Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters










Publication year range
1.
Matrix Biol ; 82: 71-85, 2019 09.
Article in English | MEDLINE | ID: mdl-30876926

ABSTRACT

Collagen fibrillogenesis and crosslinking have long been implicated in extracellular matrix (ECM)-dependent processes such as fibrosis and scarring. However, the extent to which matricellular proteins influence ECM protein production and fibrillar collagen crosslinking has yet to be determined. Here we show that thrombospondin 2 (TSP2), an anti-angiogenic matricellular protein, is an important modulator of ECM homeostasis. Specifically, through a fractionated quantitative proteomics approach, we show that loss of TSP2 leads to a unique ECM phenotype characterized by a significant decrease in fibrillar collagen, matricellular, and structural ECM protein production in the skin of TSP2 KO mice. Additionally, TSP2 KO skin displays decreased lysyl oxidase (LOX), which manifests as an increase in fibrillar collagen solubility and decreased levels of LOX-mediated fibrillar collagen crosslinking. We show that these changes are indirectly mediated by miR-29, a major regulator of ECM proteins and LOX, as miR-29 expression is increased in the TSP2 KO. Altogether, these findings indicate that TSP2 contributes to ECM production and assembly by regulating miR-29 and LOX.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , MicroRNAs/genetics , Protein-Lysine 6-Oxidase/metabolism , Thrombospondins/metabolism , Animals , Collagen/metabolism , Down-Regulation , Gene Knockout Techniques , Male , Mice , Proteomics , Thrombospondins/genetics
2.
J Thromb Haemost ; 7 Suppl 1: 35-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19630764

ABSTRACT

In the past decade, the importance of the vascular endothelium as a multifunctional regulator of vascular smooth muscle physiology and pathophysiology has been appreciated. Indeed, the endothelium responds to hemodynamic stimuli (pressure, shear stress and wall strain) and locally manufactured mediators (such as bradykinin, prostaglandins, angiotensin II and nitric oxide) that can influence blood flow, cell trafficking into tissue and angiogenesis. In this chapter, the importance of nitric oxide (NO) as a mediator of blood flow control, vascular permeability and angiogenesis will be discussed.


Subject(s)
Neovascularization, Physiologic , Nitric Oxide/physiology , Regional Blood Flow , Capillary Permeability , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans
3.
Am J Transplant ; 7(4): 769-78, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17391122

ABSTRACT

Akt is expected to be an effective target for the treatment of ischemia-reperfusion injury (I/R) due to its anti-apoptotic properties and its ability to activate the endothelial nitric oxide synthase (eNOS) enzyme. Therefore, this study was aimed to determine the efficacy of an active mutant of Akt (myr-Akt) to decrease I/R injury in a model of orthotopic liver transplantation in pigs. In addition, we analyzed the contribution of nitric oxide in the Akt-mediated effects by using an eNOS mutant (S1179DeNOS) that mimics the phosphorylation promoted by Akt in the eNOS sequence. Donors were treated with adenoviruses codifying for myr-Akt, S1179DeNOS or beta-galactosidase 24 h before liver harvesting. Then, liver grafts were orthotopically transplanted into their corresponding recipients. Levels of transaminases and lactate dehydrogenase (LDH) increased in all recipients after 24 h of transplant. However, transaminases and LDH levels were significantly lower in the myr-Akt group compared with vehicle. The percentage of apoptotic cells and the amount of activated-caspase 3 protein were also markedly reduced in myr-Akt-treated grafts after 4 days of liver transplant compared with vehicle and S1179DeNOS groups. In conclusion, myr-Akt gene therapy effectively exerts cytoprotection against hepatic I/R injury regardless of the Akt-dependent eNOS activation.


Subject(s)
Endothelial Cells/cytology , Endothelium, Vascular/physiology , Oncogene Protein v-akt/physiology , Animals , Aorta , Cattle , Cell Line , Cells, Cultured , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Hepatocytes/cytology , Hepatocytes/physiology , Humans , Kidney , Mutation , Oncogene Protein v-akt/genetics , Swine
4.
Gene Ther ; 13(18): 1342-50, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16642030

ABSTRACT

Gene delivery of angiogenic growth factors is a promising approach for the treatment of ischemic cardiovascular diseases. However, success of this new therapeutic principle is hindered by the lack of critical understanding as to how disease pathology affects the efficiency of gene delivery and/or the downstream signaling pathways of angiogenesis. Critical limb ischemia occurs in patients with advanced atherosclerosis often exhibiting deficiency in endothelial nitric oxide production. Similar to these patients, segmental femoral artery resection progresses into severe ischemic necrosis in mice deficient in endothelial nitric oxide synthase (ecNOS-KO) as well as in balb/c mice. We used these models to evaluate the influence of severe ischemia on transfection efficiency and duration of transgene expression in the skeletal muscle following plasmid injection in combination with electroporation. Subsequently, we also explored the potential therapeutic effect of the phosphomimetic mutant of ecNOS gene (NOS1177D) using optimized delivery parameters, and found significant benefit both in ecNOS-KO and balb/c mice. Our results indicate that NOS1177D gene delivery to the ischemic skeletal muscle can be efficient to reverse critical limb ischemia in pathological settings, which are refractory to treatments with a single growth factor, such as vascular endothelial growth factor.


Subject(s)
Genetic Therapy/methods , Ischemia/therapy , Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type III/genetics , Transfection/methods , Vascular Endothelial Growth Factor A/metabolism , Animals , Electroporation , Endothelium, Vascular/metabolism , Gene Expression , Genetic Vectors , Hindlimb , Humans , Ischemia/metabolism , Ischemia/pathology , Laser-Doppler Flowmetry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Neovascularization, Physiologic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/analysis , Nitric Oxide Synthase Type III/metabolism , Regional Blood Flow , Transgenes , Vasodilation
5.
Am J Physiol Gastrointest Liver Physiol ; 285(3): G652-60, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12829439

ABSTRACT

Diminished endothelial nitric oxide (NO) synthase (eNOS)-derived NO production from the hepatic vascular endothelium contributes to hepatic vasoconstriction in portal hypertension. The aim of this study was to examine the mechanism of this process by testing the influence of a constitutively active form of eNOS (S1179DeNOS) in both primary and propagated liver cells in vitro and in the sham and bile duct ligated (BDL) rat liver in vivo, using an adenoviral vector encoding green fluorescent protein (AdGFP) and S1179DeNOS (AdS1179DeNOS). AdS1179DeNOS transduction augmented basal and agonist-stimulated NO generation in nonparenchymal liver cells. Sham rats transduced in vivo with AdS1179DeNOS evidenced a decreased pressor response to incremental doses of the vasoconstrictor methoxamine compared with sham rats transduced with AdGFP. However, BDL rats transduced with AdS1179DeNOS did not display improved vasodilatory responses as evidenced by similar flow-dependent pressure increases to that observed in BDL rats transduced with AdGFP, despite similar levels of viral transgene expression. We next examined the influence of the eNOS inhibitory protein caveolin on S1179DeNOS dysfunction in cirrhotic liver. Immunogold electron microscopic analysis of caveolin in BDL liver demonstrated prominent expression not only in liver endothelial cells, but also in hepatic stellate cells. In vitro studies in the LX2 hepatic stellate cell line demonstrate that caveolin precipitates recombinant S1179DeNOS in LX2 cells, that recombinant S1179DeNOS coprecipitates caveolin, and that binding is enhanced in the presence of overexpression of caveolin. Furthermore, caveolin overexpression inhibits recombinant S1179DeNOS activity. These studies indicate that recombinant S1179DeNOS protein functions appropriately in normal liver cells and tissue but evidences dysfunction in the cirrhotic rat liver and that caveolin expression and inhibition in BDL nonparenchymal cells, including hepatic stellate cells, may account for this dysfunction.


Subject(s)
Caveolins/physiology , Nitric Oxide Synthase/metabolism , Animals , Bile Ducts , Caveolin 1 , Caveolins/metabolism , Cells, Cultured , Enzyme Activation/physiology , Gene Transfer Techniques , Ligation , Liver/cytology , Liver/metabolism , Male , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , Rats , Rats, Inbred F344 , Recombinant Proteins/metabolism , Transduction, Genetic , Vasodilation/genetics
6.
J Pharmacol Exp Ther ; 299(3): 818-24, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11714864

ABSTRACT

Endothelial nitric oxide synthase (eNOS) is important for cardiovascular homeostasis, vessel remodeling, and angiogenesis. Given the impact of endothelium- derived nitric oxide (NO) in vascular biology, much work in the past several years has focused on the control of NO synthesis by regulatory proteins that influence its function. Indeed calcium-activated calmodulin is important for regulation of NOS activity. Herein we discuss why other proteins, in addition to calmodulin, are necessary for eNOS regulation and summarize the biology of negative and positive regulators of eNOS function in vitro, in cells, and in blood vessels.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Nitric Oxide Synthase/metabolism , Animals , Caveolin 1 , Caveolins/metabolism , Enzyme Activation , HSP90 Heat-Shock Proteins/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III , Phosphorylation , Protein Processing, Post-Translational
7.
Proc Natl Acad Sci U S A ; 98(24): 14072-7, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11707586

ABSTRACT

The relative importance of lipid rafts vs. specialized rafts termed caveolae to influence signal transduction is not known. Here we show that in cells lacking caveolae, the dually acylated protein, endothelial nitric oxide synthase (eNOS), localizes to cholesterol-rich lipid raft domains of the plasma membrane. In these cells, expression of caveolin-1 (cav-1) stimulates caveolae biogenesis, promotes the interaction of cav-1 with eNOS, and the inhibition of NO release from cells. Interestingly, in cells where cav-1 does not drive caveolae assembly, despite equal levels of cav-1 and eNOS and localization of both proteins to raft domains of the plasmalemma, the physical interaction of eNOS with cav-1 is dramatically less resulting in less inhibition of NO release. Thus, cav-1 concentrated in caveolae, not in rafts, is in closer proximity to eNOS and is necessary for negative regulation of eNOS function, thereby providing the first clear example of spatial regulation of signaling in this organelle that is distinct from raft domains.


Subject(s)
Caveolae/metabolism , Caveolins/metabolism , Membrane Microdomains/metabolism , Nitric Oxide Synthase/metabolism , Signal Transduction/physiology , Animals , Caveolin 1 , Caveolins/genetics , Caveolins/physiology , Cells, Cultured , Cholesterol/metabolism , Humans , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type III , Rats , Rats, Inbred F344 , Thyroid Gland/cytology
8.
Mol Cell ; 8(3): 693-704, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11583630

ABSTRACT

The role of the protein kinase Akt in cell migration is incompletely understood. Here we show that sphingosine-1-phosphate (S1P)-induced endothelial cell migration requires the Akt-mediated phosphorylation of the G protein-coupled receptor (GPCR) EDG-1. Activated Akt binds to EDG-1 and phosphorylates the third intracellular loop at the T(236) residue. Transactivation of EDG-1 by Akt is not required for G(i)-dependent signaling but is indispensable for Rac activation, cortical actin assembly, and chemotaxis. Indeed, T236AEDG-1 mutant sequestered Akt and acted as a dominant-negative GPCR to inhibit S1P-induced Rac activation, chemotaxis, and angiogenesis. Transactivation of GPCRs by Akt may constitute a specificity switch to integrate rapid G protein-dependent signals into long-term cellular phenomena such as cell migration.


Subject(s)
Chemotaxis/physiology , Endothelium, Vascular/cytology , Immediate-Early Proteins/metabolism , Lysophospholipids , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled , Signal Transduction/physiology , Actins/metabolism , Animals , Cell Line , Endothelium, Vascular/drug effects , Enzyme Activation/physiology , Humans , Models, Biological , Neovascularization, Physiologic/physiology , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Receptors, Cell Surface , Receptors, Lysophospholipid , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 98(20): 11575-80, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11562476

ABSTRACT

One of the many biological functions of nitric oxide is the ability to protect cells from oxidative stress. To investigate the potential contribution of low steady state levels of nitric oxide generated by endothelial nitric oxide synthase (eNOS) and the mechanisms of protection against H(2)O(2), spontaneously transformed human ECV304 cells, which normally do not express eNOS, were stably transfected with a green fluorescent-tagged eNOS cDNA. The eNOS-transfected cells were found to be resistant to injury and delayed death following a 2-h exposure to H(2)O(2) (50-150 microM). Inhibition of nitric oxide synthesis abolished the protective effect against H(2)O(2) exposure. The ability of nitric oxide to protect cells depended on the presence of respiring mitochondria as ECV304+eNOS cells with diminished mitochondria respiration (rho(-)) are injured to the same extent as nontransfected ECV304 cells and recovery of mitochondrial respiration restores the ability of nitric oxide to protect against H(2)O(2)-induced death. Nitric oxide also found to have a profound effect in cell metabolism, because ECV304+eNOS cells had lower steady state levels of ATP and higher utilization of glucose via the glycolytic pathway than ECV304 cells. However, the protective effect of nitric oxide against H(2)O(2) exposure is not reproduced in ECV304 cells after treatment with azide and oligomycin suggesting that the dynamic regulation of respiration by nitric oxide represent a critical and unrecognized primary line of defense against oxidative stress.


Subject(s)
Cell Survival/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Oxidative Stress , Oxygen Consumption/drug effects , Adenosine Triphosphate/metabolism , Azides/pharmacology , Cell Line , Cells, Cultured , Glucosephosphate Dehydrogenase/metabolism , Humans , Hydrogen Peroxide/pharmacology , Kinetics , Luminescent Measurements , NADP/metabolism , Oligomycins/pharmacology
10.
J Biol Chem ; 276(32): 30359-65, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11387313

ABSTRACT

Vascular endothelial growth factor (VEGF) utilizes a phosphoinositide 3-kinase (PI 3-kinase)/Akt signaling pathway to protect endothelial cells from apoptotic death. Here we show that PI 3-kinase/Akt signaling promotes endothelial cell survival by inhibiting p38 mitogen-activated protein kinase (MAPK)-dependent apoptosis. Blockade of the PI 3-kinase or Akt pathways in conjunction with serum withdrawal stimulates p38-dependent apoptosis. Blockade of PI 3-kinase/Akt also led to enhanced VEGF activation of p38 and apoptosis. In this context, the pro-apoptotic effect of VEGF is attenuated by the p38 MAPK inhibitor SB203580. VEGF stimulation of endothelial cells or infection with an adenovirus expressing constitutively active Akt causes MEKK3 phosphorylation, which is associated with decreased MEKK3 kinase activity and down-regulation of MKK3/6 and p38 MAPK activation. Conversely, activation-deficient Akt decreases VEGF-stimulated MEKK3 phosphorylation and increases MKK/p38 activation. Activation of MKK3/6 is not dependent on Rac activation since dominant negative Rac does not decrease p38 activation triggered by inhibition of PI 3-kinase. Thus, cross-talk between the Akt and p38 MAPK pathways may regulate the level of cytoprotection versus apoptosis and is a new mechanism to explain the cytoprotective actions of Akt.


Subject(s)
Down-Regulation , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Lymphokines/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Apoptosis , Blotting, Western , Cattle , Cell Death , Cell Line , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , Imidazoles/pharmacology , Phosphatidylinositol 3-Kinases , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt , Pyridines/pharmacology , Signal Transduction , Time Factors , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , p38 Mitogen-Activated Protein Kinases
11.
Am J Pathol ; 158(5): 1757-65, 2001 May.
Article in English | MEDLINE | ID: mdl-11337373

ABSTRACT

The protective genes that mediate endothelial cell (EC) survival during angiogenesis have not been completely characterized. Here, we show that an antisense oligonucleotide to the apoptosis inhibitor survivin suppressed de novo expression of survivin in ECs by vascular endothelial cell growth factor (VEGF). In contrast, the survivin antisense oligonucleotide did not affect anti-apoptotic bcl-2 levels in endothelium. When assessed in cell death assays, antisense targeting of survivin abolished the anti-apoptotic function of VEGF against tumor necrosis factor-alpha- or ceramide-induced cell death, enhanced caspase-3 activity, promoted the generation of a approximately 17-kd active caspase-3 subunit, and increased cleavage of the caspase substrate, polyADP ribose polymerase. In contrast, the survivin antisense oligonucleotide had no effect on EC viability in the absence of VEGF. Antisense oligonucleotides to platelet-endothelial cell adhesion molecule-1 (PECAM-1, CD31), lymphocyte function-associated molecule-3 (LFA-3, CD58), or intercellular adhesion molecule-1 (ICAM-1, CD54) did not reduce the anti-apoptotic function of VEGF in endothelium. When tested on other angiogenic activities mediated by VEGF, survivin antisense treatment induced rapid regression of three-dimensional vascular capillary networks, but did not affect EC migration/chemotaxis. These data suggest that the anti-apoptotic properties of VEGF during angiogenesis are primarily mediated by the induced expression of survivin in ECS: Manipulation of this pathway may increase EC viability in compensatory angiogenesis or facilitate EC apoptosis and promote vascular regression during tumor angiogenesis.


Subject(s)
DNA, Antisense/pharmacology , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Lymphokines/pharmacology , Microtubule-Associated Proteins , Proteins/drug effects , Apoptosis/drug effects , Cell Movement/drug effects , Cells, Cultured , DNA/drug effects , DNA/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Gene Expression Regulation/drug effects , Humans , Inhibitor of Apoptosis Proteins , Neoplasm Proteins , Proteins/genetics , Proteins/metabolism , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survivin , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
J Biol Chem ; 276(28): 26674-9, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11331296

ABSTRACT

The 894G-->T polymorphism within exon 7 of the human endothelial nitric-oxide synthase (eNOS) gene codes for glutamate or aspartate, respectively, at residue 298 and has been associated with several diseases of cardiovascular origin. A recent report indicates that Asp(298)-eNOS (E298D) is cleaved intracellularly to 100- and 35-kDa fragments, suggesting a mechanism for reduced endothelial function. Here we have documented the precise cleavage site of the E298D variant as a unique aspartyl-prolyl (Asp(298)--Pro(299)) bond not seen in wild-type eNOS (Glu(298)). We show that E298D-eNOS, as isolated from cells and in vitro, is susceptible to acidic hydrolysis, and the 100-kDa fragment can be generated ex vivo by increasing temperature at low pH. Importantly, cleavage of E298D was eliminated using a sample buffer system designed to limit acidic hydrolysis of Asp--Pro bonds. These results argue against intracellular processing of E298D-eNOS and suggest that previously described fragmentation of E298D could be a product of sample preparation. We also found that eNOS turnover, NO production, and the susceptibility to cellular stress were not different in cells expressing WT versus E298D-eNOS. Finally, enzyme activities were identical for the respective recombinant enzymes. Thus, intracellular cleavage mechanisms are unlikely to account for associations between the exon 7 polymorphism and cardiovascular diseases.


Subject(s)
Nitric Oxide Synthase/genetics , Amino Acid Substitution , Aspartic Acid , Glutamic Acid , Humans , Hydrolysis , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase Type III , Protein Conformation , Structure-Activity Relationship
13.
Trends Mol Med ; 7(5): 189-91, 2001 May.
Article in English | MEDLINE | ID: mdl-11325618

ABSTRACT

Statin-based drugs are the mainstay of therapy for coronary artery disease. Recent insights into the cellular mechanisms of this class of drugs suggest that modulation of nitric oxide synthesis contributes to their beneficial actions. These effects are independent of their lipid-lowering effects and result in enhanced nitric oxide release.


Subject(s)
Anticholesteremic Agents/pharmacology , Endothelium, Vascular/drug effects , Hypolipidemic Agents/pharmacology , Lovastatin/pharmacology , Nitric Oxide Synthase/metabolism , Animals , Humans , Lipid Metabolism , Lovastatin/analogs & derivatives , Models, Biological , Nitric Oxide Synthase Type III , Oxidative Stress , Protein Processing, Post-Translational , Rats
14.
J Biol Chem ; 276(21): 17621-4, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11278264

ABSTRACT

The balance of nitric oxide (.NO) and superoxide anion (O(2)) plays an important role in vascular biology. The association of heat shock protein 90 (Hsp90) with endothelial nitric-oxide synthase (eNOS) is a critical step in the mechanisms by which eNOS generates.NO. As eNOS is capable of generating both.NO and O(2), we hypothesized that Hsp90 might also mediate eNOS-dependent O(2) production. To test this hypothesis, bovine coronary endothelial cells (BCEC) were pretreated with geldanamycin (GA, 10 microg/ml; 17.8 microm) and then stimulated with the calcium ionophore, (5 microm). GA significantly decreased -stimulated eNOS-dependent nitrite production (p < 0.001, n = 4) and significantly increased -stimulated eNOS-dependent O(2) production (p < 0.001, n = 8). increased phospho-eNOS(Ser-1179) levels by >1.6-fold over vehicle (V)-treated levels. Pretreatment with GA by itself or with increased phospho-eNOS levels. In unstimulated V-treated BCEC cultures low amounts of Hsp90 were found to associate with eNOS. Pretreatment with GA and/or increased the association of Hsp90 with eNOS. These data show that Hsp90 is essential for eNOS-dependent.NO production and that inhibition of ATP-dependent conformational changes in Hsp90 uncouples eNOS activity and increases eNOS-dependent O(2) production.


Subject(s)
Endothelium, Vascular/metabolism , HSP90 Heat-Shock Proteins/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Animals , Calcimycin/pharmacology , Cattle , Cells, Cultured , Ionophores/pharmacology , Nitric Oxide Synthase Type III
15.
J Biol Chem ; 276(22): 19672-7, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11278592

ABSTRACT

Sphingosine 1-phosphate (SPP) binds to members of the endothelial differentiation gene family (EDG) of receptors and leads to diverse signaling events including cell survival, growth, migration and differentiation. However, the mechanisms of how SPP activates these proangiogenic pathways are poorly understood. Here we show that SPP signals through the EDG-1 receptor to the heterotrimeric G protein G(i), leading to activation of the serine/threonine kinase Akt and phosphorylation of the Akt substrate, endothelial nitric-oxide synthase (eNOS). Inhibition of G(i) signaling, and phosphoinositide 3-kinase (PI 3-kinase) activity resulted in a decrease in SPP-induced endothelial cell chemotaxis. SPP also stimulates eNOS phosphorylation and NO release and these effects are also attenuated by inhibition of G(i) signaling, PI 3-kinase, and Akt. However, inhibition of NO production did not influence SPP-induced chemotaxis but effectively blocked the chemotactic actions of vascular endothelial growth factor. Thus, SPP signals through G(i) and PI 3-kinase leading to Akt activation and eNOS phosphorylation.


Subject(s)
Chemotaxis , Endothelium, Vascular/cytology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Lysophospholipids , Nitric Oxide/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Sphingosine/metabolism , Sphingosine/physiology , Animals , Blotting, Northern , Blotting, Western , Cattle , Cell Movement , Culture Media, Serum-Free/metabolism , Dose-Response Relationship, Drug , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/enzymology , Enzyme Activation , Genes, Dominant , Lung/metabolism , Lymphokines/pharmacology , Neovascularization, Physiologic , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Phosphorylation , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Receptors, Cell Surface/biosynthesis , Signal Transduction , Sphingosine/analogs & derivatives , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Virulence Factors, Bordetella/pharmacology
16.
J Biol Chem ; 276(22): 19503-11, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11278759

ABSTRACT

The Hedgehog signaling pathway is involved in early embryonic patterning as well as in cancer; however, little is known about the subcellular localization of the Hedgehog receptor complex of Patched and Smoothened. Since Hh has been found in lipid rafts in Drosophila, we hypothesized that Patched and Smoothened might also be found in these cholesterol-rich microdomains. In this study, we demonstrate that both Smoothened and Patched are in caveolin-1-enriched/raft microdomains. Immunoprecipitation studies show that Patched specifically interacts with caveolin-1, whereas Smoothened does not. Fractionation studies show that Patched and caveolin-1 can be co-isolated from buoyant density fractions that represent caveolae/raft microdomains and that Patched and caveolin-1 co-localize by confocal microscopy. Glutathione S-transferase fusion protein experiments show that the interaction between Patched and caveolin-1 involves the caveolin-1 scaffolding domain and a Patched consensus binding site. Immunocytochemistry data and fractionation studies also show that Patched seems to be required for transport of Smoothened to the membrane. Depletion of plasmalemmal cholesterol influences the distribution of the Hh receptor complex in the caveolin-enriched/raft microdomains. These data suggest that caveolin-1 may be integral for sequestering the Hh receptor complex in these caveolin-enriched microdomains, which act as a scaffold for the interactions with the Hh protein.


Subject(s)
Caveolins/metabolism , Cell Membrane/metabolism , Drosophila Proteins , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Receptors, G-Protein-Coupled , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western , COS Cells , Caveolin 1 , Caveolins/biosynthesis , Cholesterol/metabolism , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/metabolism , Humans , Immunohistochemistry , Membrane Proteins/biosynthesis , Microscopy, Confocal , Models, Biological , Molecular Sequence Data , Mutation , Patched Receptors , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction , Smoothened Receptor , Subcellular Fractions , Time Factors
17.
J Biol Chem ; 276(17): 14249-56, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11120737

ABSTRACT

Endothelial nitric-oxide synthase (eNOS) is regulated in part through specific protein interactions. Dynamin-2 is a large GTPase residing within similar membrane compartments as eNOS. Here we show that dynamin-2 binds directly with eNOS thereby augmenting eNOS activity. Double label confocal immunofluorescence demonstrates colocalization of eNOS and dynamin in both Clone 9 cells cotransfected with green fluorescent protein-dynamin and eNOS, as well as in bovine aortic endothelial cells (BAEC) expressing both proteins endogenously, predominantly in a Golgi membrane distribution. Immunoprecipitation of eNOS from BAEC lysate coprecipitates dynamin and, conversely, immunoprecipitation of dynamin coprecipitates eNOS. Additionally, the calcium ionophore, a reagent that promotes nitric oxide release, enhances coprecipitation of dynamin with eNOS in BAEC, suggesting the interaction between the proteins can be regulated by intracellular signals. In vitro studies demonstrate that glutathione S-transferase (GST)-dynamin-2 quantitatively precipitates both purified recombinant eNOS protein as well as in vitro transcribed (35)S-labeled eNOS from solution indicating a direct interaction between the proteins in vitro. Scatchard analysis of binding studies demonstrates an equilibrium dissociation constant (K(d)) of 27.6 nm. Incubation of purified recombinant eNOS protein with GST-dynamin-2 significantly increases eNOS activity as does overexpression of dynamin-2 in ECV 304 cells stably transfected with eNOS-green fluorescent protein. These studies demonstrate a direct protein-protein interaction between eNOS and dynamin-2, thereby identifying a new NOS-associated protein and providing a novel function for dynamin. These events may have relevance for eNOS regulation and trafficking within vascular endothelium.


Subject(s)
GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/metabolism , Animals , Aorta/cytology , Blotting, Western , Calcimycin/pharmacology , Cattle , Cell Line , Dose-Response Relationship, Drug , Dynamin I , Dynamins , Endothelium, Vascular/cytology , Glutathione Transferase/metabolism , Golgi Apparatus/metabolism , Ionophores/pharmacology , Kinetics , Microscopy, Confocal , Microscopy, Fluorescence , Nitric Oxide Synthase Type III , Precipitin Tests , Protein Binding , Protein Biosynthesis , Rats , Recombinant Fusion Proteins/metabolism , Transfection
18.
Nat Med ; 6(12): 1362-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11100121

ABSTRACT

Caveolin-1, the primary coat protein of caveolae, has been implicated as a regulator of signal transduction through binding of its "scaffolding domain" to key signaling molecules. However, the physiological importance of caveolin-1 in regulating signaling has been difficult to distinguish from its traditional functions in caveolae assembly, transcytosis, and cholesterol transport. To directly address the importance of the caveolin scaffolding domain in vivo, we generated a chimeric peptide with a cellular internalization sequence fused to the caveolin-1 scaffolding domain (amino acids 82-101). The chimeric peptide was efficiently taken up into blood vessels and endothelial cells, resulting in selective inhibition of acetylcholine (Ach)-induced vasodilation and nitric oxide (NO) production, respectively. More importantly, systemic administration of the peptide to mice suppressed acute inflammation and vascular leak to the same extent as a glucocorticoid or an endothelial nitric oxide synthase (eNOS) inhibitor. These data imply that the caveolin-1 scaffolding domain can selectively regulate signal transduction to eNOS in endothelial cells and that small-molecule mimicry of this domain may provide a new therapeutic approach.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Caveolins/pharmacology , Nitric Oxide/biosynthesis , Vasodilator Agents/antagonists & inhibitors , Animals , Capillary Permeability/drug effects , Caveolin 1 , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/metabolism , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Recombinant Fusion Proteins/pharmacology
19.
Am J Physiol Heart Circ Physiol ; 279(6): H2649-57, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11087217

ABSTRACT

Statin drugs can upregulate endothelial nitric oxide (NO) synthase (eNOS) in isolated endothelial cells independent of lipid-lowering effects. We investigated the effect of short-term simvastatin administration on coronary vascular eNOS and NO production in conscious dogs and canine tissues. Mongrel dogs were instrumented under general anesthesia to measure coronary blood flow (CBF). Simvastatin (20 mg. kg(-1). day(-1)) was administered orally for 2 wk; afterward, resting CBF was found to be higher compared with control (P < 0.05) and veratrine- (activator of reflex cholinergic NO-dependent coronary vasodilation) and ACh-mediated coronary vasodilation were enhanced (P < 0.05). Response to endothelium-independent vasodilators, adenosine and nitroglycerin, was not potentiated. After simvastatin administration, plasma nitrate and nitrite (NO(x)) levels increased from 5.22 +/- 1.2 to 7. 79 +/- 1.3 microM (P < 0.05); baseline and agonist-stimulated NO production in isolated coronary microvessels were augmented (P < 0.05); resting in vivo myocardial oxygen consumption (MVO(2)) decreased from 6.8 +/- 0.6 to 5.9 +/- 0.4 ml/min (P < 0.05); NO-dependent regulation of MVO(2) in response to NO agonists was augmented in isolated myocardial segments (P < 0.05); and eNOS protein increased 29% and eNOS mRNA decreased 50% in aortas and coronary vascular endothelium. Short-term administration of simvastatin in dogs increases coronary endothelial NO production to enhance NO-dependent coronary vasodilation and NO-mediated regulation of MVO(2).


Subject(s)
Anticholesteremic Agents/pharmacology , Coronary Circulation/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Nitric Oxide/biosynthesis , Simvastatin/pharmacology , Acetylcholine/pharmacology , Adenosine/pharmacology , Animals , Consciousness , Coronary Circulation/drug effects , Dogs , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Heart Rate/physiology , In Vitro Techniques , Microcirculation/physiology , Myocardium/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Nitrites/metabolism , Nitroglycerin/pharmacology , Oxygen Consumption/physiology , RNA, Messenger/analysis , Vasodilation/physiology , Vasodilator Agents/pharmacology , Veratrine/pharmacology
20.
Circ Res ; 87(8): 677-82, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-11029403

ABSTRACT

17beta-Estradiol (E(2)) is a rapid activator of endothelial nitric oxide synthase (eNOS). The product of this activation event, NO, is a fundamental determinant of cardiovascular homeostasis. We previously demonstrated that E(2)-stimulated endothelial NO release can occur without an increase in cytosolic Ca(2+). Here we demonstrate for the first time, to our knowledge, that E(2) rapidly induces phosphorylation and activation of eNOS through the phosphatidylinositol 3 (PI3)-kinase-Akt pathway. E(2) treatment (10 ng/mL) of the human endothelial cell line, EA.hy926, resulted in increased NO production, which was abrogated by the PI3-kinase inhibitor, LY294002, and the estrogen receptor antagonist ICI 182, 780. E(2) stimulated rapid Akt phosphorylation on serine 473. As has been shown for vascular endothelial growth factor, eNOS is an E(2)-activated Akt substrate, demonstrated by rapid eNOS phosphorylation on serine 1177, a critical residue for eNOS activation and enhanced sensitivity to resting cellular Ca(2+) levels. Adenoviral-mediated EA.hy926 transduction confirmed functional involvement of Akt, because a kinase-deficient, dominant-negative Akt abolished E(2)-stimulated NO release. The membrane-impermeant E(2)BSA conjugate, shown to bind endothelial cell membrane sites, also induced rapid Akt and consequent eNOS phosphorylation. Thus, engagement of membrane estrogen receptors results in rapid endothelial NO release through a PI3-kinase-Akt-dependent pathway. This explains, in part, the reduced requirement for cytosolic Ca(2+) fluxes and describes an important pathway relevant to cardiovascular pathophysiology.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide Synthase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Adenoviridae/genetics , Binding Sites/drug effects , Binding Sites/genetics , Cell Membrane/metabolism , Cells, Cultured , Chromones/pharmacology , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Genes, Dominant , Humans , Morpholines/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Estrogen/antagonists & inhibitors , Serum Albumin, Bovine/pharmacology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...