Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Exp Cell Res ; 317(2): 131-42, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20955695

ABSTRACT

AIMS: Activin A and transforming growth factor-ß1 (TGF-ß1) belong to the same family of growth and differentiation factors that modulate vascular lesion formation in distinct ways, which we wish to understand mechanistically. METHODS AND RESULTS: We investigated the expression of cell-surface receptors and activation of Smads in human vascular smooth muscle cells (SMCs) and demonstrated that activin receptor-like kinase-1 (ALK-1), ALK-4, ALK-5 and endoglin are expressed in human SMCs. As expected, TGF-ß1 activates Smad1 and Smad2 in these cells. Interestingly, activin A also induces phosphorylation of both Smads, which has not been reported for Smad1 before. Transcriptome analyses of activin A and TGF-ß1 treated SMCs with subsequent Gene-Set Enrichment Analyses revealed that many downstream gene networks are induced by both factors. However, the effect of activin A on expression kinetics of individual genes is less pronounced than for TGF-ß1, which is explained by a more rapid dephosphorylation of Smads and p38-MAPK in response to activin A. Substantial differences in expression of fibronectin, alpha-V integrin and total extracellular collagen synthesis were observed. CONCLUSIONS: Genome-wide mRNA expression analyses clarify the distinct modulation of vascular lesion formation by activin A and TGF-ß1, most significantly because activin A is non-fibrotic.


Subject(s)
Activin Receptors, Type II/metabolism , Activins/pharmacology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Phenotype , Transforming Growth Factor beta/pharmacology , Activin Receptors, Type I/metabolism , Activins/genetics , Activins/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Myocytes, Smooth Muscle/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Growth Factor/biosynthesis , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Saphenous Vein/cytology , Smad2 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Circulation ; 121(18): 2023-32, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20421523

ABSTRACT

BACKGROUND: Restenosis is the major drawback of percutaneous coronary interventions involving excessive activation and proliferation of vascular smooth muscle cells (SMCs). The nuclear receptor Nurr1 is an early response gene known mainly for its critical role in the development of dopamine neurons. In the present study, we investigated Nurr1 in human and experimental vascular restenosis. METHODS AND RESULTS: In a prospective cohort of 601 patients undergoing percutaneous coronary intervention, including stent placement, we found a strong association between Nurr1 haplotypes and in-stent restenosis risk. Furthermore, Nurr1 is specifically expressed in human in-stent restenosis and induced in cultured human SMCs in response to serum or tumor necrosis factor-alpha. Lentivirus-mediated gain- and loss-of-function experiments in SMCs demonstrated that overexpression of Nurr1 inhibited proliferation, consistent with increased expression of the key cell-cycle inhibitor p27(Kip1), whereas Nurr1 silencing enhanced SMC growth. The tumor necrosis factor-alpha-induced proinflammatory response of SMCs is inhibited by Nurr1, as reflected by reduced interleukin-1beta, tumor necrosis factor-alpha, and monocyte chemoattractant protein-1 expression. Consistent with our in vitro data, endogenous Nurr1 reduced wire injury-induced proliferation and vascular lesion formation in carotid arteries of ApoE(-/-) mice. CONCLUSIONS: Nurr1 haplotypes are associated with human restenosis risk, and Nurr1 is expressed in human in-stent restenosis. In SMCs, Nurr1 inhibits proliferation and inflammatory responses, which explains the inhibition of SMC-rich lesion formation in mice. The recently identified small-molecule drugs that enhance the activity of Nurr1 reveal this nuclear receptor as an attractive novel target for (local) intervention in restenosis.


Subject(s)
Coronary Artery Disease/genetics , Coronary Restenosis/genetics , Muscle, Smooth, Vascular/pathology , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Vasculitis/genetics , Angioplasty, Balloon, Coronary , Animals , Apolipoproteins E/genetics , Cell Division/physiology , Cells, Cultured , Coronary Artery Disease/epidemiology , Coronary Artery Disease/therapy , Coronary Restenosis/epidemiology , Coronary Restenosis/pathology , Coronary Vessels/immunology , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Disease Models, Animal , Female , Genetic Predisposition to Disease/epidemiology , Haplotypes , Humans , Linkage Disequilibrium , Mice , Mice, Mutant Strains , Muscle, Smooth, Vascular/physiology , Neovascularization, Physiologic/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Risk Factors , Stents , Vasculitis/epidemiology , Vasculitis/pathology
3.
Arterioscler Thromb Vasc Biol ; 30(8): 1591-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20413732

ABSTRACT

OBJECTIVE: 6-Mercaptopurine (6-MP), the active metabolite of the immunosuppressive prodrug azathioprine, is commonly used in autoimmune diseases and transplant recipients, who are at high risk for cardiovascular disease. Here, we aimed to gain knowledge on the action of 6-MP in atherosclerosis, with a focus on monocytes and macrophages. METHODS AND RESULTS: We demonstrate that 6-MP induces apoptosis of THP-1 monocytes, involving decreased expression of the intrinsic antiapoptotic factors B-cell CLL/Lymphoma-2 (Bcl-2) and Bcl2-like 1 (Bcl-x(L)). In addition, we show that 6-MP decreases expression of the monocyte adhesion molecules platelet endothelial adhesion molecule-1 (PECAM-1) and very late antigen-4 (VLA-4) and inhibits monocyte adhesion. Screening of a panel of cytokines relevant to atherosclerosis revealed that 6-MP robustly inhibits monocyte chemoattractant chemokine-1 (MCP-1) expression in macrophages stimulated with lipopolysaccharide (LPS). Finally, local delivery of 6-MP to the vessel wall, using a drug-eluting cuff, attenuates atherosclerosis in hypercholesterolemic apolipoprotein E*3-Leiden transgenic mice (P<0.05). In line with our in vitro data, this inhibition of atherosclerosis by 6-MP was accompanied with decreased lesion monocyte chemoattractant chemokine-1 levels, enhanced vascular apoptosis, and reduced macrophage content. CONCLUSIONS: We report novel, previously unrecognized atheroprotective actions of 6-MP in cultured monocytes/macrophages and in a mouse model of atherosclerosis, providing further insight into the effect of the immunosuppressive drug azathioprine in atherosclerosis.


Subject(s)
Apolipoprotein E3/metabolism , Atherosclerosis/prevention & control , Immunosuppressive Agents/pharmacology , Macrophages/drug effects , Mercaptopurine/pharmacology , Monocytes/drug effects , Animals , Apolipoprotein E3/genetics , Apoptosis/drug effects , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Adhesion/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Chemotaxis/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Immunosuppressive Agents/administration & dosage , Inflammation Mediators/metabolism , Integrin alpha4beta1/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mercaptopurine/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors , bcl-X Protein/metabolism
4.
Cardiovasc Res ; 87(3): 561-8, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20189954

ABSTRACT

AIMS: Structural adaptation of the vessel wall in response to sustained alterations in haemodynamic forces is known as vascular remodelling. Detailed knowledge on the mechanism underlying this vascular response is limited, and we aimed to study the function of Nur77 in smooth muscle cells (SMCs) in arterial remodelling. METHODS AND RESULTS: Carotid artery ligation in mice results in flow-induced, outward remodelling of the contralateral carotid artery, and we observed enhanced Nur77 expression during this process. Transgenic mice that express Nur77 or its dominant-negative variant, denoted as 'DeltaTA' in arterial SMCs, were exposed to carotid artery ligation, and after 4 weeks pressure-diameter relationships were measured. Structural outward remodelling is inhibited in Nur77-transgenic mice when compared with wild-type and DeltaTA-transgenic mice. The key determinants of remodelling vascular tone and macrophage accumulation were studied. No difference in contractile and relaxant responses was detected in isolated aorta, carotid, and mesenteric artery segments between transgenic and wild-type mice. SMC-specific overexpression of Nur77 in transgenic mice reduced macrophage accumulation and repressed matrix metalloproteinase (MMP)1 and MMP9 expression at early time points. MMP2 protein expression was reduced in Nur77-transgenic mice, whereas in DeltaTA-transgenic mice MMP2 expression was increased. CONCLUSION: Nur77 is induced during outward remodelling and inhibits this vascular adaptation in mice. Nur77-mediated inhibition of arterial remodelling involves a reduction in both macrophage accumulation and MMP expression levels.


Subject(s)
Carotid Arteries/enzymology , Carotid Artery Diseases/enzymology , Hemodynamics , Macrophages/pathology , Matrix Metalloproteinases/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Adaptation, Physiological , Animals , Blood Pressure , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Carotid Artery Diseases/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Gene Expression Regulation, Enzymologic , Hemodynamics/drug effects , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/genetics , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , RNA, Messenger/metabolism , Regional Blood Flow , Time Factors , Vasoconstriction , Vasoconstrictor Agents/pharmacology , Vasodilation , Vasodilator Agents/pharmacology
5.
Circulation ; 120(8): 669-76, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19667240

ABSTRACT

BACKGROUND: The cyclin-dependent kinase inhibitor p27(kip1) is a key regulator of smooth muscle cell and leukocyte proliferation in vascular disease, including in-stent restenosis. We therefore hypothesized that common genetic variations or single nucleotide polymorphisms in p27(kip1) may serve as a useful tool in risk stratification for in-stent restenosis. METHODS AND RESULTS: Three single nucleotide polymorphisms concerning the p27(kip1) gene (-838C>A, rs36228499; -79C>T, rs34330; +326G>T, rs2066827) were determined in a cohort of 715 patients undergoing coronary angioplasty and stent placement. We discovered that the p27(kip1)-838C>A single nucleotide polymorphism is associated with clinical in-stent restenosis; the -838AA genotype decreases the risk of target vessel revascularization (hazard ratio, 0.28; 95% confidence interval, 0.10 to 0.77). This finding was replicated in another cohort study of 2309 patients (hazard ratio, 0.61; 95% confidence interval, 0.40 to 0.93). No association was detected between this end point and the p27(kip1)-79C>T and +326G>T single nucleotide polymorphisms. We subsequently studied the functional importance of the -838C>A single nucleotide polymorphism and detected a 20-fold increased basal p27(kip1) transcriptional activity of the -838A allele containing promoter. CONCLUSIONS: Patients with the p27(kip1)-838AA genotype have a decreased risk of in-stent restenosis corresponding with enhanced promoter activity of the -838A allele of this cell-cycle inhibitor, which may explain decreased smooth muscle cell proliferation.


Subject(s)
Coronary Artery Disease/therapy , Coronary Restenosis/epidemiology , Coronary Restenosis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Aged , Angioplasty, Balloon, Coronary , Cell Division/physiology , Coronary Artery Disease/epidemiology , Coronary Artery Disease/genetics , Cyclin-Dependent Kinase Inhibitor p27 , Female , Genetic Predisposition to Disease/epidemiology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Risk Factors , Stents
6.
Hum Mutat ; 30(3): 417-23, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19156841

ABSTRACT

Nuclear receptor subfamily 4, group A, member 2 (NR4A2, also called Nurr1) has lately become of interest with regard to atherogenesis. We examined the association between common variation in the NR4A2 gene and cardiovascular disease in the Rotterdam Study, a prospective population-based study among persons aged > or = 55 years. Three SNPs that tag common haplotypes across a 36-kb region surrounding the NR4A2 gene were determined. Four haplotypes with frequencies >1% covered 96% of the genetic variation. In 5,650 participants without history of coronary heart disease, 729 coronary heart disease events occurred during a median follow-up time of 11.9 years. NR4A2 haplotypes were neither associated with coronary events nor with intima-media thickness (IMT), carotid plaques, or ankle-arm index (AAI). NR4A2 haplotypes showed a tendency toward associations with aortic and coronary calcification (haplo.score global simulation P values 0.076 and 0.075, respectively), which seemed to be based on haplotype 2 (individual P values were both P=0.015). Furthermore, NR4A2 haplotype 3 was associated with higher high-density lipoprotein (HDL) cholesterol and haplotype 4 with lower systolic blood pressure. In conclusion, NR4A2/NURR1 haplotypes were not associated with coronary events, carotid IMT, carotid plaques, or AAI. There was a tendency toward associations with aortic calcification and coronary calcification. Associations for NR4A2 were found with both HDL levels and blood pressure. It remains to be investigated which pathophysiological mechanisms pertain to NR4A2 function in cardiovascular disease.


Subject(s)
Cardiovascular Diseases/genetics , DNA-Binding Proteins/genetics , Haplotypes , Transcription Factors/genetics , Aged , Aorta/pathology , Blood Pressure , Calcinosis/pathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Cholesterol/analysis , Cholesterol, HDL/analysis , Coronary Vessels/pathology , Female , Gene Frequency , Genotype , Humans , Male , Middle Aged , Netherlands , Nuclear Receptor Subfamily 4, Group A, Member 2 , Odds Ratio , Polymorphism, Single Nucleotide , Prospective Studies , Tunica Intima/pathology , Tunica Media/pathology
7.
Arterioscler Thromb Vasc Biol ; 28(7): 1339-46, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18467642

ABSTRACT

OBJECTIVE: Atheroprotective blood flow induces expression of anti-inflammatory Krüppel-like factor 2 (KLF2) and activates antioxidant transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) in vascular endothelium. Previously, we obtained KLF2-induced gene expression profiles in ECs, containing several Nrf2 target genes. Our aim was to investigate the role of KLF2 in shear stress-mediated activation of Nrf2 in human umbilical vein endothelial cells (HUVECs). METHODS AND RESULTS: Expression of Nrf2 and its targets NAD(P)H dehydrogenase quinone 1 (NQO1) and heme oxygenase (HO-1) was elevated by shear and KLF2. KLF2 knockdown showed that shear-induced expression of NQO1 but not Nrf2 was dependent on KLF2. KLF2 overexpression in absence of flow resulted in more efficient activation of Nrf2 by tert-butyl hydroquinone (tBHQ) through enhanced nuclear localization, and promoted expression of a large panel of Nrf2-dependent genes resulting in superior protection against oxidative stress. Comparison of shear-, KLF2-, and Nrf2-induced transcriptomes showed that the majority of shear-modulated gene sets is influenced by KLF2 or Nrf2. CONCLUSIONS: We report that KLF2 substantially enhances antioxidant activity of Nrf2 by increasing its nuclear localization and activation. The synergistic activity of these two transcription factors forms a major contribution to the shear stress-elicited transcriptome in endothelial cells.


Subject(s)
Antioxidants/metabolism , Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , NF-E2-Related Factor 2/metabolism , Active Transport, Cell Nucleus , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Induction , Gene Expression Profiling , Gene Regulatory Networks , Heme Oxygenase-1/biosynthesis , Humans , Kruppel-Like Transcription Factors/genetics , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , NF-E2-Related Factor 2/genetics , Oxidants/pharmacology , Oxidative Stress , Pulsatile Flow , RNA Interference , RNA, Small Interfering/metabolism , Stress, Mechanical , Transfection , Up-Regulation , tert-Butylhydroperoxide/pharmacology
8.
Biochem Biophys Res Commun ; 366(4): 910-6, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18086558

ABSTRACT

NR4A nuclear receptors are induced in the liver upon fasting and regulate hepatic gluconeogenesis. Here, we studied the role of nuclear receptor Nur77 (NR4A1) in hepatic lipid metabolism. We generated mice expressing hepatic Nur77 using adenoviral vectors, and demonstrate that these mice exhibit a modulation of the plasma lipid profile and a reduction in hepatic triglyceride. Expression analysis of >25 key genes involved in lipid metabolism revealed that Nur77 inhibits SREBP1c expression. This results in decreased SREBP1c activity as is illustrated by reduced expression of its target genes stearoyl-coA desaturase-1, mitochondrial glycerol-3-phosphate acyltransferase, fatty acid synthase and the LDL receptor, and provides a mechanism for the physiological changes observed in response to Nur77. Expression of LXR target genes Abcg5 and Abcg8 is reduced by Nur77, and may suggest involvement of LXR in the inhibitory action of Nur77 on SREBP1c expression. Taken together, our study demonstrates that Nur77 modulates hepatic lipid metabolism through suppression of SREBP1c activity.


Subject(s)
DNA-Binding Proteins/metabolism , Lipid Metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Transcription Factors/metabolism , Adenoviridae , Animals , Gene Expression Regulation , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 4, Group A, Member 1 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Triglycerides/blood
9.
Am J Pathol ; 171(1): 326-37, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591977

ABSTRACT

Knowledge about the in vivo role of endothelium in chronic human atherosclerosis has mostly been derived by insights from mouse models. Therefore, we set out to establish by microarray analyses the gene expression profiles of endothelium from human large arteries, as isolated by laser microbeam microdissection, having focal atherosclerosis of the early or the advanced stage. Within individual arteries, the endothelial transcriptomes of the lesional and unaffected sides were compared pairwise, thus limiting genetic and environmental confounders. Specific endothelial signature gene sets were identified with changed expression levels in either early (n = 718) or advanced atherosclerosis (n = 403), relative to their paired plaque-free controls. Gene set enrichment analysis identified distinct sets of chemokines and differential enrichments of nuclear factor-kappaB-, p53-, and transforming growth factor-beta-related genes in advanced plaques. Immunohistochemistry validated the discriminative value of corresponding endothelial protein expression between early (fractalkine/CX3CL1, IP10/CCL10, TBX18) or advanced (BAX, NFKB2) stages of atherosclerosis and versus their plaque-free controls. The functional involvement of transforming growth factor-beta signaling in directing its downstream gene repertoire was substantiated by a consistent detection of activated SMAD2 in advanced lesions. Thus, we identified truly common, local molecular denominators of pathological changes to vascular endothelium, with a marked distinction of endothelial phenotype between early and advanced plaques.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/metabolism , Chemokines/metabolism , Endothelium, Vascular/metabolism , Gene Expression Profiling , Transforming Growth Factor beta/metabolism , Genes, p53 , Humans , Immunohistochemistry , Lasers , Microdissection , NF-kappa B/genetics , Reproducibility of Results , Signal Transduction
10.
Circulation ; 115(4): 493-500, 2007 Jan 30.
Article in English | MEDLINE | ID: mdl-17242285

ABSTRACT

BACKGROUND: Restenosis is a common complication after percutaneous coronary interventions and is characterized by excessive proliferation of vascular smooth muscle cells (SMCs). We have shown that the nuclear receptor Nur77 protects against SMC-rich lesion formation, and it has been demonstrated that 6-mercaptopurine (6-MP) enhances Nur77 activity. We hypothesized that 6-MP inhibits neointima formation through activation of Nur77. METHODS AND RESULTS: It is demonstrated that 6-MP increases Nur77 activity in cultured SMCs, which results in reduced [3H]thymidine incorporation, whereas Nur77 small interfering RNA knockdown partially restores DNA synthesis. Furthermore, we studied the effect of 6-MP in a murine model of cuff-induced neointima formation. Nur77 mRNA is upregulated in cuffed arteries, with optimal expression after 6 hours and elevated expression up to 7 days after vascular injury. Local perivascular delivery of 6-MP with a drug-eluting cuff significantly inhibits neointima formation in wild-type mice. Locally applied 6-MP does not affect inflammatory responses or apoptosis but inhibits expression of proliferating cell nuclear antigen and enhances protein levels of the cell-cycle inhibitor p27(Kip1) in the vessel wall. An even stronger inhibition of neointima formation in response to local 6-MP delivery was observed in transgenic mice that overexpressed Nur77. In contrast, 6-MP does not alter lesion formation in transgenic mice that overexpress a dominant-negative variant of Nur77 in arterial SMCs, which provides evidence for the involvement of Nur77-like factors. CONCLUSIONS: Enhancement of the activity of Nur77 by 6-MP protects against excessive SMC proliferation and SMC-rich neointima formation. We propose that activation of the nuclear receptor Nur77 is a rational approach to treating in-stent restenosis.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Coronary Restenosis/drug therapy , DNA-Binding Proteins/metabolism , Mercaptopurine/pharmacology , Muscle, Smooth, Vascular/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cells, Cultured , Coronary Restenosis/metabolism , Coronary Restenosis/pathology , DNA-Binding Proteins/genetics , Disease Models, Animal , Drug Implants , Femoral Artery/pathology , Humans , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1 , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Transcription Factors/genetics , Tunica Intima/drug effects , Tunica Intima/pathology , Umbilical Arteries/cytology
11.
Blood ; 109(10): 4249-57, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17244683

ABSTRACT

Absence of shear stress due to disturbed blood flow at arterial bifurcations and curvatures leads to endothelial dysfunction and proinflammatory gene expression, ultimately resulting in atherogenesis. KLF2 has recently been implicated as a transcription factor involved in mediating the anti-inflammatory effects of flow. We investigated the effect of shear on basal and TNF-alpha-induced genomewide expression profiles of human umbilical vein endothelial cells (HUVECs). Cluster analysis confirmed that shear stress induces expression of protective genes including KLF2, eNOS, and thrombomodulin, whereas basal expression of TNF-alpha-responsive genes was moderately decreased. Promoter analysis of these genes showed enrichment of binding sites for ATF transcription factors, whereas TNF-alpha-induced gene expression was mostly NF-kappaB dependent. Furthermore, human endothelial cells overlying atherosclerotic plaques had increased amounts of phosphorylated nuclear ATF2 compared with endothelium at unaffected sites. In HUVECs, a dramatic reduction of nuclear binding activity of ATF2 was observed under shear and appeared to be KLF2 dependent. Reduction of ATF2 with siRNA potently suppressed basal proinflammatory gene expression under no-flow conditions. In conclusion, we demonstrate that shear stress and KLF2 inhibit nuclear activity of ATF2, providing a potential mechanism by which endothelial cells exposed to laminar flow are protected from basal proinflammatory, atherogenic gene expression.


Subject(s)
Activating Transcription Factor 2/genetics , Atherosclerosis/genetics , Inflammation/genetics , Kruppel-Like Transcription Factors/physiology , Activating Transcription Factor 2/antagonists & inhibitors , Activating Transcription Factor 2/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Cluster Analysis , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Phosphorylation , Protein Kinases/metabolism , RNA, Small Interfering/pharmacology , Stress, Mechanical , Time Factors , Transcription, Genetic , Tumor Necrosis Factor-alpha/pharmacology
12.
Arterioscler Thromb Vasc Biol ; 27(3): 532-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17194892

ABSTRACT

OBJECTIVE: The flow-responsive Kruppel-like factor 2 (KLF2) is crucial for maintaining endothelial cell quiescence. Here, we describe its detailed effects on transforming growth factor-beta (TGF-beta) signaling, which normally has proatherogenic effects on endothelium. METHODS AND RESULTS: In-depth analysis of genome-wide expression data shows that prolonged lentiviral-mediated overexpression of KLF2 in human umbilical vein endothelial cells (HUVECs) diminishes the expression of a large panel of established TGF-beta-inducible genes. Both baseline and TGF-beta-induced expression levels of plasminogen activator inhibitor 1 (PAI-1) and thrombospondin-1 are greatly diminished by KLF2. Using a combination of ectopic expression, small interfering RNA-mediated knockdown, and promoter activity assays, we show that KLF2 partly inhibits the phosphorylation and subsequent nuclear accumulation of Smad2, thereby suppressing the TGF-beta-induced Smad4-mediated transcriptional activity. This is achieved through TGF-beta-independent induction of inhibitory Smad7. Additionally, a full inhibition of TGF-beta signaling is functionally achieved through a simultaneous suppression of activator protein 1 (AP-1), which is an essential cofactor for TGF-beta-dependent transcription of many genes. CONCLUSIONS: The concerted mechanism by which KLF2 inhibits TGF-beta signaling through induction of inhibitory Smad7 and attenuation of AP-1 activity provides a novel mechanism by which KLF2 contributes to sustaining a quiescent, atheroprotective status of vascular endothelium.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Signal Transduction/physiology , Smad7 Protein/metabolism , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta/metabolism , Blotting, Western , Cells, Cultured , Down-Regulation , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Kruppel-Like Transcription Factors/pharmacology , Phosphorylation , RNA Interference , Signal Transduction/drug effects , Smad7 Protein/genetics , Transcription Factor AP-1/genetics , Umbilical Veins/cytology
13.
Atherosclerosis ; 192(2): 283-90, 2007 Jun.
Article in English | MEDLINE | ID: mdl-16938301

ABSTRACT

Accumulation of lipid-laden macrophages is a hallmark of atherosclerosis. The relevance of the key transcription factor nuclear factor kappaB (NF-kappaB) for macrophage-derived foam-cell formation has not been unequivocally resolved. Transgenic mice lines were generated in which NF-kappaB activation is specifically inhibited in macrophages by overexpressing a trans-dominant, non-degradable form of IkappaBalpha (IkappaBalpha (32A/36A)) under control of the macrophage-specific SR-A promoter. Alanine substitution of serines 32 and 36 prevents degradation and retains the inactive NF-kappaB/IkappaBalpha (32A/36A) complex in the cytoplasm. Similarly, stable human THP1 monocytic cell lines were generated with integrated copies of IkappaBalpha (32A/36A) cDNA. Upon treatment with oxidized low-density lipoprotein (ox-LDL), murine peritoneal macrophages from transgenic IkappaBalpha (32A/36A) mice, as well as THP1/IkappaBalpha (32A/36A) clones, display decreased lipid loading after differentiation into macrophages. This is accompanied by increased expression of the transcription factors PPARgamma and LXRalpha as well as of the major cholesterol-efflux transporter ABCA1. Paradoxically, mRNA expression of the 'lipid-uptake' receptor CD36 is also increased. Since the net result of these changes is reduction of foam-cell formation, it is proposed that under specific inhibition of NF-kappaB activation, ABCA1-mediated cholesterol efflux prevails over CD36-mediated lipid influx.


Subject(s)
Foam Cells/physiology , Macrophages/drug effects , NF-kappa B/antagonists & inhibitors , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/biosynthesis , Animals , CD36 Antigens/biosynthesis , Cell Line , DNA-Binding Proteins/biosynthesis , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/physiology , Lipoproteins, LDL/metabolism , Liver X Receptors , Macrophages/physiology , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , Orphan Nuclear Receptors , PPAR gamma/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Scavenger Receptors, Class A/biosynthesis
14.
Cardiovasc Res ; 72(2): 231-40, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16945356

ABSTRACT

OBJECTIVE: The transcription factor KLF2 is considered an important mediator of the anti-inflammatory and anti-thrombotic properties of the endothelium. KLF2 is absent from low-shear, atherosclerosis-prone sites of the vascular tree but is induced by HMG-CoA reductase inhibitors (statins) in vitro. We studied KLF2-dependent induction of important determinants of the atheroprotective status of the endothelium to determine whether pharmacological intervention, e.g. by statins, can potentially replace shear stress. METHODS: Shear stress and statin effects in combination with TNF-alpha were determined in human umbilical vein endothelial cells by quantitative measurements of the steady-state levels and stability of mRNA for KLF2 and its downstream target genes thrombomodulin (TM) and endothelial nitric oxide synthase (eNOS). RESULTS: We demonstrate that prolonged shear stress has a potential that is superior to that of statins to induce the KLF2-dependent expression of eNOS and TM, especially in the presence of the pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha). These effects can be attributed to the sustained stabilization of KLF2 mRNA by shear, leading to an increased KLF2 protein expression and concomitant strong induction of KLF2 downstream targets. The stabilization of KLF2 mRNA is demonstrated to be dependent on signaling involving phosphoinositide 3-kinase (PI3K). CONCLUSION: The stabilization of KLF2 steady-state levels, as induced by prolonged shear stress but not by statins, may be essential for sustaining the quiescent, atheroprotective status of the vascular endothelium under inflammatory conditions.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors/metabolism , RNA, Messenger/metabolism , Atherosclerosis/immunology , Cells, Cultured , Chromones/pharmacology , Endothelial Cells/immunology , Endothelium, Vascular/immunology , Fluorescent Antibody Technique , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , In Vitro Techniques , Inflammation , Kruppel-Like Transcription Factors/genetics , Morpholines/pharmacology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oncogene Protein v-akt/metabolism , Phosphoinositide-3 Kinase Inhibitors , RNA Interference , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stress, Mechanical , Thrombomodulin/genetics , Thrombomodulin/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Eur J Cell Biol ; 85(11): 1131-44, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16959372

ABSTRACT

Members of the claudin family are involved in formation of barriers that control access to the paracellular space of epithelia. Likewise, endothelium-specific claudin-5 is involved in the function of the blood-brain barrier (BBB). Here, we assessed the role of claudin-5 in non-BBB endothelial barriers using lentiviral-driven overexpression and silencing of claudin-5 in its native environment of primary vascular endothelial cells. Effects were monitored using macromolecular tracers between 342Da and 40kDa. Measurements were made both in absence and presence of transmigrating leukocytes. Freeze-fracture preparations were analyzed for effects at the ultrastructural level. We show that overexpression of claudin-5 leads to formation of elaborate networks of junction strands, which are absent in untransduced endothelial cells. Concomitantly, a modest, non-size-selective enhancement of the barrier function was observed. In contrast, silencing of endogenous claudin-5 does not influence barrier function. The efficient sealing of the endothelium during diapedesis of monocytes or granulocytes is also claudin-5 independent. Collectively, these data provide evidence for a limited contribution of claudin-5 to the barrier function of human umbilical vein endothelial cells (HUVEC), implying that, unlike selective barriers in epithelia, the barrier of non-BBB endothelium seems largely independent of claudin-directed tight junction structures.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Membrane Proteins/metabolism , Tight Junctions/metabolism , Cell Membrane Permeability , Cells, Cultured , Claudin-5 , Endothelial Cells/ultrastructure , Fluorescent Antibody Technique , Freeze Fracturing , Gene Silencing , Humans , Lentivirus/genetics , Lentivirus/metabolism , Membrane Proteins/genetics , RNA Interference
16.
Biochem J ; 400(3): 401-10, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16886909

ABSTRACT

Lipid rafts and caveolae are biochemically similar, specialized domains of the PM (plasma membrane) that cluster specific proteins. However, they are morphologically distinct, implying different, possibly complementary functions. Two-dimensional gel electrophoresis preceding identification of proteins by MS was used to compare the relative abundance of proteins in DRMs (detergent-resistant membranes) isolated from HUVEC (human umbilical-vein endothelial cells), and caveolae immunopurified from DRM fractions. Various signalling and transport proteins were identified and additional cell-surface biotinylation revealed the majority to be exposed, demonstrating their presence at the PM. In resting endothelial cells, the scaffold of immunoisolated caveolae consists of only few resident proteins, related to structure [CAV1 (caveolin-1), vimentin] and transport (V-ATPase), as well as the GPI (glycosylphosphatidylinositol)-linked, surface-exposed protein CD59. Further quantitative characterization by immunoblotting and confocal microscopy of well-known [eNOS (endothelial nitric oxide synthase) and CAV1], less known [SNAP-23 (23 kDa synaptosome-associated protein) and BASP1 (brain acid soluble protein 1)] and novel [C8ORF2 (chromosome 8 open reading frame 2)] proteins showed different subcellular distributions with none of these proteins being exclusive to either caveolae or DRM. However, the DRM-associated fraction of the novel protein C8ORF2 (approximately 5% of total protein) associated with immunoseparated caveolae, in contrast with the raft protein SNAP-23. The segregation of caveolae from lipid rafts was visually confirmed in proliferating cells, where CAV1 was spatially separated from eNOS, SNAP-23 and BASP1. These results provide direct evidence for the previously suggested segregation of transport and signalling functions between specialized domains of the endothelial plasma membrane.


Subject(s)
Caveolae/metabolism , Endothelial Cells/metabolism , Membrane Microdomains/metabolism , Protein Transport/physiology , Proteome/metabolism , Signal Transduction/physiology , Caveolae/chemistry , Caveolin 1/metabolism , Cells, Cultured , Endothelial Cells/ultrastructure , Humans , Membrane Microdomains/chemistry , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type III/metabolism , Peptides/metabolism , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Repressor Proteins/metabolism
17.
Arterioscler Thromb Vasc Biol ; 26(10): 2288-94, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16873729

ABSTRACT

OBJECTIVE: Atherosclerosis is an inflammatory disease in which macrophage activation and lipid loading play a crucial role. In this study, we investigated expression and function of the NR4A nuclear receptor family, comprising Nur77 (NR4A1, TR3), Nurr1 (NR4A2), and NOR-1 (NR4A3) in human macrophages. METHODS AND RESULTS: Nur77, Nurr1, and NOR-1 are expressed in early and advanced human atherosclerotic lesion macrophages primarily in areas of plaque activation/progression as detected by in situ-hybridization and immunohistochemistry. Protein expression localizes to the nucleus. Primary and THP-1 macrophages transiently express NR4A-factors in response to lipopolysaccharide and tumor necrosis factor alpha. Lentiviral overexpression of Nur77, Nurr1, or NOR-1 reduces expression and production of interleukin (IL)-1beta and IL-6 proinflammatory cytokines and IL-8, macrophage inflammatory protein-1alpha and -1beta and monocyte chemoattractant protein-1 chemokines. In addition, NR4A-factors reduce oxidized-low-density lipoprotein uptake, consistent with downregulation of scavenger receptor-A, CD36, and CD11b macrophage marker genes. Knockdown of Nur77 or NOR-1 with gene-specific lentiviral short-hairpin RNAs resulted in enhanced cytokine and chemokine synthesis, increased lipid loading, and augmented CD11b expression, demonstrating endogenous NR4A-factors to inhibit macrophage activation, foam-cell formation, and differentiation. CONCLUSIONS: NR4A-factors are expressed in human atherosclerotic lesion macrophages and reduce human macrophage lipid loading and inflammatory responses, providing further evidence for a protective role of NR4A-factors in atherogenesis.


Subject(s)
Atherosclerosis/metabolism , DNA-Binding Proteins/metabolism , Inflammation/prevention & control , Lipid Metabolism , Macrophages/metabolism , Membrane Transport Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Transcription Factors/metabolism , Atherosclerosis/pathology , Chemokines/antagonists & inhibitors , Cytokines/antagonists & inhibitors , DNA-Binding Proteins/genetics , Gene Transfer Techniques , Genetic Vectors , Humans , Inflammation Mediators/antagonists & inhibitors , Lentivirus/genetics , Lipopolysaccharides/pharmacology , Lipoproteins, LDL/antagonists & inhibitors , Macrophages/drug effects , Membrane Transport Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1 , Nuclear Receptor Subfamily 4, Group A, Member 2 , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Transcription Factors/genetics
18.
Am J Pathol ; 168(6): 2027-35, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16723716

ABSTRACT

In coronary artery bypass surgery, the patency of arterial grafts is higher than that of venous grafts because of vein-graft disease, which involves excessive proliferation of venous smooth muscle cells (SMCs) and subsequent accelerated atherosclerosis. We studied the function of TR3 nuclear orphan receptor (TR3) in the early response of SMCs to mechanical strain, a major initiator of vein-graft disease. We demonstrate that TR3 expression is induced in human saphenous vein segments exposed ex vivo to whole-blood perfusion under arterial pressure. Cultured venous SMCs challenged by cyclic stretch displayed TR3 induction and enhanced DNA synthesis, whereas SMCs derived from the internal mammary artery remained quiescent. Small-interfering RNA-mediated knockdown of TR3 and adenovirus-mediated overexpression of TR3 in venous SMCs enhanced and abolished stretch-induced DNA synthesis, respectively. Accordingly, in organ cultures of wild-type murine vessel segments exposed to cyclic stretch, p27(Kip1) was down-regulated, whereas expression of this cell cycle inhibitor was unaffected by cyclic stretch in TR3-transgenic vessels, concordant with a lower proliferative response. Finally, stretch-mediated proliferation was inhibited by 6-mercaptopurine, an agonist of TR3. In conclusion, TR3 represents inhibitory mechanisms to restrict venous SMC proliferation and may contribute to prevention of vein-graft disease.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Muscle, Smooth, Vascular/cytology , Receptors, Steroid/physiology , Receptors, Thyroid Hormone/physiology , Animals , Carotid Arteries/pathology , Cell Cycle , Cell Proliferation , Cells, Cultured , Down-Regulation , Humans , Mice , Mice, Transgenic , Models, Biological , Myocytes, Smooth Muscle/cytology , Nuclear Receptor Subfamily 4, Group A, Member 1 , RNA, Small Interfering/metabolism
19.
Thromb Haemost ; 95(2): 348-53, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16493499

ABSTRACT

Tissue factor (TF) is a transmembrane protein, which is essential for initiation of the coagulation cascade. TF has been reported to play an important role in the progression of endotoxin (lipopolysaccharide, LPS)-mediated endotoxemia, being induced in numerous tissues, such as kidney, spleen and lung. We developed and validated a rabbit anti-murine TF peptide antiserum to localize TF protein in a murine sepsis model. TF protein distribution was compared to localization of TF mRNA and fibrin deposits, the ultimate resultant of procoagulant TF activity. Evident LPS mediated TF mRNA induction was observed in the tubular area at the cortico-medullar junction in the kidney, and TF activity was increased after 6 hours of endotoxemia. In the spleen, however, TF mRNA was induced in the interfollicular region upon LPS injection, corresponding to increased TF protein in the same area. The clusters of TF-protein positive cells in the spleen are predominantly granulocytes, but no TF mRNA expression was observed within these cells. Based on these observations and the presence of TF-protein positive granulocytes after splenectomy, we hypothesize that granulocytes take-up TF for transport to other locations in order to initiate fibrin formation or to induce pro-inflammatory gene expression upon interaction with factor VIIa.


Subject(s)
Gene Expression Regulation , Granulocytes/physiology , RNA, Messenger/analysis , Sepsis/genetics , Thromboplastin/genetics , Animals , Endotoxemia , Female , Fibrin/metabolism , Gene Expression Regulation/drug effects , Granulocytes/metabolism , Immune Sera , Inflammation/genetics , Kidney , Lipopolysaccharides/pharmacology , Mice , Protein Transport , Rabbits , Spleen , Thromboplastin/analysis , Thromboplastin/metabolism , Tissue Distribution
20.
Blood ; 107(11): 4354-63, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16455954

ABSTRACT

The flow-responsive transcription factor KLF2 is acquiring a leading role in the regulation of endothelial cell gene expression. A genome-wide microarray expression profiling is described employing lentivirus-mediated, 7-day overexpression of human KLF2 at levels observed under prolonged flow. KLF2 is not involved in lineage typing, as 42 endothelial-specific markers were unaffected. Rather, KLF2 generates a gene transcription profile (> 1000 genes) affecting key functional pathways such as cell migration, vasomotor function, inflammation, and hemostasis and induces a morphology change typical for shear exposure including stress fiber formation. Protein levels for thrombomodulin, endothelial nitric oxide synthase, and plasminogen activator inhibitor type-1 are altered to atheroprotective levels, even in the presence of the inflammatory cytokine TNF-alpha. KLF2 attenuates cell migration by affecting multiple genes including VEGFR2 and the potent antimigratory SEMA3F. The distribution of Weibel-Palade bodies in cultured cell populations is normalized at the single-cell level without interfering with their regulated, RalA-dependent release. In contrast, thrombin-induced release of Weibel-Palade bodies is significantly attenuated, consistent with the proposed role of VWF release at low-shear stress regions of the vasculature in atherosclerosis. These results establish that KLF2 acts as a central transcriptional switch point between the quiescent and activated states of the adult endothelial cell.


Subject(s)
Cell Differentiation/genetics , Endothelium, Vascular/cytology , Gene Expression Regulation , Kruppel-Like Transcription Factors/physiology , Cell Movement , Cell Shape , Cells, Cultured , Gene Expression Profiling/methods , Genome, Human , Humans , Umbilical Veins , von Willebrand Factor/analysis , von Willebrand Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL