Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Am J Pathol ; 193(5): 638-653, 2023 05.
Article in English | MEDLINE | ID: mdl-37080662

ABSTRACT

Vascular smooth muscle cells (VSMC) play a critical role in the development and pathogenesis of intimal hyperplasia indicative of restenosis and other vascular diseases. Fragile-X related protein-1 (FXR1) is a muscle-enhanced RNA binding protein whose expression is increased in injured arteries. Previous studies suggest that FXR1 negatively regulates inflammation, but its causality in vascular disease is unknown. In the current study, RNA-sequencing of FXR1-depleted VSMC identified many transcripts with decreased abundance, most of which were associated with proliferation and cell division. mRNA abundance and stability of a number of these transcripts were decreased in FXR1-depleted hVSMC, as was proliferation (P < 0.05); however, increases in beta-galactosidase (P < 0.05) and γH2AX (P < 0.01), indicative of senescence, were noted. Further analysis showed increased abundance of senescence-associated genes with FXR1 depletion. A novel SMC-specific conditional knockout mouse (FXR1SMC/SMC) was developed for further analysis. In a carotid artery ligation model of intimal hyperplasia, FXR1SMC/SMC mice had significantly reduced neointima formation (P < 0.001) after ligation, as well as increases in senescence drivers p16, p21, and p53 compared with several controls. These results suggest that in addition to destabilization of inflammatory transcripts, FXR1 stabilized cell cycle-related genes in VSMC, and absence of FXR1 led to induction of a senescent phenotype, supporting the hypothesis that FXR1 may mediate vascular disease by regulating stability of proliferative mRNA in VSMC.


Subject(s)
Muscle, Smooth, Vascular , Vascular Diseases , Animals , Mice , Carotid Arteries/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Hyperplasia/pathology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , RNA, Messenger/metabolism , Vascular Diseases/pathology
2.
Arterioscler Thromb Vasc Biol ; 39(10): 2014-2027, 2019 10.
Article in English | MEDLINE | ID: mdl-31462091

ABSTRACT

OBJECTIVE: Stress granules (SGs) are dynamic cytoplasmic aggregates containing mRNA, RNA-binding proteins, and translation factors that form in response to cellular stress. SGs have been shown to contribute to the pathogenesis of several human diseases, but their role in vascular diseases is unknown. This study shows that SGs accumulate in vascular smooth muscle cells (VSMCs) and macrophages during atherosclerosis. Approach and Results: Immunohistochemical analysis of atherosclerotic plaques from LDLR-/- mice revealed an increase in the stress granule-specific markers Ras-G3BP1 (GTPase-activating protein SH3 domain-binding protein) and PABP (poly-A-binding protein) in intimal macrophages and smooth muscle cells that correlated with disease progression. In vitro, PABP+ and G3BP1+ SGs were rapidly induced in VSMC and bone marrow-derived macrophages in response to atherosclerotic stimuli, including oxidized low-density lipoprotein and mediators of mitochondrial or oxidative stress. We observed an increase in eIF2α (eukaryotic translation initiation factor 2-alpha) phosphorylation, a requisite for stress granule formation, in cells exposed to these stimuli. Interestingly, SG formation, PABP expression, and eIF2α phosphorylation in VSMCs is reversed by treatment with the anti-inflammatory cytokine interleukin-19. Microtubule inhibitors reduced stress granule accumulation in VSMC, suggesting cytoskeletal regulation of stress granule formation. SG formation in VSMCs was also observed in other vascular disease pathologies, including vascular restenosis. Reduction of SG component G3BP1 by siRNA significantly altered expression profiles of inflammatory, apoptotic, and proliferative genes. CONCLUSIONS: These results indicate that SG formation is a common feature of the vascular response to injury and disease, and that modification of inflammation reduces stress granule formation in VSMC.


Subject(s)
Atherosclerosis/metabolism , Cytoplasmic Granules/genetics , DNA Helicases/genetics , Gene Expression Regulation , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , Vascular System Injuries/metabolism , Animals , Atherosclerosis/pathology , Biopsy, Needle , Cells, Cultured , Cholesterol/pharmacology , DNA Helicases/metabolism , Disease Models, Animal , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Oxidative Stress , RNA Helicases/metabolism , Random Allocation , Sensitivity and Specificity , Vascular System Injuries/pathology
3.
FASEB J ; 33(3): 3304-3316, 2019 03.
Article in English | MEDLINE | ID: mdl-30383449

ABSTRACT

Interleukin enhancer-binding factor 3 (ILF3), an RNA-binding protein, is best known for its role in innate immunity by participation in cellular antiviral responses. A role for ILF3 in angiogenesis is unreported. ILF3 expression in CD31+ capillaries of hypoxic cardiac tissue was detected by immunohistochemistry. Proangiogenic stimuli induce ILF3 mRNA and protein expression in cultured human coronary artery endothelial cells (hCAECs). Angiogenic indices, including proliferation, migration, and tube formation, are all significantly reduced in hCAECs when ILF3 is knocked down using small interfering RNA (siRNA), but are significantly increased when ILF3 is overexpressed using adenovirus. Protein and mRNA abundance of several angiogenic factors including CXCL1, VEGF, and IL-8 are decreased when ILF3 is knocked down by siRNA. These factors are increased when ILF3 is overexpressed by adenovirus. ILF3 is phosphorylated and translocates from the nucleus to the cytoplasm in response to angiogenic stimuli. Proangiogenic transcripts containing adenine and uridine-rich elements were bound to ILF3 through RNA immunoprecipitation. ILF3 stabilizes proangiogenic transcripts including VEGF, CXCL1, and IL-8 in hCAECs. Together these data suggest that in endothelial cells, the RNA stability protein, ILF3, plays a novel and central role in angiogenesis. Our working hypothesis is that ILF3 promotes angiogenesis through cytokine-inducible mRNA stabilization of proangiogenic transcripts.-Vrakas, C. N., Herman, A. B., Ray, M., Kelemen, S. E., Scalia, R., Autieri, M. V. RNA stability protein ILF3 mediates cytokine-induced angiogenesis.


Subject(s)
Neovascularization, Physiologic , Nuclear Factor 90 Proteins/metabolism , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Endothelial Cells/metabolism , Gene Knockdown Techniques , Humans , Nuclear Factor 90 Proteins/antagonists & inhibitors , Nuclear Factor 90 Proteins/genetics , Phosphorylation , Protein Transport , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Swine , Up-Regulation
4.
Cell Rep ; 24(5): 1176-1189, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067974

ABSTRACT

This work identifies the fragile-X-related protein (FXR1) as a reciprocal regulator of HuR target transcripts in vascular smooth muscle cells (VSMCs). FXR1 was identified as an HuR-interacting protein by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The HuR-FXR1 interaction is abrogated in RNase-treated extracts, indicating that their association is tethered by mRNAs. FXR1 expression is induced in diseased but not normal arteries. siRNA knockdown of FXR1 increases the abundance and stability of inflammatory mRNAs, while overexpression of FXR1 reduces their abundance and stability. Conditioned media from FXR1 siRNA-treated VSMCs enhance activation of naive VSMCs. RNA EMSA and RIP demonstrate that FXR1 interacts with an ARE and an element in the 3' UTR of TNFα. FXR1 expression is increased in VSMCs challenged with the anti-inflammatory cytokine IL-19, and FXR1 is required for IL-19 reduction of HuR. This suggests that FXR1 is an anti-inflammation responsive, HuR counter-regulatory protein that reduces abundance of pro-inflammatory transcripts.


Subject(s)
ELAV-Like Protein 1/genetics , Muscle, Smooth, Vascular/metabolism , RNA Stability , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Cells, Cultured , ELAV-Like Protein 1/metabolism , Humans , Interleukins/genetics , Interleukins/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Protein Binding , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
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
6.
J Mol Cell Cardiol ; 105: 38-48, 2017 04.
Article in English | MEDLINE | ID: mdl-28257760

ABSTRACT

The transformation of vascular smooth muscle cells [VSMC] into foam cells leading to increased plaque size and decreased stability is a key, yet understudied step in atherogenesis. We reported that Interleukin-19 (IL-19), a novel, anti-inflammatory cytokine, attenuates atherosclerosis by anti-inflammatory effects on VSMC. In this work we report that IL-19 induces expression of miR133a, a muscle-specific miRNA, in VSMC. Although previously unreported, we report that miR133a can target and reduce mRNA abundance, mRNA stability, and protein expression of Low Density Lipoprotein Receptor Adaptor Protein 1, (LDLRAP1), an adaptor protein which functions to internalize the LDL receptor. Mutations in this gene lead to LDL receptor malfunction and cause the Autosomal Recessive Hypercholesterolemia (ARH) disorder in humans. Herein we show that IL-19 reduces lipid accumulation in VSMC, and LDLRAP1 expression and oxLDL uptake in a miR133a-dependent mechanism. We show that LDLRAP1 is expressed in plaque and neointimal VSMC of mouse and human injured arteries. Transfection of miR133a and LDLRAP1 siRNA into VSMC reduces their proliferation and uptake of oxLDL. miR133a is significantly increased in plasma from hyperlipidemic compared with normolipidemic patients. Expression of miR133a in IL-19 stimulated VSMC represents a previously unrecognized link between vascular lipid metabolism and inflammation, and may represent a therapeutic opportunity to combat vascular inflammatory diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Endothelial Cells/metabolism , Interleukins/metabolism , Lipoproteins, LDL/metabolism , MicroRNAs/genetics , Myocytes, Smooth Muscle/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cell Proliferation , Cells, Cultured , Cholesterol/metabolism , Gene Expression Regulation , Humans , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Mice , RNA Interference , RNA, Messenger/genetics
7.
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
8.
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
9.
Am J Pathol ; 184(7): 2134-43, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24814101

ABSTRACT

We tested the hypothesis that IL-19, a putative member of the type 2 helper T-cell family of anti-inflammatory interleukins, can attenuate intimal hyperplasia and modulate the vascular smooth muscle cell (VSMC) response to injury. Ligated carotid artery of IL-19 knockout (KO) mice demonstrated a significantly higher neointima/intima ratio compared with wild-type (WT) mice (P = 0.04). More important, the increased neointima/intima ratio in the KO could be reversed by injection of 10 ng/g per day recombinant IL-19 into the KO mouse (P = 0.04). VSMCs explanted from IL-19 KO mice proliferated significantly more rapidly than WT. This could be inhibited by addition of IL-19 to KO VSMCs (P = 0.04 and P < 0.01). IL-19 KO VSMCs migrated more rapidly compared with WT (P < 0.01). Interestingly, there was no type 1 helper T-cell polarization in the KO mouse, but there was significantly greater leukocyte infiltrate in the ligated artery in these mice compared with WT. IL-19 KO VSMCs expressed significantly greater levels of inflammatory mRNA, including IL-1ß, tumor necrosis factor α, and monocyte chemoattractant protein-1 in response to tumor necrosis factor α stimulation (P < 0.01 for all). KO VSMCs expressed greater adhesion molecule expression and adherence to monocytes. Together, these data indicate that IL-19 is a previously unrecognized counterregulatory factor for VSMCs, and its expression is an important protective mechanism in regulation of vascular restenosis.


Subject(s)
Interleukin-10/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/pathology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Female , Hyperplasia/pathology , Interleukins , Ligation , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Recombinant Proteins/metabolism , Tunica Intima/pathology
10.
Arterioscler Thromb Vasc Biol ; 33(10): 2316-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23950143

ABSTRACT

OBJECTIVE: Interleukin-19 (IL-19) is a putative Th2, anti-inflammatory interleukin. Its expression and potential role in atherogenesis are unknown. IL-19 is not detected in normal artery and is expressed to a greater degree in plaque from symptomatic versus asymptomatic patients, suggesting a compensatory counter-regulatory function. We tested whether IL-19 could reduce atherosclerosis in susceptible mice and identified plausible mechanisms. APPROACH AND RESULTS: LDLR(-/-) mice fed an atherogenic diet and injected with either 1.0 or 10.0 ng/g per day recombinant mouse IL-19 had significantly less plaque area in the aortic arch compared with controls (P<0.0001). Weight gain, cholesterol, and triglyceride levels were not significantly different. Gene expression in splenocytes from IL-19-treated mice demonstrated immune cell Th2 polarization, with decreased expression of T-bet, interferon-γ, interleukin-1ß, and interleukin-12ß and increased expression of GATA3 and FoxP3 mRNA. A greater percentage of lymphocytes were Th2 polarized in IL-19-treated mice. Cellular characterization of plaque by immunohistochemistry demonstrated that IL-19-treated mice have significantly less macrophage infiltrate compared with controls (P<0.001). Intravital microscopy revealed significantly less leukocyte adhesion in wild-type mice injected with IL-19 and fed an atherogenic diet compared with controls. Treatment of cultured endothelial cells, vascular smooth muscle cells, and bone marrow-derived macrophages with IL-19 resulted in a significant decrease in chemokine mRNA and mRNA stability protein human antigen R. CONCLUSIONS: These data suggest that IL-19 is a potent inhibitor of experimental atherosclerosis, with diverse mechanisms including immune cell polarization, decrease in macrophage adhesion, and decrease in gene expression. This may identify IL-19 as a novel therapeutic to limit vascular inflammation.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Interleukin-10/pharmacology , Aged , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers/blood , Carotid Artery Diseases/immunology , Carotid Artery Diseases/pathology , Cells, Cultured , Cholesterol/blood , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/immunology , Female , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Interleukins/metabolism , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Recombinant Proteins/pharmacology , Spleen/drug effects , Spleen/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Time Factors , Triglycerides/blood
11.
J Biol Chem ; 287(4): 2477-84, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22158875

ABSTRACT

Heme oxygenase-1 (HO-1) has potent anti-inflammatory activity and recognized vascular protective effects. We have recently described the expression and vascular protective effects of an anti-inflammatory interleukin (IL-19), in vascular smooth muscle cells (VSMC) and injured arteries. The objective of this study was to link the anti-inflammatory effects of IL-19 with HO-1 expression in resident vascular cells. IL-19 induced HO-1 mRNA and protein in cultured human VSMC, as assayed by quantitative RT-PCR, immunoblot, and ELISA. IL-19 does not induce HO-1 mRNA or protein in human endothelial cells. IL-19 activates STAT3 in VSMC, and IL-19-induced HO-1 expression is significantly reduced by transfection of VSMC with STAT3 siRNA or mutation of the consensus STAT binding site in the HO-1 promoter. IL-19 treatment can significantly reduce ROS-induced apoptosis, as assayed by Annexin V flow cytometry. IL-19 significantly reduced ROS concentrations in cultured VSMC. The IL-19-induced reduction in ROS concentration is attenuated when HO-1 is reduced by siRNA, indicating that the IL-19-driven decrease in ROS is mediated by HO-1 expression. IL-19 reduces vascular ROS in vivo in mice treated with TNFα. This points to IL-19 as a potential therapeutic for vascular inflammatory diseases and a link for two previously unassociated protective processes: Th2 cytokine-induced anti-inflammation and ROS reduction.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Heme Oxygenase-1/biosynthesis , Interleukins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , Humans , Interleukins/genetics , Interleukins/immunology , Mice , Muscle, Smooth, Vascular/immunology , Myocytes, Smooth Muscle/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/immunology , Reactive Oxygen Species/immunology , Response Elements/physiology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vasculitis/genetics , Vasculitis/immunology , Vasculitis/metabolism
12.
Atherosclerosis ; 220(1): 45-52, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21862018

ABSTRACT

Allograft inflammatory factor-1 (AIF-1) is a cytoplasmic, scaffold signal transduction protein constitutively expressed in inflammatory cells, but inducible in vascular smooth muscle cells (VSMCs) in response to injury or inflammatory stimuli. Although several basic science and population studies have reported increased AIF-1 expression in human and experimental atherosclerosis, a direct causal effect of AIF-1 expression on development of atherosclerosis has not been reported. The purpose of this study is to establish a direct relationship between AIF-1 expression and development of atherosclerosis. AIF-1 expression is detected VSMC in atherosclerotic lesions from ApoE(-/-) mice, but not normal arteries from wild-type mice. AIF-1 expression can be induced in cultured VSMC by stimulation with oxidized LDL (ox-LDL). Transgenic mice in which AIF-1 expression is driven by the G/C modified SM22 alpha promoter to restrict AIF-1 expression to VSMC develop significantly increased atherosclerosis compared with wild-type control mice when fed a high-fat diet (P=0.022). Cultured VSMC isolated from Tg mice demonstrated significantly increased migration in response to ox-LDL compared with matched controls (P<0.001). VSMC isolated from Tg mice and cultured human VSMC which over express AIF-1 demonstrated increased expression of MMP-2 and MMP-9 mRNA and protein and increased NF-κB activation in response to ox-LDL as compared with wild-type control mice. VSMC which over express AIF-1 have significantly increased uptake of ox-LDL, and increased CD36 expression. Together, these data suggest a strong association between AIF-1 expression, NF-κB activation, and development of experimental atherosclerosis.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Calcium-Binding Proteins/metabolism , Diet, High-Fat , Microfilament Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Diseases/etiology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/pathology , CD36 Antigens/metabolism , Calcium-Binding Proteins/genetics , Cell Movement , Cells, Cultured , Disease Models, Animal , Humans , Lipoproteins, LDL/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Time Factors , Transfection
13.
Arterioscler Thromb Vasc Biol ; 31(1): 167-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20966397

ABSTRACT

OBJECTIVE: To characterize the expression and function of interleukin (IL) 19, a recently described T-helper 2 anti-inflammatory IL, on endothelial cell (EC) pathophysiological features. METHODS AND RESULTS: The expression and effects of anti-inflammatory ILs on EC activation and development of angiogenesis are uncharacterized. We demonstrate by immunohistochemistry and immunoblot that IL-19 is expressed in inflamed, but not normal, human coronary endothelium and can be induced in cultured human ECs by serum and basic fibroblast growth factor. IL-19 is mitogenic and chemotactic, and it promotes EC spreading. IL-19 activates the signaling proteins STAT3, p44/42, and Rac1. In functional ex vivo studies, IL-19 promotes cordlike structure formation of cultured ECs and enhances microvessel sprouting in the mouse aortic ring assay. IL-19 induces tube formation in gelatinous protein (Matrigel) plugs in vivo. CONCLUSIONS: To our knowledge, these data are the first to report expression of the anti-inflammatory agent, IL-19, in ECs; and the first to indicate that IL-19 is mitogenic and chemotactic for ECs and can induce the angiogenic potential of ECs.


Subject(s)
Endothelial Cells/metabolism , Inflammation/prevention & control , Interleukins/metabolism , Neovascularization, Physiologic , Animals , Blotting, Western , Cell Proliferation , Cell Shape , Cells, Cultured , Chemotaxis , Endothelial Cells/immunology , Fibroblast Growth Factor 2/metabolism , Humans , Immunohistochemistry , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction , Time Factors , rac1 GTP-Binding Protein/metabolism
14.
Am J Physiol Cell Physiol ; 296(2): C256-66, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18787073

ABSTRACT

Endothelial cell (EC) activation plays a key role in vascular inflammation, thrombosis, and angiogenesis. Allograft inflammatory factor-1 (AIF-1) is a cytoplasmic, calcium-binding, inflammation-responsive scaffold protein that has been implicated in the regulation of inflammation. The expression and function of AIF-1 in EC is uncharacterized, and the purpose of this study was to characterize AIF-1 expression and function in ECs. AIF-1 expression colocalized with CD31-positive ECs in neointima of inflamed human arteries but not normal arteries. AIF-1 is detected at low levels in unstimulated EC, but expression can be increased in response to serum and soluble factors. Stable transfection of AIF-1 small interfering RNA (siRNA) in ECs reduced AIF-1 protein expression by 73% and significantly reduced EC proliferation and migration (P < 0.05 and 0.001). Rescue of AIF-1 expression restored both proliferation and migration of siRNA-expressing ECs, and AIF-1 overexpression enhanced both of these activities, suggesting a strong association between AIF-1 expression and EC activation. Activation of mitogen-activated protein kinase p44/42 and PAK1 was significantly reduced in siRNA ECs challenged with inflammatory stimuli. Reduction of AIF-1 expression did not decrease EC tube-like structure or microvessel formation from aortic rings, but overexpression of AIF-1 did significantly increase the number and complexity of these structures. These data indicate that AIF-1 expression plays an important role in signal transduction and activation of ECs and may also participate in new vessel formation.


Subject(s)
Cell Movement , Cell Proliferation , DNA-Binding Proteins/metabolism , Endothelial Cells/metabolism , Neovascularization, Physiologic , Signal Transduction , Animals , Aorta, Thoracic/metabolism , Calcium-Binding Proteins , Cattle , Cells, Cultured , Coronary Vessels/metabolism , Endothelial Cells/enzymology , Enzyme Activation , Female , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , RNA Interference , Time Factors , Tissue Culture Techniques , Transfection , p21-Activated Kinases/metabolism
15.
Cardiovasc Res ; 81(1): 206-15, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18779232

ABSTRACT

AIMS: Allograft inflammatory factor-1 (AIF-1) is a calcium-binding, scaffold-signalling protein expressed in vascular smooth muscle cells (VSMCs) in response to injury. The effects of AIF-1 attenuation on development of intimal hyperplasia are unknown, and the molecular mechanisms of these effects remain uncharacterized. The goals of the present study were to determine whether AIF-1 knockdown reduced VSMC proliferation, migration, and intimal hyperplasia, and determine AIF-1 effects on signal transduction in VSMCs. METHODS AND RESULTS: Balloon angioplasty-injured rat carotid arteries transduced with adenovirus to overexpress AIF-1 (AdAIF-1) significantly increased, and adenovirus to knock down AIF-1 (AdsiRNA) expression significantly decreased neointimal formation compared with green fluorescent protein (AdGFP) and Adscrambled controls (P < 0.05 and P < 0.01, n = 6). Primary rat VSMCs transduced with AdAIF-1 displayed a significant increase in proliferation, and AdsiRNA-transduced VSMCs proliferated significantly more slowly than controls (P < 0.05). VSMCs transduced with AdAIF-1 show increased migration when compared with control VSMCs (P < 0.01). Rat VSMCs transduced with AdAIF-1 showed constitutive and prolonged activation of the mitogen-activated protein kinase p38, whereas AdsiRNA-treated VSMCs showed decreased p38 activation compared with AdGFP (P < 0.05). Immunohistochemical analysis of AdAIF-1-transduced carotid arteries showed increased staining with a phospho-specific p38 antibody compared with AdGFP-transduced arteries. A specific p38 inhibitor abrogated AIF-1-induced VSMC proliferation, but not AIF-1-induced migration. CONCLUSION: Taken together, AIF-1 expression plays a key role in the development of neointimal hyperplasia. AIF-1 expression enhances the activation of p38 MAP kinase. AIF-1-enhanced proliferation is p38 kinase dependent, but AIF-1-enhanced VSMC migration is p38 independent.


Subject(s)
Calcium-Binding Proteins/metabolism , Carotid Arteries/metabolism , Carotid Arteries/pathology , Tunica Intima/metabolism , Tunica Intima/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Adenoviridae/genetics , Angioplasty, Balloon/adverse effects , Animals , Calcium-Binding Proteins/genetics , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Green Fluorescent Proteins/genetics , Hyperplasia/etiology , Hyperplasia/metabolism , Hyperplasia/pathology , Male , Microfilament Proteins , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
16.
Am J Pathol ; 173(3): 901-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18669613

ABSTRACT

Anti-inflammatory cytokines may play a protective role in the progression of vascular disease. The purpose of this study was to characterize interleukin (IL)-19 expression and function in the development of intimal hyperplasia, and discern a potential mechanism of its direct effects on vascular smooth muscle cells (VSMCs). IL-19 is an immunomodulatory cytokine, the expression of which is reported to be restricted to inflammatory cells. In the present study, we found that IL-19 is not expressed in quiescent VSMCs or normal arteries but is induced in human arteries by injury and in cultured human VSMCs by inflammatory cytokines. Recombinant IL-19 significantly reduced VSMC proliferation (37.1 +/- 4.8 x 10(3) versus 72.2 +/- 6.1 x 10(3) cells/cm(2)) in a dose-dependent manner. IL-19 adenoviral gene transfer significantly reduced proliferation and neointimal formation in balloon angioplasty-injured rat carotid arteries (0.172 +/- 29.9, versus 0.333 +/- 71.9, and 0.309 +/- 56.6 microm(2)). IL-19 induced activation of STAT3 as well as the expression of the suppressor of cytokine signaling 5 (SOCS5) in VSMCs. IL-19 treatment significantly reduced the activation of p44/42 and p38 MAPKs in stimulated VSMCs. Additionally, SOCS5 was found to interact with both p44/42 and p38 MAPKs in IL-19-treated human VSMCs. This is the first description of the expression of both IL-19 and SOCS5 in VSMCs and of the functional interaction between SOCS5 and MAPKs. We propose that through induction of SOCS5 and inhibition of signal transduction, IL-19 expression in VSMCs may represent a novel, protective, autocrine response of VSMCs to inflammatory stimuli.


Subject(s)
Interleukins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Signal Transduction/physiology , Tunica Intima/pathology , Animals , Blotting, Western , Carotid Artery Injuries/metabolism , Cell Proliferation , Gene Expression , Gene Expression Regulation , Humans , Hyperplasia , Immunohistochemistry , Immunoprecipitation , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Arterioscler Thromb Vasc Biol ; 28(1): 47-53, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17991871

ABSTRACT

OBJECTIVE: Allograft Inflammatory Factor-1 (AIF-1) is a calcium binding scaffold protein which is rapidly induced in vascular smooth muscle cells (VSMCs) in response to injury and inflammation. A transgenic mouse in which AIF-1 expression was driven by a VSMC-specific SM22alpha promoter was generated to establish a direct relationship between AIF-1 expression and intimal hyperplasia. METHODS AND RESULTS: Morphological analysis of partially ligated carotid artery demonstrate a significant increase in neointimal area of AIF-1 Tg versus wild-type mice (569+/-64 um versus 256+/-49 um, P=0.004). Immunohistochemistry using antibody to the proliferation marker Ki-67 show a significantly greater number of proliferating cells in the AIF-1 Tg lesion compared with wild-type arteries (10.6%+/-1.0 versus 3.6%+/-.9, P=0.0007). AIF-1 Tg arteries also had a greater number of cells with activated signal transduction kinase p38 (55.4%+/-7.0 versus 22.6%+/-5.4, P=0.002) and PAK1 (67.5%+/-6.7 versus 35.3%+/-10.2, P=0.02) compared with wild-type. Cultured VSMCs explanted from AIF-1 Tg proliferate (55.5+/-3.6x10(3) versus 37.2+/-2.0x10(3) cells/mL, P=0.0001) and migrate more rapidly (39.2+/-3.2 versus 17.1+/-1.5 VSMCs per HPF, P=0.0003) than wild-type, and have significantly greater levels of activated p38 and PAK1 than did VSMCs from wild-type littermates (P<0.05). CONCLUSIONS: These data indicate that AIF-1 expression results in increased signal transduction, neointimal formation, and VSMC proliferation in injured mouse carotid arteries.


Subject(s)
Calcium-Binding Proteins/physiology , Carotid Artery Diseases/physiopathology , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/physiology , Tunica Intima/pathology , Animals , Calcium-Binding Proteins/genetics , Carotid Arteries/physiopathology , Cell Proliferation , Disease Models, Animal , Hyperplasia , Mice , Mice, Transgenic , Microfilament Proteins , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Promoter Regions, Genetic , Signal Transduction , Tunica Intima/injuries , Tunica Intima/physiopathology
18.
COPD ; 5(6): 329-38, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19353346

ABSTRACT

Mucous metaplasia is an important determinant of small airway obstruction in COPD. Its relationship to small airway inflammation is poorly defined. We analyzed 4 to 6 small airways in 19 COPD patients, GOLD stages 0-4, from lobectomy or lung volume reduction surgery tissue samples. To identify intracellular mucin, periodic acid fluorescent Schiff's (PAFS) stained slides were imaged by fluorescence microscopy. PAFS+ staining area, basement membrane length (L(BM)), epithelial height and area were measured. Mucin was expressed as a percentage of epithelial area. Mucin volume density (MVD) was calculated as PAFS+ area divided by the product of L(BM) and 4/pi. Airways were Giemsa stained for eosinophils and immunostained with antibodies against CD3, CD4, CD8, CD68, and neutrophil elastase (NE), and the number of positively stained cells/mm(2) was quantified in the airway wall. Mucin percent correlated with CD3(+) cell density (r = 0.553, P < 0.0001), and MVD correlated with CD3(+) (r = 0.570, P < 0.0001) and CD8(+) cell density (r = 0.279, P = 0.016). There were weak negative correlations between mucin percent as well as MVD and CD68(+) cell density (r = -0.270, P = 0.02 and r = -0.245, P = 0.036). There was no relationship between epithelial mucin content and CD4(+), NE(+), or eosinophil cell density. CD3(+) and CD8(+) lymphocytic inflammation is related to small airway mucous metaplasia in COPD and may play a causative role in its development.


Subject(s)
Bronchioles/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/pathology , Aged , Bronchioles/metabolism , Female , Humans , Inflammation , Male , Metaplasia , Middle Aged , Mucins , Pulmonary Disease, Chronic Obstructive/metabolism , Respiratory Mucosa/metabolism
19.
Am J Physiol Cell Physiol ; 290(4): C1083-91, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16291819

ABSTRACT

Allograft inflammatory factor-1 (AIF-1) is a cytoplasmic, calcium-binding, inflammation-responsive scaffold protein. Several studies have reported increased AIF-1 expression in activated macrophages and have implicated AIF-1 as a marker of activated macrophages. However, the function of AIF-1 in macrophages and the mechanism whereby it participates in macrophage activation are unknown at this time. Immunohistochemical analysis colocalized AIF-1 expression with CD68-positive macrophages in atherosclerotic human coronary arteries. Subsequent experiments were designed to determine a role for AIF-1 in macrophage activation in response to atherogenic stimuli. Stimulation of human and murine macrophages with oxidized LDL significantly increased AIF-1 expression above basal levels. Stable transfection of AIF-1 small interfering RNA (siRNA) in macrophages reduced AIF-1 protein expression by 79% and reduced macrophage proliferation by 52% (P < 0.01). Inhibition of proliferation was not due to induction of apoptosis. Sequences that did not knock down AIF-1 expression had no effect on proliferation. AIF-1 siRNA expression reduced macrophage migration by 60% (P < 0.01). Both proliferation and migration of siRNA-expressing macrophages could be restored by adenoviral expression of AIF-1 (P < 0.001 and 0.005, respectively), suggesting a tight association between AIF-1 expression and macrophage activation. Phosphorylation of Akt, p44/42 MAPK, and p38 kinase were significantly reduced in siRNA macrophages challenged with oxidized LDL (P < 0.05). Phosphorylation of p38 kinase was significantly inhibited in siRNA macrophages stimulated with T lymphocyte conditioned medium (P < 0.05). These data indicate that AIF-1 mediates atherogenesis-initiated signaling and activation of macrophages.


Subject(s)
Atherosclerosis , Cell Movement/physiology , Cell Proliferation , DNA-Binding Proteins/metabolism , Macrophages/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/physiology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Calcium-Binding Proteins , Coronary Vessels/cytology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Culture Media, Conditioned/chemistry , DNA-Binding Proteins/genetics , Humans , Lipoproteins, LDL/pharmacology , Macrophage Activation , Macrophages/cytology , Macrophages/drug effects , Mice , Microfilament Proteins , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics
20.
Am J Pathol ; 167(2): 619-26, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16049345

ABSTRACT

Allograft inflammatory factor (AIF)-1 is a cytoplasmic, calcium-binding protein whose expression in transplanted human hearts correlates with rejection and development of coronary artery vasculopathy (CAV). AIF-1 is constitutively expressed in monocytes/macrophages, but its expression in human lymphocytes has not been described. After immunohistochemical analysis of human coronary arteries with CAV, we identified AIF-1 expression in CD3-positive lymphocytes. AIF-1 was differentially expressed in peripheral blood mononuclear cells and in the T-lymphoblastoid MOLT-4 cell line exposed to various cytokines, suggesting a role for AIF-1 in T-lymphocyte activation. To determine AIF-1 function, MOLT-4 cells were stably transduced by AIF-1 retrovirus. Overexpression of AIF-1 in these cells led to a 238% increase in cell number compared to empty vector controls. AIF-1 polymerized nonmuscle actin and MOLT-4 cells overexpressing AIF-1 migrated 95% more rapidly than empty vector controls. Primary human vascular smooth muscle cells cultured in conditioned media from AIF-1-transduced MOLT-4 cells proliferated 99% more rapidly than vascular smooth muscle cells cultured in conditioned media from empty vector-transduced MOLT-4 cells. These data indicate that AIF-1 is expressed in activated T lymphocytes, that its expression enhances activation of lymphocytes, and that AIF-1 expression in activated lymphocytes may have important ramifications for activation of adjacent arterial vascular smooth muscle cells and development of CAV.


Subject(s)
Arteriosclerosis/metabolism , Calcium-Binding Proteins/metabolism , Cell Proliferation , Coronary Vessels/metabolism , Muscle, Smooth, Vascular/metabolism , T-Lymphocytes/metabolism , Actins/metabolism , Arteriosclerosis/immunology , Arteriosclerosis/pathology , Cell Movement , Cells, Cultured , Coronary Vessels/pathology , Culture Media, Conditioned , Cytokines/metabolism , DNA-Binding Proteins , Humans , Lymphocyte Activation , Macrophages/metabolism , Microfilament Proteins , Muscle, Smooth, Vascular/cytology , Myocardium/cytology , Myocardium/metabolism , Myocardium/pathology , Retroviridae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL