Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 6: 34368, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27687241

ABSTRACT

The anti-atherogenic cytokine TGF-ß inhibits macrophage foam cell formation by suppressing the expression of key genes implicated in the uptake of modified lipoproteins. We have previously shown a critical role for p38 MAPK and JNK in the TGF-ß-mediated regulation of apolipoprotein E expression in human monocytes. However, the roles of these two MAPK pathways in the control of expression of key genes involved in the uptake of modified lipoproteins in human macrophages is poorly understood and formed the focus of this study. TGF-ß activated both p38 MAPK and JNK, and knockdown of p38 MAPK or c-Jun, a key downstream target of JNK action, demonstrated their requirement in the TGF-ß-inhibited expression of several key genes implicated in macrophage lipoprotein uptake. The potential role of c-Jun and specific co-activators in the action of TGF-ß was investigated further by studies on the lipoprotein lipase gene. c-Jun did not directly interact with the minimal promoter region containing the TGF-ß response elements and a combination of transient transfection and knock down assays revealed an important role for SRC-1. These studies provide novel insights into the mechanisms underlying the TGF-ß-mediated inhibition of macrophage gene expression associated with the control of cholesterol homeostasis.

2.
Int J Biochem Cell Biol ; 46: 113-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24275094

ABSTRACT

Atherosclerosis is an inflammatory disorder of the vasculature regulated by cytokines. Amongst the cytokines, IL-33 attenuates the development of atherosclerosis in mouse model systems via several mechanisms, including inhibition of macrophage foam cell formation and promotion of a Th1 to Th2 shift. Proteases produced by macrophages, such as matrix metalloproteinases and members of ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family, play potential roles in regulating atherosclerotic plaque stability. Despite such importance, the action of IL-33 on the expression of such proteases has not been analyzed. We have therefore investigated the effect of IL-33 on the expression of ADAMTS-1, -4 and -5 in human macrophages. Immunohistochemical analysis showed that these three proteases were expressed in human atherosclerotic lesions, particularly by macrophages and, to a lesser extent, by smooth muscle cells and endothelial cells. The expression of ADAMTS-1, -4 and -5 in human macrophages was specifically inhibited by IL-33. The action of IL-33 on the expression of these ADAMTS members was mediated through its receptor ST2. IL-33 activated ERK1/2, JNK1/2 and c-Jun, but not p38 MAPK or Akt, in human macrophages. RNA interference assays using a combination of adenoviral encoding small hairpin RNA and small interfering RNA showed a requirement of ERK1/2, JNK1/2, c-Jun, PI3Kγ and PI3Kδ, but not p38α, in the IL-33-inhibited expression of these ADAMTS isoforms. These studies provide novel insights into the expression of ADAMTS-1, -4 and -5 in human atherosclerotic lesions and the regulation of their expression in human macrophages by the key anti-atherogenic cytokine IL-33.


Subject(s)
Cytokines/metabolism , Disintegrins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Metalloproteases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Thrombospondins/metabolism , Animals , Atherosclerosis/metabolism , Humans , Macrophages/metabolism , Mice , Signal Transduction
3.
Biochim Biophys Acta ; 1822(10): 1608-16, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22705205

ABSTRACT

The anti-atherogenic cytokine, TGF-ß, plays a key role during macrophage foam cell formation by modulating the expression of key genes involved in the control of cholesterol homeostasis. Unfortunately, the molecular mechanisms underlying these actions of TGF-ß remain poorly understood. In this study we examine the effect of TGF-ß on macrophage cholesterol homeostasis and delineate the role of Smads-2 and -3 during this process. Western blot analysis showed that TGF-ß induces a rapid phosphorylation-dependent activation of Smad-2 and -3 in THP-1 and primary human monocyte-derived macrophages. Small interfering RNA-mediated knockdown of Smad-2/3 expression showed that the TGF-ß-mediated regulation of key genes implicated in the uptake of modified low density lipoproteins and the efflux of cholesterol from foam cells was Smad-dependent. Additionally, through the use of virally delivered Smad-2 and/or Smad-3 short hairpin RNA, we demonstrate that TGF-ß inhibits the uptake of modified LDL by macrophages through a Smad-dependent mechanism and that the TGF-ß-mediated regulation of CD36, lipoprotein lipase and scavenger receptor-A gene expression was dependent on Smad-2. These studies reveal a crucial role for Smad signaling, particularly Smad-2, in the inhibition of foam cell formation by TGF-ß through the regulation of expression of key genes involved in the control of macrophage cholesterol homeostasis.


Subject(s)
Lipoproteins, LDL/metabolism , Macrophages/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cells, Cultured , Cholesterol/genetics , Cholesterol/metabolism , Foam Cells/metabolism , Gene Expression , Homeostasis , Humans , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Lipoproteins, LDL/genetics , Phosphorylation , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Signal Transduction , Smad2 Protein/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics
4.
Int J Biochem Cell Biol ; 43(5): 805-11, 2011 May.
Article in English | MEDLINE | ID: mdl-21334453

ABSTRACT

Atherosclerosis is an inflammatory disorder of the vasculature that is orchestrated by the action of cytokines. Macrophages play a prominent role in all stages of this disease, including foam cell formation, production of reactive oxygen species, modulation of the inflammatory response and the regulation of the stability of atherosclerotic plaques. The role of the matrix metalloproteinase family in the control of plaque stability is well established. A disintegrin and metalloproteinase with thrombospondin motif (ADAMTS) family has been implicated in several diseases and the expression of ADAMTS-4 in macrophages of atherosclerotic lesions has suggested a potential role for this protease in atherosclerosis. However, the action of cytokines on the expression of ADAMTS-4 in macrophages is poorly understood. We have investigated here the effect of transforming growth factor-ß (TGF-ß) on ADAMTS-4 expression in macrophages along with the regulatory mechanisms underlying its actions. Consistent with the anti-atherogenic role of TGF-ß, this cytokine decreased the expression of ADAMTS-4 mRNA and protein in human macrophages. Transient transfection assays showed that the -100 to +10 promoter region contained the minimal TGF-ß response elements. Small-interfering RNA-mediated knockdown revealed a critical role for Smads, p38 mitogen-activated protein kinase and c-Jun in the action of TGF-ß on ADAMTS-4 mRNA expression. These studies show for the first time that TGF-ß inhibits the expression of ADAMTS-4 in human macrophages and identifies the signalling pathways underlying this response. The inhibition of macrophage ADAMTS-4 expression is likely to contribute to the anti-atherogenic, plaque stabilisation action of TGF-ß.


Subject(s)
ADAM Proteins/metabolism , Gene Expression Regulation, Enzymologic/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/drug effects , Procollagen N-Endopeptidase/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta1/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , ADAM Proteins/genetics , ADAMTS4 Protein , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Humans , Macrophages/metabolism , Procollagen N-Endopeptidase/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transforming Growth Factor beta1/therapeutic use
5.
J Cell Biochem ; 112(2): 675-83, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268089

ABSTRACT

PPARs and LXRs are ligand-activated transcription factors that are emerging as promising therapeutic targets for limiting atherosclerosis, an inflammatory disorder orchestrated by cytokines. The potent anti-atherogenic actions of these nuclear receptors involve the regulation of glucose and lipid metabolism along with attenuation of the inflammatory response. Similarly, cholesterol-lowering drugs, statins, inhibit inflammation. Unfortunately, the mechanisms underlying such inhibitory actions of these agents in human macrophages are poorly understood and were therefore investigated in relation to IFN-γ, a key pro-atherogenic cytokine, which mediates its cellular effects mainly through STAT1. Simvastatin and PPAR agonists had no effect on the IFN-γ-induced, phosphorylation-mediated activation of STAT1 and its DNA binding but attenuated its ability to activate gene transcription. On the other hand, LXR activators attenuated both DNA binding and trans-activation potential of STAT1 induced by IFN-γ. These studies reveal differences in the mechanism of action of agonists of PPARs (and simvastatin) and LXRs on the IFN-γ-induced, STAT1-mediated gene transcription in human macrophages.


Subject(s)
Interferon-gamma/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Orphan Nuclear Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , STAT1 Transcription Factor/metabolism , Simvastatin/pharmacology , Blotting, Western , Butyrates/pharmacology , Cell Line , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Electrophoretic Mobility Shift Assay , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver X Receptors , Orphan Nuclear Receptors/agonists , Peroxisome Proliferator-Activated Receptors/agonists , Phenylurea Compounds/pharmacology , Rosiglitazone , Sulfonamides/pharmacology , Thiazolidinediones/pharmacology , Transfection
6.
J Mol Med (Berl) ; 88(12): 1203-11, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20652528

ABSTRACT

The ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) proteases are secreted enzymes that regulate extracellular matrix turnover by degrading specific matrix components. Roles for the proteases in inflammation and atherosclerosis have been suggested by a number of recent studies, and the role of ADAMTS-4 and -5 in the breakdown of aggrecan and subsequent degradation of cartilage during osteoarthritis has also been established. The ability of the ADAMTS proteases to degrade versican, the primary proteoglycan in the vasculature, is thought to be central to any hypothesized role for the proteases in atherosclerosis. In this review, we introduce the structure and function of the ADAMTS family of proteases and review the literature that links them with inflammation and atherosclerosis.


Subject(s)
ADAM Proteins/metabolism , Atherosclerosis/enzymology , ADAM Proteins/chemistry , ADAM Proteins/genetics , Animals , Cytokines/metabolism , Humans , Inflammation/enzymology , Protein Structure, Tertiary , Versicans/metabolism
7.
J Immunol ; 185(2): 1222-9, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20543107

ABSTRACT

The development of atherosclerosis, a chronic inflammatory disease characterized by the formation of arterial fibrotic plaques, has been shown to be reduced by IL-33 in vivo. However, whether IL-33 can directly affect macrophage foam cell formation, a key feature of atherosclerotic plaques, has not been determined. In this study, we investigated whether IL-33 reduces macrophage foam cell accumulation in vivo and if IL-33 reduces their formation in vitro using THP-1 and primary human monocyte-derived macrophages. In Apolipoprotein E(-/-) mice fed on a high fat diet, IL-33 treatment significantly reduced the accumulation of macrophage-derived foam cells in atherosclerotic plaques. IL-33 also reduced macrophage foam cell formation in vitro by decreasing acetylated and oxidized low-density lipoprotein uptake, reducing intracellular total and esterified cholesterol content and enhancing cholesterol efflux. These changes were associated with IL-33-mediated reduction in the expression of genes involved in modified low-density lipoprotein uptake, such as CD36, and simultaneous increase in genes involved in cholesterol efflux, including Apolipoprotein E, thereby providing a mechanism for such an action for this cytokine. IL-33 also decreased the expression of key genes implicated in cholesterol esterification and triglyceride storage, including Acyl-CoA:cholesterol acyltransferase 1 and Adipocyte differentiation-related protein. Furthermore, using bone marrow-derived macrophages from ST2(-/-) mice, we demonstrate that the IL-33 receptor, ST2, is integral to the action of IL-33 on macrophage foam cell formation. In conclusion, IL-33 has a protective role in atherosclerosis by reducing macrophage foam cell formation suggesting that IL-33 maybe a potential therapeutic agent against atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Foam Cells/drug effects , Interleukins/pharmacology , Macrophages/drug effects , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Blotting, Western , Cell Line , Cells, Cultured , Cholesterol/metabolism , Cholesterol Esters/metabolism , Female , Foam Cells/metabolism , Foam Cells/pathology , Gene Expression/drug effects , Humans , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacokinetics , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
J Immunol ; 184(10): 5827-34, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20410491

ABSTRACT

TNF-like protein 1A (TL1A), a TNF superfamily cytokine that binds to death receptor 3 (DR3), is highly expressed in macrophage foam cell-rich regions of atherosclerotic plaques, although its role in foam cell formation has yet to be elucidated. We investigated whether TL1A can directly stimulate macrophage foam cell formation in both THP-1 and primary human monocyte-derived macrophages with the underlying mechanisms involved. We demonstrated that TL1A promotes foam cell formation in human macrophages in vitro by increasing both acetylated and oxidized low-density lipoprotein uptake, by enhancing intracellular total and esterified cholesterol levels and reducing cholesterol efflux. This imbalance in cholesterol homeostasis is orchestrated by TL1A-mediated changes in the mRNA and protein expression of several genes implicated in the uptake and efflux of cholesterol, such as scavenger receptor A and ATP-binding cassette transporter A1. Furthermore, through the use of virally delivered DR3 short-hairpin RNA and bone marrow-derived macrophages from DR3 knockout mice, we demonstrate that DR3 can regulate foam cell formation and contributes significantly to the action of TL1A in this process in vitro. We show, for the first time, a novel proatherogenic role for both TL1A and DR3 that implicates this pathway as a target for the therapeutic intervention of atherosclerosis.


Subject(s)
Cell Differentiation/immunology , Foam Cells/cytology , Foam Cells/immunology , Receptors, Tumor Necrosis Factor, Member 25/physiology , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/physiology , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Biological Transport/immunology , Cell Line, Tumor , Cells, Cultured , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol Ester Transfer Proteins/metabolism , Female , Foam Cells/pathology , Humans , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Lipoproteins, LDL/metabolism , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 25/deficiency , Up-Regulation/immunology
9.
Int J Biochem Cell Biol ; 41(5): 1189-97, 2009 May.
Article in English | MEDLINE | ID: mdl-19027873

ABSTRACT

Tumour necrosis factor-alpha (TNF-alpha) is a key regulator of the immune and inflammatory responses along with numerous other cellular changes during physiological and pathophysiological conditions. The cellular actions of TNF-alpha are associated with both the activation and the inhibition of gene transcription. In contrast to gene activation, the mechanisms underlying the TNF-alpha-mediated transcriptional inhibition remain largely unclear. We have investigated this aspect using the transcription factor CCAAT/enhancer binding protein-alpha (C/EBPalpha) as a model gene. TNF-alpha decreased the expression of C/EBPalpha mRNA and protein in the human hepatoma Hep3B cell line. The activity of the proximal promoter of both the human and the Xenopus C/EBPalpha genes in transfected Hep3B cells was inhibited by TNF-alpha. Transient transfection assays using various Xenopus C/EBPalpha promoter-luciferase DNA constructs showed that a C/EBP recognition sequence was essential for the TNF-alpha response. Electrophoretic mobility shift assays showed that C/EBPalpha bound to this site and co-transfection assays revealed that it was a major activator of the promoter and its transactivation potential was reduced by TNF-alpha. The potential role of nuclear factor kappaB (NF-kappaB) in the response was also investigated in the light of its pivotal role in TNF-alpha signalling. Inhibition of NF-kappaB using pharmacological agents or by transfection of a plasmid specifying for a superrepressor attenuated the TNF-alpha-inhibited C/EBPalpha promoter activity. In addition, an involvement of NF-kappaB in DNA-protein interactions at the C/EBP recognition sequence was identified.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , Hepatocytes/physiology , NF-kappa B/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , CCAAT-Enhancer-Binding Protein-alpha/antagonists & inhibitors , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Hepatocytes/metabolism , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , NF-kappa B/pharmacology , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transfection , Tumor Necrosis Factor-alpha/physiology , Up-Regulation , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...