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










Publication year range
1.
Cardiovasc Res ; 113(2): 234-246, 2017 02.
Article in English | MEDLINE | ID: mdl-28088753

ABSTRACT

AIMS: There are conflicting reports on the role of reactive oxygen species (ROS) i.e. beneficial vs. harmful, in vascular endothelium. Here, we aim to examine whether duration of exposure to ROS and/or subcellular ROS levels are responsible for the apparently paradoxical effects of oxidants on endothelium. METHODS AND RESULTS: We have recently generated binary (Tet-ON/OFF) conditional transgenic mice (Tet-Nox2:VE-Cad-tTA) that can induce 1.8 ± 0.42-fold increase in NADPH oxidase (NOX)-derived ROS specifically in vascular endothelium upon withdrawal of tetracycline from the drinking water. Animals were divided in two groups: one exposed to high endogenous ROS levels for 8 weeks (short-term) and the other for 20 weeks (long-term). Using endothelial cells (EC) isolated from mouse hearts (MHEC), we demonstrate that both short-term and long-term increase in NOX-ROS induced AMPK-mediated activation of eNOS. Interestingly, although endothelium-dependent nitric oxide (NO)-mediated coronary vasodilation was significantly increased after short-term increase in NOX-ROS, coronary vasodilation was drastically reduced after long-term increase in ROS. We also show that short-term ROS increase induced proliferation in EC and angiogenic sprouting in the aorta. In contrast, long-term increase in cytosolic ROS resulted in nitrotyrosine-mediated inactivation of mitochondrial (mito) antioxidant MnSOD, increase in mito-ROS, loss of mitochondrial membrane potential (Δψm), decreased EC proliferation and angiogenesis. CONCLUSION: The findings suggest that NOX-derived ROS results in increased mito-ROS. Whereas short-term increase in mito-ROS was counteracted by MnSOD, long-term increase in ROS resulted in nitrotyrosine-mediated inactivation of MnSOD, leading to unchecked increase in mito-ROS and loss of Δψm followed by inhibition of endothelial function and proliferation.


Subject(s)
Coronary Vessels/enzymology , Endothelial Cells/enzymology , Mitochondria/enzymology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , AMP-Activated Protein Kinases/metabolism , Angiogenesis Inducing Agents/pharmacology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation , Cells, Cultured , Coronary Vessels/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Enzyme Activation , Genotype , In Vitro Techniques , Membrane Potential, Mitochondrial , Mice, Transgenic , Mitochondria/drug effects , NADPH Oxidases/genetics , Neovascularization, Physiologic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , Phenotype , Phosphorylation , Signal Transduction , Superoxide Dismutase/metabolism , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vasodilation , Vasodilator Agents/pharmacology
2.
Circ Res ; 115(2): 238-251, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24874427

ABSTRACT

RATIONALE: Forkhead box-O transcription factors (FoxOs) transduce a wide range of extracellular signals, resulting in changes in cell survival, cell cycle progression, and several cell type-specific responses. FoxO1 is expressed in many cell types, including endothelial cells (ECs). Previous studies have shown that Foxo1 knockout in mice results in embryonic lethality at E11 because of impaired vascular development. In contrast, somatic deletion of Foxo1 is associated with hyperproliferation of ECs. Thus, the precise role of FoxO1 in the endothelium remains enigmatic. OBJECTIVE: To determine the effect of endothelial-specific knockout and overexpression of FoxO1 on vascular homeostasis. METHODS AND RESULTS: We show that EC-specific disruption of Foxo1 in mice phenocopies the full knockout. Although endothelial expression of FoxO1 rescued otherwise Foxo1-null animals, overexpression of constitutively active FoxO1 resulted in increased EC size, occlusion of capillaries, elevated peripheral resistance, heart failure, and death. Knockdown of FoxO1 in ECs resulted in marked inhibition of basal and vascular endothelial growth factor-induced Akt-mammalian target of rapamycin complex 1 (mTORC1) signaling. CONCLUSIONS: Our findings suggest that in mice, endothelial expression of FoxO1 is both necessary and sufficient for embryonic development. Moreover, FoxO1-mediated feedback activation of Akt maintains growth factor responsive Akt/mTORC1 activity within a homeostatic range.


Subject(s)
Endothelial Cells/metabolism , Forkhead Transcription Factors/physiology , Heart Failure/genetics , Multiprotein Complexes/physiology , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins c-akt/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Enzyme Induction , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Heart Failure/physiopathology , Homeostasis , Human Umbilical Vein Endothelial Cells , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, Transgenic , Neovascularization, Physiologic/genetics , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Organ Specificity , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins , Signal Transduction/physiology , Yolk Sac/blood supply
3.
Aging (Albany NY) ; 5(7): 515-30, 2013 Jul.
Article in English | MEDLINE | ID: mdl-24018842

ABSTRACT

Age-associated decline in cardiovascular function is believed to occur from the deleterious effects of reactive oxygen species (ROS). However, failure of recent clinical trials using antioxidants in patients with cardiovascular disease, and the recent findings showing paradoxical role for NADPH oxidase-derived ROS in endothelial function challenge this long-held notion against ROS. Here, we examine the effects of endothelium-specific conditional increase in ROS on coronary endothelial function. We have generated a novel binary (Tet-ON/OFF) conditional transgenic mouse (Tet-Nox2:VE-Cad-tTA) that induces endothelial cell (EC)-specific overexpression of Nox2/gp91 (NADPH oxidase) and 1.8?0.42-fold increase in EC-ROS upon tetracycline withdrawal (Tet-OFF). We examined ROS effects on EC signaling and function. First, we demonstrate that endothelium-dependent coronary vasodilation was significantly improved in Tet-OFF Nox2 compared to Tet-ON (control) littermates. Using EC isolated from mouse heart, we show that endogenous ROS increased eNOS activation and nitric oxide (NO) synthesis through activation of the survival kinase AMPK. Coronary vasodilation in Tet-OFF Nox2 animals was CaMKK?-AMPK-dependent. Finally, we demonstrate that AMPK activation induced autophagy and thus, protected ECs from oxidant-induced cell death. Together, these findings suggest that increased ROS levels, often associated with cardiovascular conditions in advanced age, play a protective role in endothelial homeostasis by inducing AMPK-eNOS axis.


Subject(s)
Adenylate Kinase/metabolism , Aging/physiology , Coronary Vessels/physiology , Endothelium, Vascular/physiology , Oxidative Stress , Adenylate Kinase/genetics , Animals , Autophagy , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression Regulation, Enzymologic , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
4.
J Exp Med ; 209(7): 1363-77, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22689825

ABSTRACT

Regulation of vascular endothelial (VE) growth factor (VEGF)-induced permeability is critical in physiological and pathological processes. We show that tyrosine phosphorylation of VEGF receptor 2 (VEGFR2) at Y951 facilitates binding of VEGFR2 to the Rous sarcoma (Src) homology 2-domain of T cell-specific adaptor (TSAd), which in turn regulates VEGF-induced activation of the c-Src tyrosine kinase and vascular permeability. c-Src was activated in vivo and in vitro in a VEGF/TSAd-dependent manner, and was regulated via increased phosphorylation at pY418 and reduced phosphorylation at pY527. Tsad silencing blocked VEGF-induced c-Src activation, but did not affect pathways involving phospholipase Cγ, extracellular regulated kinase, and endothelial nitric oxide. VEGF-induced rearrangement of VE-cadherin-positive junctions in endothelial cells isolated from mouse lungs, or in mouse cremaster vessels, was dependent on TSAd expression, and TSAd formed a complex with VE-cadherin, VEGFR2, and c-Src at endothelial junctions. Vessels in tsad(-/-) mice showed undisturbed flow and pressure, but impaired VEGF-induced permeability, as measured by extravasation of Evans blue, dextran, and microspheres in the skin and the trachea. Histamine-induced extravasation was not affected by TSAd deficiency. We conclude that TSAd is required for VEGF-induced, c-Src-mediated regulation of endothelial cell junctions and for vascular permeability.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Capillary Permeability/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD/metabolism , Blotting, Western , Cadherins/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extravasation of Diagnostic and Therapeutic Materials/etiology , Female , Fluorescein-5-isothiocyanate/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microspheres , Phosphorylation/drug effects , Protein Binding , RNA Interference , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology
5.
PLoS One ; 6(12): e28454, 2011.
Article in English | MEDLINE | ID: mdl-22145046

ABSTRACT

BACKGROUND: ADPH oxidase-derived reactive oxygen species (ROS) play important roles in redox homeostasis and signal transduction in endothelial cells (ECs). We previously demonstrated that c-Src plays a key role in VEGF-induced, ROS-dependent selective activation of PI3K-Akt but not PLCγ-1-ERK1/2 signaling pathways. The aim of the present study was to understand how VEGFR-2-c-Src signaling axis 'senses' NADPH oxidase-derived ROS levels and couples VEGF activation of c-Src to the redox state of ECs. METHODOLOGY/PRINCIPAL FINDINGS: Using biotinylated probe that detects oxidation of cysteine thiol (cys-OH) in intracellular proteins, we demonstrate that VEGF induced oxidative modification in c-Src and VEGFR-2, and that reduction in ROS levels using siRNA against p47(phox) subunit of Rac1-dependent NADPH oxidase inhibited this phenomenon. Co-immunoprecipitation studies using human coronary artery ECs (HCAEC) showed that VEGF-induced ROS-dependent interaction between VEGFR-2 and c-Src correlated with their thiol oxidation status. Immunofluorescence studies using antibodies against internalized VEGFR-2 and c-Src demonstrated that VEGF-induced subcellular co-localization of these tyrosine kinases were also dependent on NADPH oxidsase-derived ROS. CONCLUSION/SIGNIFICANCE: These results demonstrate that VEGF induces cysteine oxidation in VEGFR-2 and c-Src in an NADPH oxidase-derived ROS-dependent manner, suggesting that VEGFR-2 and c-Src can 'sense' redox levels in ECs. The data also suggest that thiol oxidation status of VEGFR-2 and c-Src correlates with their ability to physically interact with each other and c-Src activation. Taken together, these findings suggest that prior to activating downstream c-Src-PI3K-Akt signaling pathway, VEGFR-2-c-Src axis requires an NADPH oxidase-derived ROS threshold in ECs.


Subject(s)
Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Genes, src/physiology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Blotting, Western , Cells, Cultured , Coronary Vessels/cytology , Endothelium, Vascular/cytology , Fluorescent Antibody Technique , Humans , Immunoprecipitation , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Oxidation-Reduction , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Signal Transduction
6.
Arterioscler Thromb Vasc Biol ; 30(9): 1703-10, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20702812

ABSTRACT

OBJECTIVE: To determine the functional significance of physiological reactive oxygen species (ROS) levels in endothelium-dependent nitric oxide (NO)-mediated coronary vasodilatation. METHODS AND RESULTS: Endothelium-derived NO is important in regulating coronary vascular tone. Excess ROS have been shown to reduce NO bioavailability, resulting in endothelial dysfunction and coronary diseases. NADPH oxidase is a major source of ROS in endothelial cells (ECs). By using lucigenin-based superoxide production and dichlorfluorescein diacetate (DCFH-DA) fluorescence-activated cell sorter assays, we found that mouse heart ECs from NADPH oxidase-knockdown (p47(phox-/-)) animals have reduced NADPH oxidase activity (>40%) and ROS levels (>30%) compared with wild-type mouse heart ECs. Surprisingly, a reduction in ROS did not improve coronary vasomotion; rather, endothelium-dependent vascular endothelial growth factor-mediated coronary vasodilatation was reduced by greater than 50% in p47(phox-/-) animals. Western blots and L-citrulline assays showed a significant reduction in Akt/protein kinase B (PKB) and endothelial NO synthase phosphorylation and NO synthesis, respectively, in p47(phox-/-) coronary vessels and mouse heart ECs. Adenoviral expression of constitutively active endothelial NO synthase restored vascular endothelial growth factor-mediated coronary vasodilatation in p47(phox-/-) animals. CONCLUSIONS: Endothelium-dependent vascular endothelial growth factor regulation of coronary vascular tone may require NADPH oxidase-derived ROS to activate phosphatidylinositol 3-kinase-Akt-endothelial NO synthase axis.


Subject(s)
Coronary Vessels/enzymology , Endothelium, Vascular/enzymology , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Vasodilation , Adenoviridae/genetics , Animals , Coronary Vessels/drug effects , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Enzyme Activation , Free Radical Scavengers/pharmacology , Genetic Vectors , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Transduction, Genetic , Vascular Endothelial Growth Factor A/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
7.
FEBS Lett ; 584(14): 3131-6, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20621840

ABSTRACT

The aim of the present study is to determine the role of intracellular Ca(2+) in VEGF signaling. We demonstrate that reduction in Ca(2+) by chelating compound BAPTA-AM or by IP(3)-endoplasmic reticulum blocker 2-APB selectively inhibited VEGF-induced activation of c-Src-PI3K-Akt but not ERK1/2 in human coronary artery endothelial cells (HCAEC). We also show that the selective inhibitory effects of NADPH oxidase knockdown on VEGF-mediated activation of c-Src-PI3K-Akt signaling and cell proliferation in HCAEC can be reversed by increase in intracellular Ca(2+). These results suggest an essential role for Ca(2+) in redox-dependent selective activation of c-Src-PI3K-Akt and endothelial cell proliferation.


Subject(s)
NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Calcium/metabolism , Calcium/pharmacology , Coronary Vessels/metabolism , Cytoplasm/metabolism , Egtazic Acid/analogs & derivatives , Endoplasmic Reticulum/metabolism , Endothelial Cells/metabolism , Humans , Ions/metabolism , Ions/pharmacology , NADPH Oxidases/pharmacology , Nitric Oxide Synthase Type III , Oxidation-Reduction , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/pharmacology , Rosiglitazone , Signal Transduction/physiology , Thiazolidinediones , Vascular Endothelial Growth Factors/metabolism , Vascular Endothelial Growth Factors/pharmacology
8.
Arterioscler Thromb Vasc Biol ; 28(11): 2042-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18787186

ABSTRACT

OBJECTIVE: Recently, we reported that the forkhead transcription factor, FKHR/FOXO1, is required for vascular endothelial growth factor (VEGF)-mediated upregulation of a number of genes in endothelial cells. Here, we tested the hypothesis that hepatocyte growth factor (HGF), a potent activator of PI3K-Akt in endothelial cells, is capable of depleting the nucleus of FKHR/FOXO1 and thus inhibiting VEGF induction of this class of genes. METHODS AND RESULTS: Incubation of human coronary artery endothelial cells with HGF induced prolonged PI3K/Akt-dependent phosphorylation and nuclear exclusion of FKHR/FOXO1. HGF-mediated inhibition of FKHR/FOXO1 activity resulted in secondary attenuation of VEGF-induced expression of FKHR/FOXO1-dependent genes including vascular cell adhesion molecule-1, manganese superoxide dismutase, endothelial specific molecule-1, CBP/p300 interacting transactivator with ED-rich tail-2, bone morphogenetic protein-2, matrix metalloproteinase (MMP)-10, and MGC5618. At a functional level, preincubation of HGF resulted in inhibition of VEGF-induced vascular cell adhesion molecule (VCAM)-1-mediated monocyte adhesion to endothelial cells. HGF-mediated inhibition of VEGF-inducible VCAM-1 expression and monocyte adhesion was reversed by overexpression of constitutively active phosphorylation-resistant triple mutant (TM)-FKHR. CONCLUSIONS: These findings suggest that physiological agonists of PI3K-Akt signaling pathway may modulate VEGF-FKHR/FOXO1-dependent gene expression in endothelial cells. The data underscore the importance of the "set point" of the endothelial cell when considering mechanisms of signal transduction.


Subject(s)
Endothelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Hepatocyte Growth Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Active Transport, Cell Nucleus , Cell Adhesion , Cells, Cultured , Endothelial Cells/enzymology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Humans , Monocytes/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Time Factors , Transduction, Genetic , Up-Regulation , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
9.
Endothelium ; 15(3): 143-8, 2008.
Article in English | MEDLINE | ID: mdl-18568955

ABSTRACT

Thrombomodulin (TM) is a cell surface anticoagulant glycoprotein that plays a key role in the protein C pathway. TM expression in endothelial cells may be modulated by a variety of extracellular signals. Most notably, TM has been shown to be downregulated by inflammatory mediators, such as tumor necrosis factor-alpha and lipopolysaccharide. The objective of this study was to determine the effect of thrombin on TM expression and activity. Thrombin resulted in reduced TM in primary cultures of human endothelial cells by approximately 40% at the level of mRNA, protein, and activity. These effects were blocked by the thrombin inhibitor hirudin. These results suggest that activation of the coagulation cascade may result in a positive-feedback loop consisting of thrombin-mediated repression of TM-dependent protein C activation.


Subject(s)
Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Thrombin/pharmacology , Thrombomodulin/metabolism , Cells, Cultured , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Hirudins/pharmacology , Humans , Protease Inhibitors/pharmacology , RNA, Messenger/metabolism , Thrombin/antagonists & inhibitors , Thrombomodulin/physiology , Time Factors , Umbilical Veins/cytology
10.
Biomed Pharmacother ; 62(6): 395-400, 2008.
Article in English | MEDLINE | ID: mdl-18406566

ABSTRACT

Hyperhomocysteinemia is an independent risk factor for the development of atherosclerosis, as well as for arterial and venous thrombosis. However, the mechanisms through which elevated circulating levels of homocysteine cause vascular injury and promote thrombosis remain unclear. Here, we tested the hypothesis that homocysteine (Hcy) sensitizes endothelial cells to the effect of inflammatory mediators. Human umbilical vein endothelial cells (HUVEC) were incubated with Hcy 1.0 mM for varying time points, and then treated in the absence or presence of 1.5 U/ml thrombin or 10 mg/ml lipopolysaccharide (LPS). Hcy alone had no effect on the expression of vascular cell adhesion molecule (VCAM)-1. However, Hcy enhanced thrombin- and LPS-mediated induction of VCAM-1 mRNA and protein levels. Consistent with these results, pretreatment of HUVEC with Hcy resulted in a two-fold increase in LSP-mediated induction of leukocyte adhesion. The latter effect was significantly inhibited by anti-VCAM-1 antibodies. Together, these findings suggest that Hcy sensitizes HUVEC to the effect of inflammatory mediators thrombin and LPS, at least in part through VCAM-1 expression and function.


Subject(s)
Gene Expression Regulation/drug effects , Homocysteine/physiology , Hyperhomocysteinemia/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Atherosclerosis/etiology , Cell Line , Endothelium, Vascular/metabolism , Humans , Hyperhomocysteinemia/complications , Lipopolysaccharides/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Risk Factors , Thrombin/metabolism , Thrombosis/etiology , Umbilical Veins/metabolism , Vascular Cell Adhesion Molecule-1/genetics
11.
J Biol Chem ; 282(48): 35373-85, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17908694

ABSTRACT

Vascular endothelial growth factor (VEGF) and reactive oxygen species (ROS) play critical roles in vascular physiology and pathophysiology. We have demonstrated previously that NADPH oxidase-derived ROS are required for VEGF-mediated migration and proliferation of endothelial cells. The goal of this study was to determine the extent to which VEGF signaling is coupled to NADPH oxidase activity. Human umbilical vein endothelial cells and/or human coronary artery endothelial cells were transfected with short interfering RNA against the p47(phox) subunit of NADPH oxidase, treated in the absence or presence of VEGF, and assayed for signaling, gene expression, and function. We show that NADPH oxidase activity is required for VEGF activation of phosphoinositide 3-kinase-Akt-forkhead, and p38 MAPK, but not ERK1/2 or JNK. The permissive role of NADPH oxidase on phosphoinositide 3-kinase-Akt-forkhead signaling is mediated at post-VEGF receptor levels and involves the nonreceptor tyrosine kinase Src. DNA microarrays revealed the existence of two distinct classes of VEGF-responsive genes, one that is ROS-dependent and another that is independent of ROS levels. VEGF-induced, thrombomodulin-dependent activation of protein C was dependent on NADPH oxidase activity, whereas VEGF-induced decay-accelerating factor-mediated protection of endothelial cells against complement-mediated lysis was not. Taken together, these findings suggest that NADPH oxidase-derived ROS selectively modulate some but not all the effects of VEGF on endothelial cell phenotypes.


Subject(s)
Gene Expression Regulation , NADPH Oxidases/metabolism , Oxidation-Reduction , Vascular Endothelial Growth Factor A/metabolism , Cells, Cultured , Humans , Models, Biological , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein C/metabolism , RNA, Small Interfering/metabolism , Reactive Oxygen Species , Signal Transduction , Thrombomodulin/metabolism , Time Factors
12.
J Biol Chem ; 281(46): 35544-53, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-16980307

ABSTRACT

Recently, we have shown that transient phosphorylation and inhibition of the pro-apoptotic transcription factor, forkhead, by vascular endothelial growth factor (VEGF) is essential for endothelial cell (EC) survival and proliferation. The goal of the present study was to determine whether forkhead (FKHR) also plays a positive role in agonist-mediated gene induction. Human coronary artery ECs were transduced with adenovirus overexpressing constitutively active phosphorylation-resistant triple mutant FKHR or transfected with small interference RNA (siRNA) against FKHR. The cells were then treated in the absence or presence of VEGF and assayed for gene expression using quantitative real-time PCR and Northern blots analyses. The data revealed a novel set of VEGF-responsive genes that require FKHR activity for optimal expression in ECs, including bone morphogenic protein 2, cbp/p300-interacting transactivator 2, decay accelerating factor (DAF), vascular cell adhesion molecule-1 (VCAM-1), manganese superoxide dismutase, endothelial-specific molecule-1, RING1 and YY1-binding protein, and matrix metalloproteinase-10. Consistent with a positive role for FKHR in mediating VEGF induction of DAF and VCAM-1 mRNA, siRNA against FKHR attenuated the effect of VEGF on complement-mediated EC lysis and monocyte adhesion, respectively. VEGF induction of the forkhead-dependent genes was down-regulated by the NF-kappaB inhibitor, constitutively active Ad-IkappaB, and in some cases by the nuclear factor of activated T-cells (NF-AT) inhibitor, cyclosporin. Together, these findings suggest that the VEGF-forkhead signaling axis plays an important functional role in ECs beyond the regulation of cell survival/apoptosis and cell cycle.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation/physiology , Vascular Endothelial Growth Factor A/metabolism , Adenoviridae , Cells, Cultured , Coronary Vessels/cytology , Endothelial Cells/metabolism , Forkhead Transcription Factors/genetics , Gene Deletion , Humans , RNA, Small Interfering , Transcriptional Activation
13.
Microvasc Res ; 72(3): 136-45, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16956626

ABSTRACT

Endothelial cell phenotypes are differentially regulated between different sites of the vascular tree. We tested the hypothesis that endocan, a novel soluble dermatan sulfate proteoglycan, is differentially expressed in the intact endothelium and that site-specific expression is mediated by signals in the local microenvironment. Using a combination of Northern blot analyses, Taqman RT-PCR, and in situ hybridizations, endocan was shown to be preferentially expressed in the endothelial lining of tumor xenografts, including human non-small cell lung cancer, rat glioma, and human renal cell carcinoma. In contrast, endocan mRNA was expressed at low levels in embryos between E4.5 and E18.5. Under in vitro conditions, endocan expression in human umbilical vein endothelial cells (HUVEC) was upregulated by tumor cell-conditioned medium, an effect that was inhibited by the addition of neutralizing antibody to vascular endothelial growth factor (VEGF). Moreover, treatment of HUVEC with VEGF resulted in a dose- and time-dependent increase in endocan mRNA. The results suggest that endocan is preferentially expressed in tumor endothelium in vivo and that its expression is regulated by tumor-derived factors.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Profiling , Neoplasms, Experimental/genetics , Proteoglycans/genetics , Animals , Blotting, Northern , Cell Line, Tumor , Cells, Cultured , Culture Media, Conditioned/pharmacology , Embryo, Mammalian/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation, Developmental/genetics , Humans , In Situ Hybridization , Mice , Mice, Inbred Strains , Neoplasm Proteins/genetics , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology , von Willebrand Factor/genetics
14.
FASEB J ; 19(13): 1914-6, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16172186

ABSTRACT

Endothelial cells are highly sensitive to changes in the extracellular milieu. Sepsis results in activation of inflammatory and coagulation pathways. We hypothesized that sepsis-associated mediators may alter the response capacity (so-called "set point") of endothelial cells. Human umbilical vein endothelial cells (HUVEC) were preincubated in the presence or absence of tumor necrosis factor (TNF)-alpha, lipopolysaccharide (LPS), hypoxia, hyperthermia, and/or high glucose; treated with or without thrombin for 4 h; and then processed for RNase protection assays of selected activation markers. Priming with TNF-alpha and LPS significantly inhibited thrombin-mediated induction of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, tissue factor, and E-selectin, but not platelet-derived growth factor-A or CD44. In electrophoretic mobility shift assays, thrombin-treated HUVEC demonstrated inducible binding of p65 NF-kappaB, an effect that was significantly blunted by pretreatment of cells with TNF-alpha and LPS. Consistent with these results, TNF-alpha and LPS attenuated the effect of thrombin on IkappaB phosphorylation, total cytoplasmic IkappaB, and nuclear translocation of p65 NF-kappaB. The inhibitory effect of TNF-alpha on thrombin signaling persisted for up to 24 h following removal of the cytokine. Taken together, these data suggest that inflammatory mediators prime endothelial cells to modulate subsequent thrombin response.


Subject(s)
Endothelial Cells/cytology , Endothelium, Vascular/cytology , Inflammation , Umbilical Veins/cytology , Active Transport, Cell Nucleus , Cells, Cultured , Culture Media/metabolism , Cytoplasm/metabolism , DNA Primers , Dose-Response Relationship, Drug , E-Selectin/metabolism , Endothelium, Vascular/pathology , Fever/metabolism , Humans , Hyaluronan Receptors/biosynthesis , Hypoxia/metabolism , I-kappa B Proteins/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides/metabolism , Models, Biological , Nitric Oxide Synthase/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Protein Transport , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/chemistry , Ribonucleases/metabolism , Signal Transduction , Thrombin/chemistry , Thrombin/metabolism , Time Factors , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
15.
J Biol Chem ; 280(33): 29864-73, 2005 Aug 19.
Article in English | MEDLINE | ID: mdl-15961397

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation and migration contribute significantly to atherosclerosis, postangioplasty restenosis, and transplant vasculopathy. Forkhead transcription factors belonging to the FoxO subfamily have been shown to inhibit growth and cell cycle progression in a variety of cell types. We hypothesized that forkhead proteins may play a role in VSMC biology. Under in vitro conditions, platelet-derived growth factor (PDGF)-BB, tumor necrosis factor-alpha, and insulin-like growth factor 1 stimulated phosphorylation of FoxO in human coronary artery smooth muscle cells via MEK1/2 and/or phosphatidylinositol 3-kinase-dependent signaling pathways. PDGF-BB, tumor necrosis factor-alpha, and insulin-like growth factor 1 treatment resulted in the nuclear exclusion of FoxO, whereas PDGF-BB alone down-regulated the FoxO target gene, p27(kip1), and enhanced cell survival and progression through the cell cycle. These effects were abrogated by overexpression of a constitutively active, phosphorylation-resistant mutant of the FoxO family member, TM-FKHRL1. The anti-proliferative effect of TM-FKHRL1 was partially reversed by small interfering RNA against p27(kip1). In a rat balloon carotid arterial injury model, adenovirus-mediated gene transfer of FKHRL1 caused an increase in the expression of p27(kip1) in the VSMC and inhibition of neointimal hyperplasia. These data suggest that FoxO activity inhibits VSMC proliferation and activation and that this signaling axis may represent a therapeutic target in vasculopathic disease states.


Subject(s)
Muscle, Smooth, Vascular/cytology , Nuclear Proteins/physiology , Transcription Factors/physiology , Tunica Intima/pathology , Active Transport, Cell Nucleus , Animals , Apoptosis , Becaplermin , Cell Cycle Proteins/genetics , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27 , DNA-Binding Proteins/metabolism , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors , Humans , Hyperplasia , Insulin-Like Growth Factor I/pharmacology , Nerve Tissue Proteins , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley , Signal Transduction , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/genetics
16.
J Biol Chem ; 279(48): 50537-54, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15448146

ABSTRACT

Activation and dysfunction of the endothelium underlie many vascular disorders including atherosclerosis, tumor growth, and inflammation. Endothelial cell activation is mediated by many different extra-cellular signals, which result in overlapping yet distinct patterns of gene expression. Here we show, in DNA microarray analyses, that vascular endothelial growth factor (VEGF) and thrombin result in dramatic and rapid upregulation of Down syndrome critical region (DSCR)-1 gene encoding exons 4-7, a negative feedback regulator of calcium-calcineurin-NF-AT signaling. VEGF- and thrombin-mediated induction of DSCR-1 involves the cooperative binding of NF-ATc and GATA-2/3 to neighboring consensus motifs in the upstream promoter. Constitutive expression of DSCR-1 in endothelial cells markedly impaired NF-ATc nuclear localization, proliferation, and tube formation. Under in vivo conditions, overexpression of DSCR-1 reduced vascular density in matrigel plugs and melanoma tumor growth in mice. Taken together, these findings support a model in which VEGF- and thrombin-mediated induction of endothelial cell proliferation triggers a negative feedback loop consisting of DSCR-1 gene induction and secondary inhibition of NF-AT signaling. As a natural brake in the angiogenic process, this negative pathway may lend itself to therapeutic manipulation in pathological states.


Subject(s)
Muscle Proteins/metabolism , Neovascularization, Pathologic/metabolism , Thrombin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Division/physiology , Chromosomes, Human, Pair 21 , DNA-Binding Proteins , Down Syndrome/metabolism , Endothelial Cells/metabolism , Exons , Humans , Intracellular Signaling Peptides and Proteins , Mice , Muscle Proteins/genetics , Promoter Regions, Genetic , Protein Isoforms , RNA, Messenger/metabolism
17.
J Biol Chem ; 279(42): 44030-8, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15308628

ABSTRACT

The mitochondrial antioxidant manganese superoxide dismutase (Mn-SOD) plays a critical cytoprotective role against oxidative stress. Vascular endothelial growth factor (VEGF) was shown previously to induce expression of Mn-SOD in endothelial cells by a NADPH oxidase-dependent mechanism. The goal of the current study was to determine the transcriptional mechanisms underlying this phenomenon. VEGF resulted in protein kinase C-dependent phosphorylation of IkappaB and subsequent translocation of p65 NF-kappaB into the nucleus. Overexpression of constitutively active IkappaB blocked VEGF stimulation of Mn-SOD. In transient transfection assays, VEGF increased Mn-SOD promoter activity, an effect that was dependent on a second intronic NF-kappaB consensus motif. In contrast, VEGF-mediated induction of Mn-SOD was enhanced by the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 and by dominant negative Akt and was decreased by constitutively active Akt. Overexpression of a constitutively active (phosphorylation-resistant) form of FKHRL1 (TMFKHRL1) resulted in increased Mn-SOD expression, suggesting that the negative effect of PI3K-Akt involves attenuation of forkhead activity. In co-transfection assays, the Mn-SOD promoter was transactivated by TMFKHRL1. Flavoenzyme inhibitor, diphenyleneiodonium (DPI), and antisense oligonucleotides against p47phox (AS-p47phox) inhibited VEGF stimulation of IkappaB/NF-kappaB and forkhead phosphorylation, supporting a role for NADPH oxidase activity in both signaling pathways. Like VEGF, hepatocyte growth factor (HGF) activated the PI3K-Akt-forkhead pathway. However, HGF-PI3K-Akt-forkhead signaling was insensitive to diphenyleneiodonium and AS-p47phox. Moreover, HGF failed to induce phosphorylation of IkappaB/NF-kappaB or nuclear translocation of NF-kappaB and had no effect on Mn-SOD expression. Together, these data suggest that VEGF is uniquely coupled to Mn-SOD expression through growth factor-specific reactive oxygen species (ROS)-sensitive positive (protein kinase C-NF-kappaB) and negative (PI3K-Akt-forkhead) signaling pathways.


Subject(s)
Endothelium, Vascular/enzymology , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Superoxide Dismutase/biosynthesis , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Cell Line , Cells, Cultured , Coronary Vessels , Endothelium, Vascular/cytology , Enzyme Induction/drug effects , Forkhead Transcription Factors , Humans , NADPH Oxidases/metabolism , NF-kappa B/antagonists & inhibitors , Oxidation-Reduction , Oxidative Stress/physiology , Phosphorylation , Umbilical Veins
18.
Arterioscler Thromb Vasc Biol ; 24(2): 294-300, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14656735

ABSTRACT

OBJECTIVE: Vascular endothelial growth factor (VEGF) is a potent angiogenic growth factor that promotes endothelial cell (EC) survival, migration, and permeability. The forkhead transcription factors FKHR, FKHRL1, and AFX are mammalian orthologues of DAF-16, a forkhead protein that controls longevity in Caenorhabditis elegans. In this study, we examined whether VEGF is coupled to phosphatidyl inositol 3-kinase (PI3K)/Akt/forkhead in ECs. METHODS AND RESULTS: We demonstrate that human ECs express members of the forkhead family (FKHR, FKHRL1, and AFX) and that VEGF modulates the phosphorylation, subcellular localization, and transcriptional activity of one or more of these isoforms by a PI3K/Akt signaling pathway. VEGF inhibited EC apoptosis, promoted DNA synthesis and the G(1)-to-S transition, and reduced expression of the cyclin-dependent kinase inhibitor p27(kip1). Each of these effects was blocked by the PI3K inhibitor LY294002 or by a phosphorylation-resistant mutant of FKHRL1, but not by wild-type FKHRL1. CONCLUSIONS: These results suggest that VEGF signaling in ECs is coupled to forkhead transcription factors through a PI3K/Akt-dependent pathway.


Subject(s)
DNA-Binding Proteins/physiology , Endothelium, Vascular/physiology , Nuclear Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Transcription Factors/physiology , Vascular Endothelial Growth Factor A/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Chromones/pharmacology , Coronary Vessels/chemistry , Coronary Vessels/cytology , Coronary Vessels/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Endothelium, Vascular/chemistry , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors , Gene Expression Regulation/physiology , Gene Transfer Techniques , Humans , Morpholines/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Pulmonary Artery/chemistry , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Signal Transduction/drug effects , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Umbilical Veins/chemistry
19.
J Biol Chem ; 279(2): 1513-25, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14551207

ABSTRACT

FLICE-inhibitory protein (FLIP) is a homolog of caspase-8 that lacks catalytic activity and has been shown to be important in protecting endothelial cells from apoptosis. The serine/threonine kinase Akt/PKB was recently reported to promote FLIP expression in endothelial and tumor cells. Here we examined the role of the forkhead transcription factor FOXO3a, a downstream target of Akt, in controlling FLIP regulation in endothelial cells. FOXO3a nuclear translocation was regulated by Akt in human umbilical vein endothelial cells. Transduction of a nonphosphorylatable, constitutively active mutant of FOXO3a (TM-FOXO3a) led to the down-regulation of FLIP levels. Transduction with TM-FOXO3a also increased caspase-8 activity and promoted apoptosis in endothelial cells. Conversely, transduction of a dominant-negative mutant of FOXO3a up-regulated FLIP levels and protected endothelial cells from apoptosis under serum deprivation conditions. Restoration of intracellular FLIP blocked caspase-8 activation and inhibited apoptosis in TM-FOXO3a-transduced cells. These data suggest that FOXO3a is a downstream target of Akt in endothelial cells that can promote apoptosis via FLIP down-regulation and activation of the extrinsic apoptotic pathway.


Subject(s)
Carrier Proteins/metabolism , Caspases/metabolism , DNA-Binding Proteins/physiology , Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Transcription Factors/physiology , Viral Proteins , Active Transport, Cell Nucleus , Anoikis , Apoptosis , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 8 , Caspase 9 , Cell Separation , Cell Survival , Cells, Cultured , DNA Fragmentation , DNA-Binding Proteins/chemistry , Down-Regulation , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Transcription Factors , Green Fluorescent Proteins , Growth Substances/metabolism , Humans , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Models, Biological , Mutation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt , RNA/metabolism , Ribonucleases/metabolism , Serpins/chemistry , Signal Transduction , Time Factors , Transcription Factors/chemistry , Umbilical Veins/cytology , Up-Regulation
20.
J Biol Chem ; 278(9): 6976-84, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12493764

ABSTRACT

We recently demonstrated that thrombin induces the expression of vascular adhesion molecule-1 (VCAM-1) in endothelial cells by an NF-kappaB- and GATA-dependent mechanism. In the present study, we describe the signaling pathways that mediate this response. Thrombin stimulation of the VCAM-1 gene and promoter in human umbilical vein endothelial cells was inhibited by preincubation with the phosphatidylinositol 3-kinase inhibitor, LY294002, the protein kinase C (PKC)-delta inhibitor, rottlerin, a PKC-zeta peptide inhibitor, or by overexpression of dominant negative (DN)-PKC-zeta. In electrophoretic mobility shift assays, thrombin-mediated induction of NF-kappaB p65 binding to two NF-kappaB motifs in the upstream promoter region of VCAM-1 was blocked by LY294002 and rottlerin, whereas the inducible binding of GATA-2 to a tandem GATA motif was inhibited by LY294002 and the PKC-zeta peptide inhibitor. In co-transfection assays, thrombin stimulation of a minimal promoter containing multimerized VCAM-1 NF-kappaB sites was inhibited by DN-PKC-delta but not DN-PKC-zeta. In contrast, thrombin-mediated transactivation of a minimal promoter containing tandem VCAM-1 GATA motifs was inhibited by DN-PKC-zeta but not DN-PKC-delta. Finally, thrombin failed to induce VCAM-1 expression in vascular smooth muscle cells. Taken together, these data suggest that the endothelial cell-specific effect of thrombin on VCAM-1 expression involves the coordinate activity of PKC-delta-NF-kappaB and PKC-zeta-GATA signaling pathways.


Subject(s)
NF-kappa B/metabolism , Protein Kinase C/metabolism , Thrombin/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism , Acetophenones/pharmacology , Amino Acid Motifs , Benzopyrans/pharmacology , Blotting, Northern , Cell Nucleus/metabolism , Cells, Cultured , Chromones/pharmacology , DNA-Binding Proteins/metabolism , Dimerization , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , GATA2 Transcription Factor , Genes, Dominant , Humans , Luciferases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Morpholines/pharmacology , Muscle, Smooth/cytology , Peptides/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Kinase C-delta , RNA/metabolism , RNA, Messenger/metabolism , Ribonucleases/metabolism , Signal Transduction , Thrombin/metabolism , Time Factors , Transcription Factor RelA , Transcription Factors/metabolism , Transcriptional Activation , Transfection , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...