Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 38(6): 1297-1308, 2018 06.
Article in English | MEDLINE | ID: mdl-29674474

ABSTRACT

OBJECTIVE: To test the hypothesis that loss of IL-19 (interleukin-19) exacerbates atherosclerosis. APPROACH AND RESULTS: Il19-/- mice were crossed into Ldlr-/- (low-density lipoprotein receptor knock out) mice. Double knockout (dKO) mice had increased plaque burden in aortic arch and root compared with Ldlr-/- controls after 14 weeks of high-fat diet (HFD). dKO mice injected with 10 ng/g per day rmIL-19 had significantly less plaque compared with controls. qRT-PCR and Western blot analysis revealed dKO mice had increased systemic and intraplaque polarization of T cells and macrophages to proinflammatory Th1 and M1 phenotypes, and also significantly increased TNF (tumor necrosis factor)-α expression in spleen and aortic arch compared with Ldlr-/- controls. Bone marrow transplantation suggests that immune cells participate in IL-19 protection. Bone marrow-derived macrophages and vascular smooth muscle cells isolated from dKO mice had a significantly greater expression of inflammatory cytokine mRNA and protein compared with controls. Spleen and aortic arch from dKO mice had significantly increased expression of the mRNA stability protein HuR (human antigen R). Bone marrow-derived macrophage and vascular smooth muscle cell isolated from dKO mice also had greater HuR abundance. HuR stabilizes proinflammatory transcripts by binding AU-rich elements in the 3' untranslated region. Cytokine and HuR mRNA stability were increased in dKO bone marrow-derived macrophage and vascular smooth muscle cell, which was rescued by addition of IL-19 to these cells. IL-19-induced expression of miR133a, which targets and reduced HuR abundance; miR133a levels were lower in dKO mice compared with controls. CONCLUSIONS: These data indicate that IL-19 is an atheroprotective cytokine which decreases the abundance of HuR, leading to reduced inflammatory mRNA stability.


Subject(s)
Aorta, Thoracic/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , ELAV-Like Protein 1/metabolism , Gene Deletion , Interleukin-10/deficiency , RNA Stability , RNA, Messenger/metabolism , Receptors, LDL/deficiency , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cells, Cultured , Disease Models, Animal , Disease Progression , ELAV-Like Protein 1/genetics , Female , Genetic Predisposition to Disease , Interleukin-10/administration & dosage , Interleukin-10/genetics , Interleukins , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Plaque, Atherosclerotic , RNA Stability/drug effects , RNA, Messenger/genetics , Receptors, LDL/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
Am J Physiol Cell Physiol ; 310(11): C931-41, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27053520

ABSTRACT

Neovascularization and inflammation are independent biological processes but are linked in response to injury. The role of inflammation-dampening cytokines in the regulation of angiogenesis remains to be clarified. The purpose of this work was to test the hypothesis that IL-19 can induce angiogenesis in the absence of tissue hypoxia and to identify potential mechanisms. Using the aortic ring model of angiogenesis, we found significantly reduced sprouting capacity in aortic rings from IL-19(-/-) compared with wild-type mice. Using an in vivo assay, we found that IL-19(-/-) mice respond to vascular endothelial growth factor (VEGF) significantly less than wild-type mice and demonstrate decreased capillary formation in Matrigel plugs. IL-19 signals through the IL-20 receptor complex, and IL-19 induces IL-20 receptor subunit expression in aortic rings and cultured human vascular smooth muscle cells, but not endothelial cells, in a peroxisome proliferator-activated receptor-γ-dependent mechanism. IL-19 activates STAT3, and IL-19 angiogenic activity in aortic rings is STAT3-dependent. Using a quantitative RT-PCR screening assay, we determined that IL-19 has direct proangiogenic effects on aortic rings by inducing angiogenic gene expression. M2 macrophages participate in angiogenesis, and IL-19 has indirect angiogenic effects, as IL-19-stimulated bone marrow-derived macrophages secrete proangiogenic factors that induce greater sprouting of aortic rings than unstimulated controls. Using a quantitative RT-PCR screen, we determined that IL-19 induces expression of angiogenic cytokines in bone marrow-derived macrophages. Together, these data suggest that IL-19 can promote angiogenesis in the absence of hypoxia by at least two distinct mechanisms: 1) direct effects on vascular cells and 2) indirect effects by stimulation of macrophages.


Subject(s)
Aorta, Thoracic/metabolism , Interleukin-10/metabolism , Neovascularization, Physiologic , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/immunology , Cells, Cultured , Collagen/pharmacology , Culture Media, Conditioned/metabolism , Drug Combinations , Endothelial Cells/immunology , Endothelial Cells/metabolism , Genotype , Humans , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukins , Laminin/pharmacology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/drug effects , PPAR gamma/genetics , PPAR gamma/metabolism , Phenotype , Proteoglycans/pharmacology , RNA Interference , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Time Factors , Tissue Culture Techniques , Transfection , Vascular Endothelial Growth Factor A/pharmacology
3.
Am J Physiol Cell Physiol ; 310(11): C921-30, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27053523

ABSTRACT

Vascular smooth muscle contraction is primarily regulated by phosphorylation of myosin light chain. There are also modulatory pathways that control the final level of force development. We tested the hypothesis that protein kinase C (PKC) and mitogen-activated protein (MAP) kinase modulate vascular smooth muscle activity via effects on MAP kinase phosphatase-1 (MKP-1). Swine carotid arteries were mounted for isometric force recording and subjected to histamine stimulation in the presence and absence of inhibitors of PKC [bisindolylmaleimide-1 (Bis)], MAP kinase kinase (MEK) (U0126), and MKP-1 (sanguinarine) and flash frozen for measurement of MAP kinase, PKC-potentiated myosin phosphatase inhibitor 17 (CPI-17), and caldesmon phosphorylation levels. CPI-17 was phosphorylated in response to histamine and was inhibited in the presence of Bis. Caldesmon phosphorylation levels increased in response to histamine stimulation and were decreased in response to MEK inhibition but were not affected by the addition of Bis. Inhibition of PKC significantly increased p42 MAP kinase, but not p44 MAP kinase. Inhibition of MEK with U0126 inhibited both p42 and p44 MAP kinase activity. Inhibition of MKP-1 with sanguinarine blocked the Bis-dependent increase of MAP kinase activity. Sanguinarine alone increased MAP kinase activity due to its effects on MKP-1. Sanguinarine increased MKP-1 phosphorylation, which was inhibited by inhibition of MAP kinase. This suggests that MAP kinase has a negative feedback role in inhibiting MKP-1 activity. Therefore, PKC catalyzes MKP-1 phosphorylation, which is reversed by MAP kinase. Thus the fine tuning of vascular contraction is due to the concerted effort of PKC, MAP kinase, and MKP-1.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Muscle, Smooth, Vascular/enzymology , Protein Kinase C/metabolism , Vasoconstriction , Animals , Calmodulin-Binding Proteins/metabolism , Carotid Arteries/enzymology , Dual Specificity Phosphatase 1/antagonists & inhibitors , Feedback, Physiological , In Vitro Techniques , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Swine , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
4.
Am J Pathol ; 186(5): 1361-74, 2016 05.
Article in English | MEDLINE | ID: mdl-26952642

ABSTRACT

Atherosclerosis regression is an important clinical goal, and treatments that can reverse atherosclerotic plaque formation are actively being sought. Our aim was to determine whether administration of exogenous IL-19, a Th2 cytokine, could attenuate progression of preformed atherosclerotic plaque and to identify molecular mechanisms. LDLR(-/-) mice were fed a Western diet for 12 weeks, then administered rIL-19 or phosphate-buffered saline concomitant with Western diet for an additional 8 weeks. Analysis of atherosclerosis burden showed that IL-19-treated mice were similar to baseline, in contrast to control mice which showed a 54% increase in plaque, suggesting that IL-19 halted the progression of atherosclerosis. Plaque characterization showed that IL-19-treated mice had key features of atherosclerosis regression, including a reduction in macrophage content and an enrichment in markers of M2 macrophages. Mechanistic studies revealed that IL-19 promotes the activation of key pathways leading to M2 macrophage polarization, including STAT3, STAT6, Kruppel-like factor 4, and peroxisome proliferator-activated receptor γ, and can reduce cytokine-induced inflammation in vivo. We identified a novel role for IL-19 in regulating macrophage lipid metabolism through peroxisome proliferator-activated receptor γ-dependent regulation of scavenger receptor-mediated cholesterol uptake and ABCA1-mediated cholesterol efflux. These data show that IL-19 can halt progression of preformed atherosclerotic plaques by regulating both macrophage inflammation and cholesterol homeostasis and implicate IL-19 as a link between inflammation and macrophage cholesterol metabolism.


Subject(s)
Atherosclerosis/drug therapy , Cholesterol/metabolism , Interleukin-10/pharmacology , Macrophages/metabolism , Plaque, Atherosclerotic/drug therapy , ATP Binding Cassette Transporter 1/metabolism , Animals , Biomarkers/metabolism , Diet, Western , Disease Progression , Female , Inflammation , Interleukins , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Lipid Metabolism/physiology , Macrophages/drug effects , Male , Mice, Knockout , PPAR gamma/metabolism , STAT Transcription Factors/metabolism , Transfection
5.
J Mol Cell Cardiol ; 79: 21-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450612

ABSTRACT

Hypoxia in ischemic limbs typically initiates angiogenic and inflammatory factors to promote angiogenesis in attempt to restore perfusion. There is a gap in our knowledge concerning the role of anti-inflammatory interleukins in angiogenesis, macrophage polarization, and endothelial cell activation. Interleukin-19 is a unique anti-inflammatory Th2 cytokine that promotes angiogenic effects in cultured endothelial cells (EC); the purpose of this study was to characterize a role for IL-19 in restoration of blood flow in hind-limb ischemia, and define potential mechanisms. Hind limb ischemia was induced by femoral artery ligation, and perfusion quantitated using Laser Doppler Perfusion Imaging (LDPI). Wild type mice which received i.p. injections of rIL-19 (10ng/g/day) showed significantly increased levels of perfusion compared to PBS controls. LDPI values were significantly decreased in IL-19(-/-) mice when compared to wild type mice. IL-19(-/-) mice injected with rIL-19 had significantly increased LDPI compared with PBS control mice. Significantly increased capillary density was quantitated in rIL-19 treated mice, and significantly less capillary density in IL-19(-/-) mice. Multiple cell types participate in IL-19 induced angiogenesis. IL-19 treatment of human microvascular EC induced expression of angiogenic cytokines. M2 macrophage marker and VEGF-A expression were significantly increased in macrophage and the spleen from rIL-19 injected mice, and M1 marker expression was significantly increased in the spleen from IL-19(-/-) compared with controls. Plasma VEGF-A levels are higher in rIL-19 injected mice. IL-19 decreased the expression of anti-angiogenic IL-12 in the spleen and macrophage. This study is the first to implicate IL-19 as a novel pro-angiogenic interleukin and suggests therapeutic potential for this cytokine.


Subject(s)
Cell Polarity , Endothelial Cells/metabolism , Hindlimb/blood supply , Interleukin-10/metabolism , Ischemia/pathology , Macrophages/cytology , Neovascularization, Physiologic , Animals , Capillaries/metabolism , Capillaries/pathology , Gene Expression Regulation , Hindlimb/pathology , Humans , Interleukin-10/deficiency , Interleukin-12 Subunit p40/metabolism , Interleukins , Ischemia/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Neovascularization, Physiologic/genetics , Phenotype , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...