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.
Circ Res ; 110(4): 560-8, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22267843

ABSTRACT

RATIONALE: Endothelial cells (EC) at regions exposed to disturbed flow (d-flow) are predisposed to inflammation and the subsequent development of atherosclerosis. We previously showed that thioredoxin interacting protein (TXNIP) was required for tumor necrosis factor-mediated expression of vascular cell adhesion molecule-1. OBJECTIVE: We sought to investigate the role of TXNIP in d-flow-induced cell adhesion molecule expression and leukocyte interaction with vessels, and the mechanisms by which TXNIP suppresses athero-protective gene expression. METHODS AND RESULTS: Using en face staining of mouse aorta, we found a dramatic increase of TXNIP in EC at sites exposed to d-flow as compared to steady flow. EC-specific TXNIP (EC-TXNIP) knockout mice showed significant decreases in vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 mRNA expression in the d-flow regions of mouse aorta. Intravital microscopy of mesenteric venules showed that leukocyte rolling time was decreased, whereas rolling velocity was increased significantly in EC-TXNIP knockout mice. In vitro experiments using a cutout flow chamber to generate varying flow patterns showed that increased TXNIP was required for d-flow-induced EC-monocyte adhesion. Furthermore, we found that the expression of Kruppel-like factor 2, a key anti-inflammatory transcription factor in EC, was inhibited by TXNIP. Luciferase and chromatin immunoprecipitation assays showed that TXNIP was present within a repressing complex on the Kruppel-like factor 2 promoter. CONCLUSIONS: These data demonstrate the essential role for TXNIP in mediating EC-leukocyte adhesion under d-flow, as well as define a novel mechanism by which TXNIP acts as a transcriptional corepressor to regulate Kruppel-like factor 2-dependent gene expression.


Subject(s)
Atherosclerosis/metabolism , Carrier Proteins/metabolism , Endothelial Cells/metabolism , Inflammation/metabolism , Kruppel-Like Transcription Factors/metabolism , Leukocyte Rolling , Thioredoxins/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Carrier Proteins/genetics , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Down-Regulation , Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , RNA Interference , Regional Blood Flow , Stress, Mechanical , Thioredoxins/genetics , Transfection , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
2.
Antioxid Redox Signal ; 16(6): 587-96, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-21929372

ABSTRACT

SIGNIFICANCE: The thioredoxin-interacting protein (TXNIP, also termed VDUP1 for vitamin D upregulated protein or TBP2 for thioredoxin-binding protein) was originally discovered by virtue of its strong regulation by vitamin D. Recently, TXNIP has been found to regulate the cellular reduction-oxidation (redox) state by binding to and inhibiting thioredoxin (TRX) in a redox-dependent fashion. RECENT ADVANCES: Studies of the Hcb-19 mouse, TXNIP nonsense mutated mouse, demonstrate redox-mediated roles in lipid and glucose metabolism, cardiac function, inflammation, and carcinogenesis. Exciting recent data indicate important roles for TXNIP in redox independent signaling. Specifically, sequence analysis revealed that TXNIP is a member of the classical visual/ß-arrestin superfamily, and is one of the six members of the arrestin domain-containing (ARRDC, or α-arrestin) family. CRITICAL ISSUES: Although the function of α-arrestins is not well known, recent studies suggest roles in endocytosis and protein ubiquitination through PPxY motifs in their C-terminal tails. Importantly, the ability of TXNIP to inhibit glucose uptake was found to be independent of TRX binding. Further investigation showed that several metabolic functions of TXNIP were due to the arrestin domains, thus further supporting the importance of redox independent functions of TXNIP. FUTURE DIRECTIONS: Since TXNIP transcription and protein stability are highly regulated by multiple tissue-specific stimuli, it appears that TXNIP should be a good therapeutic target for metabolic diseases.


Subject(s)
Carrier Proteins/metabolism , Thioredoxins/metabolism , Animals , Carrier Proteins/chemistry , Humans , Mice , Oxidation-Reduction , Thioredoxins/chemistry
3.
Arterioscler Thromb Vasc Biol ; 31(8): 1890-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21636804

ABSTRACT

OBJECTIVE: Thioredoxin-interacting protein (TXNIP) promotes inflammation in endothelial cells (EC) by binding to thioredoxin-1 (TRX1) in a redox-dependent manner. Formation of the TXNIP-TRX1 complex relieves inhibition of the apoptosis signal-regulating kinase 1-c-Jun N-terminal kinase-vascular cell adhesion molecule-1 pathway. Because TXNIP is an α-arrestin with numerous protein-protein interacting domains, we hypothesized that TXNIP-TRX1 trafficking should alter function of EC exposed to reactive oxygen species (ROS). METHODS AND RESULTS: In response to physiological levels of ROS (10 ng/mL tumor necrosis factor-α and 30 µmol/L H(2)O(2)), TXNIP-TRX1 translocated to the plasma membrane in human umbilical vein EC, with a peak at 30 minutes, as measured by immunofluorescence colocalization with vascular endothelial-cadherin, cell fractionation, and membrane sheet assay. TXNIP-mediated translocation of TRX1 to the membrane required TXNIP and TRX1 binding, as evidenced by inability of the ROS-insensitive TXNIP-Cys247Ser mutant to promote membrane localization. Vascular endothelial growth factor signaling required TXNIP, as shown by significant decreases in plasma membrane tyrosine phosphorylation and EC migration after TRX1 knockdown. Furthermore, TXNIP knockdown increased human umbilical vein EC apoptosis induced by tumor necrosis factor. Rescue with TXNIP-wild-type but not TXNIP-Cys247Ser prevented cell death. CONCLUSIONS: These findings suggest a novel role for the TXNIP-TRX1 complex to enable inflammation by promoting EC survival and vascular endothelial growth factor signaling under conditions of physiological oxidative stress.


Subject(s)
Carrier Proteins/metabolism , Thioredoxins/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Amino Acid Substitution , Base Sequence , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Movement/physiology , Cell Survival/physiology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Knockdown Techniques , Humans , Mutagenesis, Site-Directed , Oxidation-Reduction , Phosphorylation , Protein Interaction Domains and Motifs , Protein Transport/drug effects , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Thioredoxins/chemistry , Transcriptional Activation , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine/metabolism
4.
Arterioscler Thromb Vasc Biol ; 29(6): 895-901, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19359662

ABSTRACT

OBJECTIVE: Glucose 6-phosphate dehydrogenase (G6PD) maintains cellular NADPH levels, which are essential for cellular functions, such as vascular endothelial growth factor (VEGF)-induced angiogenesis. The molecular mechanisms regulating G6PD in angiogenesis are not fully understood. Because tyrosine phosphorylation is a key regulatory pathway for VEGF-mediated endothelial cell (EC) responses, we investigated tyrosine phosphorylation of G6PD and the role of the nonreceptor tyrosine kinase Src. METHODS AND RESULTS: VEGF increased G6PD membrane translocation as measured by a plasma membrane sheet assay, whereas tyrosine kinase inhibitor PP2 (4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo [3,4-d] pyramidine) decreased G6PD translocation by 100%. Furthermore, G6PD tyrosine phosphorylation and plasma membrane activity were increased by VEGF. In resting ECs, tyrosine kinase inhibitors PP2 and herbimycin A decreased basal G6PD activity by approximately 25%, whereas transfection with kinase inactive Src (KD-Src) decreased basal activity by approximately 30%. In mouse embryonic fibroblasts, Src-deficient (SYF) cells showed approximately 22% lower basal G6PD activity than Src-expressing S(+)YF cells. In addition, Src directly phosphorylated G6PD assayed by in vitro kinase assay. In ECs transfected with the G6PD mutants Y428F, Y507F (presumptive sites for Src-phosphorylation) or double mutant Y428F/Y507F, G6PD tyrosine phosphorylation was significantly decreased. Finally, G6PD tyrosine mutants (Y428F, Y507F, and Y428F/Y507F) decreased VEGF-mediated Akt phosphorylation and EC migration. CONCLUSIONS: G6PD activity and membrane association are regulated by Src-mediated tyrosine phosphorylation, which contributes to VEGF-mediated cellular responses in EC.


Subject(s)
Endothelial Cells/enzymology , Glucosephosphate Dehydrogenase/metabolism , Neovascularization, Physiologic , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , CSK Tyrosine-Protein Kinase , Cattle , Cell Membrane/enzymology , Cell Movement , Cells, Cultured , Endothelial Cells/drug effects , Fibroblasts/enzymology , Glucosephosphate Dehydrogenase/genetics , Humans , Mice , Mutation , Neovascularization, Physiologic/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Transport , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Tyrosine , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases
5.
Arterioscler Thromb Vasc Biol ; 28(5): 886-91, 2008 May.
Article in English | MEDLINE | ID: mdl-18292389

ABSTRACT

OBJECTIVE: The receptor tyrosine kinase Axl and its ligand Gas6 are involved in the development of renal diabetic disease. In vascular smooth muscle cells (VSMCs) Axl is activated by reactive oxygen species and stimulates migration and cell survival, suggesting a role for Axl in the vascular complications of diabetes. METHODS AND RESULTS: We investigated the effect of varying glucose concentration on Axl signaling in VSMCs. Glucose exerted powerful effects on Gas6-Axl signaling with greater activation of Akt and mTOR in low glucose, and greater activation of ERK1/2 in high glucose. Plasma membrane distribution and tyrosine phosphorylation of Axl were not affected by glucose. However, coimmunoprecipitation studies demonstrated that glucose changed the interaction of Axl with its binding partners. Specifically, binding of Axl to the p85 subunit of PI3-kinase was increased in low glucose, whereas binding to SHP-2 was increased in high glucose. Furthermore, Gas6-Axl induced migration was increased in high glucose, whereas Gas6-Axl mediated inhibition of apoptosis was greater in low glucose. CONCLUSIONS: This study demonstrates a role for glucose in altering Axl signaling through coupling to binding partners and suggests a mechanism by which Axl contributes to VSMC dysfunction in diabetes.


Subject(s)
Glucose/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Axl Receptor Tyrosine Kinase
6.
J Mol Med (Berl) ; 84(12): 997-1003, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17021908

ABSTRACT

The thioredoxin (TRX) system (TRX, TRX reductase, and NADPH) is a ubiquitous thiol oxidoreductase system that regulates cellular reduction/oxidation (redox) status. The impairment of cell redox state alters multiple cell pathways, which may contribute to the pathogenesis of cardiovascular disorders including hypertension, atherosclerosis, and heart failure. In this manuscript, we review the essential roles that TRX plays by limiting oxidative stress directly via antioxidant effects and indirectly by protein-protein interactions with key signaling molecules such as thioredoxin interacting protein (TXNIP). TRX and its endogenous regulators may represent important future targets to develop clinical therapies for diseases associated with oxidative stress.


Subject(s)
Cardiovascular System , Thioredoxins/metabolism , Animals , Humans , Models, Biological , Oxidation-Reduction , Thioredoxin-Disulfide Reductase/physiology
7.
Curr Atheroscler Rep ; 8(3): 240-4, 2006 May.
Article in English | MEDLINE | ID: mdl-16640961

ABSTRACT

Atherosclerotic lesions form preferentially at distinct sites in the arterial tree, especially at or near branch points, bifurcations, and curvatures where there is disturbed or oscillatory blood flow. In contrast, straight regions of the vasculature exhibit uniform laminar shear stress, which is atheroprotective. The ability of laminar flow to exert an anti-inflammatory effect on the endothelial cell lining of the blood vessel is revealed by preventing monocyte adhesion, proliferation, and apoptosis. Changes in endothelial cell gene expression in response to laminar shear stress reflect these changes in cell physiology with the demonstration that physiologic flow inhibits the expression of inflammatory genes. Thus, shear stress is critically important in regulating vascular physiology and pathobiology of the vessel wall via the modulation of endothelial cell gene expression.


Subject(s)
Atherosclerosis/metabolism , Gene Expression Regulation , Inflammation/genetics , Inflammation/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis , Atherosclerosis/pathology , Cell Adhesion , Cell Proliferation , Humans , Monocytes/metabolism , Oscillometry
8.
Atherosclerosis ; 183(1): 1-16, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15982657

ABSTRACT

The Rho family GTPases are regulatory molecules that link surface receptors to organisation of the actin cytoskeleton and play major roles in fundamental cellular processes. In the vasculature Rho signalling pathways are intimately involved in the regulation of endothelial barrier function, inflammation and transendothelial leukocyte migration, platelet activation, thrombosis and oxidative stress, as well as smooth muscle contraction, migration, proliferation and differentiation, and are thus implicated in many of the changes associated with atherogenesis. Indeed, it is believed that many of the beneficial, non-lipid lowering effects of statins occur as a result of their ability to inhibit Rho protein activation. Conversely, the Rho proteins can have beneficial effects on the vasculature, including the promotion of endothelial repair and the maintenance of SMC differentiation. Further identification of the mechanisms by which these proteins and their effectors act in the vasculature should lead to therapies that specifically target only the adverse effects of Rho signalling.


Subject(s)
Vascular Diseases/physiopathology , rho GTP-Binding Proteins/physiology , Atherosclerosis/enzymology , Atherosclerosis/etiology , Atherosclerosis/physiopathology , Atherosclerosis/prevention & control , Cell Differentiation , Cell Movement , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Enzyme Activation , Homeostasis , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypertension/enzymology , Hypertension/physiopathology , Models, Cardiovascular , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Oxidative Stress , Protein Prenylation/drug effects , Protein Processing, Post-Translational/drug effects , Signal Transduction , Vascular Diseases/enzymology , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/classification , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/physiology
9.
Cardiovasc Res ; 58(1): 222-30, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12667965

ABSTRACT

OBJECTIVE: Our previous studies showed that the pleiotropic cytokine leukaemia inhibitory factor (LIF) inhibits the de novo formation of experimental atherosclerotic lesions. The present study examined whether LIF also inhibits progression of pre-existing atheroma. METHODS: Balloon angioplasty was performed on the right carotid arteries of 18 rabbits immediately before placing animals on a cholesterol-enriched diet. After 4 weeks, at which time the intima:media ratio (I:M) was 0.99+/-0.12 (n=6), osmotic minipumps containing LIF (n=6) or saline control (n=6) were inserted into the peritoneal cavity of each of the remaining rabbits for a further 4 weeks. Arteries were then harvested for analysis. RESULTS: Continuous administration of LIF for the final 4 weeks of an 8-week cholesterol-enriched diet completely inhibited lesion progression in injured carotid arteries (I:M 1.05+/-0.16) compared with the saline-treated group at 8 weeks (1.62+/-0.13; P<0.05). Similarly in contralateral uninjured carotid arteries, LIF treatment prevented an increase in I:M from a baseline of 0.11+/-0.01 at 4 weeks to 0.15+/-0.02 at 8 weeks compared with 0.40+/-0.04 for the saline-treated group at 8 weeks (P<0.05). LIF reduced the number of macrophages in the neointima of uninjured arteries, but had no effect on the cellular composition of injured arteries. LIF treatment normalised smooth muscle-dependent vasoreactivity to phenylephrine and sodium nitroprusside in both injured and uninjured arteries. Expression and activity of inducible nitric oxide synthase (iNOS) were up-regulated in response to hypercholesterolemia with levels further increased following endothelial denudation. With LIF treatment, iNOS expression was increased in uninjured arteries but marginally reduced in injured arteries. LIF receptors were expressed in both uninjured and injured arteries, with LIF treatment having no significant effect on expression levels. CONCLUSION: LIF prevents progression of pre-formed atherosclerotic plaques, affecting lesion size and vascular reactivity. LIF treatment has differential effects within the artery wall, depending on the presence or absence of de-endothelialisation injury.


Subject(s)
Arteriosclerosis/drug therapy , Molecular Chaperones/therapeutic use , Proteins , Acetylcholine/pharmacology , Animals , Arteriosclerosis/metabolism , Blotting, Western/methods , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Disease Progression , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry/methods , In Vitro Techniques , Interleukin-6 , Leukemia Inhibitory Factor , Male , Models, Animal , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Rabbits , Serotonin/pharmacology , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...