasma concentrations of free amino acids (FAA), appetite hormones and prolactin as well as the hepatic expression of circadian clock-related genes in mice with CSD (CSD mice). Nocturnal increases in wheel-running activity and circadian rhythms of plasma prolactin concentrations were significantly disrupted in CSD mice. Hyperphagia with a decreased leptin/ghrelin ratio was found in CSD mice. Day-night fluctuations in plasma FAA contents were severely disrupted without affecting total FAA levels in CSD mice. Nocturnal increases in branched-chain amino acids such as Ile, Leu, and Val were further augmented in CSD mice, while daytime increases in Gly, Ala, Ser, Thr, Lys, Arg, His, Tyr, Met, Cys, Glu, and Asn were significantly attenuated. Importantly, the circadian expression of hepatic clock genes was completely unaffected in CSD mice. These findings suggest that circadian clock gene expression does not always reflect disordered behavior and sleep rhythms and that plasma FFA profiles could serve as a potential biomarker of circadian rhythm disorders.


Subject(s)
Amino Acids/blood , CLOCK Proteins/metabolism , Circadian Rhythm , Sleep Disorders, Circadian Rhythm/etiology , Sleep Disorders, Circadian Rhythm/metabolism , Stress, Psychological/complications , Stress, Psychological/metabolism , Animals , Biomarkers/blood , Gene Expression Regulation , Male , Mice , Mice, Inbred C3H , Reproducibility of Results , Sensitivity and Specificity , Sleep Disorders, Circadian Rhythm/diagnosis , Stress, Physiological , Stress, Psychological/diagnosis
17.
Biol Pharm Bull ; 37(8): 1422-7, 2014.
Article in English | MEDLINE | ID: mdl-25087965

ABSTRACT

The circadian clock is a cell-autonomous endogenous system that generates circadian rhythms in the behavior and physiology of most organisms. We previously reported that the harmala alkaloid, harmine, lengthens the circadian period of Bmal1 transcription in NIH 3T3 fibroblasts. Clock protein dynamics were examined using real-time reporter assays of PER2::LUC to determine the effects of harmine on the central clock in the suprachiasmatic nucleus (SCN). Harmine significantly lengthened the period of PER2::LUC expression in embryonic fibroblasts, in neuronal cells differentiated from neuronal progenitor cells and in SCN slices obtained from PER2::LUC mice. Although harmine did not induce the transient mRNA expression of clock genes such as Per1, Per2 and Bmal1 in embryonic fibroblasts, it significantly extended the half-life of PER2::LUC protein in neuronal cells and SCN slices. Harmine might lengthen the circadian period of the molecular clock by increasing PER2 protein stability in the SCN.


Subject(s)
Circadian Clocks/drug effects , Circadian Rhythm/drug effects , Harmine/pharmacology , Suprachiasmatic Nucleus/drug effects , ARNTL Transcription Factors/genetics , Animals , Cells, Cultured , Embryo, Mammalian , Fibroblasts , Luciferases/metabolism , Male , Mice, Transgenic , Neurons , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Suprachiasmatic Nucleus/metabolism
18.
PLoS One ; 19(1): e0296950, 2024.
Article in English | MEDLINE | ID: mdl-38285666

ABSTRACT

Nonalcoholic steatohepatitis (NASH) occurs worldwide and is characterized by lipid accumulation in hepatocytes, hepatic inflammation, fibrosis, and an increased risk of cirrhosis. Although a major proportion of NASH patients exhibit obesity and insulin resistance, 20% lack a high body mass and are categorized as "non-obese NASH". Time-restricted feeding (TRF), limiting daily food intake within certain hours, improves obesity, lipid metabolism, and liver inflammation. Here, we determined whether TRF affects NASH pathology induced by a choline-deficient high-fat diet (CDAHFD), which does not involve obesity. TRF ameliorated the increase in epididymal white adipose tissue and plasma alanine transaminase and aspartate transaminase levels after 8 weeks of a CDAHFD. Although gene expression of TNF alpha in the liver was suppressed by TRF, it did not exhibit a suppressive effect on hepatic lipid accumulation, gene expression of cytokines and macrophage markers (Mcp1, IL1b, F4/80), or fibrosis, as evaluated by Sirius red staining and western blot analysis of alpha-smooth muscle actin. A CDAHFD-induced increase in gene expression related to fibrogenesis (Collagen 1a1 and TGFß) was neither suppressed by TRF nor that of alpha-smooth muscle actin but was increased by TRF. Our results indicated that TRF has a limited suppressive effect on CDAHFD-induced NASH pathology.


Subject(s)
Choline Deficiency , Non-alcoholic Fatty Liver Disease , Humans , Animals , Mice , Non-alcoholic Fatty Liver Disease/pathology , Choline/metabolism , Diet, High-Fat/adverse effects , Actins/metabolism , Choline Deficiency/metabolism , Liver/metabolism , Liver Cirrhosis/pathology , Inflammation/pathology , Fibrosis , Obesity/complications , Lipids/adverse effects , Mice, Inbred C57BL , Disease Models, Animal
19.
Biochem Biophys Res Commun ; 432(1): 111-5, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23376072

ABSTRACT

We elucidated associations between metabolic disorders and the environmental light-dark (LD) cycle that entrains the circadian clock located in the suprachiasmatic nucleus of mammals. Mice were fed with a high-fat/high-sucrose diet for eight weeks under a normal 12h light-12h dark cycle (LD 12:12) or an ultradian 3h light-3h dark cycle (LD 3:3) that might perturb the central clock. The circadian behavioral rhythms were gradually disturbed under LD 3:3. Hyperglycemia with glucose intolerance and increases in diabetic markers, glycated albumin and hemoglobin A1c, were significantly induced without affecting body weight gain and food consumption in LD 3:3. Expression levels of hepatic gluconeogenic regulatory genes such as Pck1, G6pc, Hnf4a, and Foxo1/3/4 genes were increased under LD 3:3. Hypercholesterolemia with hepatic cholesterol accumulation was also induced in LD 3:3. Ultradian LD 3:3 cycles did not affect the adipose inflammation that is considered a major player in obesity-associated metabolic disorders. Our findings provide a link between metabolic disorders and environmental photoperiodic cycles in genetically normal animals.


Subject(s)
Circadian Clocks , Genes, Regulator/genetics , Gluconeogenesis/genetics , Glucose Intolerance/genetics , Hyperglycemia/genetics , Liver/metabolism , Photoperiod , Animals , Blood Glucose , Cholesterol/metabolism , Gene Expression Regulation , Insulin/blood , Male , Mice , Mice, Inbred ICR
20.
Biochem Biophys Res Commun ; 431(2): 332-5, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23291174

ABSTRACT

Tissue factor (TF) is involved in endotoxin-induced inflammation and mortality. We found that the circadian expression of TF mRNA, which peaked at the day to night transition (activity onset), was damped in the liver of Clock mutant mice. Luciferase reporter and chromatin immunoprecipitation analyses using embryonic fibroblasts derived from wild-type or Clock mutant mice showed that CLOCK is involved in transcription of the TF gene. Furthermore, the results of real-time luciferase reporter experiments revealed that the circadian expression of TF mRNA is regulated by clock molecules through a cell-autonomous mechanism via an E-box element located in the promoter region.


Subject(s)
CLOCK Proteins/metabolism , Circadian Rhythm/genetics , Thromboplastin/genetics , Trans-Activators , Transcription, Genetic , Animals , CLOCK Proteins/genetics , E-Box Elements , Liver/metabolism , Male , Mice , Mice, Mutant Strains , Promoter Regions, Genetic , RNA, Messenger/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL