Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
Front Immunol ; 15: 1402395, 2024.
Article in English | MEDLINE | ID: mdl-38895112

ABSTRACT

Background: Circadian rhythm disruption (CRD) is thought to increase the risk of inflammatory bowel disease. The deletion of Bmal1, a core transcription factor, leads to a complete loss of the circadian rhythm and exacerbates the severity of dextran sodium sulfate (DSS)-induced colitis in mice. However, the underlying mechanisms by which CRD and Bmal1 mediate IBD are still unclear. Methods: We used a CRD mouse model, a mouse colitis model, and an in vitro model of colonic epithelial cell monolayers. We also knocked down and overexpressed Bmal1 in Caco-2 cells by transfecting lentivirus in vitro. The collected colon tissue and treated cells were assessed and analyzed using immunohistochemistry, immunofluorescence staining, quantitative reverse transcription-polymerase chain reaction, western blot, flow cytometry, transmission electron microscopy, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling staining. Results: We found that CRD mice with downregulated Bmal1 expression were more sensitive to DSS-induced colitis and had more severely impaired intestinal barrier function than wild-type mice. Bmal1-/- mice exhibited more severe colitis, accompanied by decreased tight junction protein levels and increased apoptosis of intestinal epithelial cells compared with wild-type mice, which were alleviated by using the autophagy agonist rapamycin. Bmal1 overexpression attenuated Lipopolysaccharide-induced apoptosis of intestinal epithelial cells and impaired intestinal epithelial cells barrier function in vitro, while inhibition of autophagy reversed this protective effect. Conclusion: This study suggests that CRD leads to the downregulation of Bmal1 expression in the colon, which may exacerbate DSS-induced colitis in mice, and that Bmal1 may serve as a novel target for treating inflammatory bowel disease.


Subject(s)
ARNTL Transcription Factors , Circadian Rhythm , Colitis , Dextran Sulfate , Disease Models, Animal , Down-Regulation , Intestinal Mucosa , Mice, Knockout , Animals , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Colitis/chemically induced , Colitis/metabolism , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Humans , Circadian Rhythm/genetics , Caco-2 Cells , Mice, Inbred C57BL , Apoptosis , Male , Chronobiology Disorders/metabolism , Chronobiology Disorders/genetics
2.
Sci Rep ; 14(1): 10777, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38734687

ABSTRACT

Emerging evidence has documented that circadian rhythm disorders could be related to cardiovascular diseases. However, there is limited knowledge on the direct adverse effects of circadian misalignment on the heart. This study aimed to investigate the effect of chronic circadian rhythm disorder on heart homeostasis in a mouse model of consistent jetlag. The jetlag model was induced in mice by a serial 8-h phase advance of the light cycle using a light-controlled isolation box every 4 days for up to 3 months. Herein, we demonstrated for the first time that chronic circadian rhythm disorder established in the mouse jetlag model could lead to HFpEF-like phenotype such as cardiac hypertrophy, cardiac fibrosis, and cardiac diastolic dysfunction, following the attenuation of the Clock-sGC-cGMP-PKG1 signaling. In addition, clock gene knock down in cardiomyocytes induced hypertrophy via decreased sGC-cGMP-PKG signaling pathway. Furthermore, treatment with an sGC-activator riociguat directly attenuated the adverse effects of jetlag model-induced cardiac hypertrophy, cardiac fibrosis, and cardiac diastolic dysfunction. Our data suggest that circadian rhythm disruption could induce HFpEF-like phenotype through downregulation of the clock-sGC-cGMP-PKG1 signaling pathway. sGC could be one of the molecular targets against circadian rhythm disorder-related heart disease.


Subject(s)
CLOCK Proteins , Chronobiology Disorders , Cyclic GMP , Heart Failure , Soluble Guanylyl Cyclase , Animals , Male , Mice , Chronobiology Disorders/complications , Chronobiology Disorders/metabolism , Circadian Rhythm/physiology , CLOCK Proteins/metabolism , CLOCK Proteins/genetics , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Disease Models, Animal , Heart Failure/metabolism , Heart Failure/etiology , Heart Failure/physiopathology , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Phenotype , Signal Transduction , Soluble Guanylyl Cyclase/metabolism , Stroke Volume
3.
J Agric Food Chem ; 72(21): 12184-12197, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38745351

ABSTRACT

Oolong tea polyphenols (OTP) have attracted wide attention due to their ability to reduce inflammatory response, regulate gut microbiota, and improve cognitive function. However, exactly how the gut microbiota modulates nervous system activity is still an open question. We previously expounded that supplementing with OTP alleviated neuroinflammation in circadian rhythm disorder (CRD) mice. Here, we showed that OTP can relieve microglia activation by reducing harmful microbial metabolites lipopolysaccharide (LPS) that alleviate CRD-induced cognitive decline. Mechanistically, OTP suppressed the inflammation response by regulating the gut microbiota composition, including upregulating the relative abundance of Muribaculaceae and Clostridia_UCG-014 and downregulating Desulfovibrio, promoting the production of short-chain fatty acids (SCFAs). Moreover, the use of OTP alleviated intestinal barrier damage and decreased the LPS transport to the serum. These results further inhibited the activation of microglia, thus alleviating cognitive impairment by inhibiting neuroinflammation, neuron damage, and neurotoxicity metabolite glutamate elevation. Meanwhile, OTP upregulated the expression of synaptic plasticity-related protein postsynaptic density protein 95 (PSD-95) and synaptophysin (SYN) by elevating the brain-derived neurotrophic factor (BDNF) level. Taken together, our findings suggest that the OTP has the potential to prevent CRD-induced cognition decline by modulating gut microbiota and microbial metabolites.


Subject(s)
Camellia sinensis , Chronobiology Disorders , Cognitive Dysfunction , Gastrointestinal Microbiome , Mice, Inbred C57BL , Neuroprotective Agents , Polyphenols , Tea , Gastrointestinal Microbiome/drug effects , Animals , Polyphenols/pharmacology , Polyphenols/administration & dosage , Mice , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/etiology , Male , Tea/chemistry , Camellia sinensis/chemistry , Neuroprotective Agents/pharmacology , Chronobiology Disorders/metabolism , Chronobiology Disorders/drug therapy , Chronobiology Disorders/physiopathology , Humans , Bacteria/classification , Bacteria/drug effects , Bacteria/metabolism , Bacteria/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Microglia/drug effects , Microglia/metabolism , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Plant Extracts/chemistry
4.
Physiol Behav ; 279: 114523, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38492912

ABSTRACT

Melatonin is a neurohormone synthesized by the pineal gland to regulate the circadian rhythms and has proven to be effective in treating drug addiction and dependence. However, the effects of melatonin to modulate the drug-seeking behavior of fentanyl and its underlying molecular mechanism is elusive. This study was designed to investigate the effects of melatonin on fentanyl - induced behavioral sensitization and circadian rhythm disorders in mice. The accompanying changes in the expression of Brain and Muscle Arnt-Like (BMAL1), tyrosine hydroxylase (TH), and monoamine oxidase A (MAO-A) in relevant brain regions including the suprachiasmatic nucleus (SCN), nucleus accumbens (NAc), prefrontal cortex (PFC), and hippocampus (Hip) were investigated by western blot assays to dissect the mechanism by which melatonin modulates fentanyl - induced behavioral sensitization and circadian rhythm disorders. The present study suggest that fentanyl (0.05, 0.1 and 0.2 mg/kg) could induce behavioral sensitization and melatonin (30.0 mg/kg) could attenuate the behavioral sensitization and circadian rhythm disorders in mice. Fentanyl treatment reduced the expression of BMAL1 and MAO-A and increased that of TH in relevant brain regions. Furthermore, melatonin treatment could reverse the expression levels of BMAL1, MAO-A, and TH. In conclusion, our study demonstrate for the first time that melatonin has therapeutic potential for fentanyl addiction.


Subject(s)
Chronobiology Disorders , Melatonin , Mice , Animals , Melatonin/pharmacology , Melatonin/therapeutic use , Melatonin/metabolism , ARNTL Transcription Factors , Fentanyl/pharmacology , Fentanyl/therapeutic use , Fentanyl/metabolism , Suprachiasmatic Nucleus/metabolism , Circadian Rhythm/physiology , Chronobiology Disorders/metabolism , Monoamine Oxidase/metabolism , Monoamine Oxidase/pharmacology
5.
Mol Neurobiol ; 61(8): 6013-6030, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38265551

ABSTRACT

Although growing evidence suggests close correlations between Alzheimer's disease (AD) and circadian rhythm disruption (CRD), few studies have focused on the influence of circadian rhythm on levels of immune cells in AD. We aimed to delineate the mechanism underlying the effects of circadian related genes on T cell immune function in AD. A total of 112 brain samples were used to construct the CRD-related model by performing weighted gene co-expression network analysis and machine learning algorithms (LASSO, SVM-RFE, and RF). The ssGSEA method was used to calculate the CRDscore in order to quantify CRD status. Using single-cell transcriptome data of CSF cells, we investigated the CD4+ T cell metabolism and cell-cell communication in high- and low-risk CRD groups. Connectivity map (CMap) was applied to explore small molecule drugs targeting CRD, and the expression of the signature gene GPR4 was further validated in AD. The CRDscore algorithm, which is based on 23 circadian-related genes, can effectively classify the CRD status in AD datasets. The single-cell analysis revealed that the CD4+ T cells with high CRDscore were characterized by hypometabolism. Cell communication analysis revealed that CD4+ T cells might be involved in promoting CD8+ T cell adhesion under CRD, which may facilitate T cell infiltration into the brain parenchyma. Overall, this study indicates the potential connotation of circadian rhythm in AD, providing insights into understanding T cell metabolic reprogramming under CRD.


Subject(s)
Alzheimer Disease , CD4-Positive T-Lymphocytes , Circadian Rhythm , Single-Cell Analysis , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Humans , CD4-Positive T-Lymphocytes/metabolism , Single-Cell Analysis/methods , Circadian Rhythm/genetics , Sequence Analysis, RNA/methods , Transcriptome/genetics , Gene Regulatory Networks , Male , Chronobiology Disorders/metabolism , Chronobiology Disorders/genetics , Brain/metabolism , Brain/pathology , Metabolic Reprogramming
6.
BMC Mol Cell Biol ; 25(1): 1, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38166556

ABSTRACT

Chronic alcohol exposure increases liver damage such as lipid accumulation and hepatitis, resulting in hepatic cirrhosis. Chronic alcohol intake is known to disturb circadian rhythms in humans and animals. DEC1, a basic helix-loop-helix transcription factor, plays an important role in the circadian rhythm, inflammation, immune responses, and tumor progression. We have previously shown that Dec1 deficiency inhibits stresses such as periodontal inflammation and perivascular fibrosis of the heart. However, the significance of Dec1 deficiency in chronic alcohol consumption remains unclear. In the present study, we investigated whether the biological stress caused by chronic alcohol intake is inhibited in Dec1 knockout mice. We treated control and Dec1 knockout mice for three months by providing free access to 10% alcohol. The Dec1 knockout mice consumed more alcohol than control mice, however, we observed severe hepatic lipid accumulation and circadian rhythm disturbance in control mice. In contrast, Dec1 knockout mice exhibited little effect on these outcomes. We also investigated the expression of peroxisome proliferator-activated receptors (PPARs) and AMP-activated protein kinase (AMPK), which are involved in the regulation of fatty acid metabolism. Immunohistochemical analysis revealed increases of phosphorylation AMPK and PPARa but decreases PPARg in Dec1 knockout mice compared to that in control mice. This indicates a molecular basis for the inhibition of hepatic lipid accumulation in alcohol-treated Dec1 knockout mice. These results suggest a novel function of Dec1 in alcohol-induced hepatic lipid accumulation and circadian rhythm disorders.


Subject(s)
Chronobiology Disorders , Homeodomain Proteins , Humans , Mice , Animals , Homeodomain Proteins/metabolism , AMP-Activated Protein Kinases/metabolism , Liver/metabolism , Ethanol/metabolism , Mice, Knockout , Inflammation/metabolism , Chronobiology Disorders/metabolism , Lipids
7.
Crit Rev Food Sci Nutr ; 63(24): 7126-7147, 2023.
Article in English | MEDLINE | ID: mdl-35187990

ABSTRACT

Circadian rhythm is an intrinsic mechanism developed by organisms to adapt to external environmental signals. Nowadays, owing to the job and after-work entertainment, staying up late - Circadian rhythm disorders (CRD) are common. CRD is linked to the development of fatty liver, type 2 diabetes, and chronic gastroenteritis, which affecting the body's metabolic and inflammatory responses via multi-organ crosstalk (gut-liver-brain axis, etc.). However, studies on the mechanisms of multi-organ interactions by CRD are still weak. Current studies on therapeutic agents for CRD remain inadequate, and phytochemicals have been shown to alleviate CRD-induced syndromes that may be used for CRD-therapy in the future. Tea, a popular phytochemical-rich beverage, reduces glucolipid metabolism and inflammation. But it is immature and unclear in the mechanisms of alleviation of CRD-mediated syndrome. Here, we have analyzed the threat of CRD to hosts and their offspring' health from the perspective of the "gut-liver-brain" axis. The potential mechanisms of tea in alleviating CRD were further explored. It might be by interfering with bile acid metabolism, tryptophan metabolism, and G protein-coupled receptors, with FXR, AHR, and GPCR as potential targets. We hope to provide new perspectives on the role of tea in the prevention and mitigation of CRD.HighlightsThe review highlights the health challenges of CRD via the gut-liver-brain axis.CRD research should focus on the health effects on healthy models and its offspring.Tea may prevent CRD by regulating bile acid, tryptophan, and GPCR.Potential targets for tea prevention and mitigation of CRD include FXR, AHR and GPCR.A comprehensive assessment mechanism for tea in improving CRD should be established.


Subject(s)
Chronobiology Disorders , Diabetes Mellitus, Type 2 , Humans , Syndrome , Diabetes Mellitus, Type 2/metabolism , Tryptophan/pharmacology , Liver , Tea/chemistry , Chronobiology Disorders/metabolism , Bile Acids and Salts/metabolism , Brain
8.
J Neurodev Disord ; 14(1): 33, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35610565

ABSTRACT

BACKGROUND: Regulator of calcineurin 1 (RCAN1) is overexpressed in Down syndrome (DS), but RCAN1 levels are also increased in Alzheimer's disease (AD) and normal aging. AD is highly comorbid among individuals with DS and is characterized in part by progressive neurodegeneration that resembles accelerated aging. Importantly, abnormal RCAN1 levels have been demonstrated to promote memory deficits and pathophysiology that appear symptomatic of DS, AD, and aging. Anomalous diurnal rest-activity patterns and circadian rhythm disruptions are also common in DS, AD, and aging and have been implicated in facilitating age-related cognitive decline and AD progression. However, no prior studies have assessed whether RCAN1 dysregulation may also promote the age-associated alteration of rest-activity profiles and circadian rhythms, which could in turn contribute to neurodegeneration in DS, AD, and aging. METHODS: The present study examined the impacts of RCAN1 deficiency and overexpression on the photic entrainment, circadian periodicity, intensity and distribution, diurnal patterning, and circadian rhythmicity of wheel running in young (3-6 months old) and aged (9-14 months old) mice of both sexes. RESULTS: We found that daily RCAN1 levels in the hippocampus and suprachiasmatic nucleus (SCN) of light-entrained young mice are generally constant and that balanced RCAN1 expression is necessary for normal circadian locomotor activity rhythms. While the light-entrained diurnal period was unaltered, RCAN1-null and RCAN1-overexpressing mice displayed lengthened endogenous (free-running) circadian periods like mouse models of AD and aging. In light-entrained young mice, RCAN1 deficiency and overexpression also recapitulated the general hypoactivity, diurnal rest-wake pattern fragmentation, and attenuated amplitudes of circadian activity rhythms reported in DS, preclinical and clinical AD, healthily aging individuals, and rodent models thereof. Under constant darkness, RCAN1-null and RCAN1-overexpressing mice displayed altered locomotor behavior indicating circadian clock dysfunction. Using the Dp(16)1Yey/+ (Dp16) mouse model for DS, which expresses three copies of Rcan1, we found reduced wheel running activity and rhythmicity in both light-entrained and free-running young Dp16 mice like young RCAN1-overexpressing mice. Critically, these diurnal and circadian deficits were rescued in part or entirely by restoring Rcan1 to two copies in Dp16 mice. We also found that RCAN1 deficiency but not RCAN1 overexpression altered protein levels of the clock gene Bmal1 in the SCN. CONCLUSIONS: Collectively, this study's findings suggest that both loss and aberrant gain of RCAN1 precipitate anomalous light-entrained diurnal and circadian activity patterns emblematic of DS, AD, and possibly aging.


Subject(s)
Aging , Alzheimer Disease , Calcium-Binding Proteins , Chronobiology Disorders , DNA-Binding Proteins , Down Syndrome , Muscle Proteins , Aging/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Chronobiology Disorders/genetics , Chronobiology Disorders/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/metabolism , Female , Male , Mice , Motor Activity/physiology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Suprachiasmatic Nucleus/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
J Cardiovasc Transl Res ; 15(5): 985-997, 2022 10.
Article in English | MEDLINE | ID: mdl-35257279

ABSTRACT

Circadian rhythm disorders can accelerate atherosclerosis. This study aimed to determine the role of circadian disordered macrophages in atherosclerotic development. Mice were divided into NC group (normal circadian rhythm), L24 group (constant light), D12L12 group (weekly shift light/dark cycle), and D24 group (constant dark). Atherosclerotic progression was significantly accelerated in L24, D12L12, and D24 groups. Peritoneal macrophages from circadian disruption groups exhibited enhanced cytokine secretion and foam cell formation. Migration and proliferation of vascular smooth muscle cells (VSMCs) were increased under the conditioned medium of circadian disordered macrophages. The blockade of CD36 markedly inhibited foam cell formation. Compared with blocking CD36 or TLR4 alone, the co-inhibition of CD36 and TLR4 in macrophages further reduced cytokine secretion and more effectively inhibited VSMC migration and proliferation. In conclusion, the activation of CD36 and TLR4 in circadian disordered macrophages promotes foam cell formation and cytokine secretion and enhances VSMC migration and proliferation. Circadian rhythm disorders promote lipid uptake and cytokine secretion of macrophages by regulating CD36 and TLR4, and enhance VSMC migration and proliferation through the paracrine effect of macrophages.


Subject(s)
Atherosclerosis , Chronobiology Disorders , Animals , Mice , Atherosclerosis/metabolism , CD36 Antigens/metabolism , Cell Proliferation , Chronobiology Disorders/metabolism , Cytokines/metabolism , Foam Cells/metabolism , Lipoproteins, LDL , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Toll-Like Receptor 4/metabolism , Circadian Rhythm
10.
Food Funct ; 13(3): 1048-1061, 2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35050270

ABSTRACT

Circadian rhythm is essential to human physiological homeostasis and health. The oscillation of host circadian rhythm affects the composition and function of intestinal microbiota, meanwhile, the normal operation of host circadian rhythm depends on the diurnal changes of intestinal microbiota. The imbalance of intestinal micro-ecology or the disorder of host circadian rhythm may lead to psychiatric disorders, while the intervention of plant polysaccharides is a possible way to alleviate circadian rhythm disturbance and the related psychiatric diseases. This review discusses the interaction between host circadian rhythm and intestinal microbiota and their effects on psychiatric disorders, and proposes a possible strategy of plant polysaccharides to alleviate circadian rhythm disorders and related psychiatric disorders by regulating intestinal micro-ecology.


Subject(s)
Chronobiology Disorders/complications , Chronobiology Disorders/metabolism , Gastrointestinal Microbiome/physiology , Mental Disorders/complications , Mental Disorders/metabolism , Plants/metabolism , Polysaccharides/metabolism , Animals , Chronobiology Disorders/physiopathology , Circadian Rhythm , Cues , Homeostasis , Humans , Intestines/metabolism , Intestines/physiopathology , Mental Disorders/physiopathology , Mice , Polysaccharides/physiology
11.
Front Endocrinol (Lausanne) ; 12: 723918, 2021.
Article in English | MEDLINE | ID: mdl-34745000

ABSTRACT

Background: Hepatic lipid metabolism regulates biliary composition and influences the formation of cholesterol gallstones. The genes Hmgcr and Cyp7a1, which encode key liver enzymes, are regulated by circadian rhythm-related transcription factors. We aimed to investigate the effect of circadian rhythm disruption on hepatic cholesterol and bile acid metabolism and the incidence of cholesterol stone formation. Methods: Adult male C57BL/6J mice were fed either a lithogenic diet (LD) only during the sleep phase (time-restricted lithogenic diet feeding, TRF) or an LD ad libitum (non-time-restricted lithogenic diet feeding, nTRF) for 4 weeks. Food consumption, body mass gain, and the incidence of gallstones were assessed. Circulating metabolic parameters, lipid accumulation in the liver, the circadian expression of hepatic clock and metabolic genes, and the gut microbiota were analyzed. Results: TRF caused a dysregulation of the circadian rhythm in the mice, characterized by significant differences in the circadian expression patterns of clock-related genes. In TRF mice, the circadian rhythms in the expression of genes involved in bile acid and cholesterol metabolism were disrupted, as was the circadian rhythm of the gut microbiota. These changes were associated with high biliary cholesterol content, which promoted gallstone formation in the TRF mice. Conclusion: Disordered circadian rhythm is associated with abnormal hepatic bile acid and cholesterol metabolism in mice, which promotes gallstone formation.


Subject(s)
Chronobiology Disorders/complications , Gallstones/etiology , Gastrointestinal Microbiome , Lipid Metabolism , Liver/metabolism , Animals , Cholesterol/metabolism , Chronobiology Disorders/etiology , Chronobiology Disorders/metabolism , Chronobiology Disorders/microbiology , Circadian Rhythm/physiology , Diet/adverse effects , Gallstones/metabolism , Gallstones/microbiology , Gastrointestinal Microbiome/physiology , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Time Factors
12.
Oxid Med Cell Longev ; 2021: 8419415, 2021.
Article in English | MEDLINE | ID: mdl-34567414

ABSTRACT

The aim of this study is to compare the regulatory abilities of citrus flavonoids on the oscillating expression of circadian genes. Seven varieties of citrus fruits and twenty-five citrus flavonoids were selected and evaluated. Per2 luciferase bioluminescence report system and serum shock were used to induce circadian gene expression in mouse microglia BV-2 cells. In vivo experiments were carried out using C57BL6/J mice to evaluate the regulation of flavonoids on the oscillatory expression of liver biorhythm genes. Lipopolysaccharide was used to interfere the gene oscillating expression. QRT-PCR was performed to detect the expression of circadian rhythm-related genes, including Clock, Bmal1, Per1, Per2, Per3, Cry1, Cry2, Rev-erbα, Rev-erbß, Rorα, Dbp, and Npas2. The results show that the polymethoxyflavones (PMFs) exerted stronger circadian gene regulatory capability, while the flavonoids containing glycosides showed no biological activity. Also, all tested flavonoids decreased LPS-induced nitric oxide release, but only polymethoxyflavones inhibited circadian rhythm disorder. PMFs inhibited Nlrp3 inflammasome-related genes and proteins, including Nlrp3, IL-1ß, ASC, and Caspase1, while other flavonoids only affected IL-1ß and Caspase1 expression. This mechanism was preliminarily verified using the Nlrp3 inhibitor INF39.


Subject(s)
CLOCK Proteins/metabolism , Chronobiology Disorders/drug therapy , Circadian Rhythm/drug effects , Citrus/chemistry , Flavones/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Animals , CLOCK Proteins/genetics , Chronobiology Disorders/chemically induced , Chronobiology Disorders/metabolism , Chronobiology Disorders/pathology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Polyphenols/pharmacology
13.
Front Endocrinol (Lausanne) ; 12: 678468, 2021.
Article in English | MEDLINE | ID: mdl-34484111

ABSTRACT

Compelling evidence in rats support the idea that gestational chronodisruption induces major changes in maternal circadian rhythms and fetal development and that these changes impact adult life at many physiological levels. Using a model of chronic photoperiod shifting throughout gestation (CPS), in which pregnant female rats (Sprague-Dawley strain; n = 16 per group) were exposed to lighting schedule manipulation every 3-4 days reversing the photoperiod completely or light/dark photoperiod (12/12; LD), we explored in the adult rat male offspring body weight gain, glucose homeostasis, adipose tissue content, adipose tissue response to norepinephrine (NE), and adipose tissue proteomic in the basal condition with standard diet (SD) and in response to high-fat diet (HFD). In adult CPS male (100-200 days old; n = 8 per group), we found increasing body weight, under SD and adiposity. Also, we found an increased response to intraperitoneal glucose (IGTT). After 12 weeks of HFD, white adipose tissue depots in CPS offspring were increased further, and higher IGTT and lower intraperitoneal insulin tolerance response were found, despite the lack of changes in food intake. In in vitro experiments, we observed that adipose tissue (WAT and BAT) glycerol response to NE from CPS offspring was decreased, and it was completely abolished by HFD. At the proteomic level, in CPS adipose tissue, 275 proteins displayed differential expression, compared with LD animals fed with a standard diet. Interestingly, CPS offspring and LD fed with HFD showed 20 proteins in common (2 upregulated and 18 downregulated). Based on these common proteins, the IPA analysis found that two functional pathways were significantly altered by CPS: network 1 (AKT/ERK) and network 2 (TNF/IL4; data are available via ProteomeXchange with identifier PXD026315). The present data show that gestational chronodisruption induced deleterious effects in adipose tissue recruitment and function, supporting the idea that adipose tissue function was programmed in utero by gestational chronodisruption, inducing deficient metabolic responses that persist into adulthood.


Subject(s)
Adipose Tissue/metabolism , Circadian Rhythm/physiology , Glucose/metabolism , Photoperiod , Prenatal Exposure Delayed Effects/metabolism , Animals , Chronobiology Disorders/metabolism , Female , Homeostasis/physiology , Male , Pregnancy , Proteomics , Rats , Rats, Sprague-Dawley
14.
J Cell Mol Med ; 25(20): 9851-9862, 2021 10.
Article in English | MEDLINE | ID: mdl-34523794

ABSTRACT

Adiponectin is an adipocyte-derived hormone, which is closely associated with the development of Alzheimer's disease (AD) and has potential preventive and therapeutic significance. In the present study, we explored the relationship between adiponectin and circadian rhythm disorder in AD, the effect of adiponectin on the abnormal expression of Bmal1 mRNA/protein induced by amyloid-ß protein 31-35 (Aß31-35), and the underlying mechanism of action. We found that adiponectin-knockout mice exhibited amyloid-ß deposition, circadian rhythm disorders and abnormal expression of Bmal1. Adiponectin ameliorated the abnormal expression of the Bmal1 mRNA/protein caused by Aß31-35 by inhibiting the activity of glycogen synthase kinase 3ß (GSK3ß). These results suggest that adiponectin deficiency could induce circadian rhythm disorders and abnormal expression of the Bmal1 mRNA/protein, whilst exogenous administration of adiponectin may improve Aß31-35-induced abnormal expression of Bmal1 by inhibiting the activity of GSK3ß, thus providing a novel idea for the treatment of AD.


Subject(s)
Adiponectin/metabolism , Amyloid beta-Peptides/metabolism , Chronobiology Disorders/etiology , Chronobiology Disorders/metabolism , Peptide Fragments/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Adiponectin/genetics , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/adverse effects , Animals , Cell Line , Chronobiology Disorders/pathology , Disease Models, Animal , Disease Susceptibility , Gene Expression , Glycogen Synthase Kinase 3 beta/metabolism , Male , Mice , Mice, Knockout , Peptide Fragments/adverse effects , Protein Aggregation, Pathological/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology
15.
Endocrinol Diabetes Metab ; 4(1): e00171, 2021 01.
Article in English | MEDLINE | ID: mdl-33532613

ABSTRACT

Background: Melatonin is a hormone secreted by the pineal gland in a circadian rhythmic manner with peak synthesis at night. Melatonin signalling was suggested to play a critical role in metabolism during the circadian disruption. Methods: Melatonin-proficient (C3H-f+/+ or WT) and melatonin receptor type 1 knockout (MT1 KO) male and female mice were phase-advanced (6 hours) once a week for 6 weeks. Every week, we measured weight, food intake and basal glucose levels. At the end of the experiment, we sacrificed the animals and measured the blood's plasma for lipids profile (total lipids, phospholipids, triglycerides and total cholesterol), metabolic hormones profiles (ghrelin, leptin, insulin, glucagon, glucagon-like-peptide and resistin) and the body composition. Results: Environmental circadian disruption (ECD) did not produce any significant effects in C3H-f+/+, while it increased lipids profile in MT1 KO with the significant increase observed in total lipids and triglycerides. For metabolic hormones profile, ECD decreased plasma ghrelin and increased plasma insulin in MT1 KO females. Under control condition, MT1 KO females have significantly different body weight, fat mass, total lipids and total cholesterol than the control C3H-f+/+ females. Conclusion: Our data show that melatonin-proficient mice are not affected by ECD. When the MT1 receptors are removed, ECD induced dyslipidaemia in males and females with females experiencing the most adverse effect. Overall, our data demonstrate that MT1 signalling is an essential modulator of lipid and metabolic homeostasis during ECD.


Subject(s)
Chronobiology Disorders/etiology , Chronobiology Disorders/metabolism , Dyslipidemias/etiology , Ghrelin/metabolism , Insulin/metabolism , Leptin/metabolism , Lipid Metabolism , Receptor, Melatonin, MT1/physiology , Signal Transduction/physiology , Animals , Female , Glucagon/metabolism , Male , Mice, Knockout , Resistin/metabolism
16.
Med Oncol ; 37(10): 90, 2020 Sep 14.
Article in English | MEDLINE | ID: mdl-32926243

ABSTRACT

Endometrial cancer (EC) is one of the most common gynecologic malignancies, and the incidence rate of night shift among women workers is higher than that in the general population. Circadian rhythm disorder, mainly rhythm gene, is related to various tumor onset, including EC. This study described the sleep/night-shift features of EC patients, explored the mechanism of the circadian clock gene PER and investigated prognostic and functional values of Per1 caused by night shift. A total of 619 subjects were enrolled and divided into two groups according to night-shift duties (rhythm group and control group), analyzed for clinical risk factors and night shift features of endometrial carcinoma. Then samples were randomly selected for sequencing and western blot were performed, and the function of overexpressed PER1 in ishikawa cells was explored. We noticed that severer EC patients experienced night-shift more frequently and with longer durations. A total of 58,174 differentially expressed genes were discovered, mainly rhythm genes and related to up and downstream regulatory genes. Western blot showed that the rhythm group had elevated protein expression of BCAS4, TUBB2B and RSPO4, and decreased expression of PER1 and PER2 in night-shift. In TCGA-EC datasets, PER1 was decreased in the EC patients with a significantly positive correlation with PER2, and higher PER1 expression indicated longer survival, opposite to TUBB2B. The research of overexpressing PER1 gene in EC ishikawa cells found that PER1 can promote apoptosis, expression of TNF-a, IL-6 and PD-1/PD-L1, inhibit the tumor invasion and expression of TUBB2B gene. Together, EC severity was associated with night-shift and rhythm disorders. The rhythm relating factors PER1, TUBB2B and tumor immune factors may regulate the mechanisms of EC onset and progression.


Subject(s)
Endometrial Neoplasms , Period Circadian Proteins/metabolism , Sleep Disorders, Circadian Rhythm , Tubulin/metabolism , Adult , Aged , Chronobiology Disorders/genetics , Chronobiology Disorders/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Female , Gene Regulatory Networks , Humans , Middle Aged , Period Circadian Proteins/genetics , Sleep Disorders, Circadian Rhythm/genetics , Sleep Disorders, Circadian Rhythm/metabolism , Transcriptome , Tubulin/genetics
17.
J Parkinsons Dis ; 10(4): 1467-1476, 2020.
Article in English | MEDLINE | ID: mdl-32986681

ABSTRACT

BACKGROUND: Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) signal the environmental light to mediate circadian photoentrainment and sleep-wake cycles. There is high prevalence of circadian and sleep disruption in people with Parkinson's disease, however the underlying mechanisms of these symptoms are not clear. OBJECTIVE: Based on recent evidence of anatomical and functional loss of melanopsin ganglion cells in Parkinson's disease, we evaluate the link between melanopsin function, circadian, and sleep behavior. METHODS: The pupil light reflex and melanopsin-mediated post-illumination pupil response were measured using chromatic pupillometry in 30 optimally medicated people with Parkinson's disease and 29 age-matched healthy controls. Circadian health was determined using dim light melatonin onset, sleep questionnaires, and actigraphy. Ophthalmic examination quantified eye health and optical coherence tomography measured retinal thickness. RESULTS: The melanopsin-mediated post-illumination pupil response amplitudes were significantly reduced in Parkinson's disease (p < 0.0001) and correlated with poor sleep quality (r2 = 33; p < 0.001) and nerve fiber layer thinning (r2 = 0.40; p < 0.001). People with Parkinson's disease had significantly poorer sleep quality with higher subjective sleep scores (p < 0.05) and earlier melatonin onset (p = 0.01). Pupil light (outer retinal) response metrics, daily light exposure and outer retinal thickness were similar between the groups (p > 0.05). CONCLUSION: Our evidence-based data identify a mechanism through which inner retinal ipRGC dysfunction contributes to sleep disruption in Parkinson's disease in the presence of normal outer retinal (rod-cone photoreceptor) function. Our findings provide a rationale for designing new treatment approaches in Parkinson's disease through melanopsin photoreceptor-targeted light therapies for improving sleep-wake cycles.


Subject(s)
Chronobiology Disorders/physiopathology , Melatonin/metabolism , Parkinson Disease/physiopathology , Pupil/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Ganglion Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Rod Opsins/metabolism , Sleep Wake Disorders/physiopathology , Actigraphy , Aged , Chronobiology Disorders/etiology , Chronobiology Disorders/metabolism , Female , Humans , Male , Middle Aged , Parkinson Disease/complications , Parkinson Disease/metabolism , Sleep Wake Disorders/etiology , Sleep Wake Disorders/metabolism , Tomography, Optical Coherence
18.
Int Rev Neurobiol ; 154: 393-412, 2020.
Article in English | MEDLINE | ID: mdl-32739012

ABSTRACT

The 24-h rotational period of the earth has driven evolution of biological systems that serve to synchronize organismal physiology and behavior to this predictable environmental event. In mammals, the circadian (circa, "about" and dia, "a day") clock keeps 24-h time at the organismal and cellular level, optimizing biological function for a given time of day. The most obvious circadian output is the sleep-wake cycle, though countless bodily functions, ranging from hormone levels to cognitive function, are influenced by the circadian clock. Here we discuss the regulation of metabolic pathways by the circadian clock, discuss the evidence implicating circadian and sleep disruption in neurodegenerative diseases, and suggest some possible connections between the clock, metabolism, and neurodegenerative disease.


Subject(s)
Chronobiology Disorders/metabolism , Mitochondria/metabolism , NAD/metabolism , Neurodegenerative Diseases/metabolism , Sirtuins/metabolism , Sleep Wake Disorders/metabolism , Chronobiology Disorders/complications , Humans , Neurodegenerative Diseases/etiology , Sleep Wake Disorders/etiology
19.
Rev Endocr Metab Disord ; 21(4): 465-478, 2020 12.
Article in English | MEDLINE | ID: mdl-32691289

ABSTRACT

The last four decades, we assist to an increasing scientific interest on melatonin, a circadian hormone, a metabolic regulator which influences not only plants' metabolism and their defense against pathogens but mostly the animals and humans' metabolic pathways, their response to circadian disruption, stress and burnout syndrome. In humans, as a hormonal regulator, produced in the pineal grand as well in mitochondria, melatonin is involved in different, complex intracellular signaling pathways, with antioxidant and immune stimulating effects, proving to act as a circadian synchronizer, as a preventive and therapeutic agent in many degenerative diseases, and especially in hormone-dependent cancers. Preclinical or clinical studies showed recently the mechanisms involved in regulating the cellular activity, its role in aging and circadian disturbances and impact on degenerative diseases. Melatonin proved to have an anti-inflammatory, antiapoptotic and powerful antioxidant effect by subtle mechanisms in mitochondrial metabolic pathways. This overview includes recent and relevant literature data related to the impact of endogenous and exogeneous melatonin on the prevention of cancer progression and treatment of various degenerative diseases. Metabolomics, an emerging new omics' technology, based on high performance liquid chromatography coupled with mass spectrometry is presented as an encouraging technique to fingerprint and realize a precise evaluation and monitoring of the turnover of melatonin and its metabolites in different pathological circumstances.


Subject(s)
Aging , Chronobiology Disorders , Melatonin , Metabolic Networks and Pathways , Metabolomics , Mitochondria , Neoplasms , Neurodegenerative Diseases , Aging/metabolism , Animals , Chronobiology Disorders/drug therapy , Chronobiology Disorders/metabolism , Humans , Melatonin/pharmacology , Melatonin/physiology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism
20.
Pflugers Arch ; 472(5): 513-526, 2020 05.
Article in English | MEDLINE | ID: mdl-32363530

ABSTRACT

Circadian rhythms are generated by the circadian clock, a self-sustained internal timing system that exhibits 24-h rhythms in the body. In mammals, circadian rhythms are driven by a central clock located in suprachiasmatic nucleus and various peripheral clocks located in different tissues and organs of the body. Many cellular, behavioral, and physiological processes are regulated by the circadian clock in coordination with environmental cues. The process of metabolism is also under circadian regulation. Loss of synchronization between the internal clock and environmental zeitgebers results in disruption of the circadian rhythms that seriously impacts metabolic homeostasis leading to changed eating behavior, altered glucose and lipid metabolism, and weight gain. This in turn augments the risk of having various cardio-metabolic disorders such as obesity, diabetes, metabolic syndrome, and cardiovascular disease. This review sheds light on circadian rhythms and their role in metabolism with the identification of gaps in the current knowledge that remain to be explored in these fields. In this review, the molecular mechanisms underlying circadian rhythms have been elaborated first. Then, the focus has been kept on explaining the physiological significance of circadian rhythms in regulating metabolism. Finally, the implications for metabolism when these rhythms are disrupted due to genetic mutations or social and occupational needs enforced by modern lifestyle have been discussed.


Subject(s)
Chronobiology Disorders/metabolism , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm , Animals , Chronobiology Disorders/genetics , Chronobiology Disorders/physiopathology , Circadian Rhythm Signaling Peptides and Proteins/genetics , Gastrointestinal Microbiome , Homeostasis , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...