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










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 32(2): 247-56, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22199371

ABSTRACT

OBJECTIVE: Prediabetic states are associated with accelerated atherosclerosis, but the availability of mouse models to study connections between these diseases has been limited. The aim of this study was to test the selective role of impaired insulin receptor/insulin receptor substrate-1 signaling on atherogenesis. METHODS AND RESULTS: To address the effects of impaired insulin signaling associated with hyperinsulinemia on atherosclerosis in the absence of obesity and hyperglycemia, we generated insulin receptor (Insr)/insulin receptor substrate-1 (Insr1) double heterozygous apolipoprotein (Apoe)-knockout mice (Insr(+/-)Irs1(+/-)Apoe(-/-)) mice. Insr(+/-)Irs1(+/-)Apoe(-/-) mice fed a Western diet for 15 weeks showed elevated levels of fasting insulin compared to Insr(+/+)Irs1(+/+)Apoe(-/-) mice. There were no significant differences in glucose, triglyceride, HDL, VLDL, cholesterol levels or free fatty acid in the plasma of Insr(+/-)Irs1(+/-)Apoe(-/-) and Insr(+/+)Irs1(+/+)Apoe(-/-) mice. Atherosclerotic lesions were increased in male (brachiocephalic artery) and female (aortic tree) Insr(+/-)Irs1(+/-)Apoe(-/-) compared to Insr(+/+)Irs1(+/+)Apoe(-/-) mice. Bone marrow transfer experiments demonstrated that nonhematopoietic cells have to be Insr(+/-)Irs1(+/-) to accelerate atherosclerosis. Impaired insulin signaling resulted in decreased levels of vascular phospho-eNOS, attenuated endothelium-dependent vasorelaxation and elevated VCAM-1 expression in aortas of Insr(+/-)Irs1(+/-)Apoe(-/-) mice. In addition, phospho-ERK and vascular smooth muscle cell proliferation were significantly elevated in aortas of Insr(+/-)Irs1(+/-)Apoe(-/-) mice. CONCLUSIONS: These results demonstrate that defective insulin signaling is involved in accelerated atherosclerosis in Insr(+/-)Irs1(+/-)Apoe(-/-) mice by promoting vascular dysfunction and inflammation.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Heterozygote , Insulin Receptor Substrate Proteins/genetics , Receptor, Insulin/genetics , Signal Transduction/physiology , Animals , Apolipoproteins E/genetics , Atherosclerosis/pathology , Cell Proliferation , Disease Models, Animal , Disease Progression , Female , Insulin Receptor Substrate Proteins/physiology , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Nitric Oxide Synthase Type III/metabolism , Receptor, Insulin/physiology
2.
J Biol Chem ; 284(45): 31303-14, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19713213

ABSTRACT

12/15-Lipoxygenase (12/15LO) plays a role in the pathogenesis of atherosclerosis and diabetes and has been implicated in low density lipoprotein oxidation. Murine macrophages express high levels of 12/15LO and are key cells involved in the accumulation and efflux of oxidized low density lipoprotein in the arterial wall. During this process, macrophages up-regulate scavenger receptors that regulate lipid uptake, and ATP-binding cassette (ABC) transporters, that regulate lipid efflux. We have previously demonstrated that 12/15LO enhances the turnover and serine phosphorylation of ABCG1. In the current study, we further elucidate the mechanisms by which 12/15LO regulates ABCG1. Proteasomal inhibitors blocked the down-regulation of ABCG1 expression and resulted in accumulation of phosphorylated ABCG1. Macrophages that lack 12/15LO have enhanced transporter expression, reduced ABCG1 phosphorylation, and increased cholesterol efflux. Conversely, macrophages that overexpress 12/15LO have reduced ABCG1 expression, increased transporter phosphorylation, and reduced cholesterol efflux. 12/15LO plays a key role in activating the MAPK pathway. Inhibition of the p38 or JNK pathways with pharmacological inhibitors or dominant negative constructs blocked 12S-hydroxyeicosatetranoic acid-mediated degradation of ABCG1. Moreover, we isolated macrophages from JNK1-, JNK2-, and MKK3-deficient mice to analyze the involvement of specific MAPK pathways. JNK2- and MKK3-, but not JNK1-deficient macrophages were resistant to the down-regulation of ABCG1 protein, reduction in efflux, and increase in serine phosphorylation by 12S-hydroxyeicosatetranoic acid. These findings provide evidence that 12/15LO regulates ABCG1 expression and function through p38- and JNK2-dependent mechanisms, and that targeting these pathways may provide novel approaches for regulating cholesterol homeostasis.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Gene Expression Regulation , Lipoproteins/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 9/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Biological Transport , Female , Lipid Metabolism , Lipoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 9/genetics , Phosphorylation , p38 Mitogen-Activated Protein Kinases/genetics
3.
Arterioscler Thromb Vasc Biol ; 28(10): 1811-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18635820

ABSTRACT

OBJECTIVE: The purpose of this study was to evaluate the effect of 12/15-lipoxygenase (12/15LO) in macrophage ABCG1 expression and function associated with cholesterol efflux. METHODS AND RESULTS: 12/15LO was stably overexpressed in J774 macrophages. 12/15LO-overexpressing macrophages had a 30% reduction in HDL-mediated cholesterol efflux, corresponding with significantly reduced ABCG1 protein expression. Treatment of 12/15LO-overexpressing macrophages with a 12/15LO ribozyme to reduce 12/15LO restored HDL-mediated efflux and ABCG1 protein expression. Treating macrophages with 12/15LO unsaturated fatty acid substrates or eicosanoid products also reduced HDL-mediated cholesterol efflux. Additionally, both 12/15LO overexpression in macrophages and incubation of macrophages with eicosanoids reduced ABCG1 protein, but not mRNA, expression. However, incubation of macrophages with linoleic or arachidonic acids significantly reduced both ABCG1 mRNA and protein expression, suggesting that 12/15LO substrates and eicosanoid products differentially regulate ABCG1 expression. 12/15LO fatty acids did not decrease ABCG1 translation; however, 12/15LO fatty acids increased ABCG1 degradation when blocked by cyclohexidmide. ABCG1 degradation may be regulated through posttranslational modifications. Treatment with the 12/15LO eicosanoid product 12SHETE increased serine phosphorylation of ABCG1. CONCLUSIONS: We conclude that serine phosphorylation may increase the degradation rate of ABCG1, and as a result cause macrophage cholesterol accumulation. These findings provide evidence that 12/15LO activity in the vessel wall contributes to atherogenesis by impairing the macrophage ABCG1 cholesterol efflux pathway.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Lipoproteins/metabolism , Macrophages/enzymology , Protein Processing, Post-Translational , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Arachidonic Acid/metabolism , Cell Line , Cholesterol, HDL/metabolism , Down-Regulation , Linoleic Acid/metabolism , Lipoproteins/genetics , Mice , Phosphorylation , RNA, Catalytic/metabolism , RNA, Messenger/metabolism , Serine , Swine , Time Factors , Transfection
4.
Circulation ; 117(21): 2785-92, 2008 May 27.
Article in English | MEDLINE | ID: mdl-18490524

ABSTRACT

BACKGROUND: Patients with type 2 diabetes mellitus are at increased risk for the development of atherosclerosis. A pivotal event in the development of atherosclerosis is macrophage foam cell formation. The ATP-binding cassette (ABC) transporters ABCA1 and ABCG1 regulate macrophage cholesterol efflux and hence play a vital role in macrophage foam cell formation. We have previously found that chronic elevated glucose reduces ABCG1 expression. In the present study, we examined whether patients with type 2 diabetes mellitus had decreased ABCG1 and/or ABCA1, impaired cholesterol efflux, and increased macrophage foam cell formation. METHODS AND RESULTS: Blood was collected from patients with and without type 2 diabetes mellitus. Peripheral blood monocytes were differentiated into macrophages, and cholesterol efflux assays, immunoblots, histological analysis, and intracellular cholesteryl ester measurements were performed. Macrophages from patients with type 2 diabetes mellitus had a 30% reduction in cholesterol efflux with a corresponding 60% increase in cholesterol accumulation relative to control subjects. ABCG1 was present in macrophages from control subjects but was undetectable in macrophages from patients with type 2 diabetes mellitus. In contrast, ABCA1 expression in macrophages was similar in both control subjects and patients with type 2 diabetes mellitus. Macrophage expression of ABCG1 in both patients and control subjects was induced by treatment with the liver X receptor agonist TO-901317. Upregulation of liver X receptor dramatically reduced foam cell formation in macrophages from patients with type 2 diabetes mellitus. CONCLUSIONS: ABCG1 expression and cholesterol efflux are reduced in patients with type 2 diabetes mellitus. This impaired ABCG1-mediated cholesterol efflux significantly correlates with increased intracellular cholesterol accumulation. Strategies to upregulate ABCG1 expression and function in type 2 diabetes mellitus could have therapeutic potential for limiting the accelerated vascular disease observed in patients with type 2 diabetes mellitus.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Cholesterol, HDL/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Macrophages/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Cell Differentiation/immunology , Cells, Cultured , Cholesterol Esters/metabolism , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Female , Gene Expression , Humans , Hydrocarbons, Fluorinated/pharmacology , Macrophages/cytology , Macrophages/drug effects , Male , Middle Aged , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Sulfonamides/pharmacology
5.
Circ Res ; 102(8): 950-8, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18323526

ABSTRACT

Activated macrophages acquire a proinflammatory (classic) or antiinflammatory (alternative) phenotype that influences atherosclerosis. The present study investigated whether sphingosine-1-phosphate (S1P), with its known antiinflammatory effects, could regulate the inflammatory phenotype of lipopolysaccharide (LPS)-stimulated mouse macrophages. Activation of macrophages by LPS significantly increases proinflammatory cytokine secretion. Pretreatment of macrophages with 500 nmol/L S1P markedly reduced LPS-mediated secretion of tumor necrosis factor-alpha, monocyte chemoattractant protein-1, and interleukin-12. Such antiinflammatory actions were also evident in LPS-stimulated macrophages treated with the S1P1 receptor-specific agonist SEW2871. Pharmacological antagonism of the S1P1 receptor on macrophages using the S1P1-specific antagonist VPC44116 also blocked proinflammatory cytokine secretion in response to LPS. Studies using bone marrow-derived macrophages from S1P2-deficient mice revealed that the S1P2 receptor did not play a pivotal role in this process. Thus, activation of the S1P1 receptor in mouse macrophages limits the expression of proinflammatory cytokines. Furthermore, we demonstrated that S1P increased arginase I activity and inhibited LPS-induced inducible NO synthase activity in LPS-treated macrophages, again through S1P1 receptor activation on macrophages. Analysis of a 1.7-kb region of the murine inducible NO synthase promoter revealed the presence of putative nuclear factor kappaB, activator protein-1, and STAT-1 response elements. Using inducible NO synthase promoter-reporter constructs, we found that S1P significantly reduced the nuclear factor kappaB-mediated induction of inducible NO synthase. These findings demonstrate an important role for S1P in the regulation of macrophage phenotypic switching. Therefore, we conclude that S1P promotes the production of an alternative antiinflammatory macrophage phenotype through activation of the macrophage S1P1 receptor.


Subject(s)
Inflammation , Lysophospholipids/physiology , Macrophages/immunology , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Bone Marrow Cells , Cells, Cultured , Cytokines/biosynthesis , Lipopolysaccharides/pharmacology , Lysophospholipids/deficiency , Lysophospholipids/immunology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II , Phenotype , Receptors, Lysosphingolipid/immunology , Sphingosine/deficiency , Sphingosine/immunology , Sphingosine/physiology
6.
J Immunol ; 180(6): 4273-82, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18322240

ABSTRACT

ATP-binding cassette transporter G1 (ABCG1) effluxes cholesterol from macrophages and plays an important role in pulmonary lipid homeostasis. We hypothesize that macrophages from Abcg1(-/-) mice have increased inflammatory activity, thereby promoting acceleration of pulmonary disease. We herein demonstrate increased numbers of inflammatory cytokines and infiltrating neutrophils, eosinophils, dendritic cells, T cells, and B cells into lungs of Abcg1(-/-) mice before the onset of severe lipidosis. We further investigated the role of macrophages in causing pulmonary disease by performing bone marrow transplantations using B6 and Abcg1(-/-) bone marrow. We found that it was the macrophage, and not pneumocyte type II cells or other nonhematopoietic cells in the lung, that appeared to be the primary cell type involved in the onset of both pulmonary lipidosis and inflammation in the Abcg1(-/-) mice. Additionally, our results demonstrate that Abcg1(-/-) macrophages had elevated proinflammatory cytokine production, increased apoptotic cell clearance, and were themselves more prone to apoptosis and necrosis. However, they were quickly repopulated by monocytes that were recruited to Abcg1(-/-) lungs. In conclusion, we have shown that ABCG1 deletion in macrophages causes a striking inflammatory phenotype and initiates onset of pulmonary lipidosis in mice. Thus, our studies reveal a critical role for macrophage ABCG1 in lung inflammation and homeostasis.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Homeostasis/immunology , Inflammation Mediators/physiology , Lipoproteins/physiology , Lung/immunology , Lung/pathology , Macrophages/immunology , Macrophages/pathology , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Animals , Animals, Newborn , Homeostasis/genetics , Humans , Inflammation Mediators/metabolism , Jurkat Cells , Lipid Metabolism/genetics , Lipid Metabolism/immunology , Lipoproteins/deficiency , Lipoproteins/genetics , Lung/cytology , Lung/metabolism , Macrophages/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Diabetes ; 57(2): 484-93, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18003758

ABSTRACT

OBJECTIVES: Non-obese diabetic (NOD) mice develop spontaneous type 1 diabetes. We have shown that sphingosine-1-phosphate (S1P) reduces activation of NOD diabetic endothelium via the S1P1 receptor. In the current study, we tested the hypothesis that S1P could inhibit CD4(+) T-cell activation, further reducing inflammatory events associated with diabetes. RESEARCH DESIGN AND METHODS: CD4(+) T-cells were isolated from diabetic and nondiabetic NOD mouse splenocytes and treated in the absence or presence of S1P or the S1P1 receptor-specific agonist, SEW2871. Lymphocyte activation was examined using flow cytometry, cytokine bead assays, and a lymphocyte:endothelial adhesion assay. RESULTS: Diabetic T-cells secreted twofold more gamma-interferon (IFN-gamma) and interleukin-17 than nondiabetic lymphocytes. Pretreatment with either S1P or SEW2871 significantly reduced cytokine secretion by approximately 50%. Flow cytometry analysis showed increased expression of CD69, a marker of lymphocyte activation, on diabetic T-cells. Both S1P and SEW2871 prevented upregulation of CD69 on CD4(+) cells. Quantitative RT-PCR showed that lymphocytes from diabetic NOD mice had 2.5-fold lower hypoxia-inducible factor (HIF)-1alpha short isoform I.1 (HIF1alphaI.1) mRNA levels than control. HIF1alphaI.1 is a negative regulator of lymphocyte activation. S1P significantly increased HIF1alpha I.1 mRNA levels in both control and diabetic groups. IFN-gamma production and surface CD69 expression was significantly increased in lymphocytes of HIF1alphaI.1-deficient mice. S1P did not reduce either CD69 or IFN-gamma expression in lymphocytes from HIF1alphaI.1-deficient mice. CONCLUSIONS: S1P acts through the S1P1 receptor and HIF1alpha I.1 to negatively regulate T-cell activation, providing a potential therapeutic target for prevention of diabetes and its vascular complications.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Diabetes Mellitus, Type 1/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphocyte Activation/drug effects , Lysophospholipids/therapeutic use , Sphingosine/analogs & derivatives , T-Lymphocytes/immunology , Animals , Antigens, CD/drug effects , Antigens, Differentiation, T-Lymphocyte/drug effects , Cytokines/immunology , Diabetes Mellitus, Type 1/prevention & control , Diabetic Angiopathies/prevention & control , Flow Cytometry , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Lectins, C-Type , Mice , Mice, Inbred NOD , Mice, Knockout , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/therapeutic use , Spleen/immunology , T-Lymphocytes/drug effects
8.
Circ Res ; 99(7): 731-9, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16960101

ABSTRACT

Monocyte recruitment and adhesion to vascular endothelium are key early events in atherosclerosis. We examined the role of sphingosine-1-phosphate (S1P) on modulating monocyte/endothelial interactions in the NOD/LtJ (NOD) mouse model of type 1 diabetes. Aortas from nondiabetic and diabetic NOD mice were incubated in the absence or presence of 100 nmol/L S1P. Fluorescently labeled monocytes were incubated with the aortas. Aortas from NOD diabetic mice bound 7-fold more monocytes than nondiabetic littermates (10+/-1 monocytes bound/field for nondiabetic mice vs 74+/-12 monocytes bound/field for diabetic mice, P<0.0001). Incubation of diabetic aortas with 100 nmol/L S1P reduced monocyte adhesion to endothelium by 90%. We found expression of S1P1, S1P2, and S1P3 receptors on NOD aortic endothelial cells. The S1P1 receptor-specific agonist SEW2871 inhibited monocyte adhesion to diabetic aortas. Studies in diabetic S1P3-deficient mice revealed that the S1P3 receptor did not play a pivotal role in this process. S1P reduced endothelial VCAM-1 induction in type 1 diabetic NOD mice, most likely through inhibition of nuclear factor kappaB translocation to the nucleus. Thus, S1P activation of the S1P1 receptor functions in an antiinflammatory manner in type 1 diabetic vascular endothelium to prevent monocyte/endothelial interactions. S1P may play an important role in the prevention of vascular complications of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Endothelium, Vascular/physiopathology , Lysophospholipids/metabolism , Monocytes , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Anti-Inflammatory Agents/pharmacology , Aorta/drug effects , Aorta/metabolism , Aorta/physiopathology , Biological Transport/drug effects , Cell Adhesion/drug effects , Cell Nucleus/metabolism , Chemokine CCL2/metabolism , Diabetes Mellitus, Type 1/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Inbred NOD , Receptors, Lysosphingolipid/antagonists & inhibitors , Sphingosine/metabolism , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , Vascular Cell Adhesion Molecule-1/metabolism
9.
J Biol Chem ; 281(30): 21216-21224, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16723355

ABSTRACT

Atherosclerosis development is accelerated severalfold in patients with Type 2 diabetes. In the initial stages of disease, monocytes transmigrate into the subendothelial space and differentiate into foam cells. Scavenger receptors and ATP binding cassette (ABC) Transporters play an important role in foam cell formation as they regulate the influx and efflux of oxidized lipids. Here, we show that peritoneal macrophages isolated from Type 2 diabetic db/db mice have decreased expression of the ABC transporter ABCG1 and increased expression of the scavenger receptor CD36. We found a 2-fold increase in accumulation of esterified cholesterol in diabetic db/db macrophages compared with wild-type control macrophages. Diabetic db/db macrophages also had impaired cholesterol efflux to high density lipoprotein but not to lipid-free apo A-I, suggesting that the increased esterified cholesterol in diabetic db/db macrophages was due to a selective loss of ABCG1-mediated efflux to high density lipoprotein. Additionally, we were able to confirm down-regulation of ABCG1 using C57BL/6J peritoneal macrophages cultured in elevated glucose in vitro (25 mM glucose for 7 days), suggesting that ABCG1 expression in diabetic macrophages is regulated by chronic exposure to elevated glucose. Diabetic KK(ay) mice were also studied and were found to have decreased ABCG1 expression without an increase in CD36. These observations demonstrate that ABCG1 plays a major role in macrophage cholesterol efflux and that decreased ABCG1 function can facilitate foam cell formation in Type 2 diabetic mice.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Diabetes Mellitus, Type 2/metabolism , Foam Cells/cytology , Lipoproteins/biosynthesis , Macrophages/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Animals , CD36 Antigens/biosynthesis , DNA Primers/chemistry , Disease Models, Animal , Foam Cells/metabolism , Glucose/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA Processing, Post-Transcriptional , RNA, Small Interfering/metabolism
10.
Arterioscler Thromb Vasc Biol ; 26(6): 1260-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16543492

ABSTRACT

OBJECTIVE: 12/15 lipoxygenase (12/15LO) has been implicated as a mediator of inflammation and atherosclerosis. In the current study, we identified mechanisms through which 12/15LO mediates monocyte:endothelial interactions in vivo in apolipoprotein E-deficient mice (apoEKO), a well-characterized mouse model of atherosclerosis. METHODS AND RESULTS: In apoEKO mice that are also deficient in 12/15LO (doubleKO), monocyte adhesion to aorta in vivo was reduced by 95% in doubleKO mice compared with apoEKO mice. Inhibition of 12/15LO in apoEKO mice in vivo using CDC (Cinnamyl-3,4-Dihydroxy-a-Cyanocinnamate) prevented monocyte adhesion to aortic endothelium in apoEKO mice. Aortic endothelium of apoEKO mice had significant activation of rhoA compared with doubleKO aortic endothelium. Further, apoEKO aorta displayed significant activation of NF-kappaB. DoubleKO aorta displayed little nuclear localization of NF-kappaB. Finally, we found significant upregulation of intercellular adhesion molecule-1 (ICAM-1) on apoEKO aortic endothelium compared with doubleKO endothelium. Inhibition of rhoA and PKCalpha significantly reduced NF-kappaB activation, ICAM-1 induction, and monocyte adhesion to aorta. CONCLUSIONS: We conclude that 12/15LO products activate endothelial rhoA and PKCalpha. Activation of rhoA and PKCalpha cause activation and translocation of NF-kappaB to the nucleus, which, in turn, results in induction of ICAM-1. Induction of ICAM-1 on aortic endothelium stimulates monocyte:endothelial adhesion in vivo in apoEKO mice.


Subject(s)
Aorta/physiopathology , Apolipoproteins E/deficiency , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Endothelium, Vascular/physiopathology , Monocytes , NF-kappa B/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/deficiency , Biological Transport , Caffeic Acids/pharmacology , Cell Adhesion/drug effects , Cell Nucleus/metabolism , Enzyme Activation , Intercellular Adhesion Molecule-1/biosynthesis , Lipoxygenase Inhibitors/pharmacology , Mice , Mice, Knockout , Monocytes/drug effects , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology
11.
Arterioscler Thromb Vasc Biol ; 25(11): 2301-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16166569

ABSTRACT

BACKGROUND: 12/15-lipoxygenase (12/15-LO) activity leads to the production of the proinflammatory eicosanoids 12-S-hydroxyeicosatetraenoic acid (12SHETE) and 13-S-hydroxyoctadecadienoic acid. We have previously shown a 3.5-fold increase in endothelial intercellular adhesion molecule (ICAM)-1 expression in mice overexpressing the 12/15-LO gene. We examined whether 12/15-LO activity regulated endothelial ICAM-1 expression. METHODS AND RESULTS: Freshly isolated aortic endothelial cells (EC) from 12/15-LO transgenic mice had significantly greater nuclear factor-kappaB (NF-kappaB) activation and ICAM mRNA expression compared with C57BL/6J control. 12/15-LO transgenic EC showed elevated RhoA activity, and inhibition of RhoA using either C3 toxin or the Rho-kinase inhibitor Y-27632 blocked NF-kappaB activation, ICAM-1 induction, and monocyte adhesion. Furthermore, we show that 12SHETE activates protein kinase Calpha, which forms a complex with active RhoA and is required for NF-kappaB-dependent ICAM expression in response to 12SHETE. CONCLUSIONS: The 12/15-LO pathway stimulates ICAM-1 expression through the RhoA/protein kinase Calpha-dependent activation of NF-kappaB. These findings identify a major signaling pathway in EC through which 12/15-LO contributes to vascular inflammation and atherosclerosis.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Cell Adhesion/immunology , Intercellular Adhesion Molecule-1/genetics , Monocytes/cytology , Vasculitis/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Animals , Aorta/cytology , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Gene Expression/drug effects , Gene Expression/physiology , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Protein Kinase C-alpha/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Vasculitis/physiopathology , rhoA GTP-Binding Protein/metabolism
12.
Arterioscler Thromb Vasc Biol ; 25(5): 976-81, 2005 May.
Article in English | MEDLINE | ID: mdl-15761190

ABSTRACT

OBJECTIVE: Endothelial activation and monocyte adhesion to endothelium are key events in inflammation. Sphingosine-1-phosphate (S1P) is a sphingolipid that binds to G protein-coupled receptors on endothelial cells (ECs). We examined the role of S1P in modulating endothelial activation and monocyte-EC interactions in vivo. METHODS AND RESULTS: We injected C57BL/6J mice intravenously with tumor necrosis factor (TNF)-alpha in the presence and absence of the S1P1 receptor agonist SEW2871 and examined monocyte adhesion. Aortas from TNF-alpha-injected mice had a 4-fold increase in the number of monocytes bound, whereas aortas from TNF-alpha plus SEW2871-treated mice had few monocytes bound (P<0.0001). Using siRNA, we found that inhibiting the S1P1 receptor in vascular ECs blocked the ability of S1P to prevent monocyte-EC interactions in response to TNF-alpha. We examined signaling pathways downstream of S1P1 and found that 100 nM S1P increased phosphorylation of Akt and decreased activation of c-jun. CONCLUSIONS: Thus, we provide the first evidence that S1P signaling through the endothelial S1P1 receptor protects the vasculature against TNF-alpha-mediated monocyte-EC interactions in vivo.


Subject(s)
Cell Adhesion/drug effects , Endothelium, Vascular/cytology , Lysophospholipids/pharmacology , Monocytes/cytology , Sphingosine/analogs & derivatives , Tumor Necrosis Factor-alpha/metabolism , Vasculitis/drug therapy , Animals , Aorta/cytology , Aorta/immunology , Cell Adhesion/immunology , Cells, Cultured , Chemokines/metabolism , E-Selectin/metabolism , Endothelium, Vascular/immunology , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Monocytes/immunology , Oxadiazoles/pharmacology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Sphingosine/pharmacology , Thiophenes/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism , Vasculitis/metabolism , Vasculitis/prevention & control
13.
Circulation ; 110(14): 2024-31, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15451785

ABSTRACT

BACKGROUND: Mice lacking leukocyte type 12/15-lipoxygenase (12/15-LO) show reduced atherosclerosis in several models. 12/15-LO is expressed in a variety of cells, including vascular cells, adipocytes, macrophages, and cardiomyocytes. The purpose of this study was to determine which cellular source of 12/15-LO is important for atherosclerosis. METHODS AND RESULTS: Bone marrow from 12/15-LO-/-/apoE-/- mice was transplanted into apoE-/- mice and vice versa. Deficiency of 12/15-LO in bone marrow cells protected apoE-/- mice fed a Western diet from atherosclerosis to the same extent as complete absence of 12/15-LO, although plasma 8,12-iso-iPF2alpha-IV, a measure of lipid peroxidation, remained elevated. 12/15-LO-/-/apoE-/- mice regained the severity of atherosclerotic lesion typical of apoE-/- mice after replacement of their bone marrow cells with bone marrow from apoE-/- mice. Peritoneal macrophages obtained from wild-type but not 12/15-LO-/- mice caused endothelial activation in the presence of native LDL. Absence of 12/15-LO decreased the ability of macrophages to form foam cells when exposed to LDL. CONCLUSIONS: We conclude that macrophage 12/15-LO plays a dominant role in the development of atherosclerosis by promoting endothelial inflammation and foam cell formation.


Subject(s)
Apolipoproteins E/deficiency , Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Arteriosclerosis/enzymology , Dinoprost/analogs & derivatives , Endothelial Cells/enzymology , Foam Cells/cytology , Hyperlipoproteinemia Type II/enzymology , Macrophages, Peritoneal/enzymology , Myocytes, Smooth Muscle/enzymology , Animals , Apolipoproteins E/genetics , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/genetics , Autocrine Communication , Bone Marrow Transplantation , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Dinoprost/blood , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/genetics , Interleukin-4/pharmacology , Lipoproteins, LDL/pharmacology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , RNA, Messenger/biosynthesis , Radiation Chimera , Triglycerides/blood
14.
J Biol Chem ; 279(30): 31930-6, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15145956

ABSTRACT

We have shown that chronic elevated glucose (25 mm) increases monocyte adhesion to human aortic endothelial cells (EC). This increased adhesion is mediated primarily through induction of interleukin (IL)-8 via activation of the transcription factor AP-1 (Srinivasan, S., Yeh, M., Danziger, E. C., Hatley, M. E., Riggan, A. E., Leitinger, N., Berliner, J. A., and Hedrick, C. C. (2003) Circ. Res. 92, 371-377). In the current study, we identified the elements in the AP-1 transcriptional complex that are activated by glucose. These elements include c-Jun, c-Fos, and Fra-1. AP-1 is activated by cellular oxidative stress, and we have reported significant production of ROS by high glucose-cultured cells. We examined signaling pathways upstream of AP-1 in EC that lead to AP-1 activation by HG. EC cultured in 25 mm glucose had a 2-fold increase in p38 phosphorylation compared with control normal glucose-cultured EC. Inhibition of the p38 pathway using 5 microm SB203580 significantly reduced glucose-mediated IL-8 mRNA production by 60%. Furthermore, blocking p38 pathway activation using a dominant-negative p38 construct significantly reduced glucose-mediated monocyte adhesion by 50%. Thus, glucose-stimulated monocyte adhesion is primarily regulated through phosphorylation of p38 with subsequent activation of AP-1, leading to IL-8 production. To study this pathway in the setting of diabetes, we used the db/db mouse. P38 phosphorylation was increased in diabetic db/db mice compared with control mice. We found a dramatic elevation in plasma levels of KC, the mouse ortholog of IL-8 in diabetic db/db mice (1800 +/- 100 pg/ml KC in db/db versus 300 +/- 75 pg/ml in C57BL/6J control mice, p < 0.0001). Inhibition of the p38 pathway in diabetic db/db mice significantly reduced monocyte adhesion by 50%. Taken together, these data indicate that chronic elevated glucose in diabetes activates the p38 MAP kinase pathway to increase inflammatory IL-8 gene induction and monocyte/endothelial adhesion.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/immunology , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Glucose/pharmacology , Interleukin-8/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Animals , Base Sequence , Cell Adhesion/drug effects , Cells, Cultured , DNA Primers/genetics , Diabetes Mellitus, Type 2/genetics , Endothelium, Vascular/drug effects , Humans , In Vitro Techniques , Interleukin-8/genetics , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Biological , Monocytes/drug effects , Monocytes/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , p38 Mitogen-Activated Protein Kinases
15.
Arterioscler Thromb Vasc Biol ; 24(5): 851-7, 2004 May.
Article in English | MEDLINE | ID: mdl-15001458

ABSTRACT

OBJECTIVE: We have previously reported increased monocyte adhesion to human aortic endothelial cells (HAECs) cultured in 25 mmol/L glucose (HG) compared with normal glucose (NG) (5.5 mmol/L). In this study, we explored mechanisms that contribute to increased monocyte adhesion by elevated glucose. METHODS AND RESULTS: We found that HAECs cultured in HG have increased production of the chemokine interleukin-6 (IL-6). We examined whether IL-6 directly modulated monocyte adhesion to EC. Inhibition of IL-6 using a neutralizing antibody significantly reduced glucose-mediated monocyte adhesion by 50%, and addition of IL-6 directly to human EC stimulated monocyte adhesion. PPARalpha has been reported to negatively regulate expression of IL-6 in vascular cells, so we examined PPARalpha-associated signaling in EC. A known PPARalpha agonist, Wy14,643, prevented glucose-mediated IL-6 production by EC and reduced glucose-mediated monocyte adhesion by 40%. HG-cultured HAEC had a 50% reduction in expression of PPARalpha compared with control EC. Primary aortic EC isolated from PPARalpha knockout (KO) mice showed increased monocyte adhesion compared with EC isolated from control mice. PPARalpha KO EC also had increased production of IL-6. Finally, we measured IL-6 levels in diabetic db/db mice and found significant 6-fold elevations in IL-6 levels in db/db EC. CONCLUSIONS: These data indicate that IL-6 production is increased in diabetes and contributes to early vascular inflammatory changes. PPARalpha protects EC from glucose-mediated monocyte adhesion, in part through regulation of IL-6 production.


Subject(s)
Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Glucose/pharmacology , Interleukin-6/physiology , Monocytes/drug effects , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Aorta , Cell Adhesion/drug effects , Cell Adhesion/physiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/pharmacology , Interleukin-8/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Monocytes/cytology , Monocytes/metabolism , Pioglitazone , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Proteins/pharmacology , Thiazolidinediones/pharmacology , Transcription Factors/agonists , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription Factors/genetics
16.
J Biol Chem ; 279(10): 9440-50, 2004 Mar 05.
Article in English | MEDLINE | ID: mdl-14676201

ABSTRACT

We have shown that the 12/15-lipoxygenase (12/15-LO) product 12S-hydroxyeicosatetraenoic acid increases monocyte adhesion to human endothelial cells (EC) in vitro. Recent studies have implicated 12/15-LO in mediating atherosclerosis in mice. We generated transgenic mice on a C57BL/6J (B6) background that modestly overexpressed the murine 12/15-LO gene (designated LOTG). LOTG mice had 2.5-fold elevations in levels of 12S-hydroxyeicosatetraenoic acid and a 2-fold increase in expression of 12/15-LO protein in vivo. These mice developed spontaneous aortic fatty streak lesions on a chow diet. Thus, we examined effects of 12/15-LO expression on early events leading to atherosclerosis in these mice. We found that, under basal unstimulated conditions, LOTG EC bound more monocytes than B6 control EC (18 +/- 2 versus 7 +/- 1 monocytes/field, respectively; p < 0.0001). Inhibition of 12/15-LO activity in LOTG EC using a 12/15-LO ribozyme completely blocked monocyte adhesion in LOTG mice. Thus, 12/15-LO activity is required for monocyte/EC adhesion in the vessel wall. Expression of ICAM-1 in aortic endothelia of LOTG mice was increased severalfold. VCAM-1 expression was not changed. In a series of blocking studies, antibodies to alpha(4) and beta(2) integrins in WEHI monocytes blocked monocyte adhesion to both LOTG and B6 control EC. Inhibition of ICAM-1, VCAM-1, and connecting segment-1 fibronectin in EC significantly reduced adhesion of WEHI monocytes to LOTG EC. In summary, these data indicate that EC from LOTG mice are "pre-activated" to bind monocytes. Monocyte adhesion in LOTG mice is mediated through beta(2) integrin and ICAM-1 interactions as well as through VLA-4 and connecting segment-1 fibronectin/VCAM-1 interactions. Thus, 12/15-LO mediates monocyte/EC interactions in the vessel wall in atherogenesis at least in part through molecular regulation of expression of endothelial adhesion molecules.


Subject(s)
Arteriosclerosis/enzymology , Lipoxygenase/genetics , Animals , Arteriosclerosis/pathology , Cell Adhesion , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Enzyme Activation , Inflammation/metabolism , Inflammation/pathology , Intercellular Adhesion Molecule-1/metabolism , Lipoxygenase/metabolism , Male , Mice , Mice, Transgenic , Monocytes/enzymology , Monocytes/pathology , Vascular Cell Adhesion Molecule-1/metabolism
17.
Endocrinology ; 144(12): 5227-31, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12960000

ABSTRACT

Chronic elevated glucose levels and activation of the renal renin-angiotensin system have been implicated in the pathogenesis of diabetic nephropathy. We tested the ability of lisofylline (LSF), a novel antiinflammatory compound, to prevent extracellular matrix (ECM) accumulation and growth factor production by human mesangial cells (HMCs) cultured in chronic elevated glucose (HG) or angiotensin II (AngII). HMCs were cultured in normal glucose (NG) (5.5 mm) and in HG (25 mm) for 7 d or with 10-7 m AngII for 4 h with or without LSF. Levels of the ECM protein fibronectin and TGF-beta in media were shown to increase in HG compared with NG. LSF decreased HG-induced fibronectin and TGF-beta production to control levels. Increased expression of collagen type IV and laminin was observed in AngII-cultured HMCs. LSF protected HMCs from the AngII induction of these key matrix proteins. cAMP-responsive binding element phosphorylation was significantly higher in both HG and AngII-cultured HMCs. LSF reduced phosphorylation of both cAMP-responsive binding element and p38 MAPK compared with control. These data demonstrate that LSF protects HMCs from HG- and AngII-mediated ECM deposition by the reduction of matrix protein secretion possibly through regulation of TGF-beta production and modulation of the p38 MAPK pathway. These results suggest that LSF may provide therapeutic benefit for prevention or treatment of diabetic nephropathy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Extracellular Matrix/drug effects , Glomerular Mesangium/cytology , Hyperglycemia/drug therapy , Pentoxifylline/analogs & derivatives , Pentoxifylline/pharmacology , Angiotensin II/pharmacology , Cells, Cultured , Connective Tissue Growth Factor , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/genetics , Fibronectins/metabolism , Gene Expression/drug effects , Glomerular Mesangium/drug effects , Glomerular Mesangium/metabolism , Glucose/pharmacology , Humans , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Transforming Growth Factor beta/metabolism , Vasoconstrictor Agents/pharmacology , p38 Mitogen-Activated Protein Kinases
18.
J Biol Chem ; 278(28): 25369-75, 2003 Jul 11.
Article in English | MEDLINE | ID: mdl-12734208

ABSTRACT

Atherosclerosis is a major complication of diabetes. Up to 16 weeks of age, the db/db mouse is insulin-resistant and hyperglycemic and is a good model of Type 2 diabetes. After approximately 16 weeks of age, the mice develop pancreatic beta cell failure that can progress to a Type 1 diabetes phenotype. We have previously shown that glucose increases production of endothelial 12/15 lipoxygenase (12/15LO) products in vitro. In young 10-week-old Type 2 diabetic db/db mice, we found significant elevations in levels of urinary 12/15LO products, 12S-hydroxyeicosatetraenoic acid (12S-HETE) and 13S-hydroxyoctadecaenoic acid (13S-HODE) in vivo compared with C57BLKS/J mice. Using isolated primary aortic endothelial cells (ECs) from db/db mice and WEHI78/24 mouse monocyte cells in static adhesion assays, we found increased WEHI monocyte adhesion to db/db ECs (14 +/- 2 monocytes/field for db/db ECs versus 4 +/- 1 monocytes/field for C57BLKS/J ECs, p < 0.002). Thus, ECs from db/db mice appear to be "pre-activated" to bind monocytes. Analysis of db/db ECs revealed a 2-fold elevation in 12/15LO protein compared with C57BLKS/J EC. To determine that 12/15LO products were responsible for the increased monocyte adhesion observed with db/db ECs, we inhibited expression of murine 12/15LO using either an adenovirus expressing a ribozyme to 12/15LO (AdRZ) or with the 12/15LO inhibitor cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate. Treatment of db/db ECs for 48 h with AdRZ or 4 h with 10 microm cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate significantly reduced monocyte adhesion to db/db endothelium (p < 0.009). Thus, inhibition of the murine 12/15LO in db/db mice significantly reduced monocyte/endothelial interactions. We also found that adhesion of monocytes to diabetic db/db ECs was mediated by interactions of alpha4beta1 integrin on monocytes with endothelial vascular cell adhesion molecule 1 and connecting segment 1 fibronectin and interactions of beta2 integrins with endothelial intercellular adhesion molecule 1. In summary, regulation of the 12/15LO pathway is important for mediating early vascular changes in diabetes. Modulation of the 12/15LO pathway in the vessel wall may provide therapeutic benefit for early vascular inflammatory events in diabetes.


Subject(s)
Arachidonate 12-Lipoxygenase/biosynthesis , Arachidonate 15-Lipoxygenase/biosynthesis , Endothelium, Vascular/metabolism , Monocytes/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/urine , Animals , Aorta/metabolism , Cell Adhesion , Eicosanoids/metabolism , Fibronectins/metabolism , Flow Cytometry , Immunoassay , Inflammation , Islets of Langerhans/metabolism , Linoleic Acids/urine , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Phenotype , Reactive Oxygen Species
19.
Circ Res ; 92(4): 371-7, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12600878

ABSTRACT

We have shown that glucose increases monocyte adhesion to human aortic endothelial cells (HAECs) in vitro.1 In the present study, we examined mechanisms by which glucose stimulates monocyte:endothelial interactions. HAECs cultured for 7 days in 25 mmol/L glucose had a 2-fold elevation in interleukin-8 (IL-8) secretion over control cells cultured in 5.5 mmol/L glucose (P<0.001). Use of a neutralizing antibody to IL-8 prevented glucose-mediated monocyte adhesion. Both glucose and IL-8 activated beta1 integrin on the HAEC surface, suggesting that both activate the alpha5beta1 integrin complex on the endothelial surface. The alpha5beta1 integrin complex is important for anchoring connecting segment-1 fibronectin on the HAEC surface for monocyte adhesion. Analysis of the human IL-8 promoter revealed binding sites for NF-kappaB and AP-1 as well as several aligned carbohydrate response elements (also known as E-boxes). Glucose dramatically stimulated IL-8 promoter activity. Using mutated IL-8 promoter constructs and EMSA, we found that the AP-1 element and the glucose-response element were responsible for much of the glucose-mediated activation of IL-8 transcription. Interestingly, inhibition of reactive oxygen species (ROS) production through use of pharmacological uncouplers of the mitochondrial electron transport chain significantly reduced glucose-mediated induction of IL-8 expression. These data indicate that glucose regulates monocyte:endothelial interactions through stimulation of IL-8 and ROS production and activation of the alpha5beta1 integrin complex on HAECs.


Subject(s)
Endothelium, Vascular/drug effects , Glucose/pharmacology , Interleukin-8/biosynthesis , Monocytes/drug effects , Animals , Binding Sites/genetics , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , Diabetes Mellitus, Type 2/physiopathology , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-8/genetics , Mice , Mice, Inbred C57BL , Mice, Obese , Mitochondria/drug effects , Mitochondria/metabolism , Monocytes/cytology , Promoter Regions, Genetic/genetics , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Thenoyltrifluoroacetone/pharmacology , Transcription Factor AP-1/metabolism , Uncoupling Agents/pharmacology
20.
Am J Physiol Lung Cell Mol Physiol ; 282(3): L457-67, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11839539

ABSTRACT

After alveolar formation, >20% of interstitial lung fibroblasts undergo apoptosis, a process that is of critical importance for normal lung maturation. The immature lung contains two morphologically distinct fibroblast populations, lipid-filled interstitial fibroblasts (LIF) and non-LIF (NLIF), which differ with respect to contractile protein content, proliferative capacity, and expression of mRNAs for fibronectin and types I and III collagen, but not tropoelastin. After alveolarization, apoptosis occurs in only one fibroblast population, the LIF. Using flow cytometry to analyze fibroblasts stained with a lipophilic, fluorescent dye, we identified a subset, designated LIF(-), that contained fewer lipid droplets. Unlike LIF that retain lipid, LIF(+), the LIF(-) do not undergo apoptosis after alveolarization. In LIF(+), apoptosis was correlated with downregulation of insulin-like growth factor I receptor (IGF-IR) mRNA and cell surface protein expression. Treatment with anti-IGF-IR decreased total lung fibroblast survival (P = 0.05) as did treatment with the phosphatidylinositol 3-kinase inhibitor LY-294002 and the ras-raf-mitogen-activated protein kinase inhibitor PD-98059 (P < 0.002), which block IGF-I/insulin receptor survival pathways. These observations implicate downregulation of IGF-IR expression in fibroblast apoptosis after alveolar formation.


Subject(s)
Aging/metabolism , Animals, Newborn/metabolism , Apoptosis/physiology , Fibroblasts/physiology , Pulmonary Alveoli/embryology , Pulmonary Alveoli/growth & development , Receptor, IGF Type 1/metabolism , Animals , Animals, Newborn/growth & development , Cell Membrane/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Chromones/pharmacology , Down-Regulation , Embryonic and Fetal Development , Enzyme Inhibitors/pharmacology , Fetus/cytology , Fetus/metabolism , Fibroblasts/classification , Flavonoids/pharmacology , Intracellular Membranes/metabolism , Lipid Metabolism , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Pulmonary Alveoli/cytology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...