Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
FASEB J ; 31(6): 2612-2624, 2017 06.
Article in English | MEDLINE | ID: mdl-28274989

ABSTRACT

NADPH oxidase-generated reactive oxygen species (ROS) from immune cells are well known to be important for pathogen killing in response to TLR ligands. Here, we investigated a new aspect of NADPH oxidase in the TLR2/6-induced release of the immunologically relevant GM-CSF by endothelial cells. Stimulation of human endothelial cells with TLR2/6 agonist, MALP-2 (macrophage-activating lipopeptide of 2 kDa), induced NADPH oxidase activation and ROS formation. Inhibition by ROS scavengers and NADPH oxidase inhibitors blocked MALP-2-induced GM-CSF release. NADPH oxidase activators or ROS donors alone did not result in GM-CSF secretion; however, additional superoxide supply augmented MALP-2-induced GM-CSF secretion and restored GM-CSF levels after NADPH oxidase inhibition. MALP-2-dependent NF-ĸB activation was suppressed by NADPH oxidase inhibition, and inhibition of NF-κB completely blunted MALP-2-induced GM-CSF release. Vascular explants from mice that were deficient for the NADPH oxidase subunit p47 phox showed diminished intimal superoxide production and GM-CSF release after ex vivo stimulation with MALP-2. Moreover, an increase in circulating progenitor cells after MALP-2 injection was completely abolished in p47phox-knockout mice. Finally, MALP-2 stimulation increased mRNA expression of the major subunit NADPH oxidase, (Nox)2, in endothelial cells, and Nox2 inhibition prevented MALP-2-induced GM-CSF release. Our findings identify a Nox2-containing NADPH oxidase as a crucial regulator of the immunologic important growth factor GM-CSF after TLR2/6 stimulation in endothelial cells.-Schuett, J., Schuett, H., Oberoi, R., Koch, A.-K., Pretzer, S., Luchtefeld, M., Schieffer, B., Grote, K. NADPH oxidase NOX2 mediates TLR2/6-dependent release of GM-CSF from endothelial cells.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 6/metabolism , ATPases Associated with Diverse Cellular Activities , Animals , Cell Survival , Cells, Cultured , DNA Helicases , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Lipopeptides/pharmacology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidases/genetics , NF-kappa B , Phosphorylation , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics
2.
PLoS One ; 11(7): e0160145, 2016.
Article in English | MEDLINE | ID: mdl-27467817

ABSTRACT

OBJECTIVE: It is well known that atherosclerotic inflammatory vascular disease is critically driven by oxidized lipids and cytokines. In this regard, tumor necrosis factor (TNF)-α is known as a crucial mediator of early pro-atherosclerotic events. Epidemiologic data suggest that blockade of TNF-α has beneficial effects on vascular outcomes in patients with rheumatoid arthritis, however, detailed mechanistic studies are still lacking. This study aims to elucidate effects of TNF-α blockade by adalimumab-which is approved for several inflammatory disorders-on endothelial activation and monocyte adhesion under pro-atherosclerotic conditions. METHODS AND RESULTS: Phorbol myristate acetate (PMA) differentiated THP-1 macrophages were stimulated with oxidized low density lipoprotein and subsequent analysis of this conditioned media (oxLDL CM) revealed a strong release of TNF-α. The TNF-α rich supernatant led to activation of human umbilical vein endothelial cells (HUVEC) as shown by enhanced expression of major adhesion molecules, such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1) and E-selectin which was suppressed by the TNF-α inhibitor adalimumab. Accordingly, adalimumab effectively prevented THP-1 monocyte adhesion to endothelial cells under static as well as under flow conditions. Furthermore, adalimumab suppressed endothelial leakage as shown by Evan's blue diffusion across a confluent endothelial monolayer. Of note, after intraperitoneal injection we detected abundant deposition of fluorophore-labelled adalimumab in atherosclerotic plaques of hypercholesterolemic mice. CONCLUSION: Our results show that adalimumab prevents major inflammatory effects of TNF-α on endothelial activation, endothelial monocyte adhesion, endothelial leakage and therefore extends the therapeutic options of adalimumab to limit vascular inflammation.


Subject(s)
Adalimumab/pharmacology , Cell Adhesion/drug effects , Endothelial Cells/drug effects , Monocytes/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Cell Line , Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells , Humans , Lipoproteins, LDL/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Tumor Necrosis Factor-alpha/metabolism
3.
PLoS One ; 10(9): e0137924, 2015.
Article in English | MEDLINE | ID: mdl-26367277

ABSTRACT

BACKGROUND: Lipocalin (LCN) 2 is associated with multiple acute and chronic inflammatory diseases but the underlying molecular and cellular mechanisms remain unclear. Here, we investigated whether LCN2 is released from macrophages and contributes to pro-atherosclerotic processes and whether LCN2 plasma levels are associated with the severity of coronary artery disease progression in humans. METHODS AND RESULTS: In an autocrine-paracrine loop, tumor necrosis factor (TNF)-α promoted the release of LCN2 from murine bone-marrow derived macrophages (BMDM) and vice versa. Moreover, LCN2 stimulation of BMDM led to up-regulation of M1 macrophage markers. In addition, enhanced migration of monocytic J774A.1 cells towards LCN2 was observed. Furthermore, LCN2 increased the expression of the scavenger receptors Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) as well as scavenger receptor class A-1 (SRA-1) and induced the conversion of macrophages to foam cells. In atherosclerotic lesions of low density lipoprotein receptor-deficient (ldlr-/-) mice fed a high fat, high cholesterol diet, LCN2 was found to be co-localized with macrophages in the shoulder region of the atherosclerotic plaque. In addition, LCN2 plasma levels were significantly increased in plasma samples of these mice. Finally, LCN2 plasma levels correlated with the severity of coronary artery disease (CAD) in patients as determined by coronary angiography. CONCLUSIONS: Here we demonstrated that LCN2 plays a pivotal role in processes involved in atherogenesis by promoting polarization and migration of monocytic cells and development of macrophages towards foam cells. Moreover, LCN2 may be used as a prognostic marker to determine the status of CAD progression.


Subject(s)
Bone Marrow Cells/metabolism , Coronary Artery Disease/blood , Foam Cells/metabolism , Lipocalins/blood , Oncogene Proteins/blood , Proto-Oncogene Proteins/blood , Acute-Phase Proteins/genetics , Animals , Bone Marrow Cells/pathology , Cell Line , Coronary Angiography , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/genetics , Female , Foam Cells/pathology , Humans , Lipocalin-2 , Lipocalins/genetics , Male , Mice , Mice, Knockout , Oncogene Proteins/genetics , Proto-Oncogene Proteins/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Scavenger Receptors, Class E/genetics , Scavenger Receptors, Class E/metabolism
4.
PLoS One ; 8(2): e55265, 2013.
Article in English | MEDLINE | ID: mdl-23408963

ABSTRACT

OBJECTIVES: We here investigated whether experimental gingivitis enhances systemic markers of inflammation which are also known as surrogate markers of atherosclerotic plaque development. BACKGROUND: Gingivitis is a low-level oral infection induced by bacterial deposits with a high prevalence within Western populations. A potential link between the more severe oral disease periodontitis and cardiovascular disease has already been shown. METHODS: 37 non-smoking young volunteers with no inflammatory disease or any cardiovascular risk factors participated in this single-subject interventional study with an intra-individual control. Intentionally experimental oral inflammation was induced by the interruption of oral hygiene for 21 days, followed by a 21-days resolving phase after reinitiation of oral hygiene. Primary outcome measures at baseline, day 21 and 42 were concentrations of hsCRP, IL-6, and MCP-1, as well as adhesion capacity and oxLDL uptake of isolated blood monocytes. RESULTS: The partial cessation of oral hygiene procedures was followed by the significant increase of gingival bleeding (34.0%, P<0.0001). This local inflammation was associated with a systemic increase in hsCRP (0.24 mg/L, P = 0.038), IL-6 (12.52 ng/L, P = 0.0002) and MCP-1 (9.10 ng/l, P = 0.124) in peripheral blood samples between baseline and day 21, which decreased at day 42. Monocytes showed an enhanced adherence to endothelial cells and increased foam cell formation after oxLDL uptake (P<0.050) at day 21 of gingivitis. CONCLUSIONS: Bacterial-induced gingival low-level inflammation induced a systemic increase in inflammatory markers. Dental hygiene almost completely reversed this experimental inflammatory process, suggesting that appropriate dental prophylaxis may also limit systemic markers of inflammation in subjects with natural gingivitis. International Clinical Trials Register Platform of the World Health Organization, registry number: DRKS00003366, URL: http://apps.who.int/trialsearch/Default.aspx.


Subject(s)
Biomarkers/blood , Gingivitis/physiopathology , Inflammation/physiopathology , Adult , Base Sequence , Cells, Cultured , DNA Primers , Dental Plaque/microbiology , Female , Gingivitis/blood , Gingivitis/microbiology , Humans , Inflammation/blood , Male , Polymerase Chain Reaction , Reference Values , Young Adult
5.
Hepatology ; 57(5): 1716-24, 2013 May.
Article in English | MEDLINE | ID: mdl-23212706

ABSTRACT

UNLABELLED: Oxidized low-density lipoprotein (oxLDL) has been reported as an inhibitor of hepatitis C virus (HCV) cell entry, making it the only known component of human lipid metabolism with an antiviral effect on HCV. However, several questions remain open, including its effect on full-length cell-culture-grown HCV (HCVcc) of different genotypes or on other steps of the viral replication cycle, its mechanism of action, and whether endogenous oxLDL shares the anti-HCV properties of in vitro-generated oxLDL. We combined molecular virology tools with oxLDL serum measurements in different patient cohorts to address these questions. We found that oxLDL inhibits HCVcc at least as potently as HCV pseudoparticles. There was moderate variation between genotypes, with genotype 4 appearing the most oxLDL sensitive. Intracellular RNA replication and assembly and release of new particles were unaffected. HCV particles entering target cells lost oxLDL sensitivity with time kinetics parallel to anti-SR-BI (scavenger receptor class B type I), but significantly earlier than anti-CD81, suggesting that oxLDL acts by perturbing interaction between HCV and SR-BI. Finally, in chronically HCV-infected individuals, endogenous serum oxLDL levels did not correlate with viral load, but in HCV-negative sera, high endogenous oxLDL had a negative effect on HCV infectivity in vitro. CONCLUSION: oxLDL is a potent pangenotype HCV entry inhibitor that maintains its activity in the context of human serum and targets an early step of HCV entry.


Subject(s)
Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis C, Chronic/blood , Lipoproteins, LDL/pharmacology , Virus Replication/drug effects , CD36 Antigens/physiology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Cells, Cultured , DNA, Viral/genetics , Genotype , Hepacivirus/drug effects , Humans , In Vitro Techniques , Lipoproteins, LDL/blood , Liver Neoplasms/pathology , Liver Neoplasms/virology , Viral Load , Virion/physiology , Virus Replication/physiology
6.
PLoS One ; 7(12): e51608, 2012.
Article in English | MEDLINE | ID: mdl-23300554

ABSTRACT

BACKGROUND: While the impact of inflammation as the substantial driving force of atherosclerosis has been investigated in detail throughout the years, the influence of negative regulators of pro-atherogenic pathways on plaque development has remained largely unknown. Suppressor of cytokine signaling (SOCS)-1 potently restricts transduction of various inflammatory signals and, thereby modulates T-cell development, macrophage activation and dendritic cell maturation. Its role in atherogenesis, however has not been elucidated so far. METHODS AND RESULTS: Loss of SOCS-1 in the low-density lipoprotein receptor deficient murine model of atherosclerosis resulted in a complex, systemic and ultimately lethal inflammation with increased generation of Ly-6C(hi) monocytes and activated macrophages. Even short-term exposure of these mice to high-cholesterol dieting caused enhanced atherosclerotic plaque development with accumulation of M1 macrophages, Ly-6C positive cells and neutrophils. CONCLUSION: Our data not only imply that SOCS-1 is athero-protective but also emphasize the fundamental, regulatory importance of SOCS-1 in inflammation-prone organisms.


Subject(s)
Antigens, Ly/metabolism , Atherosclerosis/etiology , Cytokines/metabolism , Inflammation/etiology , Monocytes/pathology , Receptors, LDL/physiology , Suppressor of Cytokine Signaling Proteins/physiology , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , DNA-Binding Proteins/physiology , Diet, High-Fat , Female , Flow Cytometry , Immunoenzyme Techniques , Inflammation/metabolism , Inflammation/pathology , Macrophage Activation , Male , Mice , Mice, Knockout , Monocytes/metabolism , Neutrophils/metabolism , Neutrophils/pathology , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein
7.
Arterioscler Thromb Vasc Biol ; 32(2): 281-90, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22075248

ABSTRACT

OBJECTIVE: Transsignaling of interleukin (IL)-6 is a central pathway in the pathogenesis of disorders associated with chronic inflammation, such as Crohn disease, rheumatoid arthritis, and inflammatory colon cancer. Notably, IL-6 also represents an independent risk factor for coronary artery disease (CAD) in humans and is crucially involved in vascular inflammatory processes. METHODS AND RESULTS: In the present study, we showed that treatment with a fusion protein of the natural IL-6 transsignaling inhibitor soluble glycoprotein 130 (sgp130) and IgG1-Fc (sgp130Fc) dramatically reduced atherosclerosis in hypercholesterolemic Ldlr(-/-) mice without affecting weight gain and serum lipid levels. Moreover, sgp130Fc treatment even led to a significant regression of advanced atherosclerosis. Mechanistically, endothelial activation and intimal smooth muscle cell infiltration were decreased in sgp130Fc-treated mice, resulting in a marked reduction of monocyte recruitment and subsequent atherosclerotic plaque progression. Of note, patients with CAD exhibited significantly lower plasma levels of endogenous sgp130, suggesting that a compromised counterbalancing of IL-6 transsignaling may contribute to atherogenesis in humans. CONCLUSIONS: These data clarify, for the first time, the critical involvement of, in particular, the transsignaling of IL-6 in CAD and warrant further investigation of sgp130Fc as a novel therapeutic for the treatment of CAD and related diseases.


Subject(s)
Atherosclerosis/physiopathology , Coronary Artery Disease/physiopathology , Disease Progression , Interleukin-6/physiology , Signal Transduction/physiology , Aged , Animals , Atherosclerosis/prevention & control , Coronary Artery Disease/blood , Coronary Artery Disease/prevention & control , Cytokine Receptor gp130/blood , Cytokine Receptor gp130/pharmacology , Cytokine Receptor gp130/therapeutic use , Disease Models, Animal , Female , Humans , Hypercholesterolemia/blood , Hypercholesterolemia/physiopathology , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptors, LDL/deficiency , Receptors, LDL/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Signal Transduction/drug effects
8.
PLoS One ; 6(5): e19427, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21573245

ABSTRACT

BACKGROUND: Elevated levels of acute phase proteins (APP) are often found in patients with cardiovascular diseases. In a previous study, we demonstrated the importance of the IL-6-gp130 axis -as a key regulator of inflammatory acute phase signaling in hepatocytes-for the development of atherosclerosis. BACKGROUND/PRINCIPAL FINDINGS: Gp130-dependent gene expression was analyzed in a previously established hepatocyte-specific gp130 knockout mouse model. We performed whole transcriptome analysis in isolated hepatocytes to measure tissue specific responses after proinflammatory stimulus with IL-6 across different time points. Our analyses revealed an unexpected small gene cluster that requires IL-6 stimulus for early activation. Several of the genes in this cluster are involved in different cell defense mechanisms. Thus, stressors that trigger both general stress and inflammatory responses lead to activation of a stereotypic innate cellular defense response. Furthermore, we identified a potential biomarker Lipocalin (LCN) 2 for the gp130 dependent early inflammatory response. CONCLUSIONS/SIGNIFICANCE: Our findings suggest a complex network of tightly linked genes involved in the early activation of different parts of the innate immune response including acute phase proteins, complement and coagulation cascade.


Subject(s)
Acute-Phase Proteins/genetics , Cytokine Receptor gp130/metabolism , Immunity, Innate/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Cytokine Receptor gp130/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Hepatocytes/drug effects , Hepatocytes/metabolism , Immunity, Innate/genetics , Interleukin-6/pharmacology , Lipocalin-2 , Lipocalins/genetics , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/genetics , Polymerase Chain Reaction , Serum Amyloid A Protein/genetics , Signal Transduction/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics
9.
Trends Cardiovasc Med ; 21(8): 211-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22902067

ABSTRACT

The CC chemokine receptor 7 (CCR7) and its ligands CCL19 and CCL21 essentially contribute to both immunity and tolerance by directing T cells and antigen-presenting dendritic cells (DCs) to and within lymph organs. In the pathogenesis of atherosclerosis, the accumulation of cholesterol in the subendothelial space of the vessel wall represents the initial step of plaque development in which DCs acquire and process low-density lipoprotein cholesterol as antigen in the vessel wall and then migrate to draining lymph nodes and present this antigen to naive T cells. Deletion of CCR7 receptor in murine atherosclerosis not only results in a reduced atherosclerotic plaque content but also leads to a disturbed entry and exit of T cells within the inflamed vessel wall. These observations are consistent with the notion that CCR7-dependent T cell priming in secondary lymphoid organs and CCR7-dependent recirculation of T cells between secondary lymphoid organs and inflamed tissue is pivotal for atherosclerotic plaque development and may represent an interesting target for innovative immune-modulatory therapy.


Subject(s)
Atherosclerosis/immunology , Receptors, CCR7/metabolism , Signal Transduction , Animals , Atherosclerosis/therapy , Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Chemotaxis , Cholesterol/immunology , Dendritic Cells/immunology , Humans , Immunity, Innate , Immunotherapy/methods , Ligands , T-Lymphocytes/immunology
10.
Circulation ; 122(16): 1621-8, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20921438

ABSTRACT

BACKGROUND: Atherosclerosis is a systemic inflammatory disease characterized by the formation of atherosclerotic plaques. Both innate immunity and adaptive immunity contribute to atherogenesis, but the mode of interaction is poorly understood. Chemokine receptor 7 (CCR7) is critically involved in the transition from innate to adaptive immune activation by coordinating the migration to and positioning of antigen-presenting dendritic cells and T cells in secondary lymphoid organs. More recently, it was shown that CCR7 is also responsible for T-cell migration into inflamed tissues and T-cell egress from these tissues via the afferent lymph. Thus, we investigated the influence of a systemic CCR7 deficiency on atherogenesis in atherosclerosis-prone low-density lipoprotein receptor (ldlr) knockout mice. METHODS AND RESULTS: CCR7 deficiency resulted in reduced atherosclerotic plaque development. CCR7(-/-) T cells showed impaired entry and exit behavior from atherosclerotic lesions. Oxidized low-density lipoprotein, a key molecule for atherogenesis with antigenic features, was used to pulse dendritic cells and to expand T cells ex vivo. Adoptive transfer of C57BL/6 wild-type T cells but not ccr7(-/-)-derived T cells primed with oxidized low-density lipoprotein-pulsed dendritic cells resulted in a reconstitution of atherogenesis in ccr7(-/-)/ldlr(-/-) mice. CONCLUSION: These results demonstrate that both CCR7-dependent T-cell priming in secondary lymphoid organs and CCR7-dependent recirculation of T cells between secondary lymphoid organs and inflamed tissue are crucially involved in atherosclerotic plaque development.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Receptors, CCR7/deficiency , Adaptive Immunity/physiology , Animals , Aortic Diseases/metabolism , Aortic Diseases/physiopathology , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Body Weight/physiology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , Cell Movement/physiology , Disease Models, Animal , Immunity, Innate/physiology , Lipoproteins, LDL/pharmacology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR7/genetics
11.
Hypertension ; 54(5): 1035-42, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19805641

ABSTRACT

Inflammation and vascular remodeling are hallmarks of atherosclerosis, hypertension, and restenosis after angioplasty. Here we investigated the role of the hepatocyte gp130-dependent systemic acute phase response on vascular remodeling after carotid artery ligation. Mice with a hepatocyte-specific gp130 knockout on an apolipoprotein E(-/-) background (gp130-) were compared with control mice (gp130(flox)). Vascular remodeling was induced by permanent ligation of the left common carotid artery. This, in turn, activated the systemic acute phase reaction in gp130(flox) mice, as measured by serum amyloid A plasma levels, which was completely abrogated in gp130- mice (P<0.05). Morphometric analysis of the carotid artery revealed severe neointima formation and media thickening 28 days after ligation in gp130(flox) mice, which was suppressed in gp130- mice (P<0.01). Serial sections from gp130- carotid segments showed significantly less smooth muscle cell (SMC) proliferation and monocyte recruitment (P<0.01). To evaluate the impact of the gp130-dependent systemic acute phase response on SMCs, hepatocytes from gp130(flox) and gp130- mice were stimulated with interleukin 6. Interleukin 6-induced secretion of serum amyloid A was completely abolished in gp130- hepatocytes (P<0.01). Moreover, when stimulated with supernatants from gp130- hepatocytes, SMCs showed significantly less migration and proliferation compared with supernatants from gp130(flox) hepatocytes (P<0.01). Recombinant serum amyloid A induced SMC migration and proliferation (P<0.05) and serum amyloid A injection after carotid artery ligation restored vascular remodeling in gp130- mice (P<0.01). These results imply a critical role for the gp130-dependent systemic acute phase response for vascular inflammation and SMC migration, as well as proliferation, and, subsequently, for vascular remodeling.


Subject(s)
Carotid Artery, Common/surgery , Cytokine Receptor gp130/deficiency , Muscle, Smooth, Vascular/physiology , Neovascularization, Physiologic/physiology , Serum Amyloid A Protein/pharmacology , Animals , Carotid Artery, Common/physiology , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Hepatocytes , Ligation , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Neovascularization, Physiologic/drug effects , Probability , Random Allocation , Sensitivity and Specificity , Tunica Intima/cytology , Tunica Intima/metabolism , Tunica Media/cytology , Tunica Media/metabolism
12.
Thromb Haemost ; 102(2): 215-22, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19652871

ABSTRACT

The importance of inflammation as a driver of pathology is no longer confined to autoimmune and infectious diseases. In line with convincing experimental data as well as abundant clinical findings the current view of atherosclerosis points to inflammation as a critical regulator of atherosclerotic plaque formation and progression leading to the fatal clinical endpoints myocardial infarction, stroke or sudden cardiac death. The underlying mechanisms have been a matter of intense research during the last decades. In this regard, the interleukin-6 (IL-6) cytokines and their signalling events have been shown to contribute to both, atherosclerotic plaque development and plaque destabilisation via a variety of mechanisms. These involve the release of other pro-inflammatory cytokines, oxidation of lipoproteins by phospholipases, stimulation of acute phase protein secretion, the release of prothrombotic mediators, and the activation of matrix metalloproteinases. Moreover, the formation of reactive oxygen species generated by vascular enzyme systems may play a critical role in the regulation of IL-6 indicating a cross talk between vasoactive substances i.e. angiotensin II or adrenalin and pro-inflammatory cytokines such as IL-6. In this review we will summarise and discuss the underlying molecular and cellular mechanisms how IL-6 as an early and central regulator of inflammation contributes to atherosclerosis and how this knowledge can be integrated into the clinical context.


Subject(s)
Atherosclerosis/etiology , Interleukin-6/physiology , Acute-Phase Reaction/physiopathology , Animals , Atherosclerosis/physiopathology , Atherosclerosis/therapy , Cytokine Receptor gp130/physiology , Disease Models, Animal , Humans , Mice , Models, Cardiovascular , Renin-Angiotensin System/physiology , Signal Transduction/physiology
13.
J Mol Cell Cardiol ; 45(3): 394-403, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18671980

ABSTRACT

Cell-based therapy after myocardial infarction (MI) is a promising therapeutic option but the relevant cell subsets and dosage requirements are poorly defined. We hypothesized that cell therapy for myocardial infarction is improved by ex vivo expansion and high-dose transplantation of defined hematopoietic progenitor cells (HPCs). Since beta-catenin promotes self-renewal of stem cells we evaluated the therapeutic efficacy of beta-catenin-mediated ex vivo expansion of mouse HPCs in a mouse model of myocardial ischemia/reperfusion followed by intraarterial cell delivery. The impact of cell dose was determined by comparing a low-dose (LD, 5 x 10(5) cells) vs. a high-dose (HD, 1 x 10(7) cells) cell transplantation regimen of beta-catenin-HPCs. The impact of beta-catenin modification of HPCs was determined by comparing control-transduced HPCs (GFP-HPCs) vs. transgenic beta-catenin-HPCs. HD beta-catenin-HPCs significantly improved LV function and end-systolic and end-diastolic dimensions as compared to saline and LD beta-catenin-HPCs. Furthermore, while treatment with HD GFP-HPC resulted in a modest cardiac improvement the application of beta-catenin-HPCs was superior, resulting in a significant improvement in EF, FS and LVESD over saline and control GFP-HPC treatment. Although myocardial engraftment of HPCs was only transient, as determined by cell quantification after dye labeling, beta-catenin-HPC treatment significantly decreased infarct size, reduced cardiomyocyte apoptosis and increased capillary angiogenesis in vitro and in vivo. Ex vivo expanded HPCs improve cardiac function and remodeling post MI in a cell number- and beta-catenin-dependent manner.


Subject(s)
Heart/physiopathology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardium/metabolism , Transduction, Genetic , beta Catenin/genetics , Animals , Cells, Cultured , Disease Models, Animal , Heart Function Tests , Leukocyte Count , Mice , Myocardial Infarction/pathology , Myocardium/pathology , Ventricular Remodeling/physiology , beta Catenin/physiology
14.
Eur Heart J ; 29(16): 1956-65, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18565968

ABSTRACT

AIMS: To evaluate the impact of a combined treatment of angiotensin II type 1 (AT(1))-receptor blockade and 3-hydroxy-3-methyl-glutaryl-CoA-reductase inhibition (statin) on the secretory phospholipase A(2) type IIA (sPLA(2)-IIA) and oxidized low density lipoprotein (oxLDL) in patients with coronary artery disease (CAD). METHODS AND RESULTS: Sixty patients with angiographically documented CAD and a history of arterial hypertension were randomized in a double-blinded fashion to pravastatin (PRAV, 40 mg/day, n = 30) or PRAV plus irbesartan (PRAV+IRB, 40 mg/day+300 mg/day, n = 30) and were treated for 3 months. Blood pressure (BP) and cholesterol fractions were determined at baseline and after 3 months. SPLA(2) activity as primary endpoint, sPLA(2)-IIA protein, oxLDL levels, and high-sensitivity (hs)-C-reactive protein were measured by an enzyme-linked immunabsorbent assay. In both treatment groups, systolic BP levels and circulating HDL and LDL levels were reduced to the same extent. The combined treatment of PRAV+IRB significantly decreased sPLA(2)-IIA activity and sPLA(2)-IIA-protein concentration compared with PRAV treatment alone (P < 0.05). In addition, PRAV+IRB significantly reduced oxLDL levels compared with PRAV treatment alone (P < 0.05). This effect was independent of changes in LDL cholesterol levels. CONCLUSION: These findings are consistent with the notion that the combined treatment of pravastatin with irbesartan reduced sPLA(2)-IIA-activity, sPLA(2)-IIA-protein concentration, and oxLDL in patients with CAD suggesting a novel anti-atherogenic effect by combining AT(1)-receptor blockade with statin treatment.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Coronary Artery Disease/drug therapy , Group II Phospholipases A2/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Lipoproteins, LDL/blood , Adult , Aged , Analysis of Variance , Biphenyl Compounds/administration & dosage , C-Reactive Protein/analysis , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Humans , Irbesartan , Male , Middle Aged , Pravastatin/administration & dosage , Tetrazoles/administration & dosage , Treatment Outcome
15.
J Exp Med ; 204(8): 1935-44, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17664290

ABSTRACT

Liver-derived acute phase proteins (APPs) emerged as powerful predictors of cardiovascular disease and cardiovascular events, but their functional role in atherosclerosis remains enigmatic. We report that the gp130 receptor, which is a key component of the inflammatory signaling pathway within hepatocytes, influences the risk of atherosclerosis in a hepatocyte-specific gp130 knockout. Mice on an atherosclerosis-prone genetic background exhibit less aortic atherosclerosis (P < 0.05) with decreased plaque macrophages (P < 0.01). Translating these findings into humans, we show that genetic variation within the human gp130 homologue, interleukin 6 signal transducer (IL6ST), is significantly associated with coronary artery disease (CAD; P < 0.05). We further show a significant association of atherosclerotic disease at the ostium of the coronary arteries (P < 0.005) as a clinically important and heritable subphenotype in a large sample of families with myocardial infarction (MI) and a second independent population-based cohort. Our results reveal a central role of a hepatocyte-specific, gp130-dependent acute phase reaction for plaque development in a murine model of atherosclerosis, and further implicate IL6ST as a genetic susceptibility factor for CAD and MI in humans. Thus, the acute phase reaction should be considered an important target for future drug development in the management of CAD.


Subject(s)
Atherosclerosis/metabolism , Cytokine Receptor gp130/physiology , Animals , Aorta/metabolism , Coronary Vessels/metabolism , Cytokine Receptor gp130/metabolism , Genetic Predisposition to Disease , Hepatocytes/metabolism , Humans , Inflammation , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Polymorphism, Genetic , Risk
16.
Arterioscler Thromb Vasc Biol ; 27(5): 1184-90, 2007 May.
Article in English | MEDLINE | ID: mdl-17347483

ABSTRACT

OBJECTIVE: Thrombus formation after atherosclerotic plaque rupture critically involves the platelet collagen receptor glycoprotein (GP) VI. We investigated the impact of EXP3179, an active metabolite of the angiotensin II type 1 (AT1)-receptor antagonist Losartan (LOS) on GPVI-dependent platelet activation. METHODS AND RESULTS: EXP3179 and LOS but not EXP3174--the major AT1-receptor blocking metabolite of LOS--dose-dependently inhibited collagen-I (P<0.01) and GPVI-dependent platelet aggregation (P<0.01) analyzed by optical aggregometry. Platelet activation was further determined by flow cytometry measuring the expression of platelet PAC-1, an epitope of the activated fibrinogen-receptor complex. EXP3179 and LOS inhibited collagen-I (P<0.01) and GPVI-dependent PAC-1 expression (P<0.01). EXP3179 and LOS but not EXP3174 decreased the adhesion of GPVI-receptor expressing Chinese hamster ovarian cells on collagen-I under arterial shear conditions determined by flow chamber analysis (P<0.01 and P<0.05). EXP3179 also reduced human atherosclerotic plaque material-induced platelet aggregation (P<0.01) in vitro and murine platelet adhesion after acute vessel injury in vivo as determined by intravital microscopy (P<0.01). CONCLUSION: EXP3179 acts as a specific inhibitor of the platelet collagen receptor GPVI independent of AT1-receptor antagonism. Further investigations may clarify its individual potential as a novel pharmacological approach to specifically inhibit atherothrombotic events by GPVI-receptor blockade.


Subject(s)
Atherosclerosis/complications , Losartan/analogs & derivatives , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/metabolism , Thrombosis , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Atherosclerosis/blood , Flow Cytometry , Humans , Losartan/therapeutic use , Microscopy, Fluorescence , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptor, Angiotensin, Type 1/drug effects , Thrombosis/blood , Thrombosis/etiology , Thrombosis/prevention & control , Treatment Outcome
17.
Circ Res ; 100(6): 894-903, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17332431

ABSTRACT

Accumulating evidence suggests a critical role of increased reactive oxygen species production for left ventricular (LV) remodeling and dysfunction after myocardial infarction (MI). An increased myocardial activity of the NAD(P)H oxidase, a major oxidant enzyme system, has been observed in human heart failure; however, the role of the NAD(P)H oxidase for LV remodeling and dysfunction after MI remains to be determined. MI was induced in wild-type (WT) mice (n=46) and mice lacking the cytosolic NAD(P)H oxidase component p47(phox) (p47(phox)-/- mice) (n=32). Infarct size was similar among the groups. NAD(P)H oxidase activity was markedly increased in remote LV myocardium of WT mice after MI as compared with sham-operated mice (83+/-8 versus 16.7+/-3.5 nmol of O(2)(-) x microg(-1) x min(-1); P<0.01) but not in p47(phox)-/- mice after MI (13.5+/-3.6 versus 15.5+/-3.5 nmol of O(2)(-) x microg(-1) x min(-1)), as assessed by electron-spin resonance spectroscopy using the spin probe CP-H. Furthermore, increased myocardial xanthine oxidase activity was observed in WT, but not in p47(phox)-/- mice after MI, suggesting NAD(P)H oxidase-dependent xanthine oxidase activation. Myocardial reactive oxygen species production was increased in WT mice, but not in p47(phox)-/- mice, after MI. LV cavity dilatation and dysfunction 4 weeks after MI were markedly attenuated in p47(phox)-/- mice as compared with WT mice, as assessed by echocardiography (LV end-diastolic diameter: 4.5+/-0.2 versus 6.3+/-0.3 mm, P<0.01; LV ejection fraction, 35.8+/-2.5 versus 22.6+/-4.4%, P<0.05). Furthermore, cardiomyocyte hypertrophy, apoptosis, and interstitial fibrosis were substantially reduced in p47(phox)-/- mice as compared with WT mice. Importantly, the survival rate was markedly higher in p47(phox)-/- mice as compared with WT mice after MI (72% versus 48%; P<0.05). These results suggest a pivotal role of NAD(P)H oxidase activation and its subunit p47(phox) for LV remodeling/dysfunction and survival after MI. The NAD(P)H oxidase system represents therefore a potential novel therapeutic target to prevent cardiac failure after MI.


Subject(s)
Myocardial Infarction/physiopathology , Myocardium/metabolism , NADPH Oxidases/metabolism , Ventricular Dysfunction, Left/enzymology , Ventricular Remodeling , Animals , Apoptosis/genetics , Disease Models, Animal , Disease Progression , Electron Spin Resonance Spectroscopy , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Heart Function Tests , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardium/enzymology , NADPH Oxidases/genetics , Nitric Oxide/metabolism , Superoxides/metabolism , Survival Rate , Ventricular Dysfunction, Left/genetics , Ventricular Remodeling/drug effects , Ventricular Remodeling/genetics , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/metabolism
18.
Cell ; 128(3): 589-600, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17289576

ABSTRACT

Postpartum cardiomyopathy (PPCM) is a disease of unknown etiology and exposes women to high risk of mortality after delivery. Here, we show that female mice with a cardiomyocyte-specific deletion of stat3 develop PPCM. In these mice, cardiac cathepsin D (CD) expression and activity is enhanced and associated with the generation of a cleaved antiangiogenic and proapoptotic 16 kDa form of the nursing hormone prolactin. Treatment with bromocriptine, an inhibitor of prolactin secretion, prevents the development of PPCM, whereas forced myocardial generation of 16 kDa prolactin impairs the cardiac capillary network and function, thereby recapitulating the cardiac phenotype of PPCM. Myocardial STAT3 protein levels are reduced and serum levels of activated CD and 16 kDa prolactin are elevated in PPCM patients. Thus, a biologically active derivative of the pregnancy hormone prolactin mediates PPCM, implying that inhibition of prolactin release may represent a novel therapeutic strategy for PPCM.


Subject(s)
Cardiomyopathies/metabolism , Cathepsin D/metabolism , Pregnancy Complications, Cardiovascular/metabolism , Prolactin/metabolism , Puerperal Disorders/metabolism , STAT3 Transcription Factor/metabolism , Animals , Bromocriptine/pharmacology , Bromocriptine/therapeutic use , Cardiomyopathies/prevention & control , Cathepsin D/blood , Disease Models, Animal , Female , Heart Transplantation , Humans , Hypertrophy, Left Ventricular , Lactation/blood , Lipoproteins, LDL/blood , Male , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Neovascularization, Pathologic , Oxidative Stress , Peptide Fragments/blood , Postpartum Period/metabolism , Pregnancy , Pregnancy Complications, Cardiovascular/prevention & control , Prolactin/antagonists & inhibitors , Prolactin/blood , Puerperal Disorders/prevention & control , STAT3 Transcription Factor/blood , STAT3 Transcription Factor/genetics
19.
Atherosclerosis ; 194(1): 62-70, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17069818

ABSTRACT

Accumulation and modification of low density lipoproteins (LDL) within the vessel wall represent key events in atherogenesis. Secretory phospholipase A2 type IIA (sPLA2-IIA) modulates the enzymatic process of LDL-modification and was recently identified as an independent predictor of coronary events in patients with coronary artery disease (CAD). Angiotensin II (ANG II) type 1 (AT1)-receptor blockade reduces LDL-modification and atherosclerotic plaque formation in rodent and primate models of atherosclerosis. Therefore, we assessed whether ANG II via its AT1-receptor enhances sPLA2-IIA-dependent lipid peroxidation in vitro and in patients with CAD. Stimulation of rat aortic smooth muscle cells with ANG II (10(-7) mol/L) enhanced sPLA2-IIA protein expression, activity as well as LDL-peroxidation, determined by western blot, activity assay and malondialdehyde (MDA)-assay and diene formation, respectively, and were blunted by AT1-receptor blockade (Losartan, 10(-5) mol/L). In addition, ANG II-induced sPLA2 activity and LDL-peroxidation were abolished by the sPLA2-IIa activity inhibitor LY311727 (10(-5) mol/L). To evaluate a potential clinical implication, patients (n=18) with angiographically documented CAD were treated with the AT1-receptor blocker Irbesartan (IRB; 300 mg/d) for 12 weeks. Blood samples were obtained from patients pre- and post-treatment and from healthy volunteers. SPLA2-IIA serum level and activity, circulating antibodies against oxidized LDL (oxLDL), oxLDL and MDA were determined in patients and found to be significantly increased compared to healthy volunteers. IRB therapy reduced these markers of inflammation, whereas total cholesterol, HDL- and LDL-fractions remained unchanged. ANG II may elicit pro-atherosclerotic effects via type IIA sPLA2-dependent LDL-modifications. Chronical AT1-receptor blockade reduces sPLA2-IIA level and activity and subsequently lipid peroxidation. Theses findings represent a novel anti-atherosclerotic mechanism and imply that AT1-receptor blockade elicits anti-atherosclerotic potencies even in the absence of plasma cholesterol reduction.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Biphenyl Compounds/administration & dosage , Coronary Artery Disease/drug therapy , Group II Phospholipases A2/metabolism , Lipid Peroxidation/drug effects , Receptor, Angiotensin, Type 1/metabolism , Tetrazoles/administration & dosage , Animals , Aorta/cytology , Cells, Cultured , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Female , Group II Phospholipases A2/antagonists & inhibitors , Group II Phospholipases A2/blood , Humans , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/pathology , Indoles/pharmacology , Irbesartan , Lipid Peroxidation/physiology , Lipoproteins, LDL/metabolism , Losartan/pharmacology , Male , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics
20.
Cardiovasc Res ; 71(3): 596-605, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16843452

ABSTRACT

OBJECTIVE: Renin-angiotensin system (RAS) activation leads to increased production of NAD(P)H oxidase-derived reactive oxygen species (ROS), and both have been implicated in the initiation and progression of arterial hypertension, atherosclerosis, and cardiac hypertrophy. The cytosolic subunit p47phox is critically involved in agonist-induced NAD(P)H oxidase activation. Here, we investigated the role of p47phox in blood pressure control, endothelium-dependent relaxation, cardiac hypertrophy, RAS activation, and renal oxidative stress under resting conditions. METHODS AND RESULTS: Mice deficient in p47phox (on C57BL/6 background) developed significantly higher systolic blood pressure levels compared to C57BL/6 wild-type animals (136.0+/-3.0 mmHg vs. 112.2+/-2.6, P<0.01, n=16) as measured by the tail cuff method from week 6 up to week 12 post partum. The increase in blood pressure in p47phox-/- mice was associated with an impaired endothelium-dependent relaxation (P<0.005 vs. wild-type, n=11). At the age of 12 weeks p47phox-/- mice showed increased plasma renin activity as analyzed by radioimmunoassay (14.5+/-1.8 ng/mL/h vs. 9.6+/-1.7 ng/mL/h, P<0.05, n=10) and enhanced angiotensin converting enzyme (ACE) activity in the kidney and aorta as measured by Hip-His-Leu cleavage (7.6+/-0.8 vs. 4.8+/-0.9 nmol/L His-Leu/mg protein, P<0.05, n=5) compared to wild-type mice. No differences in oxygen radical formation was determined in kidney samples by lucigenin- and luminol-enhanced chemiluminescence or by electron spin resonance spectroscopy. Consistently, treatment with the radical scavenger tempol did not lower blood pressure in p47phox-/- mice, whereas ACE and angiotensin II type I receptor inhibition normalized blood pressure. CONCLUSION: Deficiency of the NAD(P)H oxidase subunit p47phox leads to RAS activation, which subsequently contributes to blood pressure increase in a ROS-independent manner.


Subject(s)
Blood Pressure/physiology , NADPH Oxidases/physiology , Renin-Angiotensin System/physiology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Antioxidants/pharmacology , Blood Pressure/drug effects , Cardiomegaly/physiopathology , Cyclic N-Oxides/pharmacology , Endothelium, Vascular/physiopathology , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/deficiency , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Renin/blood , Reverse Transcriptase Polymerase Chain Reaction/methods , Spin Labels
SELECTION OF CITATIONS
SEARCH DETAIL
...