Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
FEBS Lett ; 2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38825601

ABSTRACT

Mitochondrial biogenesis requires precise regulation of both mitochondrial-encoded and nuclear-encoded genes. Nuclear receptor Nur77 is known to regulate mitochondrial metabolism in macrophages and skeletal muscle. Here, we compared genome-wide Nur77 binding site and target gene expression in these two cell types, which revealed conserved regulation of mitochondrial genes and enrichment of motifs for the transcription factor Yin-Yang 1 (YY1). We show that Nur77 and YY1 interact, that YY1 increases Nur77 activity, and that their binding sites are co-enriched at mitochondrial ribosomal protein gene loci in macrophages. Nur77 and YY1 co-expression synergistically increases Mrpl1 expression as well as mitochondrial abundance and activity in macrophages but not skeletal muscle. As such, we identify a macrophage-specific Nur77-YY1 interaction that enhances mitochondrial metabolism.

2.
Adv Sci (Weinh) ; 11(7): e2304690, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38064171

ABSTRACT

Clearance of multiple rounds of apoptotic cells (ACs) through continual efferocytosis is critical in the maintenance of organ function, the resolution of acute inflammation, and tissue repair. To date, little is known about the nature of mechanisms and factors that govern this fundamental process. Herein, the authors reported that breakdown of ACs leads to upregulation of 12-lipoxygenase in macrophages. This enzyme converts docosahexaenoic acid to maresin conjugates in tissue regeneration (MCTRs). The levels of these autacoids are elevated at sites of high apoptotic burden in vivo and in efferocytosing macrophages in vitro. Abrogation of MCTR production using genetic approaches limits the ability of macrophages to perform continual efferocytosis both in vivo and in vitro, an effect that is rescued by add-back of MCTRs. Mechanistically, MCTR-mediated priming of macrophages for continual efferocytosis is dependent on alterations in Rac1 signalling and glycolytic metabolism. Inhibition of Rac1 abolishes the ability of MCTRs to increase glucose uptake and efferocytosis in vitro, whereas inhibition of glycolysis limits the MCTR-mediated increases in efferocytosis and tissue repair. Together, these findings demonstrate that upregulation of MCTRs by efferocytosing macrophages plays a central role in the regulation of continual efferocytosis via the autocrine and paracrine modulation of metabolic pathways.


Subject(s)
Efferocytosis , Phagocytosis , Macrophages/metabolism , Signal Transduction , Glycolysis
3.
J Nat Prod ; 86(11): 2546-2553, 2023 11 24.
Article in English | MEDLINE | ID: mdl-37879110

ABSTRACT

The methyl ester of resolvin D5n-3 DPA, a lipid mediator biosynthesized from the omega-3 fatty acid n-3 docosapentaenoic acid, was stereoselectively prepared in 8% yield over 12 steps (longest linear sequence). The key steps for the introduction of the two stereogenic secondary alcohols were an organocatalyzed oxyamination and the Midland Alpine borane reduction. For the assembly of the carbon chain, the Sonogashira cross-coupling reaction and the Takai olefination were utilized. The physical properties, including retention time in liquid chromatography and tandem mass spectra, of the synthetic material were matched against material from human peripheral blood and mouse infectious exudates. Synthetic RvD5n-3 DPA, obtained just prior to biological experiments, displayed potent leukocyte-directed activities, upregulating the ability of neutrophils and macrophages to phagocytose bacteria, known as hallmark bioactions of specialized pro-resolving endogenous mediators.


Subject(s)
Docosahexaenoic Acids , Macrophages , Animals , Mice , Humans , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/chemistry , Phagocytosis , Neutrophils , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Chromatography, Liquid , Inflammation
4.
Biochem Pharmacol ; 207: 115348, 2023 01.
Article in English | MEDLINE | ID: mdl-36400250

ABSTRACT

We recently found that the G protein coupled receptor GPR101 mediates the phagocyte-directed pro-resolving activities of RvD5n-3 DPA (n-3 docosapentaenoic acid-derived Resolvin D5). Herein, we investigated the endogenous role of this pro-resolving receptor in modulating macrophage biology using a novel mouse line where the expression of Gpr101 was conditionally deleted in macrophages (MacGpr101KO). Peritoneal macrophages obtained from naïve MacGpr101KO mice displayed a marked shift in the expression of phenotypic and activation markers, including the Interleukin (IL)-10 and IL-23 receptors. Loss of Gpr101 on macrophages was also associated with a significant disruption in their cellular metabolism and a decreased ability to migrate towards the chemoattractant Mcp-1. The alterations in macrophage phenotype observed in Gpr101 deficient macrophages were maintained following inflammatory challenge. This was linked with an increased inflammatory response in the Gpr101 deficient animals and a reduced ability of phagocytes, including macrophages, to clear bacteria. Loss of Gpr101 on macrophages disrupted host pro-resolving responses to zymosan challenge with MacGpr101KO mice exhibiting significantly higher neutrophil numbers and a delay in the resolution interval when compared with control mice. These observations were linked with a marked dysregulation in peritoneal lipid mediator concentrations in Gpr101 deficient mice, with a downregulation of pro-resolving mediators including MaR2n-3 DPA, Resolvin (Rv) D3 and RvE3. Together these findings identify Gpr101 as a novel regulator of both macrophage phenotype and function, modulating key biological activities in both limiting the propagation of inflammation and expediting its resolution.


Subject(s)
Inflammation , Macrophages , Receptors, G-Protein-Coupled , Animals , Mice , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/metabolism , Immunity , Macrophages/metabolism , Phenotype , Receptors, G-Protein-Coupled/genetics
5.
Nat Commun ; 13(1): 2070, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440539

ABSTRACT

Deubiquitylating enzymes (DUBs) play an essential role in targeted protein degradation and represent an emerging therapeutic paradigm in cancer. However, their therapeutic potential in pancreatic ductal adenocarcinoma (PDAC) has not been explored. Here, we develop a DUB discovery pipeline, combining activity-based proteomics with a loss-of-function genetic screen in patient-derived PDAC organoids and murine genetic models. This approach identifies USP25 as a master regulator of PDAC growth and maintenance. Genetic and pharmacological USP25 inhibition results in potent growth impairment in PDAC organoids, while normal pancreatic organoids are insensitive, and causes dramatic regression of patient-derived xenografts. Mechanistically, USP25 deubiquitinates and stabilizes the HIF-1α transcription factor. PDAC is characterized by a severely hypoxic microenvironment, and USP25 depletion abrogates HIF-1α transcriptional activity and impairs glycolysis, inducing PDAC cell death in the tumor hypoxic core. Thus, the USP25/HIF-1α axis is an essential mechanism of metabolic reprogramming and survival in PDAC, which can be therapeutically exploited.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Glycolysis/genetics , Humans , Mice , Pancreatic Neoplasms/metabolism , Tumor Microenvironment/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Pancreatic Neoplasms
6.
EBioMedicine ; 79: 103974, 2022 May.
Article in English | MEDLINE | ID: mdl-35430453

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is a progressive degenerative disorder that leads to joint destruction. Available treatments only target the inflammatory component with minimal impact on joint repair. We recently uncovered a previously unappreciated family of pro-resolving mediators, the maresin conjugate in tissue regeneration (MCTR), that display both immunoregulatory and tissue-protective activities. Thus, we queried whether the production of these autacoids is disrupted in RA patients and whether they can be useful in treating joint inflammation and promoting joint repair. METHODS: Using a highly phenotyped RA cohort we evaluated plasma MCTR concentrations and correlated these to clinical markers of disease activity. To evaluate the immunoregulatory and tissue reparative activities we employed both in vivo models of arthritis and organ culture models. FINDINGS: Herein, we observed that plasma MCTR3 concentrations were negatively correlated with joint disease activity and severity in RA patients. Evaluation of the mechanisms engaged by this mediator in arthritic mice demonstrated that MCTR3 reprograms monocytes to confer enduring joint protective properties. Single cell transcriptomic profiling and flow cytometric evaluation of macrophages from mice treated with MCTR3-reprogrammed monocytes revealed a role for Arginase-1 (Arg-1) in mediating their joint reparative and pro-resolving activities. Arg-1 inhibition reversed both the anti-arthritic and tissue reparative actions of MCTR3-reprogrammed monocytes. INTERPRETATION: Our findings demonstrate that circulating MCTR3 levels are negatively correlated with disease in RA. When administered to mice in vivo, MCTR3 displayed both anti-inflammatory and joint reparative activities, protecting both cartilage and bone in murine arthritis. These activities were, at least in part, mediated via the reprogramming of mononuclear phagocyte responses. FUNDING: This work was supported by funding from the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation programme (grant no: 677542) and the Barts Charity (grant no: MGU0343) to J.D. J.D. is also supported by a Sir Henry Dale Fellowship jointly funded by the Wellcome Trust and the Royal Society (grant 107613/Z/15/Z).


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Animals , Anti-Inflammatory Agents/pharmacology , Arginase/genetics , Arthritis, Rheumatoid/drug therapy , Humans , Macrophages , Mice , Monocytes
7.
Front Immunol ; 12: 733019, 2021.
Article in English | MEDLINE | ID: mdl-34531873

ABSTRACT

The mechanisms that lead to disease onset and propagation in patients with chronic rhinosinusitis (CRS) are not fully elucidated. Maresins (MaR) are a family of essential fatty acid-derived lipid mediators that play a central role in the regulation of inflammation with several studies demonstrating that these mediators display protective activities in airway inflammation. Therefore, in the present studies we evaluated whether concentrations of these mediators were altered in both peripheral blood and nasal secretions from CRS patients. Herein, we focused on patients with CRS that also develop nasal polyps (CRSwNP), given that therapeutic options for the treatment of these patients are limited. Thereby, insights into disease mechanisms in these patients may help design more effective treatments. For this purpose, we compared maresin concentrations from CRSwNP patients with those found in healthy volunteers or patients with an upper respiratory tract infection (URTI), as a self-resolving inflammatory condition. Using liquid chromatography tandem mass spectrometry, we found that MaR concentrations were significantly decreased in plasma from patients with CRSwNP when compared to healthy volunteers. MaR concentrations were observed to be significantly upregulated in nasal secretions from patients with CRSwNP when compared with both healthy volunteers and URTI subjects. Concentration of these mediators in both plasma and nasal secretions from CRSwNP patients were positively correlated with quality-of-life scores in these patients. Assessment of the concentrations of other pro-resolving and pro-inflammatory lipid mediators (LM) demonstrated that there was a general shift in LM levels in both plasma and nasal secretions from CRSwNP when compared with healthy volunteers and URTI subjects. Of note, incubation of peripheral blood cells from CRSwNP patients with MaR1 downregulated the expression of activation markers on peripheral blood phagocytes, including CD41 and CD62P, markers of platelet-leukocyte heterotypic aggregates. Together these findings demonstrate that both local and systemic LM concentrations, in particularly those of the MaR family, become altered in patients with CRSwNP. They also suggest that therapeutics designed around MaR1 may be useful in regulating the activation of phagocytes in patients with CRSwNP thereby potentially also limiting the local inflammatory response in these patients.


Subject(s)
Docosahexaenoic Acids/blood , Nasal Mucosa/metabolism , Nasal Polyps/blood , Rhinitis/blood , Sinusitis/blood , Adult , Case-Control Studies , Chromatography, Liquid , Chronic Disease , Female , Humans , Male , Middle Aged , Nasal Polyps/diagnosis , Rhinitis/diagnosis , Secretory Pathway , Sinusitis/diagnosis , Tandem Mass Spectrometry
9.
ACS Pharmacol Transl Sci ; 4(3): 1188-1194, 2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34151208

ABSTRACT

Specialized pro-resolving mediators (SPMs) are enzymatically oxygenated derivatives of polyunsaturated fatty acids that function as central immunoregulators in mammals. Among them are resolvins (Rvs) that stimulate the clearance of harmful stimuli and limit pro-inflammatory processes. Because of their beneficial features and their high potency, SPMs are promising molecules for anti-inflammatory therapy. Besides mammals, also marine algae form lipid mediators such as prostaglandins and leukotrienes. In particular, microalgae are attractive candidates for the production of bioactive high-value metabolites. Here, we identified the diatom Cylindrotheca closterium as a prolific producer of SPMs. The diatom forms RvE3 and novel structurally related eicosanoids, including 14S/R,17R,18R-trihydroxy-eicosatetraenoic acid, which displays inflammation-resolving and anti-inflammatory bioactivities. This pro-resolving compound might enable advancements in anti-inflammatory therapy in mammals.

10.
Circ Res ; 127(5): 664-676, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32434457

ABSTRACT

RATIONALE: The alarmin S100A9 has been identified as a potential therapeutic target in myocardial infarction. Short-term S100A9 blockade during the inflammatory phase post-myocardial infarction inhibits systemic and cardiac inflammation and improves cardiac function long term. OBJECTIVE: To evaluate the impact of S100A9 blockade on postischemic cardiac repair. METHODS AND RESULTS: We assessed cardiac function, hematopoietic response, and myeloid phagocyte dynamics in WT (wild type) C57BL/6 mice with permanent coronary artery ligation, treated with the specific S100A9 blocker ABR-238901 for 7 or 21 days. In contrast to the beneficial effects of short-term therapy, extended S100A9 blockade led to progressive deterioration of cardiac function and left ventricle dilation. The treatment reduced the proliferation of Lin-Sca-1+c-Kit+ hematopoietic stem and progenitor cells in the bone marrow and the production of proreparatory CD150+CD48-CCR2+ hematopoietic stem cells. Monocyte trafficking from the spleen to the myocardium and subsequent phenotype switching to reparatory Ly6CloMerTKhi macrophages was also impaired, leading to inefficient efferocytosis, accumulation of apoptotic cardiomyocytes, and a larger myocardial scar. The transcription factor Nur77 (Nr4a1 [nuclear receptor subfamily 4 group A member 1]) mediates the transition from inflammatory Ly6Chi monocytes to reparatory Ly6Clo macrophages. S100A9 upregulated the levels and activity of Nur77 in monocytes and macrophages in vitro and in Ly6Chi/int monocytes in vivo, and S100A9 blockade antagonized these effects. Finally, the presence of reparatory macrophages in the myocardium was also impaired in S100A9-/- mice with permanent myocardial ischemia, leading to depressed cardiac function long term. CONCLUSIONS: We show that S100A9 plays an important role in both the inflammatory and the reparatory immune responses to myocardial infarction. Long-term S100A9 blockade negatively impacts cardiac recovery and counterbalances the beneficial effects of short-term therapy. These results define a therapeutic window targeting the inflammatory phase for optimal effects of S100A9 blockade as potential immunomodulatory treatment in acute myocardial infarction.


Subject(s)
Calgranulin B/metabolism , Hematopoietic Stem Cells/metabolism , Inflammation/metabolism , Inflammation/prevention & control , Myocardial Infarction/metabolism , Myocardium/metabolism , Neutrophils/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis , Calgranulin A/blood , Calgranulin B/blood , Calgranulin B/genetics , Cell Proliferation , Disease Models, Animal , Hematopoiesis , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Myocardial Infarction/drug therapy , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardium/immunology , Myocardium/pathology , Neutrophils/immunology , Neutrophils/pathology , Phagocytosis , Phenotype , RAW 264.7 Cells , Signal Transduction , Ventricular Function, Left , Ventricular Remodeling
11.
J Cell Sci ; 133(3)2020 02 12.
Article in English | MEDLINE | ID: mdl-31964713

ABSTRACT

Endothelial YAP/TAZ (YAP is also known as YAP1, and TAZ as WWTR1) signaling is crucial for sprouting angiogenesis and vascular homeostasis. However, the underlying molecular mechanisms that explain how YAP/TAZ control the vasculature remain unclear. This study reveals that the focal adhesion protein deleted-in-liver-cancer 1 (DLC1) is a direct transcriptional target of the activated YAP/TAZ-TEAD complex. We find that substrate stiffening and VEGF stimuli promote expression of DLC1 in endothelial cells. In turn, DLC1 expression levels are YAP and TAZ dependent, and constitutive activation of YAP is sufficient to drive DLC1 expression. DLC1 is needed to limit F-actin fiber formation, integrin-based focal adhesion lifetime and integrin-mediated traction forces. Depletion of endothelial DLC1 strongly perturbs cell polarization in directed collective migration and inhibits the formation of angiogenic sprouts. Importantly, ectopic expression of DLC1 is sufficient to restore migration and angiogenic sprouting in YAP-depleted cells. Together, these findings point towards a crucial and prominent role for DLC1 in YAP/TAZ-driven endothelial adhesion remodeling and collective migration during angiogenesis.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Adaptor Proteins, Signal Transducing , Endothelial Cells , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Endothelial Cells/metabolism , GTPase-Activating Proteins/genetics , Humans , Morphogenesis , Neovascularization, Pathologic , Phosphoproteins/genetics , Phosphoproteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics
12.
J Clin Invest ; 130(1): 359-373, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31793912

ABSTRACT

N-3 docosapentaenoic acid-derived resolvin D5 (RvD5n-3 DPA) is diurnally regulated in peripheral blood and exerts tissue-protective actions during inflammatory arthritis. Here, using an orphan GPCR screening approach coupled with functional readouts, we investigated the receptor(s) involved in mediating the leukocyte-directed actions of RvD5n-3 DPA and identified GPR101 as the top candidate. RvD5n-3 DPA bound to GPR101 with high selectivity and stereospecificity, as demonstrated by a calculated KD of approximately 6.9 nM. In macrophages, GPR101 knockdown limited the ability of RvD5n-3 DPA to upregulate cyclic adenosine monophosphate, phagocytosis of bacteria, and efferocytosis. Inhibition of this receptor in mouse and human leukocytes abrogated the pro-resolving actions of RvD5n-3 DPA, including the regulation of bacterial phagocytosis in neutrophils. Knockdown of the receptor in vivo reversed the protective actions of RvD5n-3 DPA in limiting joint and gut inflammation during inflammatory arthritis. Administration of RvD5n-3 DPA during E. coli-initiated inflammation regulated neutrophil trafficking to the site of inflammation, increased bacterial phagocytosis by neutrophils and macrophages, and accelerated the resolution of infectious inflammation. These in vivo protective actions of RvD5n-3 DPA were limited when Gpr101 was knocked down. Together, our findings demonstrate a fundamental role for GPR101 in mediating the leukocyte-directed actions of RvD5n-3 DPA.


Subject(s)
Arthritis/drug therapy , Docosahexaenoic Acids/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli/immunology , Macrophages/immunology , Neutrophils/immunology , Receptors, G-Protein-Coupled/agonists , Animals , Arthritis/genetics , Arthritis/immunology , Arthritis/pathology , CHO Cells , Cricetulus , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/pathology , Gene Knockdown Techniques , Humans , Macrophages/pathology , Male , Mice , Neutrophils/pathology , Phagocytosis/drug effects , Phagocytosis/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology
13.
Cell Rep ; 24(8): 2127-2140.e7, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30134173

ABSTRACT

Activation of macrophages by inflammatory stimuli induces reprogramming of mitochondrial metabolism to support the production of pro-inflammatory cytokines and nitric oxide. Hallmarks of this metabolic rewiring are downregulation of α-ketoglutarate formation by isocitrate dehydrogenase (IDH) and accumulation of glutamine-derived succinate, which enhances the inflammatory response via the activity of succinate dehydrogenase (SDH). Here, we identify the nuclear receptor Nur77 (Nr4a1) as a key upstream transcriptional regulator of this pro-inflammatory metabolic switch in macrophages. Nur77-deficient macrophages fail to downregulate IDH expression and accumulate higher levels of succinate and other TCA cycle-derived metabolites in response to inflammatory stimulation in a glutamine-independent manner. Consequently, these macrophages produce more nitric oxide and pro-inflammatory cytokines in an SDH-dependent manner. In vivo, bone marrow Nur77 deficiency exacerbates atherosclerosis development and leads to increased circulating succinate levels. In summary, Nur77 induces an anti-inflammatory metabolic state in macrophages that protects against chronic inflammatory diseases such as atherosclerosis.


Subject(s)
Gene Expression Regulation/genetics , Inflammation/metabolism , Macrophages/metabolism , Mitochondria/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Humans
14.
J Biol Chem ; 293(39): 15070-15083, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30111591

ABSTRACT

Gene targeting via homologous recombination can occasionally result in incomplete disruption of the targeted gene. Here, we show that a widely used Nur77-deficient transgenic mouse model expresses a truncated protein encoding for part of the N-terminal domain of nuclear receptor Nur77. This truncated Nur77 protein is absent in a newly developed Nur77-deficient mouse strain generated using Cre-Lox recombination. Comparison of these two mouse strains using immunohistochemistry, flow cytometry, and colony-forming assays shows that homologous recombination-derived Nur77-deficient mice, but not WT or Cre-Lox-derived Nur77-deficient mice, suffer from liver immune cell infiltrates, loss of splenic architecture, and increased numbers of bone marrow hematopoietic stem cells and splenic colony-forming cells with age. Mechanistically, we demonstrate that the truncated Nur77 N-terminal domain protein maintains the stability and activity of hypoxia-inducible factor (HIF)-1, a transcription factor known to regulate bone marrow homeostasis. Additionally, a previously discovered, but uncharacterized, human Nur77 transcript variant that encodes solely for its N-terminal domain, designated TR3ß, can also stabilize and activate HIF-1α. Meta-analysis of publicly available microarray data sets shows that TR3ß is highly expressed in human bone marrow cells and acute myeloid leukemia samples. In conclusion, our study provides evidence that a transgenic mouse model commonly used to study the biological function of Nur77 has several major drawbacks, while simultaneously identifying the importance of nongenomic Nur77 activity in the regulation of bone marrow homeostasis.


Subject(s)
Bone Marrow Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Protein Domains/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Flow Cytometry , Gene Expression Regulation/genetics , Homeostasis/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/chemistry
15.
Cardiovasc Res ; 114(12): 1617-1628, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29850786

ABSTRACT

Aims: Cardiac remodelling and heart failure are promoted by persistent sympathetic activity. We recently reported that nuclear receptor Nur77 may protect against sympathetic agonist-induced cardiac remodelling in mice. The sympathetic co-transmitter neuropeptide Y (NPY) is co-released with catecholamines and is a known cardiac modulator and predictor of heart failure mortality. Recently, transcriptome analyses revealed NPY as a putative target of Nur77. In this study, we assess whether Nur77 modulates adverse cardiac remodelling via NPY signalling. Methods and results: Nur77 represses NPY expression in the PC12 adrenal chromaffin cell line. Accordingly, NPY levels are higher in adrenal gland, plasma, and heart from Nur77-KO compared to wild-type mice. Conditioned medium from Nur77-silenced chromaffin cells and serum from Nur77-KO mice induce marked hypertrophy in cultured neonatal rat cardiomyocytes, which is inhibited by the NPY type 1 receptor (NPY1R) antagonist BIBO3304. In cardiomyocytes from Nur77-KO mice, intracellular Ca2+ is increased partially via the NPY1R. This is independent from elevated circulating NPY since cardiomyocyte-specific Nur77-deficient mice (CM-KO) do not have elevated circulating NPY, but do exhibit BIBO3304-sensitive, increased cardiomyocyte intracellular Ca2+. In vivo, this translates to NPY1R antagonism attenuating cardiac calcineurin activity and isoproterenol-induced cardiomyocyte hypertrophy and fibrosis in full-body Nur77-KO mice, but not in CM-KO mice. Conclusions: The cardioprotective action of Nur77 can be ascribed to both inhibition of circulating NPY levels and to cardiomyocyte-specific modulation of NPY-NPY1R signalling. These results reveal the underlying mechanism of Nur77 as a promising modifier gene in heart failure.


Subject(s)
Adrenal Glands/metabolism , Cardiomegaly/prevention & control , Myocytes, Cardiac/metabolism , Neuropeptide Y/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Sympathetic Nervous System/metabolism , Ventricular Remodeling , Animals , Calcineurin/metabolism , Calcium Signaling , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Female , Fibrosis , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Neuropeptide Y/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , PC12 Cells , Rats , Rats, Wistar , Receptors, Neuropeptide Y/metabolism , Sympathetic Nervous System/physiopathology
16.
Haematologica ; 103(6): 1073-1082, 2018 06.
Article in English | MEDLINE | ID: mdl-29545340

ABSTRACT

Tissue Factor is a cell-surface glycoprotein expressed in various cells of the vasculature and is the principal regulator of the blood coagulation cascade and hemostasis. Notably, aberrant expression of Tissue Factor is associated with cardiovascular pathologies such as atherosclerosis and thrombosis. Here, we sought to identify factors that regulate Tissue Factor gene expression and activity. Tissue Factor gene expression is regulated by various transcription factors, including activating protein-1 and nuclear factor-κ B. The peptidyl-prolyl isomerase Pin1 is known to modulate the activity of these two transcription factors, and we now show that Pin1 augments Tissue Factor gene expression in both vascular smooth muscle cells and activated endothelial cells via activating protein-1 and nuclear factor-κ B signaling. Furthermore, the cytoplasmic domain of Tissue Factor contains a well-conserved phospho-Ser258-Pro259 amino-acid motif recognized by Pin1. Using co-immunoprecipitation and solution nuclear magnetic resonance spectroscopy, we show that the WW-domain of Pin1 directly binds the cytoplasmic domain of Tissue Factor. This interaction occurs via the phospho-Ser258-Pro259 sequence in the Tissue Factor cytoplasmic domain and results in increased protein half-life and pro-coagulant activity. Taken together, our results establish Pin1 as an upstream regulator of Tissue Factor-mediated coagulation, thereby opening up new avenues for research into the use of specific Pin1 inhibitors for the treatment of diseases characterized by pathological coagulation, such as thrombosis and atherosclerosis.


Subject(s)
Coagulants/metabolism , Gene Expression , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Thromboplastin/genetics , Thromboplastin/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Multiprotein Complexes/metabolism , NF-kappa B/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Proteolysis , Thromboplastin/chemistry , Transcription Factor AP-1/metabolism
17.
PLoS One ; 12(2): e0172721, 2017.
Article in English | MEDLINE | ID: mdl-28231341

ABSTRACT

Cellular cholesterol metabolism is subject to tight regulation to maintain adequate levels of this central lipid molecule. Herein, the sterol-responsive Liver X Receptors (LXRs) play an important role owing to their ability to reduce cellular cholesterol load. In this context, identifying the full set of LXR-regulated genes will contribute to our understanding of their role in cholesterol metabolism. Using global transcriptional analysis we report here the identification of RNF145 as an LXR-regulated target gene. We demonstrate that RNF145 is regulated by LXRs in both human and mouse primary cells and cell lines, and in vivo in mice. Regulation of RNF145 by LXR depends on a functional LXR-element in its proximal promotor. Consistent with LXR-dependent regulation of Rnf145 we show that regulation is lost in macrophages and fibroblasts from Lxrαß(-/-) mice, and also in vivo in livers of Lxrα(-/-) mice treated with the LXR synthetic ligand T0901317. RNF145 is closely related to RNF139/TRC8, an E3 ligase implicated in control of SREBP processing. However, silencing of RNF145 in HepG2 or HeLa cells does not impair SREBP1/2 processing and sterol-responsive gene expression in these cells. Similar to TRC8, we demonstrate that RNF145 is localized to the ER and that it possesses intrinsic E3 ubiquitin ligase activity. In summary, we report the identification of RNF145 as an ER-resident E3 ubiquitin ligase that is transcriptionally controlled by LXR.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Liver X Receptors/genetics , Membrane Proteins/genetics , Transcription, Genetic , Animals , Cell Line , Cholesterol/metabolism , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver/drug effects , Liver/metabolism , Liver X Receptors/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic , Sulfonamides/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
18.
Arterioscler Thromb Vasc Biol ; 37(3): 423-432, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28082258

ABSTRACT

OBJECTIVE: The sterol-responsive nuclear receptors, liver X receptors α (LXRα, NR1H3) and ß (LXRß, NR1H2), are key determinants of cellular cholesterol homeostasis. LXRs are activated under conditions of high cellular sterol load and induce expression of the cholesterol efflux transporters ABCA1 and ABCG1 to promote efflux of excess cellular cholesterol. However, the full set of genes that contribute to LXR-stimulated cholesterol efflux is unknown, and their identification is the objective of this study. APPROACH AND RESULTS: We systematically compared the global transcriptional response of macrophages to distinct classes of LXR ligands. This allowed us to identify both common and ligand-specific transcriptional responses in macrophages. Among these, we identified endonuclease-exonuclease-phosphatase family domain containing 1 (EEPD1/KIAA1706) as a direct transcriptional target of LXRs in human and murine macrophages. EEPD1 specifically localizes to the plasma membrane owing to the presence of a myristoylation site in its N terminus. Accordingly, the first 10 amino acids of EEPD1 are sufficient to confer plasma membrane localization in the context of a chimeric protein with GFP. Functionally, we report that silencing expression of EEPD1 blunts maximal LXR-stimulated Apo AI-dependent efflux and demonstrate that this is the result of reduced abundance of ABCA1 protein in human and murine macrophages. CONCLUSIONS: In this study, we identify EEPD1 as a novel LXR-regulated gene in macrophages and propose that it promotes cellular cholesterol efflux by controlling cellular levels and activity of ABCA1.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cell Membrane/enzymology , Cholesterol/metabolism , Endodeoxyribonucleases/metabolism , Liver X Receptors/metabolism , Macrophages/enzymology , ATP Binding Cassette Transporter 1/genetics , Animals , Apolipoprotein A-I/metabolism , Biological Transport , COS Cells , Cell Membrane/drug effects , Chlorocebus aethiops , Endodeoxyribonucleases/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , HeLa Cells , Hep G2 Cells , Humans , Ligands , Liver X Receptors/agonists , Liver X Receptors/deficiency , Liver X Receptors/genetics , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , RNA Interference , Transcriptome , Transfection
19.
BMC Genomics ; 17: 162, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26932821

ABSTRACT

BACKGROUND: The nuclear orphan receptor Nur77 (NR4A1, TR3, or NGFI-B) has been shown to modulate the inflammatory response of macrophages. To further elucidate the role of Nur77 in macrophage physiology, we compared the transcriptome of bone marrow-derived macrophages (BMM) from wild-type (WT) and Nur77-knockout (KO) mice. RESULTS: In line with previous observations, SDF-1α (CXCL12) was among the most upregulated genes in Nur77-deficient BMM and we demonstrated that Nur77 binds directly to the SDF-1α promoter, resulting in inhibition of SDF-1α expression. The cytokine receptor CX3CR1 was strongly downregulated in Nur77-KO BMM, implying involvement of Nur77 in macrophage tolerance. Ingenuity pathway analyses (IPA) to identify canonical pathways regulation and gene set enrichment analyses (GSEA) revealed a potential role for Nur77 in extracellular matrix homeostasis. Nur77-deficiency increased the collagen content of macrophage extracellular matrix through enhanced expression of several collagen subtypes and diminished matrix metalloproteinase (MMP)-9 activity. IPA upstream regulator analyses discerned the small GTPase Rac1 as a novel regulator of Nur77-mediated gene expression. We identified an inhibitory feedback loop with increased Rac1 activity in Nur77-KO BMM, which may explain the augmented phagocytic activity of these cells. Finally, we predict multiple chronic inflammatory diseases to be influenced by macrophage Nur77 expression. GSEA and IPA associated Nur77 to osteoarthritis, chronic obstructive pulmonary disease, rheumatoid arthritis, psoriasis, and allergic airway inflammatory diseases. CONCLUSIONS: Altogether these data identify Nur77 as a modulator of macrophage function and an interesting target to treat chronic inflammatory disease.


Subject(s)
Extracellular Matrix/metabolism , Immune Tolerance , Inflammation/metabolism , Macrophages/cytology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phagocytosis , Animals , CX3C Chemokine Receptor 1 , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Collagen/metabolism , Gene Expression Regulation , Homeostasis , Inflammation/genetics , Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Neuropeptides/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Promoter Regions, Genetic , RAW 264.7 Cells , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Transcriptome , rac1 GTP-Binding Protein/metabolism
20.
Biochim Biophys Acta ; 1863(2): 219-28, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26548523

ABSTRACT

FHL2 belongs to the LIM-domain only proteins and contains four and a half LIM domains, each of which are composed of two zinc finger structures. FHL2 exhibits specific interaction with proteins exhibiting diverse functions, including transmembrane receptors, transcription factors and transcription co-regulators, enzymes, and structural proteins. The function of these proteins is regulated by FHL2, which modulates intracellular signal transduction pathways involved in a plethora of cellular tasks. The present review summarizes the current knowledge on the protein interactome of FHL2 and provides an overview of the functional implication of these interactions in apoptosis, migration, and regulation of nuclear receptor function. FHL2 was originally identified in the heart and there is extensive literature available on the role of FHL2 in the cardiovascular system, which is also summarized in this review.


Subject(s)
Cardiovascular Diseases/metabolism , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Protein Interaction Mapping/methods , Transcription Factors/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , LIM-Homeodomain Proteins/chemistry , Models, Biological , Models, Molecular , Muscle Proteins/chemistry , Protein Structure, Tertiary , Transcription Factors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...