Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
FASEB J ; 36(7): e22356, 2022 07.
Article in English | MEDLINE | ID: mdl-35704036

ABSTRACT

The circadian clock controls the physiological function of tissues through the regulation of thousands of genes in a cell-type-specific manner. The core cellular circadian clock is a transcription-translation negative feedback loop, which can recruit epigenetic regulators to facilitate temporal control of gene expression. Histone methyltransferase, mixed lineage leukemia gene 3 (MLL3) was reported to be required for the maintenance of circadian oscillations in cultured cells. Here, we test the role of MLL3 in circadian organization in whole animals. Using mice expressing catalytically inactive MLL3, we show that MLL3 methyltransferase activity is in fact not required for circadian oscillations in vitro in a range of tissues, nor for the maintenance of circadian behavioral rhythms in vivo. In contrast to a previous report, loss of MLL3-dependent methylation did not affect the global levels of H3K4 methylation in liver, indicating substantial compensation from other methyltransferases. Furthermore, we found little evidence of genomic repositioning of H3K4me3 marks. We did, however, observe repositioning of H3K4me1 from intronic regions to intergenic regions and gene promoters; however, there were no changes in H3K4me1 mark abundance around core circadian clock genes. Output functions of the circadian clock, such as control of inflammation, were largely intact in MLL3-methyltransferase-deficient mice, although some gene-specific changes were observed, with sexually dimorphic loss of circadian regulation of specific cytokines. Taken together, these observations indicate that MLL3-directed histone methylation is not essential for core circadian clock function; however, it may influence the inflammatory response.


Subject(s)
Circadian Clocks , Animals , Circadian Clocks/genetics , Circadian Rhythm , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Methylation , Mice , Protein Processing, Post-Translational
2.
Elife ; 112022 03 14.
Article in English | MEDLINE | ID: mdl-35285799

ABSTRACT

The mammalian circadian clock exerts control of daily gene expression through cycles of DNA binding. Here, we develop a quantitative model of how a finite pool of BMAL1 protein can regulate thousands of target sites over daily time scales. We used quantitative imaging to track dynamic changes in endogenous labelled proteins across peripheral tissues and the SCN. We determine the contribution of multiple rhythmic processes coordinating BMAL1 DNA binding, including cycling molecular abundance, binding affinities, and repression. We find nuclear BMAL1 concentration determines corresponding CLOCK through heterodimerisation and define a DNA residence time of this complex. Repression of CLOCK:BMAL1 is achieved through rhythmic changes to BMAL1:CRY1 association and high-affinity interactions between PER2:CRY1 which mediates CLOCK:BMAL1 displacement from DNA. Finally, stochastic modelling reveals a dual role for PER:CRY complexes in which increasing concentrations of PER2:CRY1 promotes removal of BMAL1:CLOCK from genes consequently enhancing ability to move to new target sites.


Subject(s)
Circadian Clocks , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Circadian Clocks/genetics , Circadian Rhythm/genetics , Mammals/metabolism
3.
FASEB J ; 35(10): e21843, 2021 10.
Article in English | MEDLINE | ID: mdl-34464475

ABSTRACT

Robust inflammatory responses are critical to survival following respiratory infection, with current attention focused on the clinical consequences of the Coronavirus pandemic. Epigenetic factors are increasingly recognized as important determinants of immune responses, and EZH2 is a prominent target due to the availability of highly specific and efficacious antagonists. However, very little is known about the role of EZH2 in the myeloid lineage. Here, we show EZH2 acts in macrophages to limit inflammatory responses to activation, and in neutrophils for chemotaxis. Selective genetic deletion in macrophages results in a remarkable gain in protection from infection with the prevalent lung pathogen, pneumococcus. In contrast, neutrophils lacking EZH2 showed impaired mobility in response to chemotactic signals, and resulted in increased susceptibility to pneumococcus. In summary, EZH2 shows complex, and divergent roles in different myeloid lineages, likely contributing to the earlier conflicting reports. Compounds targeting EZH2 are likely to impair mucosal immunity; however, they may prove useful for conditions driven by pulmonary neutrophil influx, such as adult respiratory distress syndrome.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/immunology , Inflammation/immunology , Macrophages/immunology , Neutrophils/immunology , Animals , Cells, Cultured , Macrophages/cytology , Mice, Inbred C57BL , Neutrophils/cytology
4.
Eur Respir J ; 56(6)2020 12.
Article in English | MEDLINE | ID: mdl-32586876

ABSTRACT

BACKGROUND: The circadian clock powerfully regulates inflammation and the clock protein REV-ERBα is known to play a key role as a repressor of the inflammatory response. Asthma is an inflammatory disease of the airways with a strong time of day rhythm. Airway hyper-responsiveness (AHR) is a dominant feature of asthma; however, it is not known if this is under clock control. OBJECTIVES: To determine if allergy-mediated AHR is gated by the clock protein REV-ERBα. METHODS: After exposure to the intra-nasal house dust mite (HDM) allergen challenge model at either dawn or dusk, AHR to methacholine was measured invasively in mice. MAIN RESULTS: Wild-type (WT) mice show markedly different time of day AHR responses (maximal at dusk/start of the active phase), both in vivo and ex vivo, in precision cut lung slices. Time of day effects on AHR were abolished in mice lacking the clock gene Rev-erbα, indicating that such effects on asthma response are likely to be mediated via the circadian clock. We suggest that muscarinic receptors one (Chrm 1) and three (Chrm 3) may play a role in this pathway. CONCLUSIONS: We identify a novel circuit regulating a core process in asthma, potentially involving circadian control of muscarinic receptor expression, in a REV-ERBα dependent fashion. CLINICAL IMPLICATION: These insights suggest the importance of considering the timing of drug administration in clinic trials and in clinical practice (chronotherapy).


Subject(s)
Asthma , Circadian Clocks , Animals , Circadian Rhythm , Inflammation , Mice , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
5.
Proc Natl Acad Sci U S A ; 117(3): 1543-1551, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31900362

ABSTRACT

The circadian clock regulates many aspects of immunity. Bacterial infections are affected by time of day, but the mechanisms involved remain undefined. Here we show that loss of the core clock protein BMAL1 in macrophages confers protection against pneumococcal pneumonia. Infected mice show both reduced weight loss and lower bacterial burden in circulating blood. In vivo studies of macrophage phagocytosis reveal increased bacterial ingestion following Bmal1 deletion, which was also seen in vitro. BMAL1-/- macrophages exhibited marked differences in actin cytoskeletal organization, a phosphoproteome enriched for cytoskeletal changes, with reduced phosphocofilin and increased active RhoA. Further analysis of the BMAL1-/- macrophages identified altered cell morphology and increased motility. Mechanistically, BMAL1 regulated a network of cell movement genes, 148 of which were within 100 kb of high-confidence BMAL1 binding sites. Links to RhoA function were identified, with 29 genes impacting RhoA expression or activation. RhoA inhibition restored the phagocytic phenotype to that seen in control macrophages. In summary, we identify a surprising gain of antibacterial function due to loss of BMAL1 in macrophages, associated with a RhoA-dependent cytoskeletal change, an increase in cell motility, and gain of phagocytic function.


Subject(s)
ARNTL Transcription Factors/antagonists & inhibitors , ARNTL Transcription Factors/genetics , Cell Movement/drug effects , Disease Resistance/genetics , Macrophages/drug effects , Phagocytosis/drug effects , Pneumonia, Pneumococcal/metabolism , Actins/metabolism , Animals , Circadian Clocks/genetics , Circadian Clocks/physiology , Cytoskeleton , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Streptococcus pneumoniae/pathogenicity , rhoA GTP-Binding Protein/metabolism
6.
Proc Natl Acad Sci U S A ; 117(2): 1139-1147, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31879343

ABSTRACT

Pulmonary inflammatory responses lie under circadian control; however, the importance of circadian mechanisms in the underlying fibrotic phenotype is not understood. Here, we identify a striking change to these mechanisms resulting in a gain of amplitude and lack of synchrony within pulmonary fibrotic tissue. These changes result from an infiltration of mesenchymal cells, an important cell type in the pathogenesis of pulmonary fibrosis. Mutation of the core clock protein REVERBα in these cells exacerbated the development of bleomycin-induced fibrosis, whereas mutation of REVERBα in club or myeloid cells had no effect on the bleomycin phenotype. Knockdown of REVERBα revealed regulation of the little-understood transcription factor TBPL1. Both REVERBα and TBPL1 altered integrinß1 focal-adhesion formation, resulting in increased myofibroblast activation. The translational importance of our findings was established through analysis of 2 human cohorts. In the UK Biobank, circadian strain markers (sleep length, chronotype, and shift work) are associated with pulmonary fibrosis, making them risk factors. In a separate cohort, REVERBα expression was increased in human idiopathic pulmonary fibrosis (IPF) lung tissue. Pharmacological targeting of REVERBα inhibited myofibroblast activation in IPF fibroblasts and collagen secretion in organotypic cultures from IPF patients, thus suggesting that targeting of REVERBα could be a viable therapeutic approach.


Subject(s)
CLOCK Proteins/antagonists & inhibitors , Circadian Clocks/physiology , Fibroblasts/drug effects , Pulmonary Fibrosis/drug therapy , Animals , Bleomycin/adverse effects , CLOCK Proteins/genetics , CLOCK Proteins/therapeutic use , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Idiopathic Pulmonary Fibrosis , Integrins , Lung/pathology , Male , Mesenchymal Stem Cells , Mice , Mice, Knockout , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , TATA Box Binding Protein-Like Proteins/metabolism , Transcriptome
7.
FASEB J ; 33(5): 6226-6238, 2019 05.
Article in English | MEDLINE | ID: mdl-30794439

ABSTRACT

Pulmonary airway epithelial cells (AECs) form a critical interface between host and environment. We investigated the role of the circadian clock using mice bearing targeted deletion of the circadian gene brain and muscle ARNT-like 1 (Bmal1) in AECs. Pulmonary neutrophil infiltration, biomechanical function, and responses to influenza infection were all disrupted. A circadian time-series RNA sequencing study of laser-captured AECs revealed widespread disruption in genes of the core circadian clock and output pathways regulating cell metabolism (lipids and xenobiotics), extracellular matrix, and chemokine signaling, but strikingly also the gain of a novel rhythmic transcriptome in Bmal1-targeted cells. Many of the rhythmic components were replicated in primary AECs cultured in air-liquid interface, indicating significant cell autonomy for control of pulmonary circadian physiology. Finally, we found that metabolic cues dictate phasing of the pulmonary clock and circadian responses to immunologic challenges. Thus, the local circadian clock in AECs is vital in lung health by coordinating major cell processes such as metabolism and immunity.-Zhang, Z. Hunter, L., Wu, G., Maidstone, R., Mizoro, Y., Vonslow, R., Fife, M., Hopwood, T., Begley, N., Saer, B., Wang, P., Cunningham, P., Baxter, M., Durrington, H., Blaikley, J. F., Hussell, T., Rattray, M., Hogenesch, J. B., Gibbs, J., Ray, D. W., Loudon, A. S. I. Genome-wide effect of pulmonary airway epithelial cell-specific Bmal1 deletion.


Subject(s)
ARNTL Transcription Factors/genetics , Alveolar Epithelial Cells/metabolism , Transcriptome , Alveolar Epithelial Cells/microbiology , Animals , Cells, Cultured , Circadian Clocks , Female , Gene Deletion , Humans , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Xenobiotics/metabolism
8.
FASEB J ; 33(1): 126-139, 2019 01.
Article in English | MEDLINE | ID: mdl-29965797

ABSTRACT

The circadian clock is a critical regulator of immune function. We recently highlighted a role for the circadian clock in a mouse model of pulmonary inflammation. The epithelial clock protein Bmal1 was required to regulate neutrophil recruitment in response to inflammatory challenge. Bmal1 regulated glucocorticoid receptor (GR) recruitment to the neutrophil chemokine, CXC chemokine ligand 5 (CXCL5), providing a candidate mechanism. We now show that clock control of pulmonary neutrophilia persists without rhythmic glucocorticoid availability. Epithelial GR-null mice had elevated expression of proinflammatory chemokines in the lung under homeostatic conditions. However, deletion of GR in the bronchial epithelium blocked rhythmic CXCL5 production, identifying GR as required to confer circadian control to CXCL5. Surprisingly, rhythmic pulmonary neutrophilia persisted, despite nonrhythmic CXCL5 responses, indicating additional circadian control mechanisms. Deletion of GR in myeloid cells alone did not prevent circadian variation in pulmonary neutrophilia and showed reduced neutrophilic inflammation in response to dexamethasone treatment. These new data show GR is required to confer circadian control to some inflammatory chemokines, but that this alone is insufficient to prevent circadian control of neutrophilic inflammation in response to inhaled LPS, with additional control mechanisms arising in the myeloid cell lineage.-Ince, L. M., Zhang, Z., Beesley, S., Vonslow, R. M., Saer, B. R., Matthews, L. C., Begley, N., Gibbs, J. E., Ray, D. W., Loudon, A. S. I. Circadian variation in pulmonary inflammatory responses is independent of rhythmic glucocorticoid signaling in airway epithelial cells.


Subject(s)
Circadian Rhythm/immunology , Epithelial Cells/immunology , Macrophages, Peritoneal/immunology , Neutrophils/immunology , Pneumonia/immunology , Receptors, Glucocorticoid/physiology , Respiratory System/immunology , Animals , Cells, Cultured , Chemokine CXCL5/metabolism , Circadian Rhythm/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glucocorticoids/pharmacology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/drug therapy , Pneumonia/metabolism , Pneumonia/pathology , Respiratory System/drug effects , Respiratory System/metabolism , Respiratory System/pathology , Signal Transduction
9.
J Clin Invest ; 128(10): 4454-4471, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30179226

ABSTRACT

The glucocorticoid receptor (GR) is a major drug target in inflammatory disease. However, chronic glucocorticoid (GC) treatment leads to disordered energy metabolism, including increased weight gain, adiposity, and hepatosteatosis - all programs modulated by the circadian clock. We demonstrated that while antiinflammatory GC actions were maintained irrespective of dosing time, the liver was significantly more GC sensitive during the day. Temporal segregation of GC action was underpinned by a physical interaction of GR with the circadian transcription factor REVERBa and co-binding with liver-specific hepatocyte nuclear transcription factors (HNFs) on chromatin. REVERBa promoted efficient GR recruitment to chromatin during the day, acting in part by maintaining histone acetylation, with REVERBa-dependent GC responses providing segregation of carbohydrate and lipid metabolism. Importantly, deletion of Reverba inverted circadian liver GC sensitivity and protected mice from hepatosteatosis induced by chronic GC administration. Our results reveal a mechanism by which the circadian clock acts through REVERBa in liver on elements bound by HNF4A/HNF6 to direct GR action on energy metabolism.


Subject(s)
Chromatin/metabolism , Circadian Clocks/drug effects , Fatty Liver/metabolism , Glucocorticoids/adverse effects , Liver/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Animals , Chromatin/genetics , Chromatin/pathology , Circadian Clocks/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Glucocorticoids/pharmacology , HEK293 Cells , Humans , Liver/pathology , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
10.
Sci Rep ; 8(1): 3782, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29491349

ABSTRACT

Resistance to the intestinal parasitic helminth Trichuris muris requires T-helper 2 (TH2) cellular and associated IgG1 responses, with expulsion typically taking up to 4 weeks in mice. Here, we show that the time-of-day of the initial infection affects efficiency of worm expulsion, with strong TH2 bias and early expulsion in morning-infected mice. Conversely, mice infected at the start of the night show delayed resistance to infection, and this is associated with feeding-driven metabolic cues, such that feeding restriction to the day-time in normally nocturnal-feeding mice disrupts parasitic expulsion kinetics. We deleted the circadian regulator BMAL1 in antigen-presenting dendritic cells (DCs) in vivo and found a loss of time-of-day dependency of helminth expulsion. RNAseq analyses revealed that IL-12 responses to worm antigen by circadian-synchronised DCs were dependent on BMAL1. Therefore, we find that circadian machinery in DCs contributes to the TH1/TH2 balance, and that environmental, or genetic perturbation of the DC clock results in altered parasite expulsion kinetics.


Subject(s)
ARNTL Transcription Factors/physiology , Circadian Rhythm , Dendritic Cells/immunology , Lymph Nodes/immunology , Th2 Cells/immunology , Trichuriasis/immunology , Trichuris/pathogenicity , Animals , Cells, Cultured , Dendritic Cells/parasitology , Lymph Nodes/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , Th2 Cells/parasitology , Trichuriasis/parasitology
11.
J Clin Invest ; 128(6): 2281-2296, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29533925

ABSTRACT

Recent studies reveal that airway epithelial cells are critical pulmonary circadian pacemaker cells, mediating rhythmic inflammatory responses. Using mouse models, we now identify the rhythmic circadian repressor REV-ERBα as essential to the mechanism coupling the pulmonary clock to innate immunity, involving both myeloid and bronchial epithelial cells in temporal gating and determining amplitude of response to inhaled endotoxin. Dual mutation of REV-ERBα and its paralog REV-ERBß in bronchial epithelia further augmented inflammatory responses and chemokine activation, but also initiated a basal inflammatory state, revealing a critical homeostatic role for REV-ERB proteins in the suppression of the endogenous proinflammatory mechanism in unchallenged cells. However, REV-ERBα plays the dominant role, as deletion of REV-ERBß alone had no impact on inflammatory responses. In turn, inflammatory challenges cause striking changes in stability and degradation of REV-ERBα protein, driven by SUMOylation and ubiquitination. We developed a novel selective oxazole-based inverse agonist of REV-ERB, which protects REV-ERBα protein from degradation, and used this to reveal how proinflammatory cytokines trigger rapid degradation of REV-ERBα in the elaboration of an inflammatory response. Thus, dynamic changes in stability of REV-ERBα protein couple the core clock to innate immunity.


Subject(s)
Circadian Clocks/immunology , Circadian Rhythm/immunology , Homeostasis/immunology , Immunity, Innate , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , Pneumonia/immunology , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Homeostasis/genetics , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Pneumonia/genetics , Pneumonia/pathology , Proteolysis , Sumoylation/genetics , Sumoylation/immunology
12.
Nat Med ; 20(8): 919-26, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25064128

ABSTRACT

The circadian system is an important regulator of immune function. Human inflammatory lung diseases frequently show time-of-day variation in symptom severity and lung function, but the mechanisms and cell types underlying these effects remain unclear. We show that pulmonary antibacterial responses are modulated by a circadian clock within epithelial club (Clara) cells. These drive circadian neutrophil recruitment to the lung via the chemokine CXCL5. Genetic ablation of the clock gene Bmal1 (also called Arntl or MOP3) in bronchiolar cells disrupts rhythmic Cxcl5 expression, resulting in exaggerated inflammatory responses to lipopolysaccharide and an impaired host response to Streptococcus pneumoniae infection. Adrenalectomy blocks rhythmic inflammatory responses and the circadian regulation of CXCL5, suggesting a key role for the adrenal axis in driving CXCL5 expression and pulmonary neutrophil recruitment. Glucocorticoid receptor occupancy at the Cxcl5 locus shows circadian oscillations, but this is disrupted in mice with bronchiole-specific ablation of Bmal1, leading to enhanced CXCL5 expression despite normal corticosteroid secretion. The therapeutic effects of the synthetic glucocorticoid dexamethasone depend on intact clock function in the airway. We now define a regulatory mechanism that links the circadian clock and glucocorticoid hormones to control both time-of-day variation and the magnitude of pulmonary inflammation and responses to bacterial infection.


Subject(s)
ARNTL Transcription Factors/immunology , Chemokine CXCL5/immunology , Circadian Clocks/immunology , Glucocorticoids/pharmacology , Pneumonia, Pneumococcal/immunology , Streptococcus pneumoniae , ARNTL Transcription Factors/genetics , Animals , Cells, Cultured , Chemokine CXCL5/biosynthesis , Circadian Rhythm/physiology , Dexamethasone/pharmacology , Epithelial Cells/immunology , Humans , Lipopolysaccharides/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Period Circadian Proteins/immunology , Pneumonia, Pneumococcal/genetics , Receptors, Glucocorticoid/immunology , Uteroglobin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...