Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Am J Reprod Immunol ; 86(3): e13436, 2021 09.
Article in English | MEDLINE | ID: mdl-33934423

ABSTRACT

PROBLEM: Circadian rhythms are involved not only in the repair and regeneration of the immune system, but may also be associated with regulation of inflammation and immune responses. Rev-erbα could constitute a link between immunity and circadian rhythms since it is a transcription factor that regulates circadian rhythms and has functions in multiple physiological and pathological processes. Decidual macrophages (dMφs) play crucial roles in immune balance at the maternal-fetal interface, and abnormal macrophage polarization is related to adverse pregnancy outcomes, such as infertility, recurrent spontaneous abortion, and preterm labor. However, whether Rev-erbα could modulate the polarization of macrophages is unknown. METHODS OF STUDY: In this study, we analyzed the phenotype of dMφs and the expression of Rev-erbα in dMφs from normal pregnancies and miscarriages. The effect of Rev-erbα on macrophage polarization was evaluated by its knockdown or pharmacological activation. The mechanism by which the Rev-erbα agonist SR9009 regulates macrophage polarization was also estimated. RESULTS: A type-1 macrophage (M1)-like dominance was observed in dMφs from human miscarriages, with a decreased expression of Rev-erbα compared to that from normal pregnancies. Rev-erbα knockdown promoted M1 polarization in macrophages differentiated from the THP1 cell line, whereas pharmacological activation of Rev-erbα by SR9009 induced type-2 macrophage (M2)-like polarization in dMφs. Furthermore, we found that SR9009 induced M2 polarization in macrophages differentiated from the U937 cell line via the PI3K/Akt signaling pathway. CONCLUSION: Rev-erbα may play an essential role in macrophage polarization. These findings might help elucidate the role of Rev-erbα in regulating the differentiation and functions of macrophages and suggest a therapeutic target for pregnancy loss and pregnancy complications.


Subject(s)
Decidua/metabolism , Macrophages/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy Complications/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Circadian Rhythm/physiology , Decidua/immunology , Female , Humans , Macrophages/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , Pregnancy , Pregnancy Complications/immunology , Signal Transduction/physiology
2.
Biochem Biophys Res Commun ; 529(4): 916-921, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32819599

ABSTRACT

Hepatic ischemia-reperfusion (I/R) injury is a complex pathophysiological process that often times occurs in liver transplantation, hepatectomy, and ischemic shock. Aberrant activation of inflammatory responses has been implicated in hepatic I/R injury. In this study, we aimed to investigate the role of circadian clock gene Rev-erbα (a well-known regulator of inflammation) in hepatic I/R injury. We first showed that Rev-erbα ablation sensitized mice to hepatic I/R injury as evidenced by higher levels of plasma alanine aminotransferase and aspartate aminotransferase, an increased histological score, as well as enhanced hepatic myeloperoxidase activity in Rev-erbα-/- mice. More severe hepatic I/R injury in Rev-erbα-/- mice was accompanied by higher expression of pro-inflammatory cytokines, exacerbated activation of Nlrp3 inflammasome, and more extensive infiltration of inflammatory cells. Moreover, pharmacological activation of Rev-erbα by SR9009 significantly alleviated the hepatic damage and inflammatory responses. In addition, I/R operation started at ZT18 (corresponding to low Rev-erbα expression) caused more severe liver damage and inflammatory responses in wild-type mice as compared to operation started at ZT6 (corresponding to high Rev-erbα expression), supporting a protective effect of Rev-erbα on hepatic I/R injury. Collectively, Rev-erbα protects hepatic I/R injury probably via repression of inflammatory responses, and targeting Rev-erbα may be a promising approach for management of hepatic I/R injury.


Subject(s)
Circadian Clocks/immunology , Liver/metabolism , Macrophages/immunology , Neutrophils/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Reperfusion Injury/metabolism , Alanine Transaminase/genetics , Alanine Transaminase/immunology , Animals , Aspartate Aminotransferases/genetics , Aspartate Aminotransferases/immunology , Circadian Clocks/drug effects , Circadian Clocks/genetics , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Liver/immunology , Liver/pathology , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , Peroxidase/genetics , Peroxidase/immunology , Pyrrolidines/pharmacology , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Thiophenes/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
3.
Sci Immunol ; 4(40)2019 10 04.
Article in English | MEDLINE | ID: mdl-31586012

ABSTRACT

Many gut functions are attuned to circadian rhythm. Intestinal group 3 innate lymphoid cells (ILC3s) include NKp46+ and NKp46- subsets, which are RORγt dependent and provide mucosal defense through secretion of interleukin-22 (IL-22) and IL-17. Because ILC3s highly express some key circadian clock genes, we investigated whether ILC3s are also attuned to circadian rhythm. We noted circadian oscillations in the expression of clock and cytokine genes, such as REV-ERBα, IL-22, and IL-17, whereas acute disruption of the circadian rhythm affected cytokine secretion by ILC3s. Because of prominent and rhythmic expression of REV-ERBα in ILC3s, we also investigated the impact of constitutive deletion of REV-ERBα, which has been previously shown to inhibit the expression of a RORγt repressor, NFIL3, while also directly antagonizing DNA binding of RORγt. Development of the NKp46+ ILC3 subset was markedly impaired, with reduced cell numbers, RORγt expression, and IL-22 production in REV-ERBα-deficient mice. The NKp46- ILC3 subsets developed normally, potentially due to compensatory expression of other clock genes, but IL-17 secretion paradoxically increased, probably because RORγt was not antagonized by REV-ERBα. We conclude that ILC3s are attuned to circadian rhythm, but clock regulator REV-ERBα also has circadian-independent impacts on ILC3 development and functions due to its roles in the regulation of RORγt.


Subject(s)
Circadian Rhythm/immunology , Immunity, Innate/immunology , Intestines/immunology , Lymphocytes/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Animals , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology
4.
Proc Natl Acad Sci U S A ; 116(37): 18528-18536, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31455731

ABSTRACT

T helper 17 (Th17) cells produce interleukin-17 (IL-17) cytokines and drive inflammatory responses in autoimmune diseases such as multiple sclerosis. The differentiation of Th17 cells is dependent on the retinoic acid receptor-related orphan nuclear receptor RORγt. Here, we identify REV-ERBα (encoded by Nr1d1), a member of the nuclear hormone receptor family, as a transcriptional repressor that antagonizes RORγt function in Th17 cells. REV-ERBα binds to ROR response elements (RORE) in Th17 cells and inhibits the expression of RORγt-dependent genes including Il17a and Il17f Furthermore, elevated REV-ERBα expression or treatment with a synthetic REV-ERB agonist significantly delays the onset and impedes the progression of experimental autoimmune encephalomyelitis (EAE). These results suggest that modulating REV-ERBα activity may be used to manipulate Th17 cells in autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Th17 Cells/immunology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Genetic Loci , HEK293 Cells , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Mice , Mice, Transgenic , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , RNA-Seq , Response Elements/genetics , Th17 Cells/metabolism , Thiophenes/pharmacology , Thiophenes/therapeutic use
5.
Int Immunopharmacol ; 73: 312-320, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31129418

ABSTRACT

Progressive lung injury and pulmonary inflammation can be induced by an intraperitoneal injection of lipopolysaccharide (LPS). Interleukin-1ß (IL-1ß) is a key pro-inflammatory cytokine that can further exaggerate inflammation, which is cleaved and activated by the NALP3 inflammasome. Although the nuclear receptor Rev-erbα attenuates the level of LPS-induced pulmonary inflammation, the mechanism remains unclear. In this study, we investigated the influence of LPS-induced production of IL-1ß and Rev-erbα on the development of lung inflammation. Herein, we demonstrate that Rev-erbα reduces IL-1ß production and lung injury following an intraperitoneal injection of LPS, which is dependent on the NF-κB/NALP3 pathway. Thus, Rev-erbα is able to decrease the extent of acute lung injury by regulating IL-1ß production. This mechanism may represent a potential novel therapeutic approach for lung injury.


Subject(s)
Acute Lung Injury/immunology , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Glycine/analogs & derivatives , Glycine/pharmacology , Interleukin-1beta/immunology , Isoquinolines/pharmacology , Lipopolysaccharides , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group D, Member 1/agonists , Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors , RAW 264.7 Cells , Signal Transduction , Thiophenes/pharmacology
6.
Nat Commun ; 10(1): 377, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670689

ABSTRACT

The circadian clock regulates immune responses to microbes and affects pathogen replication, but the underlying molecular mechanisms are not well understood. Here we demonstrate that the circadian components BMAL1 and REV-ERBα influence several steps in the hepatitis C virus (HCV) life cycle, including particle entry into hepatocytes and RNA genome replication. Genetic knock out of Bmal1 and over-expression or activation of REV-ERB with synthetic agonists inhibits the replication of HCV and the related flaviruses dengue and Zika via perturbation of lipid signaling pathways. This study highlights a role for the circadian clock component REV-ERBα in regulating flavivirus replication.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Clocks/genetics , Flavivirus/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Virus Replication/drug effects , ARNTL Transcription Factors/immunology , ARNTL Transcription Factors/pharmacology , Cell Line , Circadian Clocks/immunology , DNA Replication , Dengue , Dengue Virus/drug effects , Dengue Virus/genetics , Flavivirus/drug effects , Flavivirus/metabolism , Flavivirus/pathogenicity , Gene Expression Regulation/genetics , Genes, Essential/genetics , Hepacivirus/drug effects , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C , Hepatocytes/immunology , Hepatocytes/virology , Humans , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/pharmacology , Proteomics , RNA, Messenger/metabolism , Virus Internalization/drug effects , Zika Virus/drug effects , Zika Virus/genetics , Zika Virus Infection
7.
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
8.
Biochem Biophys Res Commun ; 469(2): 151-7, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26616049

ABSTRACT

Under physiological conditions, astrocytes maintain homeostasis in the CNS. Following inflammation and injury to the CNS, however, activated astrocytes produce neurotoxic molecules such as cytokines and chemokines, amplifying the initial molecular-cellular events evoked by inflammation and injury. Nuclear receptors REV-ERBα and REV-ERBß (REV-ERBs) are crucial in the regulation of inflammation- and metabolism-related gene transcription. The current study sought to elucidate a role of REV-ERBs in rat C6 astroglial cells on the expression of inflammatory molecules following stimulation with the neuroinflammatory cytokine tumor necrosis factor (TNF). Stimulation of C6 cells with TNF (10 ng/ml) significantly increased the mRNA expression of CCL2, interleukin-6 (IL-6), inducible nitric oxide synthase (iNOS), and matrix metalloprotease (MMP)-9, but not fibroblast growth factor-2 (FGF-2), cyclooxygenase-2 (COX-2) and MMP-2. Treatment with either REV-ERB agonists GSK4112 or SR9009 significantly blocked TNF-induced upregulation of CCL2 mRNA and MMP-9 mRNA, but not IL-6 mRNA and iNOS mRNA expression. Furthermore, treatment with RGFP966, a selective histone deacetylase 3 (HDAC3) inhibitor, potently reversed the inhibitory effects of GSK4112 on TNF-induced expression of MMP-9 mRNA, but not CCL2 mRNA. Expression of Rev-erbs mRNA in C6 astroglial cells, primary cultured rat cortical and spinal astrocytes was confirmed by reverse transcription polymerase chain reaction. Together, the findings demonstrate an anti-inflammatory effect, downregulating of MMP-9 and CCL2 transcription, of astroglial REV-ERBs activation through HDAC3-dependent and HDAC3-independent mechanisms.


Subject(s)
Cell Line/immunology , Cytokines/immunology , Immunologic Factors/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , Receptors, Cytoplasmic and Nuclear/immunology , Repressor Proteins/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Rats
9.
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
10.
J Biol Chem ; 288(15): 10692-702, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23449984

ABSTRACT

Nuclear receptors modulate macrophage effector functions, which are imperative for clearance or survival of mycobacterial infection. The adopted orphan nuclear receptor Rev-erbα is a constitutive transcriptional repressor as it lacks AF2 domain and was earlier shown to be present in macrophages. In the present study, we highlight the differences in the relative subcellular localization of Rev-erbα in monocytes and macrophages. The nuclear localization of Rev-erbα in macrophages is subsequent to monocyte differentiation. Expression analysis of Rev-erbα elucidated it to be considerably more expressed in M1 phenotype in comparison with M2. Rev-erbα overexpression augments antimycobacterial properties of macrophage by keeping IL10 in a basal repressed state. Further, promoter analysis revealed that IL10 promoter harbors a Rev-erbα binding site exclusive to humans and higher order primates and not mouse, demonstrating a species barrier in its functionality. This direct gene repression is mediated by recruitment of co-repressors NCoR and HDAC3. In addition, our data elucidate that its overexpression reduced the survival of intracellular pathogen Mycobacterium tuberculosis by enhancing phagosome lysosome maturation, an event resulting from IL10 repression. Thus, these findings suggest that Rev-erbα bestows protection against mycobacterial infection by direct gene repression of IL10 and thus provide a novel target in modulating macrophage microbicidal properties.


Subject(s)
Gene Expression Regulation , Interleukin-10/biosynthesis , Macrophages/metabolism , Mycobacterium tuberculosis/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Response Elements , Animals , Cell Line , Female , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Male , Mice , Mycobacterium tuberculosis/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , Species Specificity , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/pathology
11.
Proc Natl Acad Sci U S A ; 109(2): 582-7, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22184247

ABSTRACT

Diurnal variation in inflammatory and immune function is evident in the physiology and pathology of humans and animals, but molecular mechanisms and mediating cell types that provide this gating remain unknown. By screening cytokine responses in mice to endotoxin challenge at different times of day, we reveal that the magnitude of response exhibited pronounced temporal dependence, yet only within a subset of proinflammatory cytokines. Disruption of the circadian clockwork in macrophages (primary effector cells of the innate immune system) by conditional targeting of a key clock gene (bmal1) removed all temporal gating of endotoxin-induced cytokine response in cultured cells and in vivo. Loss of circadian gating was coincident with suppressed rev-erbα expression, implicating this nuclear receptor as a potential link between the clock and inflammatory pathways. This finding was confirmed in vivo and in vitro through genetic and pharmacological modulation of REV-ERBα activity. Circadian gating of endotoxin response was lost in rev-erbα(-/-) mice and in cultured macrophages from these animals, despite maintenance of circadian rhythmicity within these cells. Using human macrophages, which show circadian clock gene oscillations and rhythmic endotoxin responses, we demonstrate that administration of a synthetic REV-ERB ligand, or genetic knockdown of rev-erbα expression, is effective at modulating the production and release of the proinflammatory cytokine IL-6. This work demonstrates that the macrophage clockwork provides temporal gating of systemic responses to endotoxin, and identifies REV-ERBα as the key link between the clock and immune function. REV-ERBα may therefore represent a unique therapeutic target in human inflammatory disease.


Subject(s)
Circadian Rhythm/immunology , Gene Expression Regulation/immunology , Immunity, Innate/immunology , Interleukin-6/immunology , Nuclear Receptor Subfamily 1, Group D, Member 1/immunology , ARNTL Transcription Factors/genetics , Analysis of Variance , Animals , Endotoxins/toxicity , Humans , Macrophages/immunology , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL