Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
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
2.
Biochem Biophys Res Commun ; 560: 14-20, 2021 06 30.
Article in English | MEDLINE | ID: mdl-33965785

ABSTRACT

Clock genes express circadian rhythms in most organs. These rhythms are organized throughout the whole body, regulated by the suprachiasmatic nucleus (SCN) in the brain. Disturbance of these clock gene expression rhythms is a risk factor for diseases such as obesity. In the present study, to explore the role of clock genes in developing diabetes, we examined the effect of streptozotocin (STZ)-induced high glucose on Period1 (Per1) gene expression rhythm in the liver and the olfactory bub (OB) in the brain. We found a drastic increase of Per1 expression in both tissues after STZ injection while blood glucose content was low. After a rapid expression peak, Per1 expression showed no rhythm. Associated with an increase of glucose content, behavior became arrhythmic. Finally, we succeeded in detecting an increase of Per1 expression in mice hair follicles on day 1 after STZ administration, before the onset of symptoms. These results show that elevated Per1 expression by STZ plays an important role in the aggravation of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Liver/metabolism , Olfactory Bulb/metabolism , Period Circadian Proteins/biosynthesis , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Drinking/drug effects , Gene Expression , Hair/drug effects , Hair/metabolism , Locomotion , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Periodicity , Streptozocin
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.
Neurosci Lett ; 743: 135563, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33359046

ABSTRACT

N-myc downstream-regulated gene 2 (NDRG2), a member of the NDRG family, has multiple functions in cell proliferation, differentiation, and stress responses, and is predominantly expressed by astrocytes in the central nervous system. Previous studies including ours demonstrated that NDRG2 is involved in various central nervous system pathologies. However, the significance of NDRG2 in neurodevelopment is not fully understood. Here, we investigated the expression profile of NDRG2 during postnatal brain development, the role of NDRG2 in social behavior, and transcriptome changes in the brain of NDRG2-deficient mice. NDRG2 expression in the brain increased over time from postnatal day 1 to adulthood. Deletion of NDRG2 resulted in abnormal social behavior, as indicated by reduced exploratory activity toward a novel mouse in a three-chamber social interaction test. Microarray analysis identified genes differentially expressed in the NDRG2-deficient brain, and upregulated gene expression of Bmp4 and Per2 was confirmed by quantitative PCR analysis. Expression of both these genes and the encoded proteins increased over time during postnatal brain development, similar to NDRG2. Gene expression of Bmp4 and Per2 was upregulated in cultured astrocytes isolated from NDRG2-deficient mice. These results suggest that NDRG2 contributes to brain development required for proper social behavior by modulating gene expression in astrocytes.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Social Behavior , Animals , Bone Morphogenetic Protein 4/biosynthesis , Bone Morphogenetic Protein 4/genetics , Cells, Cultured , Gene Expression , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Period Circadian Proteins/biosynthesis , Period Circadian Proteins/genetics
5.
Int J Mol Sci ; 21(18)2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32967200

ABSTRACT

Circadian oscillations of mRNAs and proteins are the main features of circadian clock genes. Among them, Period1 (Per1) is a key component in negative-feedback regulation, which shows a robust diurnal oscillation and the importance of circadian rhythm and translational regulation of circadian clock genes has been recognized. In the present study, we investigated the 5'-untranslated region (5'-UTR) of the mouse core clock gene, Per1, at the posttranscriptional level, particularly its translational regulation. The 5'-UTR of Per1 was found to promote its translation via an internal ribosomal entry site (IRES). We found that polypyrimidine tract-binding protein 1 (PTBP1) binds to the 5'-UTR of Per1 and positively regulates the IRES-mediated translation of Per1 without affecting the levels of Per1 mRNA. The reduction of PTBP1 level also decreased the endogenous levels of the PER1 protein but not of its mRNA. As for the oscillation of PER1 expression, the disruption of PTBP1 levels lowered the PER1 expression but not the phase of the oscillation. PTBP1 also changed the amplitudes of the mRNAs of other circadian clock genes, such as Cryptochrome 1 (Cry1) and Per3. Our results suggest that the PTBP1 is important for rhythmic translation of Per1 and it fine-tunes the overall circadian system.


Subject(s)
5' Untranslated Regions , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Period Circadian Proteins/biosynthesis , Polypyrimidine Tract-Binding Protein/metabolism , Protein Biosynthesis , Animals , Cryptochromes/biosynthesis , Cryptochromes/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Mice , NIH 3T3 Cells , Period Circadian Proteins/genetics , Polypyrimidine Tract-Binding Protein/genetics
6.
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
7.
CNS Neurosci Ther ; 26(3): 343-354, 2020 03.
Article in English | MEDLINE | ID: mdl-31411808

ABSTRACT

INTRODUCTION: The occurrence of circadian rhythm disorder in patients with Alzheimer's disease (AD) is closely related to the abnormal deposition of amyloid-ß (Aß), and d-Ser2-oxyntomodulin (Oxy) is a protease-resistant oxyntomodulin analogue that has been shown to exert neuroprotective effects. AIMS: This study aimed to explore whether Oxy, a new GLP-1R/GCGR dual receptor agonist, can improve the Aß-induced disrupted circadian rhythm and the role of GLP-1R. METHODS: A mouse wheel-running experiment was performed to explore the circadian rhythm, and western blotting and real-time PCR were performed to assess the expression of the circadian clock genes Bmal1 and Per2. Furthermore, a lentivirus encoding an shGLP-1R-GFP-PURO was used to interfere with GLP-1R gene expression and so explore the role of GLP-1R. RESULTS: The present study has confirmed that Oxy could restore Aß31-35-induced circadian rhythm disorders and improve the abnormal expression of Bmal1 and Per2. After interfering the GLP-1R gene, we found that Oxy could not improve the Aß31-35-induced circadian rhythm disorder and abnormal expression of clock genes. CONCLUSION: This study demonstrated that Oxy could improve Aß31-35-induced circadian rhythm disorders, and GLP-1R plays a critical role. This study thus describes a novel target that may be potentially used in the treatment of AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Chronobiology Disorders/chemically induced , Chronobiology Disorders/drug therapy , Oxyntomodulin/therapeutic use , Peptide Fragments/toxicity , ARNTL Transcription Factors/biosynthesis , Animals , Cell Line , Chronobiology Disorders/metabolism , Glucagon-Like Peptide-1 Receptor/biosynthesis , Male , Mice , Mice, Inbred C57BL , Period Circadian Proteins/biosynthesis
8.
Neuropsychobiology ; 77(1): 1-7, 2019.
Article in English | MEDLINE | ID: mdl-30110692

ABSTRACT

INTRODUCTION: The aim of this study was to expand on this field of work by examining, within a cohort of pregnant women with diagnosed clinical anxiety, the mRNA expression of a panel of genes associated with the cortisol pathway and comparing them to controls. METHODS: Placental samples were obtained from 24 pregnant women, 12 with a diagnosed anxiety disorder and 12 with no psychiatric history, within 30 min of delivery. Differential expression analysis of 85 genes known to be involved in glucocorticoid synthesis, metabolism or signalling was conducted for the: (1) full sample, (2) those at term without labour (5 cases, 7 controls) and (3) those at term with labour (7 cases, 5 controls). Correlation analyses between gene expression and measures of anxiety and depressive symptom severity were also conducted. RESULTS: No robust difference in placental gene expression between pregnant women with and without anxiety disorder was found nor did we detect robust differences by labour status. However, correlational analyses putatively showed a decrease in PER1 expression was associated with an increase in anxiety symptom severity, explaining up to 32% of the variance in anxiety symptom severity. DISCUSSION: Overall, the strongest correlation was found between a decrease in placental PER1 expression and increased anxiety scores. Labour status was found to have a profound effect on mRNA expression. The placental samples obtained from women following labour produced greater numbers of significant differences in mRNA species expression suggesting that in long-standing anxiety the placenta may respond differently under conditions of chronic stress.


Subject(s)
Anxiety/genetics , Anxiety/metabolism , Gene Expression , Hydrocortisone/biosynthesis , Placenta/metabolism , Signal Transduction , Adult , Case-Control Studies , Depression/metabolism , Female , Humans , Labor, Obstetric/metabolism , Period Circadian Proteins/biosynthesis , Period Circadian Proteins/genetics , Pregnancy , Young Adult
9.
Neurotox Res ; 35(2): 421-431, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30328585

ABSTRACT

A single injection of LPS produced low-grade neuroinflammation leading to Parkinson's disease (PD) in mice several months later. Whether such a phenomenon occurs in rats and whether such low-grade neuroinflammation would aggravate rotenone (ROT) neurotoxicity and disrupts circadian clock gene/protein expressions were examined in this study. Male rats were given two injections of LPS (2.5-7.5 mg/kg), and neuroinflammation and dopamine neuron loss were evident 3 months later. Seven months after a single LPS (5 mg/kg) injection, rats received low doses of ROT (0.5 mg/kg, sc, 5 times/week for 4 weeks) to examine low-grade neuroinflammation on ROT toxicity. LPS plus ROT produced more pronounced non-motor and motor dysfunctions than LPS or ROT alone in behavioral tests, and decreased mitochondrial complex 1 activity, together with aggravated neuroinflammation and neuron loss. The expressions of clock core genes brain and muscle Arnt-like protein-1 (Bmal1), locomotor output cycles kaput (Clock), and neuronal PAS domain protein-2 (Npas2) were decreased in LPS, ROT, and LPS plus ROT groups. The expressions of circadian feedback genes Periods (Per1 and Per2) were also decreased, but Cryptochromes (Cry1 and Cry2) were unaltered. The circadian clock target genes nuclear receptor Rev-Erbα (Nr1d1), and D-box-binding protein (Dbp) expressions were also decreased. Consistent with the transcript levels, circadian clock protein BMAL1, CLOCK, NR1D1, and DBP were also decreased. Thus, LPS-induced chronic low-grade neuroinflammation potentiated ROT neurotoxicity and disrupted circadian clock gene/protein expression, suggesting a role of disrupted circadian in PD development and progression. Graphical Abstract ᅟ.


Subject(s)
Circadian Clocks/physiology , Inflammation Mediators/metabolism , Nerve Degeneration/metabolism , Period Circadian Proteins/biosynthesis , Rotenone/toxicity , Animals , Circadian Clocks/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Gene Expression , Inflammation/chemically induced , Inflammation/metabolism , Insecticides/toxicity , Lipopolysaccharides/toxicity , Male , Nerve Degeneration/chemically induced , Period Circadian Proteins/genetics , Rats , Rats, Sprague-Dawley
10.
Biochem Biophys Res Commun ; 508(3): 871-876, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30538043

ABSTRACT

Exercise increases skeletal muscle health in part by altering the types of genes that are transcribed. Previous work suggested that glucocorticoids signal through the protein Regulated in Development and DNA Damage 1 (REDD1) to regulate gene expression following acute aerobic exercise. The present study shows that expression of the core clock gene, Period1, is among those modulated by the glucocorticoid-REDD1 signaling pathway in skeletal muscle. We also provide evidence that Aldosterone and Epinephrine contribute to the regulation of Period1 expression via REDD1. These data show that adrenal stress hormones signal through REDD1 to regulate skeletal muscle gene expression, specifically those of the core clock, following acute aerobic exercise.


Subject(s)
Gene Expression Regulation , Glucocorticoids/pharmacology , Muscle, Skeletal/metabolism , Period Circadian Proteins/genetics , Physical Conditioning, Animal , Transcription Factors/genetics , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Aldosterone/pharmacology , Animals , Cells, Cultured , Corticosterone/pharmacology , Dexamethasone/pharmacology , Epinephrine/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction , Muscle Fibers, Skeletal/drug effects , Period Circadian Proteins/biosynthesis , Receptors, Glucocorticoid/metabolism , Transcription Factors/biosynthesis , Transcription Factors/physiology
11.
Sci Rep ; 8(1): 15547, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341352

ABSTRACT

Animals create implicit memories of the time of day that significant events occur then anticipate the recurrence of those conditions at the same time on subsequent days. We tested the hypothesis that implicit time memory for daily encounters relies on the setting of the canonical circadian clockwork in brain areas involved in the formation or expression of context memories. We conditioned mice to avoid locations paired with a mild foot shock at one of two Zeitgeber times set 8 hours apart. Place avoidance was exhibited only when testing time matched the prior training time. The suprachiasmatic nucleus, dorsal striatum, nucleus accumbens, cingulate cortex, hippocampal complex, and amygdala were assessed for clock gene expression. Baseline phase dependent differences in clock gene expression were found in most tissues. Evidence for conditioned resetting of a molecular circadian oscillation was found only in the striatum (dorsal striatum and nucleus accumbens shell), and specifically for Per2 expression. There was no evidence of glucocorticoid stress response in any tissue. The results are consistent with a model where temporal conditioning promotes a selective Per2 response in dopamine-targeted brain regions responsible for sensorimotor integration, without resetting the entire circadian clockwork.


Subject(s)
Circadian Clocks , Conditioning, Psychological , Corpus Striatum/physiology , Gene Expression , Period Circadian Proteins/biosynthesis , RNA, Messenger/biosynthesis , Animals , Gene Expression Profiling , Mice, Inbred C57BL , Models, Neurological , Time
12.
Proc Natl Acad Sci U S A ; 115(30): 7825-7830, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29991600

ABSTRACT

Misalignment between internal circadian rhythmicity and externally imposed behavioral schedules, such as occurs in shift workers, has been implicated in elevated risk of metabolic disorders. To determine underlying mechanisms, it is essential to assess whether and how peripheral clocks are disturbed during shift work and to what extent this is linked to the central suprachiasmatic nuclei (SCN) pacemaker and/or misaligned behavioral time cues. Investigating rhythms in circulating metabolites as biomarkers of peripheral clock disturbances may offer new insights. We evaluated the impact of misaligned sleep/wake and feeding/fasting cycles on circulating metabolites using a targeted metabolomics approach. Sequential plasma samples obtained during a 24-h constant routine that followed a 3-d simulated night-shift schedule, compared with a simulated day-shift schedule, were analyzed for 132 circulating metabolites. Nearly half of these metabolites showed a 24-h rhythmicity under constant routine following either or both simulated shift schedules. However, while traditional markers of the circadian clock in the SCN-melatonin, cortisol, and PER3 expression-maintained a stable phase alignment after both schedules, only a few metabolites did the same. Many showed reversed rhythms, lost their rhythms, or showed rhythmicity only under constant routine following the night-shift schedule. Here, 95% of the metabolites with a 24-h rhythmicity showed rhythms that were driven by behavioral time cues externally imposed during the preceding simulated shift schedule rather than being driven by the central SCN circadian clock. Characterization of these metabolite rhythms will provide insight into the underlying mechanisms linking shift work and metabolic disorders.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Fasting/blood , Gene Expression Regulation/physiology , Hydrocortisone/blood , Melatonin/blood , Period Circadian Proteins/biosynthesis , Adult , Female , Humans , Male
13.
PLoS One ; 13(6): e0199777, 2018.
Article in English | MEDLINE | ID: mdl-29940038

ABSTRACT

BACKGROUND: TGF-ß signaling is a cellular pathway that functions in most cells and has been shown to play a role in multiple processes, such as the immune response, cell differentiation and proliferation. Recent evidence suggests a possible interaction between TGF-ß signaling and the molecular circadian oscillator. The current study aims to characterize this interaction in the zebrafish at the molecular and behavioral levels, taking advantage of the early development of a functional circadian clock and the availability of light-entrainable clock-containing cell lines. RESULTS: Smad3a, a TGF-ß signaling-related gene, exhibited a circadian expression pattern throughout the brain of zebrafish larvae. Both pharmacological inhibition and indirect activation of TGF-ß signaling in zebrafish Pac-2 cells caused a concentration dependent disruption of rhythmic promoter activity of the core clock gene Per1b. Inhibition of TGF-ß signaling in intact zebrafish larvae caused a phase delay in the rhythmic expression of Per1b mRNA. TGF-ß inhibition also reversibly disrupted, phase delayed and increased the period of circadian rhythms of locomotor activity in zebrafish larvae. CONCLUSIONS: The current research provides evidence for an interaction between the TGF-ß signaling pathway and the circadian clock system at the molecular and behavioral levels, and points to the importance of TGF-ß signaling for normal circadian clock function. Future examination of this interaction should contribute to a better understanding of its underlying mechanisms and its influence on a variety of cellular processes including the cell cycle, with possible implications for cancer development and progression.


Subject(s)
Circadian Clocks/physiology , Gene Expression Regulation/physiology , Period Circadian Proteins/biosynthesis , Signal Transduction/physiology , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Line , Female , Male , Period Circadian Proteins/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
14.
FASEB J ; 32(10): 5626-5639, 2018 10.
Article in English | MEDLINE | ID: mdl-29733691

ABSTRACT

Mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs) are two closely related hormone-activated transcription factors that regulate major pathophysiologic functions. High homology between these receptors accounts for the crossbinding of their corresponding ligands, MR being activated by both aldosterone and cortisol and GR essentially activated by cortisol. Their coexpression and ability to bind similar DNA motifs highlight the need to investigate their respective contributions to overall corticosteroid signaling. Here, we decipher the transcriptional regulatory mechanisms that underlie selective effects of MRs and GRs on shared genomic targets in a human renal cellular model. Kinetic, serial, and sequential chromatin immunoprecipitation approaches were performed on the period circadian protein 1 ( PER1) target gene, providing evidence that both receptors dynamically and cyclically interact at the same target promoter in a specific and distinct transcriptional signature. During this process, both receptors regulate PER1 gene by binding as homo- or heterodimers to the same promoter region. Our results suggest a novel level of MR-GR target gene regulation, which should be considered for a better and integrated understanding of corticosteroid-related pathophysiology.-Le Billan, F., Amazit, L., Bleakley, K., Xue, Q.-Y., Pussard, E., Lhadj, C., Kolkhof, P., Viengchareun, S., Fagart, J., Lombès, M. Corticosteroid receptors adopt distinct cyclical transcriptional signatures.


Subject(s)
Gene Expression Regulation , Nucleotide Motifs , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Response Elements , Signal Transduction , Transcription, Genetic , Cell Line , Humans , Period Circadian Proteins/biosynthesis , Period Circadian Proteins/genetics , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics
15.
Proc Natl Acad Sci U S A ; 115(16): 4276-4281, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610316

ABSTRACT

Endogenous circadian clocks control 24-h physiological and behavioral rhythms in mammals. Here, we report a real-time in vivo fluorescence recording system that enables long-term monitoring of circadian rhythms in the brains of freely moving mice. With a designed reporter of circadian clock gene expression, we tracked robust Cry1 transcription reporter rhythms in the suprachiasmatic nucleus (SCN) of WT, Cry1-/- , and Cry2-/- mice in LD (12 h light, 12 h dark) and DD (constant darkness) conditions and verified that signals remained stable for over 6 mo. Further, we recorded Cry1 transcriptional rhythms in the subparaventricular zone (SPZ) and hippocampal CA1/2 regions of WT mice housed under LD and DD conditions. By using a Cre-loxP system, we recorded Per2 and Cry1 transcription rhythms specifically in vasoactive intestinal peptide (VIP) neurons of the SCN. Finally, we demonstrated the dynamics of Per2 and Cry1 transcriptional rhythms in SCN VIP neurons following an 8-h phase advance in the light/dark cycle.


Subject(s)
Circadian Rhythm/physiology , Cryptochromes/biosynthesis , Fiber Optic Technology/methods , Fluorometry/methods , Neurons/metabolism , Period Circadian Proteins/biosynthesis , Suprachiasmatic Nucleus/metabolism , Animals , Bacterial Proteins/analysis , Bacterial Proteins/genetics , CA1 Region, Hippocampal/metabolism , CA2 Region, Hippocampal/metabolism , Cells, Cultured , Circadian Rhythm/genetics , Cryptochromes/deficiency , Cryptochromes/genetics , Dependovirus/genetics , Fiber Optic Technology/instrumentation , Fluorometry/instrumentation , Genes, Reporter , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hypothalamus, Anterior/metabolism , Longitudinal Studies , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Movement , Neurons/chemistry , Neurons/classification , Optical Fibers , Organ Specificity , Period Circadian Proteins/genetics , Photoperiod , Suprachiasmatic Nucleus/cytology , Transcription, Genetic , Vasoactive Intestinal Peptide/analysis
16.
Eur J Neurosci ; 48(8): 2696-2717, 2018 10.
Article in English | MEDLINE | ID: mdl-29396876

ABSTRACT

Neuronal oscillations of the brain, such as those observed in the cortices and hippocampi of behaving animals and humans, span across wide frequency bands, from slow delta waves (0.1 Hz) to ultra-fast ripples (600 Hz). Here, we focus on ultra-slow neuronal oscillators in the hypothalamic suprachiasmatic nuclei (SCN), the master daily clock that operates on interlocking transcription-translation feedback loops to produce circadian rhythms in clock gene expression with a period of near 24 h (< 0.001 Hz). This intracellular molecular clock interacts with the cell's membrane through poorly understood mechanisms to drive the daily pattern in the electrical excitability of SCN neurons, exhibiting an up-state during the day and a down-state at night. In turn, the membrane activity feeds back to regulate the oscillatory activity of clock gene programs. In this review, we emphasise the circadian processes that drive daily electrical oscillations in SCN neurons, and highlight how mathematical modelling contributes to our increasing understanding of circadian rhythm generation, synchronisation and communication within this hypothalamic region and across other brain circuits.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Models, Theoretical , Neurons/physiology , Period Circadian Proteins/biosynthesis , Suprachiasmatic Nucleus/physiology , Animals , Gene Expression , Humans , Period Circadian Proteins/genetics , Time Factors
17.
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
18.
Int J Oncol ; 52(1): 279-287, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115399

ABSTRACT

Recent studies have demonstrated that abnormal expression of the clock gene PER2 is closely associated with the development of a variety of cancer types. However, the expression of PER2 in oral squamous cell carcinoma (OSCC), a common malignant tumor in humans, and its correlations with the clinicopathological parameters and survival time of OSCC patients and the altered expression of important tumor-related genes remain unclear. In the present study, we detected the mRNA and protein expression levels of PER2, PIK3CA, PTEN, P53, P14ARF and caspase­8 in OSCC tissues and cancer-adjacent oral mucosa by reverse transcription-quantitative PCR (RT-qPCR), western blotting and immunohistochemistry. The results showed that the PER2, PTEN, P53, P14ARF and caspase­8 mRNA and protein expression levels in OSCC were significantly reduced compared with those in cancer-adjacent tissues. Additionally, the PIK3CA protein expression level was significantly increased in OSCC tissues, whereas the mRNA level was not. Decreased expression of PER2 was significantly associated with advanced clinical stage and the presence of lymphatic metastasis in OSCC patients. Patients with PER2­negative expression had a significantly shorter survival time than those with PER2­positive expression. PER2 expression was negatively correlated with PIK3CA and P53 levels, and positively correlated with PTEN, P14ARF and caspase­8 levels. In summary, the results of this study suggest that loss of PER2 expression is closely associated with the genesis and development of OSCC and that PER2 may be an important prognostic biomarker in OSCC. PER2 may serve an antitumor role via the P53/P14ARF, PIK3CA/AKT and caspase­8 pathways.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Mouth Neoplasms/genetics , Period Circadian Proteins/genetics , Adult , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Period Circadian Proteins/biosynthesis , Prognosis , Squamous Cell Carcinoma of Head and Neck
19.
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
20.
Brain Res ; 1671: 93-101, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28709906

ABSTRACT

Shiftworkers are exposed to several adverse health conditions, one being eating at night. Food consumption at an unnatural time-of-day is thought to be one of the main factors responsible for the increased risk of developing metabolic diseases, such as obesity and diabetes mellitus. The underlying mechanism is considered to include disruption of the circadian organization of physiology, leading to disruption of metabolism. When food is consumed at night, the hypothalamus, a brain region central to homeostasis, receives contradicting input from the central clock and the systemic circulation. This study investigated how timing of feeding affects hypothalamic function by studying, in different hypothalamic nuclei, expression of clock genes and key neuropeptide genes involved in energy metabolism, including orexin, melanin-concentrating hormone (MCH) and neuropeptide Y. Animals with food available ad libitum showed diurnal variation in the expression of clock genes Per1 and Per2 in the perifornical area and arcuate nucleus. Clock gene rhythms were lost in both nuclei when food was restricted to the light (i.e., sleep) period. Neuropeptide genes did not display significant daily variation in either feeding groups, except for orexin-receptor 1 in ad libitum animals. Analysis of genes involved in glutamatergic and GABAergic signaling did not reveal diurnal variation in expression, nor effects of feeding time. In conclusion, feeding at the 'wrong' time-of-day not only induces desynchronization between brain and body clocks but also within the hypothalamus, which may contribute further to the underlying pathology of metabolic dysregulation.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm/physiology , Feeding Behavior/physiology , Gene Expression Regulation , Hypothalamus/physiology , Neuropeptides/genetics , Suprachiasmatic Nucleus Neurons/physiology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Brain/metabolism , Energy Metabolism , Food Deprivation , Hypothalamus/metabolism , Male , Motor Activity/physiology , Neuropeptides/biosynthesis , Neuropeptides/metabolism , Period Circadian Proteins/biosynthesis , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Photoperiod , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...