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










Publication year range
1.
J Cell Physiol ; 2015 May 07.
Article in English | MEDLINE | ID: mdl-25953328

ABSTRACT

The transcription of the ATP-binding cassette transporter A1 (ABCA1) gene, which plays a key anti-atherogenic role, is known to be induced by agonists of liver X receptors (LXRs). LXRs form obligate heterodimers with retinoid X receptors (RXRs) and interact with their recognition sequences in the regulatory regions of key genes implicated in the control of cholesterol, fatty acid and glucose homeostasis. We have previously shown a novel role for c-Jun N-terminal kinase (JNK) and phosphoinositide 3-kinase (PI3K) in the LXRs-mediated induction of macrophage gene expression. Protein kinase C (PKC) is often found to regulate the action of nuclear receptors and cross talk between this kinase family and JNK and/or PI3K has been shown in several settings. We have therefore investigated a potential role for PKC in the action of LXR/RXR agonist 22-(R)-hydroxycholesterol (22-(R)-HC)/9-cis-retinoic acid (9cRA) in THP-1 macrophages, including the induction of ABCA1 expression. The pan PKC inhibitor bisindoylmaleimide was found to attenuate the induction of ABCA1 protein expression, the activation of the JNK signaling pathway and the stimulation of activator protein-1 (AP-1) DNA binding activity in macrophages treated with 22-(R)-HC and 9cRA. The role of PKC in the action of these ligands was confirmed further by the use of more isotype-specific inhibitors. These studies therefore reveal a potentially important role for PKC in the action of 22-(R)-HC and 9cRA in human macrophages. This article is protected by copyright. All rights reserved.

2.
J Cell Biochem ; 116(9): 2032-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25752819

ABSTRACT

The transcription of the ATP-binding cassette transporter A1 (ABCA1) gene, which plays a key anti-atherogenic role, is known to be induced by agonists of liver X receptors (LXRs). LXRs form obligate heterodimers with retinoid X receptors (RXRs) and interact with their recognition sequences in the regulatory regions of key genes implicated in the control of cholesterol, fatty acid and glucose homeostasis. We have previously shown a novel role for c-Jun N-terminal kinase (JNK) and phosphoinositide 3-kinase (PI3K) in the LXRs-mediated induction of macrophage gene expression. Protein kinase C (PKC) is often found to regulate the action of nuclear receptors and cross talk between this kinase family and JNK and/or PI3K has been shown in several settings. We have, therefore, investigated a potential role for PKC in the action of LXR/RXR agonist 22-(R)-hydroxycholesterol (22-(R)-HC)/9-cis-retinoic acid (9cRA) in THP-1 macrophages, including the induction of ABCA1 expression. The pan PKC inhibitor bisindoylmaleimide was found to attenuate the induction of ABCA1 protein expression, the activation of the JNK signaling pathway and the stimulation of activator protein-1 (AP-1) DNA binding activity in macrophages treated with 22-(R)-HC and 9cRA. The role of PKC in the action of these ligands was confirmed further by the use of more isotype-specific inhibitors. These studies, therefore, reveal a potentially important role for PKC in the action of 22-(R)-HC and 9cRA in human macrophages.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Hydroxycholesterols/pharmacology , Macrophages/drug effects , Protein Kinase C/metabolism , Tretinoin/pharmacology , ATP Binding Cassette Transporter 1/antagonists & inhibitors , Alitretinoin , Cell Line , Gene Expression Regulation/drug effects , Humans , Indoles/pharmacology , Liver X Receptors , MAP Kinase Signaling System/drug effects , Macrophages/cytology , Maleimides/pharmacology , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/metabolism , Retinoid X Receptors/agonists , Retinoid X Receptors/metabolism
3.
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
4.
Cell Signal ; 23(3): 542-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21070853

ABSTRACT

Liver X receptors (LXRs) are ligand-dependent transcription factors that are activated by metabolites of cholesterol, oxysterols, and a number of synthetic agonists. LXRs play potent anti-atherogenic roles in part by stimulating the efflux of cholesterol from macrophage foam cells. The LXR-induced expression of ATP-binding cassette transporter (ABC)-A1 and Apolipoprotein E (ApoE) in macrophages is essential for the stimulation of cholesterol efflux and the prevention of atherosclerotic development. Unfortunately, the signaling pathways underlying such regulation are poorly understood and were therefore investigated in human macrophages. The expression of ApoE and ABCA1 induced by synthetic or natural LXR ligands [TO901317, GW3965, and 22-(R)-hydroxycholesterol (22-(R)-HC), respectively] was attenuated by inhibitors of c-Jun N-terminal kinase (JNK) (curcumin and SP600125) and phosphoinositide 3-kinase (PI3K) (LY294002). Similar results were obtained with ABCG1 and LXR-α, two other LXR target genes. LXR agonists activated several components of the JNK pathway (SEK1, JNK and c-Jun) along with AKT, a downstream target for PI3K. In addition, dominant negative mutants of JNK and PI3K pathways inhibited the LXR-agonists-induced activity of the ABCA1 and LXR-α gene promoters in transfected cells. LXR agonists also induced the binding of activator protein-1 (AP-1), a key transcription factor family regulated by JNK, to recognition sequences present in the regulatory regions of the ApoE and ABCA1 genes. These studies reveal a novel role for JNK and PI3K/AKT signaling in the LXR-regulated expression in macrophages of several key genes implicated in atherosclerosis.


Subject(s)
JNK Mitogen-Activated Protein Kinases/physiology , Macrophages/enzymology , Orphan Nuclear Receptors/metabolism , Phosphatidylinositol 3-Kinases/physiology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Cells, Cultured , Cholesterol/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Liver X Receptors , Macrophages/drug effects , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Transcription Factor AP-1/metabolism , Transcriptional Activation
5.
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
6.
Int J Biochem Cell Biol ; 42(1): 113-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19800021

ABSTRACT

Elevated circulating levels of acute phase proteins (APP) are associated with inflammation and inflammatory disorders such as cardiovascular disease. APP are mainly synthesised by hepatocytes and their transcription is induced by pro-inflammatory cytokines such as interleukin-1 (IL-1). The molecular mechanisms underlying the IL-1-induced expression of key transcription factors implicated in the regulation of APP are poorly understood. We have investigated this aspect using the CCAAT/enhancer binding protein-delta (C/EBPdelta) as a model gene. IL-1 induced the expression of C/EBPdelta mRNA and protein in the human hepatoma Hep3B cell line, a widely employed model system for studies on cytokine signalling in relation to the expression of APP. The IL-1-mediated induction of C/EBPdelta expression was attenuated in the presence of pharmacological inhibitors against c-Jun N-terminal kinase (JNK) (curcumin and SP600125), casein kinase 2 (CK2) (apigenin) and nuclear factor-kappaB (NF-kappaB) (NF-kappaB activation inhibitor). RNA interference assays showed significant attenuation of the IL-1-induced expression of C/EBPdelta following knockdown of the p50 and p65 subunits of NF-kappaB. IL-1 induced NF-kappaB DNA binding and activation by this transcription factor and this was attenuated by curcumin and apigenin. Taken together, these results suggest a potentially crucial role for NF-kappaB in the IL-1-induced expression of C/EBPdelta, and thereby downstream APP genes regulated by this transcription factor.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Interleukin-1beta/pharmacology , NF-kappa B/metabolism , Signal Transduction/drug effects , Apigenin/pharmacology , Binding Sites , CCAAT-Enhancer-Binding Protein-delta/metabolism , Casein Kinase II/antagonists & inhibitors , Cell Line, Tumor , Curcumin/pharmacology , DNA/metabolism , Enzyme Activation/drug effects , Gene Knockdown Techniques , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Time Factors , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
7.
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
8.
J Mol Med (Berl) ; 86(8): 887-97, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18437331

ABSTRACT

Casein kinase 2 (CK2) is a highly conserved serine-threonine kinase that uses both adenosine triphosphate and guanosine triphosphate as phosphate donors. This constitutively active and ubiquitously expressed enzyme is often present as a tetrameric holoenzyme complex of two catalytic subunits (alpha and/or alpha') and two regulatory beta subunits. The enzyme is known to phosphorylate more than 300 substrates and controls a wide range of processes, including the regulation of cell cycle, apoptosis, transformation, and circadian rhythm. Several lines of recent evidence also suggest a potentially important role for CK2 in the control of the inflammatory response. This review will give an overview of CK2 and its regulation and describe the evidence implicating its role in inflammation.


Subject(s)
Casein Kinase II/physiology , Inflammation/enzymology , Animals , Apoptosis , Catalytic Domain , Humans , Inflammation/metabolism , Phosphorylation , Signal Transduction , Transcription Factors/metabolism
9.
Cytokine Growth Factor Rev ; 17(6): 487-99, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17056295

ABSTRACT

Transforming growth factor-beta (TGF-beta) plays a pivotal role in a range of biological processes, including the control of cellular proliferation and differentiation, regulation of tissue repair and extracellular matrix accumulation, and modulation of the immune and inflammatory responses. The role of TGF-beta in the pathogenesis of atherosclerosis, which is widely perceived as a form of chronic inflammation, has been the subject of debate for a number of years. A pro-atherogenic role was suspected because of its ability to promote fibrosis and to inhibit endothelial regeneration. However, several recent studies have shown that TGF-beta limits atherosclerosis by modulating a number of processes, including the accumulation of lipids in the vessel wall and the inflammatory response. This review will discuss the role of TGF-beta in atherosclerosis along with the molecular mechanisms underlying its action during the pathogenesis of the disease.


Subject(s)
Atherosclerosis/etiology , Transforming Growth Factor beta/physiology , Animals , Atherosclerosis/physiopathology , Blood Vessels/physiopathology , Endothelial Cells/physiology , Humans , Macrophages/physiology , Models, Cardiovascular , Monocytes/physiology , Muscle, Smooth, Vascular/physiopathology , Signal Transduction
10.
Arterioscler Thromb Vasc Biol ; 26(6): 1323-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16601234

ABSTRACT

OBJECTIVE: The cytokine transforming growth factor-beta (TGF-beta) and apolipoprotein E (apoE) play potent antiatherogenic roles. Despite such importance, the mechanisms underlying the regulation of apoE expression by TGF-beta have not been characterized and were therefore investigated. METHODS AND RESULTS: Using THP-1 cell line as a model system, with key findings confirmed in primary cultures, we show that TGF-beta induces the expression of apoE, and this is prevented by pharmacological inhibitors of c-Jun N-terminal kinase (JNK), p38 kinase, and casein kinase 2 (CK2). In support for an important role for these pathways, TGF-beta activates JNK, p38 kinase, and CK2, and dominant-negative (DN) forms of these proteins inhibit the cytokine-induced apoE expression. TGF-beta also increases the phosphorylation and expression of c-Jun, a downstream target for JNK action and a component of activator protein-1 (AP-1), and DN c-Jun inhibits the induction of apoE expression in response to the cytokine. AP-1 DNA binding was also induced by TGF-beta, and the action of p38 kinase, JNK, and CK2 converged on the activation of c-Jun/AP-1. CONCLUSIONS: These studies reveal a novel role for JNK, p38 kinase, CK2, and c-Jun/AP-1 in the TGF-beta-induced expression of apoE.


Subject(s)
Apolipoproteins E/genetics , Casein Kinase II/physiology , Gene Expression Regulation, Enzymologic , JNK Mitogen-Activated Protein Kinases/physiology , Transforming Growth Factor beta/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Apolipoproteins E/metabolism , Casein Kinase II/genetics , Cell Line, Tumor , DNA/metabolism , Enzyme Activation , Gene Expression Regulation/physiology , Genes, Dominant , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/metabolism , Monocytes/metabolism , Phosphorylation , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
11.
J Biol Chem ; 278(20): 17741-51, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12609974

ABSTRACT

Interferon-gamma (IFN-gamma) is a pleiotropic cytokine that modulates the immune function, cell proliferation, apoptosis, macrophage activation, and numerous other cellular responses. These biological actions of IFN-gamma are characterized by both the activation and the inhibition of gene transcription. Unfortunately, in contrast to gene activation, the mechanisms through which the cytokine suppresses gene transcription remain largely unclear. We show here for the first time that exposure of macrophages to IFN-gamma leads to a dramatic induction in the expression of the inducible cAMP early repressor (ICER), a potent inhibitor of gene transcription. In addition, a synergistic action of IFN-gamma and calcium in the activation of ICER expression was identified. The IFN-gamma-mediated activation of ICER expression was not blocked by H89, bisindoylmaleimide, SB202190, PD98059, W7, and AG490, which inhibit protein kinase A, protein kinase C, p38 mitogen-activated protein kinase, extracellular signal-regulated kinase, calcium-calmodulin-dependent protein kinase, and Janus kinase-2, respectively. In contrast, apigenin, a selective casein kinase 2 (CK2) inhibitor, was found to inhibit response. Consistent with this finding, IFN-gamma stimulated CK2 activity and the level of phosphorylated cAMP response element-binding protein, which is known to induce ICER gene transcription, and this response was inhibited in the presence of apigenin. These studies, therefore, identify a previously uncharacterized pathway, involving the IFN-gamma-mediated stimulation of CK2 activity, activation of cAMP response element-binding protein, and increased production of ICER, which may then play an important role in the inhibition of macrophage gene transcription by this cytokine.


Subject(s)
Cyclic AMP/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Interferon-gamma/pharmacology , Macrophages/enzymology , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins , Transcription, Genetic , 8-Bromo Cyclic Adenosine Monophosphate/metabolism , Animals , Apigenin , Blotting, Western , Calcimycin/pharmacology , Calcium/metabolism , Casein Kinase II , Cell Line , Cloning, Molecular , Colforsin/pharmacology , Cyclic AMP Response Element Modulator , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/metabolism , Flavonoids/pharmacology , Genetic Vectors , Imidazoles/pharmacology , Indoles/pharmacology , Interferon-gamma/metabolism , Macrophages/metabolism , Maleimides/pharmacology , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Protein Binding , Protein Kinase C/antagonists & inhibitors , Pyridines/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sulfonamides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thapsigargin/pharmacology , Time Factors , Transcriptional Activation , Tyrphostins/pharmacology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...