Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nature ; 592(7856): 763-767, 2021 04.
Article in English | MEDLINE | ID: mdl-33762728

ABSTRACT

Systemic insulin sensitivity shows a diurnal rhythm with a peak upon waking1,2. The molecular mechanism that underlies this temporal pattern is unclear. Here we show that the nuclear receptors REV-ERB-α and REV-ERB-ß (referred to here as 'REV-ERB') in the GABAergic (γ-aminobutyric acid-producing) neurons in the suprachiasmatic nucleus (SCN) (SCNGABA neurons) control the diurnal rhythm of insulin-mediated suppression of hepatic glucose production in mice, without affecting diurnal eating or locomotor behaviours during regular light-dark cycles. REV-ERB regulates the rhythmic expression of genes that are involved in neurotransmission in the SCN, and modulates the oscillatory firing activity of SCNGABA neurons. Chemogenetic stimulation of SCNGABA neurons at waking leads to glucose intolerance, whereas restoration of the temporal pattern of either SCNGABA neuron firing or REV-ERB expression rescues the time-dependent glucose metabolic phenotype caused by REV-ERB depletion. In individuals with diabetes, an increased level of blood glucose after waking is a defining feature of the 'extended dawn phenomenon'3,4. Patients with type 2 diabetes with the extended dawn phenomenon exhibit a differential temporal pattern of expression of REV-ERB genes compared to patients with type 2 diabetes who do not have the extended dawn phenomenon. These findings provide mechanistic insights into how the central circadian clock regulates the diurnal rhythm of hepatic insulin sensitivity, with implications for our understanding of the extended dawn phenomenon in type 2 diabetes.


Subject(s)
Circadian Rhythm , GABAergic Neurons/physiology , Insulin Resistance , Liver/physiology , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Animals , Blood Glucose , Circadian Clocks , Diabetes Mellitus, Type 2 , Female , Glucose/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Photoperiod , Suprachiasmatic Nucleus/cytology , Synaptic Transmission
2.
Theranostics ; 10(9): 4168-4182, 2020.
Article in English | MEDLINE | ID: mdl-32226546

ABSTRACT

REV-ERBα (NR1D1) is a circadian clock component that functions as a transcriptional repressor. Due to its role in direct modulation of metabolic genes, REV-ERBα is regarded as an integrator of cell metabolism with circadian clock. Accordingly, REV-ERBα is first proposed as a drug target for treating sleep disorders and metabolic syndromes (e.g., dyslipidaemia, hyperglycaemia and obesity). Recent years of studies uncover a rather broad role of REV-ERBα in pathological conditions including local inflammatory diseases, heart failure and cancers. Moreover, REV-ERBα is involved in regulation of circadian drug metabolism that has implications in chronopharmacology. In the meantime, recent years have witnessed discovery of an array of new REV-ERBα ligands most of which have pharmacological activities in vivo. In this article, we review the regulatory role of REV-ERBα in various types of diseases and discuss the underlying mechanisms. We also describe the newly discovered ligands and the old ones together with their targeting potential. Despite well-established pharmacological effects of REV-ERBα ligands in animals (preclinical studies), no progress has been made regarding their translation to clinical trials. This implies certain challenges associated with drug development of REV-ERBα ligands. In particular, we discuss the potential challenges related to drug safety (or adverse effects) and bioavailability. For new drug development, it is advocated that REV-ERBα should be targeted to treat local diseases and a targeting drug should be locally distributed, avoiding the adverse effects on other tissues.


Subject(s)
Circadian Clocks , Nuclear Receptor Subfamily 1, Group D, Member 1 , Animals , Drug Development , Humans , Ligands , Mice , Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Signal Transduction
3.
Drug Metab Dispos ; 47(11): 1333-1342, 2019 11.
Article in English | MEDLINE | ID: mdl-31515204

ABSTRACT

Flavin-containing monooxygenase 5 (FMO5) is a phase I enzyme that plays an important role in xenobiotic metabolism. Here, we aimed to characterize diurnal rhythms of Fmo5 expression and activity in mouse liver and to investigate the potential roles of clock genes (Bmal1, Rev-erbα, and E4bp4) in the generation of diurnal rhythms. Fmo5 mRNA and protein showed robust diurnal rhythms, with peak values at zeitgeber time (ZT) 10/14 and trough values at ZT2/22 in mouse liver. Consistently, a diurnal rhythm was observed for in vitro microsomal Baeyer-Villiger oxidation of pentoxifylline (PTX), a specific reaction catalyzed by Fmo5. Pharmacokinetic studies revealed a more extensive Baeyer-Villiger oxidation of PTX at dosing time of ZT14 than at ZT2, consistent with the diurnal pattern of Fmo5 protein. Fmo5 expression was downregulated and its rhythm was blunted in Bmal1 -/- and Rev-erbα -/- mice. Positive regulation of Fmo5 by Bmal1 and Rev-erbα was confirmed in primary mouse hepatocytes and/or Hepa1-6 cells. Furthermore, Fmo5 expression was upregulated and its rhythm was attenuated in E4bp4 -/- mice. Negative regulation of Fmo5 by E4bp4 was validated using primary mouse hepatocytes. Combined luciferase reporter and chromatin immunoprecipitation assays demonstrated that Bmal1 (a known Rev-erbα activator) activated Fmo5 transcription via direct binding to an E-box (-1822/-1816 bp) in the promoter, whereas E4bp4 (a known Rev-erbα target gene) inhibited Fmo5 transcription by binding to two D-boxes (-1726/-1718 and -804/-796 bp). In conclusion, circadian clock genes control diurnal expression of Fmo5 through transcriptional actions on E-box and D-box cis-elements. SIGNIFICANCE STATEMENT: Hepatic Fmo5 displayed diurnal rhythmicities in expression and activity in mice. We uncovered the molecular mechanism by which the rhythmic Fmo5 expression was generated. Fmo5 promoter presents E-box and D-box binding elements for transcriptional actions from circadian clock proteins such as Bmal1, E4bp4, and Dbp. These findings have implications for understanding clock-controlled drug metabolism and for facilitating the practice of chronotherapeutics.


Subject(s)
Circadian Rhythm/physiology , Gene Expression Regulation, Enzymologic , Liver/enzymology , Oxygenases/genetics , Animals , Basic-Leucine Zipper Transcription Factors/physiology , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Oxygenases/metabolism , Promoter Regions, Genetic , Transcription, Genetic
4.
Acta Biochim Biophys Sin (Shanghai) ; 51(9): 908-914, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31411318

ABSTRACT

Perturbation of the circadian rhythm damages the biological characteristics of cells and leads to their dysfunction. Rev-erbα, an important gene in the transcription-translation loop of circadian rhythm, is involved in regulating the balance between pro-inflammation and anti-inflammation. The disruption of this balance in human endometrial stroma cells (hESCs) destroys their biological behavior function in maintaining the menstrual cycle and embryonic implantation. Whether pharmacological modulation of Rev-erbα affects the inflammation of hESCs remains unclear. In this study, we treated hESCs with lipopolysaccharide (LPS) and found that LPS treatment increased the mRNA levels of pro-inflammatory cytokines, such as interleukin (IL)-1ß, IL-6, IL-8, IL-18, and TNFα, and the secretion of IL-6. SR9009, a Rev-erbα agonist, significantly alleviated the LPS-induced production of pro-inflammatory cytokines in hESCs. Meanwhile, knockdown of Rev-erbα increased the expressions of IL-1ß, IL-6, and IL-8, accompanied by an increased mRNA level of the core clock gene Bmal1. Western blot analysis showed that SR9009 inhibited the expression of toll-like receptor 4 (TLR4) and the activation of NF-κB induced by LPS. All these findings suggested that pharmacological activation of Rev-erbα attenuated the LPS-induced inflammatory response of hESCs by suppressing TLR4-regulated NF-κB activation. This study may provide a strategy for preventing inflammation-related endometrial dysfunction and infertility or recurrent implantation failure.


Subject(s)
Cytokines/immunology , Endometritis/immunology , Endometrium/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Stromal Cells/immunology , Endometritis/chemically induced , Endometrium/cytology , Endometrium/pathology , Female , Humans , Lipopolysaccharides , NF-kappa B/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Stromal Cells/cytology , Stromal Cells/pathology , Toll-Like Receptor 4/metabolism
5.
Autophagy ; 15(10): 1854-1856, 2019 10.
Article in English | MEDLINE | ID: mdl-31318631

ABSTRACT

Most essential physiological functions in mammals show a 24-h rhythmic pattern, which includes sleep-wake, feeding-non-feeding cycles and energy metabolism. Recent studies indicate that macroautophagy/autophagy also displays a robust circadian rhythmicity following the daily feeding pattern in adult mammals. We discovered that MiT-TFE transcription factors TFEB and TFE3, master regulators of autophagy and lysosomal biogenesis, are activated in a circadian manner and drive the expression of NR1D1/REV-ERBα, a key component of the core clockwork, thus revealing a molecular link between the nutrient-driven circadian cycle and the light-induced molecular clock. The dynamic balance between TFEB and TFE3 activation and NR1D1 expression is responsible for the modulation and oscillation of autophagy and metabolism genes.


Subject(s)
Autophagy/physiology , Circadian Rhythm/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Biological Clocks/genetics , Mice , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Time Factors
6.
Sci Rep ; 9(1): 10171, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31308426

ABSTRACT

The circadian clock regulates behavioural and physiological processes in a 24-h cycle. The nuclear receptors REV-ERBα and REV-ERBß are involved in the cell-autonomous circadian transcriptional/translational feedback loops as transcriptional repressors. A number of studies have also demonstrated a pivotal role of REV-ERBs in regulation of metabolic, neuronal, and inflammatory functions including bile acid metabolism, lipid metabolism, and production of inflammatory cytokines. Given the multifunctional role of REV-ERBs, it is important to elucidate the mechanism through which REV-ERBs exert their functions. To this end, we established a Rev-erbα/Rev-erbß double-knockout mouse embryonic stem (ES) cell model and analyzed the circadian clock and clock-controlled output gene expressions. A comprehensive mRNA-seq analysis revealed that the double knockout of both Rev-erbα and Rev-erbß does not abrogate expression rhythms of E-box-regulated core clock genes but drastically changes a diverse set of other rhythmically-expressed output genes. Of note, REV-ERBα/ß deficiency does not compromise circadian expression rhythms of PER2, while REV-ERB target genes, Bmal1 and Npas2, are significantly upregulated. This study highlight the relevance of REV-ERBs as pivotal output mediators of the mammalian circadian clock.


Subject(s)
Circadian Clocks/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Animals , Circadian Clocks/physiology , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Embryonic Stem Cells/physiology , Gene Expression/genetics , Gene Expression Regulation/genetics , Mammals/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Transcription Factors/metabolism , Transcriptional Activation/genetics
7.
EMBO J ; 38(12)2019 06 17.
Article in English | MEDLINE | ID: mdl-31126958

ABSTRACT

Autophagy and energy metabolism are known to follow a circadian pattern. However, it is unclear whether autophagy and the circadian clock are coordinated by common control mechanisms. Here, we show that the oscillation of autophagy genes is dependent on the nutrient-sensitive activation of TFEB and TFE3, key regulators of autophagy, lysosomal biogenesis, and cell homeostasis. TFEB and TFE3 display a circadian activation over the 24-h cycle and are responsible for the rhythmic induction of genes involved in autophagy during the light phase. Genetic ablation of TFEB and TFE3 in mice results in deregulated autophagy over the diurnal cycle and altered gene expression causing abnormal circadian wheel-running behavior. In addition, TFEB and TFE3 directly regulate the expression of Rev-erbα (Nr1d1), a transcriptional repressor component of the core clock machinery also involved in the regulation of whole-body metabolism and autophagy. Comparative analysis of the cistromes of TFEB/TFE3 and REV-ERBα showed an extensive overlap of their binding sites, particularly in genes involved in autophagy and metabolic functions. These data reveal a direct link between nutrient and clock-dependent regulation of gene expression shedding a new light on the crosstalk between autophagy, metabolism, and circadian cycles.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Circadian Clocks , Energy Metabolism , Nutrients/physiology , Animals , Autophagy/drug effects , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Binding Sites , Cells, Cultured , Circadian Clocks/drug effects , Circadian Clocks/genetics , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Energy Metabolism/drug effects , Energy Metabolism/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Nutrients/pharmacology , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription Factors/physiology
8.
Proc Natl Acad Sci U S A ; 116(25): 12147-12152, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31127047

ABSTRACT

The nuclear receptors REV-ERBα and -ß link circadian rhythms and metabolism. Like other nuclear receptors, REV-ERB activity can be regulated by ligands, including naturally occurring heme. A putative ligand, SR9009, has been reported to elicit a range of beneficial effects in healthy as well as diseased animal models and cell systems. However, the direct involvement of REV-ERBs in these effects of SR9009 has not been thoroughly assessed, as experiments were not performed in the complete absence of both proteins. Here, we report the generation of a mouse model for conditional genetic deletion of REV-ERBα and -ß. We show that SR9009 can decrease cell viability, rewire cellular metabolism, and alter gene transcription in hepatocytes and embryonic stem cells lacking both REV-ERBα and -ß. Thus, the effects of SR9009 cannot be used solely as surrogate for REV-ERB activity.


Subject(s)
Cell Proliferation/drug effects , Nuclear Receptor Subfamily 1, Group D, Member 1/drug effects , Pyrrolidines/pharmacology , Receptors, Cytoplasmic and Nuclear/drug effects , Repressor Proteins/drug effects , Thiophenes/pharmacology , Animals , Cell Respiration/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Repressor Proteins/physiology
9.
Brain Behav Immun ; 78: 116-130, 2019 05.
Article in English | MEDLINE | ID: mdl-30682503

ABSTRACT

The orphan nuclear receptors REV-ERBα and REV-ERBß (REV-ERBs) are crucial in the regulation of inflammatory-related gene transcription in astroglioma cells, but their role in nociceptive transduction has yet to be elaborated. Spinal dorsal horn astrocytes contribute to the maintenance of chronic pain. Treatment of cultured spinal astrocytes with specific REV-ERBs agonists SR9009 or GSK4112 significantly prevented lipopolysaccharide (LPS)-induced mRNA upregulation of pronociceptive molecules interleukin-1ß (IL-1ß) mRNA, interleukin-6 (IL-6) mRNA and matrix metalloprotease-9 (MMP-9) mRNA, but not CCL2 mRNA expression. Treatment with SR9009 also blocked tumor necrosis factor-induced IL-1ß mRNA, IL-6 mRNA and MMP-9 mRNA. In addition, treatment with SR9009 significantly blocked LPS-induced upregulation of IL-1ß protein, IL-6 protein and MMP-9 activity. The inhibitory effects of SR9009 on LPS-induced expression of pronociceptive molecules were blocked by knockdown of REV-ERBs expression with short interference RNA, confirming that SR9009 exerts its effect through REV-ERBs. Intrathecal LPS treatment in male mice induces hind paw mechanical hypersensitivity, and upregulation of IL-1ß mRNA, IL-6 mRNA and glial fibrillary acidic protein (GFAP) expression in spinal dorsal horn. Intrathecal pretreatment of SR9009 prevented the onset of LPS-induced mechanical hypersensitivity, cytokine expression and GFAP expression. Intrathecal injection of SR9009 also ameliorated mechanical hypersensitivity during the maintenance phase of complete Freund's adjuvant-induced inflammatory pain and partial sciatic nerve ligation-, paclitaxel-, and streptozotocin-induced neuropathy in mice. The current findings suggest that spinal astrocytic REV-ERBs could be critical in the regulation of nociceptive transduction through downregulation of pronociceptive molecule expression. Thus, spinal REV-ERBs could be an effective therapeutic target in the treatment of chronic pain.


Subject(s)
Astrocytes/metabolism , Nociceptors/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Animals , Cytokines/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Hyperalgesia/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Male , Mice , Neuralgia/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Primary Cell Culture , Pyrrolidines/pharmacology , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Spinal Cord/metabolism , Spinal Cord Dorsal Horn/metabolism , Thiophenes/pharmacology , Tumor Necrosis Factor-alpha/metabolism
10.
Thorax ; 74(4): 413-416, 2019 04.
Article in English | MEDLINE | ID: mdl-30301818

ABSTRACT

The importance of circadian factors in managing patients is poorly understood. We present two retrospective cohort studies showing that lungs reperfused between 4 and 8 AM have a higher incidence (OR 1.12; 95% CI 1.03 to 1.21; p=0.01) of primary graft dysfunction (PGD) in the first 72 hours after transplantation. Cooling of the donor lung, occurring during organ preservation, shifts the donor circadian clock causing desynchrony with the recipient. The clock protein REV-ERBα directly regulates PGD biomarkers explaining this circadian regulation while also allowing them to be manipulated with synthetic REV-ERB ligands.


Subject(s)
Circadian Clocks/physiology , Lung Transplantation/methods , Primary Graft Dysfunction/prevention & control , Adult , Aged , Animals , Female , Humans , Macrophages, Alveolar/metabolism , Male , Mice, Knockout , Middle Aged , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Organ Preservation/methods , Primary Graft Dysfunction/etiology , Retrospective Studies , Risk Factors , Time Factors , Tissue Donors , Transplant Recipients
11.
Eur J Neurosci ; 48(3): 1924-1943, 2018 08.
Article in English | MEDLINE | ID: mdl-30028550

ABSTRACT

Nuclear receptor subfamily 1, group D, member 1 (Nr1d1) (also known as Rev-erb alpha) has been linked to circadian rhythm regulation, mood-related behaviour and disorders associated with social deficits. Recent work from our laboratory found striking decreases in Nr1d1 in the nucleus accumbens (NAc) in the maternal condition and indirect evidence that Nr1d1 was interacting with numerous addiction and reward-related genes to modulate social reward. In this study, we applied our insights from the maternal state to nonparental adult mice to determine whether decreases in Nr1d1 expression in the NAc via adeno-associated viral (AAV) vectors and short hairpin RNA (shRNA)-mediated gene knockdown were sufficient to modulate social behaviours and mood-related behaviours. Knockdown of Nr1d1 in the NAc enhanced sociability and reduced anxiety, but did not affect depressive-like traits in female mice. In male mice, Nr1d1 knockdown had no significant behavioural effects. Microarray analysis of Nr1d1 knockdown in females identified changes in circadian rhythm and histone deacetylase genes and suggested possible drugs, including histone deacetylase inhibitors, that could mimic actions of Nr1d1 knockdown. Quantitative real-time PCR (qPCR) analysis confirmed expression upregulation of gene period circadian clock 1 (Per1) and period circadian clock 2 (Per2) with Nr1d1 knockdown. The evidence for roles for opioid-related genes opioid receptor, delta 1 (Oprd1) and preproenkephalin (Penk) was also found. Together, these results suggest that Nr1d1 in the NAc modulates sociability and anxiety-related behaviour in a sex-specific manner, and circadian, histone deacetylase and opioid-related genes may be involved in the expression of these behavioural phenotypes.


Subject(s)
Anxiety/physiopathology , Circadian Rhythm , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Nucleus Accumbens/physiology , Social Behavior , Animals , Female , Gene Knockdown Techniques , Male , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Reward
12.
PLoS One ; 13(5): e0196787, 2018.
Article in English | MEDLINE | ID: mdl-29723273

ABSTRACT

The nuclear receptors REV-ERBα and REV-ERBß have been demonstrated to be core members of the circadian clock and participate in the regulation of a diverse set of metabolic functions. Due to their overlapping tissue expression patterns and gene expression profiles, REV-ERBß is thought to be redundant to REV-ERBα. Recent work has highlighted REV-ERBα's role in the regulation of skeletal muscle oxidative capacity and mitochondrial biogenesis. Considering the similarity between the REV-ERBs and the hypothesized overlap in function, we sought to determine whether REV-ERBß-deficiency presented with a similar skeletal muscle phenotype as REV-ERBα-deficiency. Ectopic overexpression in C2C12 cells demonstrated that REV-ERBß drives mitochondrial biogenesis and the expression of genes involved in fatty acid oxidation. Intriguingly, knock down of REV-ERBß in C2C12 cultures also resulted in mitochondrial biogenesis and increased expression of genes involved in fatty acid ß-oxidation. To determine whether these effects occurred in vivo, we examined REV-ERBß-deficient mice and observed a similar increase in expression of genes involved in mitochondrial biogenesis and fatty acid ß-oxidation. Consistent with these results, REV-ERBß-deficient mice exhibited an altered metabolic phenotype compared to wild-type littermate controls when measured by indirect calorimetry. This likely compensated for the increased food consumption that occurred, possibly aiding in the maintenance of their weight over time. Since feeding behaviors are a direct circadian output, this study suggests that REV-ERBß may have more subtle effects on circadian behaviors than originally identified. Furthermore, these data implicate REV-ERBß in the control of skeletal muscle metabolism and energy expenditure and suggest that development of REV-ERBα versus REV-ERBß selective ligands may have therapeutic utility in the treatment of metabolic syndrome.


Subject(s)
Circadian Rhythm/physiology , Energy Metabolism/physiology , Mitochondria, Muscle/physiology , Muscle, Skeletal/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Repressor Proteins/physiology , Animals , Body Weight , Calorimetry, Indirect , Cell Line , Circadian Rhythm/genetics , Energy Metabolism/genetics , Fatty Acids/metabolism , Feeding Behavior/physiology , Female , Gene Expression Regulation , Male , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Organelle Biogenesis , Oxidation-Reduction , Oxidative Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/deficiency , Repressor Proteins/genetics
13.
Sci Rep ; 7(1): 14383, 2017 10 30.
Article in English | MEDLINE | ID: mdl-29085009

ABSTRACT

The nuclear receptor Rev-erb-α modulates hepatic lipid and glucose metabolism, adipogenesis and thermogenesis. We have previously demonstrated that Rev-erb-α is also an important regulator of skeletal muscle mitochondrial biogenesis and function, and autophagy. As such, Rev-erb-α over-expression in skeletal muscle or its pharmacological activation improved mitochondrial respiration and enhanced exercise capacity. Here, in gain- and loss-of function studies, we show that Rev-erb-α also controls muscle mass. Rev-erb-α-deficiency in skeletal muscle leads to increased expression of the atrophy-related genes (atrogenes), associated with reduced muscle mass and decreased fiber size. By contrast, in vivo and in vitro Rev-erb-α over-expression results in reduced atrogenes expression and increased fiber size. Finally, Rev-erb-α pharmacological activation blocks dexamethasone-induced upregulation of atrogenes and muscle atrophy. This study identifies Rev-erb-α as a promising pharmacological target to preserve muscle mass.


Subject(s)
Muscular Atrophy/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Adipogenesis , Animals , Autophagy , Cell Differentiation , Liver/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Diseases/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Repressor Proteins/genetics , Transcriptional Activation
14.
JCI Insight ; 2(17)2017 09 07.
Article in English | MEDLINE | ID: mdl-28878135

ABSTRACT

A cure for heart failure remains a major unmet clinical need, and current therapies targeting neurohomonal and hemodynamic regulation have limited efficacy. The pathological remodeling of the myocardium has been associated with a stereotypical gene expression program, which had long been viewed as the consequence and not the driver of the disease until very recently. Despite the advance, there is no therapy available to reverse the already committed gene program. Here, we demonstrate that transcriptional repressor REV-ERB binds near driver transcription factors across the genome. Pharmacological activation of REV-ERB selectively suppresses aberrant pathologic gene expression and prevents cardiomyocyte hypertrophy. In vivo, REV-ERBα activation prevents development of cardiac hypertrophy, reduces fibrosis, and halts progression of advanced heart failure in mouse models. Thus, to our knowledge, modulation of gene networks by targeting REV-ERBα represents a novel approach to heart failure therapy.


Subject(s)
Heart Failure/prevention & control , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Transcription, Genetic , Animals , Cardiotonic Agents/pharmacology , Gene Expression , Gene Regulatory Networks , Heart Failure/genetics , Humans , Hypertrophy/chemically induced , Male , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenylephrine/pharmacology , Rats , Rats, Sprague-Dawley
15.
Proc Natl Acad Sci U S A ; 113(20): 5730-5, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27143721

ABSTRACT

Certain components and functions of the immune system, most notably cytokine production and immune cell migration, are under circadian regulation. Such regulation suggests that circadian rhythms may have an effect on disease onset, progression, and resolution. In the vesicular stomatitis virus (VSV)-induced encephalitis model, the replication, caudal penetration, and survivability of intranasally applied VSV depends on both innate and adaptive immune mechanisms. In the current study, we investigated the effect of circadian time of infection on the progression and outcome of VSV-induced encephalitis and demonstrated a significant decrease in the survival rate in mice infected at the start of the rest cycle, zeitgeber time 0 (ZT0). The lower survival rate in these mice was associated with higher levels of circulating chemokine (C-C motif) ligand 2 (CCL2), a greater number of peripherally derived immune cells accumulating in the olfactory bulb (OB), and increased production of proinflammatory cytokines, indicating an immune-mediated pathology. We also found that the acrophase of molecular circadian clock component REV-ERBα mRNA expression in the OB coincides with the start of the active cycle, ZT12, when VSV infection results in a more favorable outcome. This result led us to hypothesize that REV-ERBα may mediate the circadian effect on survival following VSV infection. Blocking REV-ERBα activity before VSV administration resulted in a significant increase in the expression of CCL2 and decreased survival in mice infected at the start of the active cycle. These data demonstrate that REV-ERBα-mediated inhibition of CCL2 expression during viral-induced encephalitis may have a protective effect.


Subject(s)
Circadian Rhythm/immunology , Encephalitis/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Vesiculovirus/immunology , Adaptive Immunity , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Encephalitis/virology , Gene Expression , Male , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Mortality , Up-Regulation/immunology
16.
Yakugaku Zasshi ; 136(2): 297-308, 2016.
Article in Japanese | MEDLINE | ID: mdl-26831808

ABSTRACT

Animal defense mechanisms against both endogenous and exogenous toxic compounds function mainly through receptor-type transcription factors, including the constitutive androstane receptor (CAR). Following xenobiotic stimulation, CAR translocates into the nucleus and transactivates its target genes including oxygenic and conjugative enzymes and transporters in hepatocytes. We identified subcellular localization signals in the rat CAR: two nuclear localization signals (NLS1 and 2); two nuclear export signals (NES1 and 2); and a cytoplasmic retention region. The nuclear import of CAR is regulated by the importin-Ran system and microtubule network. Five splice variants (SV1-5) were identified in rat liver in addition to wild-type CAR. When expressed in immortalized cells, their artificial transcripts were inactive as transcription factors. A CAR mutant with three consecutive alanine residues inserted into the ligand-binding domain of CAR showed ligand-dependent activation of target genes in immortalized cells, which is in marked contrast to the constitutive transactivating nature of wild-type CAR. Using this assay system, androstenol and clotrimazole, both of which are inverse agonists of CAR, were classified as an antagonist and weak agonist, respectively. A member of the DEAD box DNA/RNA helicase family (DP97) and protein arginine methyltransferase 5 (PRMT5) were found to be gene (or promotor)-specific coactivators of CAR. The expression of the CAR gene might be under the control of clock genes mediated by the nuclear receptor Rev-erb-α.


Subject(s)
Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/physiology , Active Transport, Cell Nucleus , Androstenols , Animals , Cell Nucleus/metabolism , Circadian Rhythm/genetics , Clotrimazole , Constitutive Androstane Receptor , Gene Expression , Humans , Karyopherins/physiology , Mice , Microtubules/physiology , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Rats , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology , ran GTP-Binding Protein/physiology
17.
Diabetes Obes Metab ; 17 Suppl 1: 106-14, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26332975

ABSTRACT

Several hormones are regulated by circadian rhythms to adjust the metabolism to the light/dark cycles and feeding/activity patterns throughout the day. Circadian rhythms are mainly governed by the central clock located in the suprachiasmatic nucleus but also by clocks present in peripheral organs, like the endocrine pancreas. Plasma glucose levels and the main pancreatic hormones insulin and glucagon also exhibit daily variations. Alterations in circadian rhythms are associated with metabolic disturbances and pathologies such as obesity and diabetes. The molecular components of central and peripheral clocks and their regulatory mechanisms are well established. Among the different clock genes, Rev-erbα is considered one of the key links between circadian rhythms and metabolism. Rev-erbα is a critical part of a negative feedback loop in the core circadian clock and modulates the clock oscillatory properties. In addition, Rev-erbα plays an important role in the regulation of lipid and glucose metabolism, thermogenesis, adipocyte and muscle differentiation as well as mitochondrial function. In the endocrine pancreas, Rev-erbα regulates insulin and glucagon secretion and pancreatic ß-cell proliferation. In the present review, we discuss all these subjects and, particularly, the role of the clock gene Rev-erbα in the endocrine pancreas.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm/physiology , Islets of Langerhans/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Adipocytes/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Feedback, Physiological , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Lipid Metabolism/genetics , Mitochondria/physiology , Muscle, Skeletal/metabolism , Thermogenesis/genetics
18.
Trends Endocrinol Metab ; 25(11): 586-92, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25066191

ABSTRACT

Rev-erbα is a nuclear receptor that links circadian rhythms to transcriptional control of metabolic pathways. Rev-erbα is a potent transcriptional repressor and plays an important role in the core mammalian molecular clock while also serving as a key regulator of clock output in metabolic tissues including liver and brown adipose tissue (BAT). Recent findings have shed new light on the role of Rev-erbα and its paralog Rev-erbß in rhythm generation, as well as additional regulatory roles for Rev-erbα in other tissues that contribute to energy expenditure, inflammation, and behavior. This review highlights physiological functions of Rev-erbα and ß in multiple tissues and discusses the therapeutic potential and challenges of targeting these pathways in human disease.


Subject(s)
Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Animals , Biological Clocks/genetics , Circadian Rhythm/genetics , Energy Metabolism/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation , Humans , Lipid Metabolism/genetics , Muscle, Skeletal/metabolism
19.
J Reprod Dev ; 60(5): 362-70, 2014.
Article in English | MEDLINE | ID: mdl-25007867

ABSTRACT

The nuclear receptor REV-ERBα (encoded by NR1D1) has a critical role in metabolism and physiology as well as circadian rhythm. Here, we investigated the possible contribution of clock genes including NR1D1 to the secretion of prostaglandin F2α (PGF2α) from bovine uterine stromal (USCs) and epithelial cells (UECs) by modulating the expression of PTGS2. The circadian oscillation of clock genes in the cells was weak compared with that reported in rodents, but the expression of BMAL1, PER1, and NR1D1 was changed temporally by treatment with ovarian steroids. Significant expression of clock genes including NR1D1 was detected in USCs exposed to progesterone. NR1D1 was also significantly expressed in UECs exposed to estradiol. The expression of PTGS2 was suppressed in USCs exposed to progesterone, while the expression was initially suppressed in UECs exposed to estradiol and then increased after long-term exposure to estradiol. BMAL1 knockdown with specific siRNA caused a significant decrease in the transcript levels of NR1D1 and PTGS2 in USCs, but not in UECs. The production of PGF2α also decreased in USCs after BMAL1 knockdown, while its level did not significantly change in UECs. The transcript level of PTGS2 was increased by treatment with the antagonist of REV-ERBα in both cell types, but the agonist was ineffective. In these two cell types treated with the agonist or antagonist, the PGF2α production coincided well with the PTGS2 expression. Collectively, these results indicate that REV-ERBα plays an inhibitory role in the expression of PTGS2 in both bovine USCs and UECs treated with ovarian steroids.


Subject(s)
Cattle/metabolism , Cyclooxygenase 2/genetics , Endometrium/metabolism , Gene Expression/drug effects , Gonadal Steroid Hormones/pharmacology , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Animals , Circadian Rhythm , Cyclooxygenase 2/physiology , Dinoprost/metabolism , Endometrium/chemistry , Epithelial Cells/drug effects , Estradiol/pharmacology , Female , Heme/pharmacology , Isoquinolines/pharmacology , Mice , Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Progesterone/pharmacology , RNA, Messenger/analysis , Rats , Stromal Cells/metabolism , Thiophenes/pharmacology , Transfection
20.
Immunity ; 40(2): 178-86, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24560196

ABSTRACT

Immune parameters change with time of day and disruption of circadian rhythms has been linked to inflammatory pathologies. A circadian-clock-controlled immune system might allow an organism to anticipate daily changes in activity and feeding and the associated risk of infection or tissue damage to the host. Responses to bacteria have been shown to vary depending on time of infection, with mice being more at risk of sepsis when challenged ahead of their activity phase. Studies highlight the extent to which the molecular clock, most notably the core clock proteins BMAL1, CLOCK, and REV-ERBα, control fundamental aspects of the immune response. Examples include the BMAL1:CLOCK heterodimer regulating toll-like receptor 9 (TLR9) expression and repressing expression of the inflammatory monocyte chemokine ligand (CCL2) as well as REV-ERBα suppressing the induction of interleukin-6. Understanding the daily rhythm of the immune system could have implications for vaccinations and how we manage infectious and inflammatory diseases.


Subject(s)
Circadian Rhythm/physiology , Immunity , Nuclear Receptor Subfamily 1, Group D, Member 1/physiology , Animals , Circadian Rhythm/genetics , Circadian Rhythm/immunology , Gene Expression Regulation , Humans , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology
SELECTION OF CITATIONS
SEARCH DETAIL