Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Signal ; 49: 30-38, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29793020

ABSTRACT

Endothelial cell activation by proinflammatory stimuli drives leukocyte recruitment through enhanced expression of counter-receptors such as vascular cell adhesion molecule-1 (VCAM-1). We previously demonstrated that activation of the receptor tyrosine kinase EphA2 with its ligand ephrin-A1 induces VCAM-1 expression. Here, we sought to characterize the proinflammatory signaling pathways involved. Analysis of over-represented transcription factors in ephrin-A1-induced genes identified multiple potential transcriptional regulators, including the Rel family members nuclear factor-κB (NF-κB/p65) and nuclear factor of activated T-cells (NFAT). While ephrin-A1 failed to induce endothelial NF-κB activation, NF-κB inhibitors prevented ephrin-A1-induced VCAM-1 expression, suggesting basal NF-κB activity is required. In contrast, ephrin-A1 induced a robust EphA2-dependent increase in NFAT activation, and mutation of the NF-κB/NFAT-binding sites in the VCAM-1 promoter blunted ephrin-A1-induced promoter activity. NFAT activation classically occurs through calcium-dependent calcineurin activation, and inhibiting NFAT signaling with calcineurin inhibitors (cyclosporine A, FK506) or direct NFAT inhibitors (A-285222) was sufficient to block ephrin-A1-induced VCAM-1 expression. Consistent with robust NFAT activation, ephrin-A1-induced an EphA2-dependent calcium influx in endothelial cells that was required for ephrin-A1-induced NFAT activation and VCAM-1 expression. This work provides the first data showing EphA2-dependent calcium influx and NFAT activation and identifies NFAT as a novel EphA2-dependent proinflammatory pathway in endothelial activation.


Subject(s)
Calcium/metabolism , Ephrin-A2/metabolism , NFATC Transcription Factors/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Calcium Signaling/drug effects , Cell Line , Cell Nucleus/metabolism , Cyclosporine/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ephrin-A2/antagonists & inhibitors , Ephrin-A2/genetics , Gene Expression Regulation/drug effects , Histone Deacetylases/metabolism , Humans , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , RNA Interference , RNA, Small Interfering/metabolism , Receptor, EphA2 , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Pharmacol Res ; 67(1): 42-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23098817

ABSTRACT

Despite significant advancements in treatment regimens, cardiovascular disease remains a worldwide leader of morbidity, mortality, and healthcare cost. A large percentage of cardiovascular disease is directly attributable to the process of atherosclerosis, a chronic inflammatory disease of the vessel wall. In the hunt for novel therapeutic targets in cardiovascular disease, neuronal guidance molecules are emerging as significant regulators of cardiovascular remodeling and inflammation. The Eph family of neuronal guidance molecules comprises the largest family of receptor tyrosine kinases in the mammalian genome. While best characterized in embryogenesis and carcinogenesis, Eph receptors and their ephrin ligands are becoming increasingly recognized as important players in chronic inflammatory diseases and immune function. Herein we discuss the current evidence for how Eph/ephrin interactions, particularly EphA2/ephrinA1 and EphB/ephrinB2, affect inflammation and cardiovascular disease.


Subject(s)
Ephrins/physiology , Receptors, Eph Family/physiology , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Endothelial Cells/physiology , Humans , Immunity , Inflammation/metabolism , Inflammation/physiopathology , Leukocytes/physiology
3.
Mol Biol Cell ; 24(3): 398-408, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23171552

ABSTRACT

Shear stress generated by distinct blood flow patterns modulates endothelial cell phenotype to spatially restrict atherosclerotic plaque development. Signaling through p21-activated kinase (PAK) mediates several of the deleterious effects of shear stress, including enhanced NF-κB activation and proinflammatory gene expression. Whereas shear stress activates PAK in endothelial cells on a fibronectin matrix, basement membrane proteins limit shear-induced PAK activation and inflammation through a protein kinase A-dependent pathway; however, the mechanisms underlying this regulation were unknown. We show that basement membrane proteins limit membrane recruitment of PAK2, the dominant isoform in endothelial cells, by blocking its interaction with the adaptor protein Nck. This uncoupling response requires protein kinase A-dependent nitric oxide production and subsequent PAK2 phosphorylation on Ser-20 in the Nck-binding domain. Of importance, shear stress does not stimulate nitric oxide production in endothelial cells on fibronectin, resulting in enhanced PAK activation, NF-κB phosphorylation, ICAM-1 expression, and monocyte adhesion. These data demonstrate that differential flow-induced nitric oxide production regulates matrix-specific PAK signaling and describe a novel mechanism of nitric oxide-dependent NF-κB inhibition.


Subject(s)
NF-kappa B/metabolism , Nitric Oxide/metabolism , p21-Activated Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Aorta/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Basement Membrane/metabolism , Biomechanical Phenomena , Cattle , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/pathology , Enzyme Activation , Extracellular Matrix/metabolism , Humans , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction , Stress, Physiological
4.
Int J Vasc Med ; 2012: 569654, 2012.
Article in English | MEDLINE | ID: mdl-22489274

ABSTRACT

A clear relationship between diabetes and cardiovascular disease has been established for decades. Despite this, the mechanisms by which diabetes contributes to plaque formation remain in question. Some of this confusion derives from studies in type 2 diabetics where multiple components of metabolic syndrome show proatherosclerotic effects independent of underlying diabetes. However, the hyperglycemia that defines the diabetic condition independently affects atherogenesis in cell culture systems, animal models, and human patients. Endothelial cell biology plays a central role in atherosclerotic plaque formation regulating vessel permeability, inflammation, and thrombosis. The current paper highlights the mechanisms by which hyperglycemia affects endothelial cell biology to promote plaque formation.

5.
Arterioscler Thromb Vasc Biol ; 32(3): 686-95, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22247258

ABSTRACT

OBJECTIVE: Endothelial cell activation results in altered cell-cell interactions with adjacent endothelial cells and with infiltrating leukocytes. Eph receptors and their ephrin ligands regulate cell-cell interactions during tissue remodeling, and multiple proinflammatory mediators induce endothelial EphA receptor and ephrinA ligand expression. Therefore, we sought to elucidate the role of EphA receptors and ephrinA ligands in endothelial cell activation and atherosclerosis. METHODS AND RESULTS: Quantitative reverse transcription-polymerase chain reaction screening for EphA/ephrinA expression in atherosclerosis-prone macrovascular endothelium identified EphA2, EphA4, and ephrinA1 as the dominant isoforms. Endothelial activation with oxidized low-density lipoprotein and proinflammatory cytokines induced EphA2 and ephrinA1 expression and sustained EphA2 activation, whereas EphA4 expression was unaffected. Atherosclerotic plaques from mice and humans showed enhanced EphA2 and ephrinA1 expression colocalizing in the endothelial cell layer. EphA2 activation with recombinant Fc-ephrinA1 induced proinflammatory gene expression (eg vascular cell adhesion molecule-1, E-selectin) and stimulated monocyte adhesion, whereas inhibiting EphA2 (small interfering RNA, pharmacological inhibitors) abrogated both ephrinA1-induced and oxidized low-density lipoprotein-induced vascular cell adhesion molecule-1 expression. CONCLUSION: The current data suggest that enhanced EphA2 signaling during endothelial cell activation perpetuates proinflammatory gene expression. Coupled with EphA2 expression in mouse and human atherosclerotic plaques, these data implicate EphA2 as a novel proinflammatory mediator and potential regulator of atherosclerotic plaque development.


Subject(s)
Atherosclerosis/immunology , Endothelial Cells/immunology , Inflammation Mediators/metabolism , Inflammation/immunology , Receptor, EphA2/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cell Adhesion , Cells, Cultured , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/antagonists & inhibitors , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Phenotype , RNA Interference , Receptor, EphA2/antagonists & inhibitors , Receptor, EphA2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transfection
6.
Circ Res ; 106(8): 1394-403, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20224042

ABSTRACT

RATIONALE: Atherosclerosis is initiated by blood flow patterns that activate inflammatory pathways in endothelial cells. Activation of inflammatory signaling by fluid shear stress is highly dependent on the composition of the subendothelial extracellular matrix. The basement membrane proteins laminin and collagen found in normal vessels suppress flow-induced p21 activated kinase (PAK) and nuclear factor (NF)-kappaB activation. By contrast, the provisional matrix proteins fibronectin and fibrinogen found in wounded or inflamed vessels support flow-induced PAK and NF-kappaB activation. PAK mediates both flow-induced permeability and matrix-specific activation of NF-kappaB. OBJECTIVE: To elucidate the mechanisms regulating matrix-specific PAK activation. METHODS AND RESULTS: We now show that matrix composition does not affect the upstream pathway by which flow activates PAK (integrin activation, Rac). Instead, basement membrane proteins enhance flow-induced protein kinase (PK)A activation, which suppresses PAK. Inhibiting PKA restored flow-induced PAK and NF-kappaB activation in cells on basement membrane proteins, whereas stimulating PKA inhibited flow-induced activation of inflammatory signaling in cells on fibronectin. PKA suppressed inflammatory signaling through PAK inhibition. Activating PKA by injection of the prostacyclin analog iloprost reduced PAK activation and inflammatory gene expression at sites of disturbed flow in vivo, whereas inhibiting PKA by PKA inhibitor (PKI) injection enhanced PAK activation and inflammatory gene expression. Inhibiting PAK prevented the enhancement of inflammatory gene expression by PKI. CONCLUSIONS: Basement membrane proteins inhibit inflammatory signaling in endothelial cells via PKA-dependent inhibition of PAK.


Subject(s)
Basement Membrane/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/enzymology , Inflammation/enzymology , Mechanotransduction, Cellular , p21-Activated Kinases/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Cattle , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Endothelial Cells/drug effects , Enzyme Activation , Enzyme Activators/administration & dosage , Humans , Iloprost/administration & dosage , Inflammation/drug therapy , Inflammation/physiopathology , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Integrins/metabolism , Male , Mechanotransduction, Cellular/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Pulsatile Flow , Regional Blood Flow , Stress, Mechanical , Time Factors , Transfection , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...