Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Cephalalgia ; 44(4): 3331024241247845, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38676534

ABSTRACT

BACKGROUND: Cluster headache is a primary headache disorder characterized by bouts with circadian and circannual patterns. The CLOCK gene has a central role in regulating circadian rhythms. Here, we investigate the circannual CLOCK expression in a population of cluster headache patients in comparison to matched controls. METHODS: Patients with cluster headache were sampled two to four times over at least one year, both in or outside bouts, one week after each solstice and equinox. The expression of CLOCK was measured by quantitative real-time polymerase chain reaction (RT-PCR) in the peripheral blood. RESULTS: This study included 50 patients and 58 matched controls. Among the patient population, composed of 42/50 males (84%) with an average age of 44.6 years, 45/50 (90%) suffered from episodic cluster headache. Two to four samples were collected from each patient adding up to 161 samples, 36 (22.3%) of which were collected within a bout. CLOCK expression for cluster headache patients was considerably different from that of the control population in winter (p-value mean = 0.006283), spring (p-value mean = 0.000006) and summer (p-value mean = 0.000064), but not in autumn (p-value mean = 0.262272). For each season transition, the variations in CLOCK expression were more pronounced in the control group than in the cluster headache population. No statistically significant differences were found between bout and non-bout samples. No individual factors (age, sex, circadian chronotype, smoking and coffee habits or history of migraine) were related to CLOCK expression. CONCLUSIONS: We observed that CLOCK expression in cluster headache patients fluctuates less throughout the year than in the control population. Bout activity and lifestyle factors do not seem to influence CLOCK expression.


Subject(s)
CLOCK Proteins , Cluster Headache , Humans , Cluster Headache/genetics , Male , Female , Adult , CLOCK Proteins/genetics , CLOCK Proteins/biosynthesis , Middle Aged , Circadian Rhythm , Seasons
2.
FASEB J ; 35(10): e21886, 2021 10.
Article in English | MEDLINE | ID: mdl-34473369

ABSTRACT

The cardiac circadian clock is responsible for the modulation of different myocardial processes, and its dysregulation has been linked to disease development. How this clock machinery is regulated in the heart remains an open question. Because noradrenaline (NE) can act as a zeitgeber in cardiomyocytes, we tested the hypothesis that adrenergic signaling resets cardiac clock gene expression in vivo. In its anti-phase with Clock and Bmal1, cardiac Per1 abundance increased during the dark phase, concurrent with the rise in heart rate and preceded by an increase in NE levels. Sympathetic denervation altered Bmal1 and Clock amplitude, while Per1 was affected in both amplitude and oscillatory pattern. We next treated mice with a ß-adrenergic receptor (ß-AR) blocker. Strikingly, the ß-AR blockade during the day suppressed the nocturnal increase in Per1 mRNA, without altering Clock or Bmal1. In contrast, activating ß-AR with isoproterenol (ISO) promoted an increase in Per1 expression, demonstrating its responsiveness to adrenergic input. Inhibitors of ERK1/2 and CREB attenuated ISO-induced Per1 expression. Upstream of ERK1/2, PI3Kγ mediated ISO induction of Per1 transcription, while activation of ß2-AR, but not ß1-AR induced increases in ERK1/2 phosphorylation and Per1 expression. Consistent with the ß2-induction of Per1 mRNA, ISO failed to activate ERK1/2 and elevate Per1 in the heart of ß2-AR-/- mice, whereas a ß2-AR antagonist attenuated the nocturnal rise in Per1 expression. Our study established a link between NE/ß2-AR signaling and Per1 oscillation via the PI3Ky-ERK1/2-CREB pathway, providing a new framework for understanding the physiological mechanism involved in resetting cardiac clock genes.


Subject(s)
Gene Expression Regulation , MAP Kinase Signaling System , Myocardium/metabolism , Period Circadian Proteins/biosynthesis , Receptors, Adrenergic, beta-2/metabolism , ARNTL Transcription Factors/biosynthesis , ARNTL Transcription Factors/genetics , Adrenergic beta-2 Receptor Antagonists/pharmacology , Animals , CLOCK Proteins/biosynthesis , Isoproterenol/pharmacology , Male , Mice , Mice, Knockout , Period Circadian Proteins/genetics , Receptors, Adrenergic, beta-2/genetics
3.
Alcohol Clin Exp Res ; 45(3): 530-542, 2021 03.
Article in English | MEDLINE | ID: mdl-33606281

ABSTRACT

INTRODUCTIONS: Binge drinking is a deadly pattern of alcohol consumption. Evidence suggests that genetic variation in clock genes is strongly associated with alcohol misuse; however, the neuroanatomical basis for such a relationship is unknown. The shell region of the nucleus accumbens (NAcSh) is well known to play a role in binge drinking. Hence, we examined whether clock genes in the NAcSh regulate binge drinking. METHODS: To address this question, 2 experiments were performed on male C57BL/6J mice. In the first experiment, mice exposed to alcohol or sucrose under the 4-day drinking-in-the-dark (DID) paradigm were euthanized at 2 different time points on day 4 [7 hours after light (pre-binge drinking) or dark (post-binge drinking) onset]. The brains were processed for RT-PCR to examine the expression of circadian clock genes (Clock, Per1, and Per2) in the NAcSh and suprachiasmatic nucleus (SCN). In the second experiment, mice were exposed to alcohol, sucrose, or water as described above. On day 4, 1 hour prior to the onset of alcohol exposure, mice were bilaterally infused with either a mixture of circadian clock gene antisense oligodeoxynucleotides (AS-ODNs; antisense group) or nonsense/random ODNs (R-ODNs; control group) through surgically implanted cannulas above the NAcSh. Alcohol/sucrose/water consumption was measured for 4 hours. Blood alcohol concentration was measured to confirm binge drinking. Microinfusion sites were histologically verified using cresyl violet staining. RESULTS: As compared to sucrose, mice euthanized post-binge drinking (not pre-binge drinking) on day 4 displayed a greater expression of circadian genes in the NAcSh but not in the SCN. Knockdown of clock genes in the NAcSh caused a significantly lower volume of alcohol to be consumed on day 4 than in the control treatment. No differences were found in sucrose or water consumption. CONCLUSIONS: Our results suggest that clock genes in the NAcSh play a crucial role in binge drinking.


Subject(s)
Binge Drinking/metabolism , CLOCK Proteins/biosynthesis , Nucleus Accumbens/metabolism , Oligonucleotides, Antisense/administration & dosage , Period Circadian Proteins/biosynthesis , Animals , Binge Drinking/genetics , CLOCK Proteins/antagonists & inhibitors , CLOCK Proteins/genetics , Circadian Clocks/drug effects , Circadian Clocks/physiology , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Down-Regulation/drug effects , Down-Regulation/physiology , Ethanol/administration & dosage , Male , Mice , Mice, Inbred C57BL , Microinjections/methods , Nucleus Accumbens/drug effects , Period Circadian Proteins/antagonists & inhibitors , Period Circadian Proteins/genetics
4.
Mediators Inflamm ; 2020: 8192187, 2020.
Article in English | MEDLINE | ID: mdl-32879620

ABSTRACT

miRNA-10a is rhythmically expressed and regulates genes involved in lipid and glucose metabolism. However, the effects of miRNA-10a on obesity and glucose intolerance, as well as on the diurnal pattern of expression of circadian clock genes, remain unknown. We explored the effects of miRNA-10a-5p on insulin resistance and on the diurnal patterns of serum triglycerides and gut microbiota in high-fat diet- (HFD-) fed mice. The results showed that oral administration of miRNA-10a-5p significantly prevented body weight gain and improved glucose tolerance and insulin sensitivity in HFD-fed mice. Administration of miRNA-10a-5p also maintained the diurnal rhythm of Clock, Per2, and Cry1 expression, as well as serum glucose and triglyceride levels. Surprisingly, the diurnal oscillations of three genera of microbes, Oscillospira, Ruminococcus, and Lachnospiraceae, disrupted by HFD feeding, maintained by administration of miRNA-10a-5p. Moreover, a strong positive correlation was found between hepatic Clock expression and relative abundance of Lachnospiraceae, both in control mice (r = 0.877) and in mice administered miRNA-10a-5p (r = 0.853). Furthermore, we found that along with changes in Lachnospiraceae abundance, butyrate content in the feces maintained a diurnal rhythm after miRNA-10a-5p administration in HFD-fed mice. In conclusion, we suggest that miRNA-10a-5p may improve HFD-induced glucose intolerance and insulin resistance through the modulation of the diurnal rhythm of Lachnospiraceae and its metabolite butyrate. Therefore, miRNA-10a-5p may have preventative properties in subjects with metabolic disorders.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Insulin Resistance , MicroRNAs/genetics , Triglycerides/genetics , Animals , Blood Glucose/metabolism , Body Weight , CLOCK Proteins/biosynthesis , Clostridiales , Cryptochromes/biosynthesis , Glucose/metabolism , Glucose Intolerance/metabolism , Glucose Tolerance Test , Lipid Metabolism , Lipids/blood , Liver/metabolism , Male , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , Obesity/metabolism , Period Circadian Proteins/biosynthesis , RNA, Messenger/metabolism , Ruminococcus , Triglycerides/biosynthesis , Weight Gain
5.
Front Immunol ; 11: 614960, 2020.
Article in English | MEDLINE | ID: mdl-33552079

ABSTRACT

Dim light at night (dLAN) has become a pervasive part of the modern world, and growing evidence shows its association with increased health risks. Though this link is attributed to a disturbed circadian clock, the underlying mechanisms that can explain how circadian disruption from dLAN causes negative health effects remain unclear. Here, we exposed rats to a light-dark cycle (12:12 h) with low-intensity light at night (~2 lx) for 2 and 5 weeks and explored the steady-state pattern of circulating immune cells and renal immune-related markers, which are well controlled by the circadian clock. After 5 weeks, dLAN impaired the daily variation in several types of white blood cells, especially monocytes and T cells. Two-week dLAN caused a reduction in blood monocytes and altered gene expression of macrophage marker Cd68 and monocyte-attracting chemokine Ccl2 in the kidney. Interestingly, dLAN decreased renal 3-nitrotyrosine levels and resulted in up-regulation of the main endogenous antioxidant pathways, indicating a disturbance in the renal redox balance and an activation of compensatory mechanisms. These effects paralleled the altered renal expression of the molecular clock components and increased plasma corticosterone levels. Together, our results show that chronic exposure to dLAN weakened the circadian control of daily variation of circulating immune cells and disturbed renal immune and redox homeostasis. Consequences of this dLAN-disturbed immune balance on the ability of the immune system to cope with other challenges should by clarified in further studies.


Subject(s)
Circadian Rhythm/immunology , Immune System/radiation effects , Kidney/immunology , Light/adverse effects , Photoperiod , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/genetics , CLOCK Proteins/biosynthesis , CLOCK Proteins/genetics , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Chemokines/biosynthesis , Chemokines/genetics , Corticosterone/blood , Gene Expression Regulation/radiation effects , Homeostasis/radiation effects , Immunophenotyping , Kidney/metabolism , Kidney Cortex/enzymology , Leukocyte Count , Male , Melatonin/blood , Oxidation-Reduction , Rats , Rats, Wistar , Respiratory Burst , Superoxide Dismutase/analysis
6.
Am J Physiol Renal Physiol ; 317(6): F1536-F1548, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31588796

ABSTRACT

The sodium-chloride cotransporter (NCC) in the distal convoluted tubule contributes importantly to sodium balance and blood pressure (BP) regulation. NCC phosphorylation determines transport activity and has a diurnal rhythm influenced by glucocorticoids. Disturbing this rhythm induces "nondipping" BP, an abnormality that increases cardiovascular risk. The receptor through which glucocorticoids regulate NCC is not known. In this study, we found that acute administration of corticosterone to male C57BL6 mice doubled NCC phosphorylation without affecting total NCC abundance in both adrenalectomized and adrenal-intact mice. Corticosterone also increased the whole kidney expression of canonical clock genes: period circadian protein homolog 1 (Per1), Per2, cryptochrome 1, and aryl hydrocarbon receptor nuclear translocator-like protein 1. In adrenal-intact mice, chronic blockade of glucocorticoid receptor (GR) with RU486 did not change total NCC but prevented corticosterone-induced NCC phosphorylation and activation of clock genes. Blockade of mineralocorticoid receptor (MR) with spironolactone reduced the total pool of NCC but did not affect stimulation by corticosterone. The diurnal rhythm of NCC phosphorylation, measured at 6-h intervals, was blunted by chronic GR blockade, and a similar dampening of diurnal variation was seen in GR heterozygous null mice. These effects on NCC phosphorylation did not reflect altered rhythmicity of plasma corticosterone or serum and glucocorticoid-induced kinase 1 activity. Both mineralocorticoids and glucocorticoids emerge as regulators of NCC, acting via distinct receptor pathways. MR activation provides maintenance of the NCC protein pool; GR activation dynamically regulates NCC phosphorylation and establishes the diurnal rhythm of NCC activity. This study has implications for circadian BP homeostasis, particularly in individuals with abnormal glucocorticoid signaling as is found in chronic stress and corticosteroid therapy.


Subject(s)
Circadian Rhythm/physiology , Kidney/metabolism , Receptors, Glucocorticoid/metabolism , Sodium Chloride Symporters/metabolism , Adrenalectomy , Animals , CLOCK Proteins/biosynthesis , CLOCK Proteins/genetics , Circadian Rhythm/drug effects , Corticosterone/blood , Corticosterone/pharmacology , Kidney/drug effects , Male , Mice , Mice, Inbred C57BL , Mifepristone/pharmacology , Mineralocorticoid Receptor Antagonists/pharmacology , Phosphorylation/drug effects , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/drug effects , Spironolactone/pharmacology
7.
J Neurosci ; 39(40): 7958-7967, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31420455

ABSTRACT

The transcription factor, myocyte enhancer factor-2 (MEF2), is required for normal circadian behavior in Drosophila; however, its role in the mammalian circadian system has not been established. Of the four mammalian Mef2 genes, Mef2d is highly expressed in the suprachiasmatic nucleus (SCN) of the hypothalamus, a region critical for coordinating peripheral circadian clocks. Using both conventional and brain-specific Mef2d KO (Mef2d-/-) mouse lines, we demonstrate that MEF2D is essential for maintaining the length of the circadian free-running period of locomotor activity and normal sleep patterns in male mice. Crossing Mef2d-/- with Per2::luc reporter mice, we show that these behavioral changes are achieved without altering the endogenous period of the master circadian oscillator in the SCN. Together, our data suggest that alterations in behavior in Mef2d-/- mice may be the result of an effect on SCN output, rather than an effect on timekeeping within the SCN itself. These findings add to the growing body of evidence that MEF2 proteins play important roles in the brain.SIGNIFICANCE STATEMENT These studies are the first to show a role for MEF2 proteins in the brain outside of the hippocampus, and our findings suggest that these proteins may play diverse roles in the CNS. It is important to continue to build on our understanding of the roles of proteins acting in the SCN because SCN dysfunction underlies jet lag in humans and influences the response to shift work schedules, which are now known as risk factors for the development of cancer. Our work on MEF2D could be the basis for opening new lines of research in the development and regulation of circadian rhythms.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Sleep/genetics , Sleep/physiology , Animals , Behavior, Animal , CLOCK Proteins/biosynthesis , CLOCK Proteins/genetics , Light , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , RNA/biosynthesis , RNA/genetics , Sleep Wake Disorders/genetics , Sleep Wake Disorders/psychology , Suprachiasmatic Nucleus/physiology
8.
Exp Cell Res ; 382(1): 111451, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31173767

ABSTRACT

Trimethylamine N-oxide (TMAO) promotes atherosclerosis in association with the functions of endothelial cells. Clock and Bmal1, as two main components of molecular circadian clock, play important regulatory roles during progression of atherogenesis. However, whether Clock and Bmal1 are involved in the regulation of endothelial proliferation disturbed by TMAO are unclear. We observed that cell proliferation of human umbilical vein endothelial cells (HUVECs) was inhibited after exposed to TMAO for 24 h. Besides, TMAO caused increased expression of lncRNA-NEAT1, Clock and Bmal1, and inhibited MAPK pathways. While MAPK pathways were blocked, the expression of Clock and Bmal1 was elevated. NEAT1 showed a circadian rhythmic expression in HUVECs, and its overexpression reduced cell proliferation. Knockdown or overexpression of NEAT1 might decrease or increase the expression of Clock and Bmal1 respectively, while raised or suppressed the expression of MAPK pathways correspondingly. Asparagus extract (AE) was found to improve the TMAO-reduced HUVECs proliferation. Moreover, it ameliorated the disorders of NEAT1, Clock, Bmal1, and MAPK signaling pathways induced by TMAO. Therefore, our findings indicated that NEAT1 regulating Clock-Bmal1 via MAPK pathways was involved in TMAO-repressed HUVECs proliferation, and AE improved endothelial proliferation by TMAO, proposing a novel mechanism for cardiovascular disease prevention.


Subject(s)
Asparagaceae/chemistry , Circadian Rhythm/drug effects , Gene Expression Regulation/physiology , Methylamines/toxicity , Plant Extracts/pharmacology , RNA, Long Noncoding/physiology , ARNTL Transcription Factors/antagonists & inhibitors , ARNTL Transcription Factors/biosynthesis , ARNTL Transcription Factors/genetics , Atherosclerosis/genetics , Atherosclerosis/physiopathology , CLOCK Proteins/biosynthesis , CLOCK Proteins/genetics , Cell Division/drug effects , Circadian Rhythm/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Methylamines/pharmacology , Plant Stems/chemistry , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
9.
BMC Neurosci ; 20(1): 24, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138137

ABSTRACT

BACKGROUND: Mutants which carry mutations in genes encoding mitochondrial ligases MUL1 and PARKIN are convenient Drosophila models of Parkinson's disease (PD). In several studies it has been shown that in Parkinson's disease sleep disturbance occurs, which may be the result of a disturbed circadian clock. RESULTS: We found that the ROS level was higher, while the anti-oxidant enzyme SOD1 level was lower in mul1A6 and park1 mutants than in the white mutant used as a control. Moreover, mutations of both ligases affected circadian rhythms and the clock. The expression of clock genes per, tim and clock and the level of PER protein were changed in the mutants. Moreover, expression of ATG5, an autophagy protein also involved in circadian rhythm regulation, was decreased in the brain and in PDF-immunoreactive large ventral lateral clock neurons. The observed changes in the molecular clock resulted in a longer period of locomotor activity rhythm, increased total activity and shorter sleep at night. Finally, the lack of both ligases led to decreased longevity and climbing ability of the flies. CONCLUSIONS: All of the changes observed in the brains of these Drosophila models of PD, in which mitochondrial ligases MUL1 and PARKIN do not function, may explain the mechanisms of some neurological and behavioural symptoms of PD.


Subject(s)
Brain/metabolism , Circadian Clocks/physiology , Circadian Rhythm/physiology , Drosophila Proteins/physiology , Locomotion/physiology , Parkinson Disease/physiopathology , Sleep/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Animals, Genetically Modified/physiology , CLOCK Proteins/biosynthesis , Disease Models, Animal , Drosophila , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Longevity/physiology , Motor Skills/physiology , Mutation , Neurons/metabolism , Parkinson Disease/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Ubiquitin-Protein Ligases/genetics
10.
Elife ; 82019 02 05.
Article in English | MEDLINE | ID: mdl-30720431

ABSTRACT

Sleep depriving mice affects clock-gene expression, suggesting that these genes contribute to sleep homeostasis. The mechanisms linking extended wakefulness to clock-gene expression are, however, not well understood. We propose CIRBP to play a role because its rhythmic expression is i) sleep-wake driven and ii) necessary for high-amplitude clock-gene expression in vitro. We therefore expect Cirbp knock-out (KO) mice to exhibit attenuated sleep-deprivation-induced changes in clock-gene expression, and consequently to differ in their sleep homeostatic regulation. Lack of CIRBP indeed blunted the sleep-deprivation incurred changes in cortical expression of Nr1d1, whereas it amplified the changes in Per2 and Clock. Concerning sleep homeostasis, KO mice accrued only half the extra REM sleep wild-type (WT) littermates obtained during recovery. Unexpectedly, KO mice were more active during lights-off which was accompanied with faster theta oscillations compared to WT mice. Thus, CIRBP adjusts cortical clock-gene expression after sleep deprivation and expedites REM-sleep recovery.


Subject(s)
CLOCK Proteins/biosynthesis , Gene Expression , RNA-Binding Proteins/metabolism , Sleep Deprivation , Sleep, REM , Animals , Gene Knockout Techniques , Mice, Inbred C57BL , Mice, Knockout , RNA-Binding Proteins/genetics
11.
Exp Gerontol ; 117: 61-71, 2019 03.
Article in English | MEDLINE | ID: mdl-30415070

ABSTRACT

Gene expression is much altered in aging. We observed age-dependent decline of core clock genes' expression in the whole body of the fruit fly. We hypothesized that inducible overexpression of clock genes (cry, per, tim, cyc and Clk) in the nervous system can improve healthspan of D. melanogaster. We studied the lifespan of transgenic Drosophila and showed life extension for cry, per, cyc and tim genes. It was also the significant positive changes in the stress-resistance of flies overexpressing core clock genes in conditions of hyperthermia, hyperoxia, starvation and persistent lighting. The overexpression of per and cry restore circadian rhythms of locomotor activity. The results presented support the hypotheses that the compensation of circadian oscillator genes expression can improve the healthspan in Drosophila melanogaster.


Subject(s)
CLOCK Proteins/biosynthesis , Drosophila Proteins/biosynthesis , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation/physiology , Longevity/genetics , Neurons/metabolism , Aging/genetics , Aging/physiology , Animals , Animals, Genetically Modified , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Female , Fever/genetics , Fever/physiopathology , Locomotion/genetics , Locomotion/physiology , Longevity/physiology , Male , Oxidative Stress/genetics , Oxidative Stress/physiology , Photic Stimulation , Starvation/genetics , Starvation/physiopathology
12.
FASEB J ; 33(3): 4473-4489, 2019 03.
Article in English | MEDLINE | ID: mdl-30566374

ABSTRACT

Calorie restriction (CR) delays aging and affects the circadian clocks by reprogramming circadian rhythms in gene expression. To expand on the circadian mechanisms in CR, we assayed rhythms in the protein translation by analyzing polysome-associated mRNAs in the liver of mice fed ad libitum (AL) and CR diets. Global comparison of the diets revealed that <1% of transcripts were differentially abundant in the polysomes. In contrast, the large differential, up to 10%, was detected when CR and AL diets were compared at individual times throughout the day. Most transcripts that were rhythmic under AL lost their rhythms, and many new transcripts gained rhythms under CR. Only a small fraction of transcripts, including the circadian clock genes, were rhythmic under both diets. Thus, CR strongly reprograms translation. CR affected translation of enzymes regulating long-chain acetyl-coenzyme A (Acyl-CoA) metabolism. The expression of the Acyl-CoA thioesterase (ACOT) family was induced upon CR, leading to the increased transcriptional activity of peroxisome proliferator-activated receptor α, the transcriptional factor regulated by the ACOT products. We propose that the differential translation induced by CR leads to a temporal partition and reprogramming of metabolic processes and provides a link between CR, lipid metabolism, and the circadian clock.-Makwana, K., Gosai, N., Poe, A., Kondratov, R. V. Calorie restriction reprograms diurnal rhythms in protein translation to regulate metabolism.


Subject(s)
Caloric Restriction , Circadian Rhythm/physiology , Gene Expression Regulation/physiology , Protein Biosynthesis , Acyl Coenzyme A/metabolism , Adaptation, Physiological , Aging/metabolism , Animals , Blood Glucose/analysis , CLOCK Proteins/biosynthesis , CLOCK Proteins/genetics , Fasting , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , PPAR alpha/metabolism , Polyribosomes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Thiolester Hydrolases/metabolism , Transcription, Genetic
13.
Exp Neurol ; 306: 45-54, 2018 08.
Article in English | MEDLINE | ID: mdl-29660304

ABSTRACT

Postoperative cognitive dysfunction (POCD) is a common clinical phenomenon characterized by cognitive deficits in patients after anesthesia and surgery. Advanced age is a significant independent risk factor for POCD. We previously reported that in young mice, sleep-wake rhythm is involved in the isoflurane-induced memory impairment. In present study, we sought to determine whether advanced age increased the risk of POCD through aggravated and prolonged post-anesthetic circadian disruption in the elderly. We constructed POCD model by submitting the mice to 5-h 1.3% isoflurane anesthesia from Zeitgeber Time (ZT) 14 to ZT19. Under novel object recognition assay (NOR) and Morris water maze (MWM) test, We found 5-h isoflurane anesthesia impaired the cognition of young mice for early 3 days after anesthesia but damaged the aged for at least 1 week. With Mini-Mitter continuously monitoring, a 3.22 ±â€¯0.75 h gross motor activity acrophase delay was manifested in young mice on D1, while in the aged mice, the gross motor activity phase shift lasted for 3 days, consistent with the body temperature rhythm trends of change. Melatonin has been considered as an effective remedy for circadian rhythm shift. In aged mice, melatonin was pretreated intragastrically at the dose of 10 mg/kg daily for 7 consecutive days before anesthesia. We found that melatonin prevented isoflurane-induced cognitive impairments by restoring the locomotor activity and temperature circadian rhythm via clock gene resynchronization. Overall, these results indicated that Long-term isoflurane anesthesia induced more aggravated and prolonged memory deficits and circadian rhythms disruption in aged mice. Melatonin could prevent isoflurane-induced cognitive impairments by circadian rhythm resynchronization.


Subject(s)
Anesthetics, Inhalation/toxicity , Circadian Rhythm , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/physiopathology , Isoflurane/toxicity , Aging/psychology , Animals , Body Temperature/drug effects , CLOCK Proteins/biosynthesis , CLOCK Proteins/genetics , Cognitive Dysfunction/therapy , Maze Learning , Melatonin/therapeutic use , Memory Disorders/chemically induced , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Postoperative Complications/drug therapy , Postoperative Complications/physiopathology , Postoperative Complications/psychology , Recognition, Psychology
14.
Psychopharmacology (Berl) ; 235(3): 627-640, 2018 03.
Article in English | MEDLINE | ID: mdl-29151193

ABSTRACT

Major depressive disorder (MDD) is a growing problem worldwide. Though, the etiology remains unresolved, circadian rhythm disturbances are frequently observed in MDD and thus is speculated to play a key role herein. The present study focuses on circadian rhythm disturbances in the chronic mild stress (CMS) animal model of depression and examined whether the atypical antidepressant, agomelatine, which is mediating its action via melatonergic and serotonergic receptors, is capable of resynchronizing the perturbed rhythm. Melatonin is often used as a marker of the circadian phase, but the functional and behavioral output is dictated on a cellular level by the molecular clock, driven by the clock genes. We applied in situ hybridization histochemistry to measure the expression levels of the core clock genes, period (Per) 1 and 2 and bone and muscle ARNT-like protein 1 (Bmal1), in multiple brain regions believed to be implicated in depression. Agomelatine showed an antidepressant-like effect in the sucrose consumption test and an anxiolytic-like profile in the elevated zero maze. We found that CMS increased nighttime melatonin release in rats and that agomelatine attenuated this effect. Stress was shown to have a time and region-specific effect on clock gene expression in the brain. Treatment with agomelatine failed to normalize clock gene expression, and the observed modifying effect on gene expression did not associate with the antidepressant-like effect. This suggests that the antidepressant actions of agomelatine are mainly independent of circadian rhythm synchronization and, in this regard, not superior to traditional antidepressants tested in our model.


Subject(s)
Acetamides/therapeutic use , Antidepressive Agents/therapeutic use , CLOCK Proteins/biosynthesis , Circadian Rhythm/drug effects , Depression/drug therapy , Disease Models, Animal , Acetamides/pharmacology , Animals , Antidepressive Agents/pharmacology , CLOCK Proteins/genetics , Circadian Rhythm/physiology , Depression/genetics , Depression/metabolism , Male , Melatonin/pharmacology , Period Circadian Proteins/biosynthesis , Rats , Rats, Wistar , Treatment Outcome
15.
Alcohol Clin Exp Res ; 41(12): 2007-2014, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28960346

ABSTRACT

BACKGROUND: Alcoholic liver disease (ALD) is commonly associated with intestinal permeability. An unanswered question is why only a subset of heavy alcohol drinkers develop endotoxemia. Recent studies suggest that circadian disruption is the susceptibility factor for alcohol-induced gut leakiness to endotoxins. The circadian protein PER2 is increased after exposure to alcohol and siRNA knockdown of PER2 in vitro blocks alcohol-induced intestinal barrier dysfunction. We have shown that blocking CYP2E1 (i.e., important for alcohol metabolism) with siRNA inhibits the alcohol-induced increase in PER2 and suggesting that oxidative stress may mediate alcohol-induced increase in PER2 in intestinal epithelial cells. The aim of this study was to elucidate whether a mechanism incited by alcohol-derived oxidative stress mediates the transcriptional induction of PER2 and subsequent intestinal hyperpermeability. METHODS: Caco-2 cells were exposed to 0.2% alcohol with or without pretreatment with modulators of oxidative stress or PKA activity. Permeability of the Caco-2 monolayer was assessed by transepithelial electrical resistance. Protein expression was measured by Western blot and mRNA with real-time polymerase chain reaction. Wild-type C57BL/6J mice were fed with alcohol diet (29% of total calories, 4.5% v/v) for 8 weeks. Western blot was used to analyze PER2 expression in mouse proximal colon tissue. RESULTS: Alcohol increased oxidative stress, caused Caco-2 cell monolayer dysfunction, and increased levels of the circadian clock proteins PER2 and CLOCK. These effects were mitigated by pretreatment of Caco-2 cells with an antioxidant scavenger. Alcohol-derived oxidative stress activated cAMP response element-binding (CREB) via the PKA pathway and increased PER2 mRNA and protein. Inhibiting CREB prevented the increase in PER2 and Caco-2 cell monolayer hyperpermeability. CONCLUSIONS: Taken together, these data suggest that strategies to reduce alcohol-induced oxidative stress may alleviate alcohol-mediated circadian disruption and intestinal leakiness, critical drivers of ALD.


Subject(s)
Chronobiology Disorders/chemically induced , Cyclic AMP Response Element-Binding Protein/metabolism , Ethanol/adverse effects , Intestinal Mucosa/metabolism , Intestines/drug effects , CLOCK Proteins/biosynthesis , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Free Radical Scavengers/pharmacology , Humans , Oxidative Stress/drug effects , Period Circadian Proteins/biosynthesis , Permeability/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism
16.
Pharmacol Biochem Behav ; 162: 46-54, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28624585

ABSTRACT

Over the past 20years, substantive research has firmly implicated the lateral habenula in myriad neural processes including addiction, depression, and sleep. More recently, evidence has emerged suggesting that the lateral habenula is a component of the brain's intrinsic daily or circadian timekeeping system. This system centers on the master circadian pacemaker in the suprachiasmatic nuclei of the hypothalamus that is synchronized to the external world through environmental light information received directly from the eye. Rhythmic clock gene expression in suprachiasmatic neurons drives variation in their electrical activity enabling communication of temporal information, and the organization of circadian rhythms in downstream targets. Here, we review the evidence implicating the lateral habenula as part of an extended neural circadian system. We consider findings suggesting that the lateral habenula is a recipient of circadian signals from the suprachiasmatic nuclei as well as light information from the eye. Further we examine the proposition that the lateral habenula itself expresses intrinsic clock gene and neuronal rhythms. We then speculate on how circadian information communicated from the lateral habenula could influence activity and function in downstream targets such as the ventral tegmental area and raphe nuclei.


Subject(s)
Circadian Rhythm/physiology , Habenula/physiology , Animals , CLOCK Proteins/biosynthesis , Habenula/cytology , Humans , Suprachiasmatic Nucleus Neurons/cytology , Suprachiasmatic Nucleus Neurons/physiology
17.
Cancer Biother Radiopharm ; 32(4): 139-146, 2017 May.
Article in English | MEDLINE | ID: mdl-28514207

ABSTRACT

Abnormal autophagy regulation affects the chemoresistance of ovarian cancer, during which the circadian gene clock may play a major role. In this study, RNA interference plasmid pSUPER-Clock and overexpression plasmid pcDNA3.1-Clock of CLOCK were used to stably transfect the SKOV3/DDP cells by lipofection. Upon screening, the in vitro transfected cell lines with pSUPER-Clock, the autophagy level, and G0/G1 phase cells were significantly reduced, and the expression levels of Clock, LC3, P-gp, and MRP2 were inhibited. In contrast, the autophagy level and G0/G1 phase cells in cell lines transfected with pcDNA3.1-Clock were significantly increased, and the expressions of Clock, LC3, P-gp, and MRP2 were enhanced. In comparison with the untransfected control group showed the percentage of apoptotic cells in SKOV3/DDP cell lines of Clock interfering expression group after cisplatin treatment was significantly increased while the survival was substantially reduced. These results indicated that inhibiting the circadian gene Clock expression can reverse the cisplatin resistance of ovarian cancer SKOV3/DDP cell lines by affecting the protein expression of drug resistance genes during which autophagy plays an important role. The CLOCK gene may be designated as a novel candidate for targeted gene therapy in drug-resistant ovarian cancer.


Subject(s)
CLOCK Proteins/genetics , Circadian Clocks/genetics , Ovarian Neoplasms/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antineoplastic Agents/pharmacology , Autophagy/genetics , CLOCK Proteins/biosynthesis , Cell Line, Tumor , Cell Proliferation/genetics , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Humans , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/biosynthesis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Transfection
18.
Exp Eye Res ; 160: 21-30, 2017 07.
Article in English | MEDLINE | ID: mdl-28434813

ABSTRACT

Teleost fish continues to grow their eyes throughout life with the body size. In Astatotilapia burtoni, the fish retina increases by adding new retinal cells at the ciliary marginal zone (CMZ) and in the outer nuclear layer (ONL). Cell proliferation at both sites exhibits a daily rhythm in number of dividing cells. To understand how this diurnal rhythm of new cell production is controlled in retinal progenitor cells, we studied the transcription pattern of clock genes in retina, including clock1a, clock1b, bmal1a (brain and muscle ARNT-Like), and per1b (period1b). We found that these genes have a strong diurnal rhythmic transcription during light-dark cycles but not in constant darkness. An oscillation in pcna transcription was also observed during light-dark cycles, but again not in constant darkness. Our results also indicate an association between Clock proteins and the upstream region of pcna (proliferating cellular nuclear antigen) gene. A luciferase reporter assay conducted in an inducible clock knockdown cell line further demonstrated that the mutation on predicted E-Boxes in pcna promoter region significantly attenuated the transcriptional activation induced by Clock protein. These results suggested that the diurnal rhythmic expression of clock genes in A. burtoni retina could be light dependent and might contribute to the daily regulation of the proliferation of the retina progenitors through key components of cell cycle machinery, for instance, pcna.


Subject(s)
CLOCK Proteins/genetics , Gene Expression Regulation , Proliferating Cell Nuclear Antigen/genetics , RNA/genetics , Retina/metabolism , Animals , Blotting, Western , CLOCK Proteins/biosynthesis , Cell Division , Cell Line , Cell Proliferation , Cichlids , Circadian Rhythm/physiology , Immunohistochemistry , In Situ Hybridization , Light , Mice , Models, Animal , Photoperiod , Proliferating Cell Nuclear Antigen/metabolism , Real-Time Polymerase Chain Reaction , Retina/cytology , Stem Cells/cytology , Stem Cells/metabolism , Transcription, Genetic
19.
PLoS Genet ; 12(12): e1006512, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27941970

ABSTRACT

Altered daily patterns of hormone action are suspected to contribute to metabolic disease. It is poorly understood how the adrenal glucocorticoid hormones contribute to the coordination of daily global patterns of transcription and metabolism. Here, we examined diurnal metabolite and transcriptome patterns in a zebrafish glucocorticoid deficiency model by RNA-Seq, NMR spectroscopy and liquid chromatography-based methods. We observed dysregulation of metabolic pathways including glutaminolysis, the citrate and urea cycles and glyoxylate detoxification. Constant, non-rhythmic glucocorticoid treatment rescued many of these changes, with some notable exceptions among the amino acid related pathways. Surprisingly, the non-rhythmic glucocorticoid treatment rescued almost half of the entire dysregulated diurnal transcriptome patterns. A combination of E-box and glucocorticoid response elements is enriched in the rescued genes. This simple enhancer element combination is sufficient to drive rhythmic circadian reporter gene expression under non-rhythmic glucocorticoid exposure, revealing a permissive function for the hormones in glucocorticoid-dependent circadian transcription. Our work highlights metabolic pathways potentially contributing to morbidity in patients with glucocorticoid deficiency, even under glucocorticoid replacement therapy. Moreover, we provide mechanistic insight into the interaction between the circadian clock and glucocorticoids in the transcriptional regulation of metabolism.


Subject(s)
CLOCK Proteins/biosynthesis , Circadian Clocks/genetics , E-Box Elements/genetics , Glucocorticoids/genetics , Metabolic Networks and Pathways/genetics , Animals , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Citric Acid/metabolism , Gene Expression Regulation , Glucocorticoids/biosynthesis , Glucocorticoids/deficiency , High-Throughput Nucleotide Sequencing , Hormones/genetics , Hormones/metabolism , Humans , Magnetic Resonance Spectroscopy , Transcription, Genetic , Transcriptome/genetics , Urea/metabolism , Zebrafish
20.
Genet Mol Res ; 15(4)2016 Oct 24.
Article in English | MEDLINE | ID: mdl-27813571

ABSTRACT

Marine animals exhibit a variety of biological rhythms, such as solar and lunar-related cycles; however, our current molecular understanding of biological rhythms in marine animals is quite limited. Identifying and understanding the expression patterns of clock genes from available transcriptomes will help elucidate biological rhythms in marine species. Here, we perform a comprehensive survey of phototransduction and circadian genes using the mantle transcriptome of the scallop Patinopecten yessoensis and compare the results with those from three other bivalves. The comparison reveals the presence of transcripts for most of the core members of the phototransduction and circadian networks seen in terrestrial model species in the four marine bivalves. Matches were found for all 37 queried genes, and the expressed transcripts from the deep sequencing data matched 8 key insect and mammalian circadian genes. This demonstrates the high level of conservation of the timekeeping mechanism from terrestrial species to marine bivalves. The results provide a valuable gene resource for studies of "marine rhythms" and also further our understanding of the diversification and evolution of rhythms in marine species.


Subject(s)
Bivalvia/genetics , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Transcriptome/genetics , Animals , Aquatic Organisms/genetics , Aquatic Organisms/growth & development , Biological Evolution , Bivalvia/growth & development , CLOCK Proteins/biosynthesis , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Light Signal Transduction/genetics , Molecular Sequence Annotation
SELECTION OF CITATIONS
SEARCH DETAIL
...