Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Radiother Oncol ; 121(2): 342-343, 2016 11.
Article in English | MEDLINE | ID: mdl-27814982
2.
Drug Des Devel Ther ; 10: 2691-9, 2016.
Article in English | MEDLINE | ID: mdl-27616880

ABSTRACT

OBJECTIVE: There is increasing evidence supporting the role of platelets in atherosclerotic vascular disease. The G-protein-coupled receptor P2Y12 is a central mediator of platelet activation and aggregation but has also been linked to platelet-independent vascular disease. Ticagrelor is an oral P2Y12 antagonist that is used as a standard treatment in patients after acute myocardial infarction. However, the effects of ticagrelor on advanced atherosclerosis have not been investigated. MATERIALS AND METHODS: Twenty-week-old apolipoprotein-E-deficient mice received standard chow or standard chow supplemented with 0.15% ticagrelor (approximately 270 mg/kg/day) for 25 weeks. The lesion area was evaluated in the aortic sinus by Movat's pentachrome staining and lesion composition, thickness of the fibrous cap, and size of the necrotic core evaluated by morphometry. RAW 264.7 macrophages were serum starved and treated with ticagrelor in vitro for the detection and quantification of apoptosis. In addition, oxLDL uptake in RAW 264.7 macrophages was evaluated. RESULTS: A trend toward the reduction of total lesion size was detected. However, data did not reach the levels of significance (control, n=11, 565,881 µm(2) [interquartile range {IQR} 454,778-603,925 µm(2)] versus ticagrelor, n=13, 462,595 µm(2) [IQR 379,740-546,037 µm(2)]; P=0.1). A significant reduction in the relative area of the necrotic core (control, n=11, 0.46 [IQR 0.4-0.51] versus ticagrelor, n=13, 0.34 [IQR 0.31-0.39]; P=0.008), and a significant increase in fibrous caps thickness (control, n=11, 3.7 µm [IQR 3.4-4.2 µm] versus ticagrelor, n=13, 4.7 [IQR 4.3-5.5 µm], P=0.04) were seen in ticagrelor-treated mice. In vitro studies demonstrated a reduction in apoptotic RAW 264.7 macrophages (control 0.07±0.03 versus ticagrelor 0.03±0.03; P=0.0002) when incubated with ticagrelor. Uptake of oxLDL in RAW 264.7 was significantly reduced when treated with ticagrelor (control 9.2 [IQR 5.3-12.9] versus ticagrelor 6.4 [IQR 2.5-9.5], P=0.02). CONCLUSION: The present study demonstrates for the first time a plaque-stabilizing effect of ticagrelor in a model of advanced vascular disease, potentially induced by a reduction of oxLDL uptake or an inhibition of apoptosis as seen in vitro.


Subject(s)
Adenosine/analogs & derivatives , Atherosclerosis/drug therapy , Disease Models, Animal , Plaque, Atherosclerotic/drug therapy , Adenosine/administration & dosage , Adenosine/therapeutic use , Administration, Oral , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Apoptosis/drug effects , Atherosclerosis/pathology , Dose-Response Relationship, Drug , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Structure-Activity Relationship , Ticagrelor
3.
Drug Des Devel Ther ; 9: 5203-11, 2015.
Article in English | MEDLINE | ID: mdl-26392754

ABSTRACT

OBJECTIVE: Thrombin has multiple proatherogenic effects including platelet activation and the induction of inflammatory processes. Recently, the cytokine oncostatin M has been shown to have proinflammatory effects. This study was designed to investigate the effects of thrombin inhibition on the initiation and progression of atherosclerosis and on the expression of oncostatin M. METHODS: Apolipoprotein E-deficient mice at different ages were fed the thrombin inhibitor dabigatran etexilate. The mean lesion area was measured in the aortic sinus and in the innominate artery. CD45-positive cells within the aortic tissue were measured by flow cytometry. Oncostatin M expression was measured in the tissue sections by immunocytochemistry. RESULTS: Treatment with dabigatran etexilate resulted in a significant reduction of the mean area of atherosclerotic lesions in the aortic sinus in both the young mice (11,176±1,500 µm(2) (control) versus 3,822±836 µm(2) (dabigatran etexilate), P<0.05) and selectively in the older mice at 28 weeks (234,099±13,500 µm(2) (control) versus 175,226±16,132 µm(2) (dabigatran etexilate), P<0.05). There were also fewer CD45-positive cells within the aortas of the dabigatran-treated mice and enhanced NO production in endothelial cells pretreated with dabigatran. In addition, the expression of oncostatin M was reduced in the lesions of dabigatran etexilate-treated mice. CONCLUSION: Inhibition of thrombin by dabigatran retards the development of early lesions and the progression of some established lesions in ApoE-/- mice. It improves endothelial function and retards macrophage accumulation within the vascular wall. Dabigatran also inhibits the expression of oncostatin M, and this suggests that oncostatin M may play a role in the initiation and progression of atherosclerosis.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Dabigatran/pharmacology , Oncostatin M/metabolism , Age Factors , Animals , Antithrombins/pharmacology , Atherosclerosis/pathology , Disease Models, Animal , Disease Progression , Flow Cytometry , Leukocyte Common Antigens/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sinus of Valsalva/pathology
4.
Drug Des Devel Ther ; 9: 3935-42, 2015.
Article in English | MEDLINE | ID: mdl-26251572

ABSTRACT

INTRODUCTION: Besides their blood pressure-lowering effects, olmesartan medoxomil and amlodipine besylate exhibit additional anti-inflammatory mechanisms in atherosclerosic disease. Most of the studies investigating the effects of atherosclerosis focused on early atherosclerotic lesions, whereas lesions in human disease, at the time when medical treatment is started, are already well established. Therefore, we set up a model of advanced atherosclerosis and investigated the effects of olmesartan medoxomil, amlodipine besylate, and the combination of both on atherosclerotic lesion size and lesion composition. MATERIALS AND METHODS: Olmesartan medoxomil (1 mg/kg/day), amlodipine besylate (1.5 mg/kg/day), and the combination of both was added to chow and was fed to apolipoprotein E-deficient (ApoE(-/-)) mice at 25 weeks of age. Mice were sacrificed after 25 weeks of drug administration and perfused with formalin. Innominate arteries were dissected out and paraffin embedded. Serial sections were generated, and lesion sizes and their composition - such as minimal thickness of the fibrous cap, size of the necrotic core, and presence of calcification - were analyzed. Electrophoretic mobility shift assays were used to detect DNA-binding activity of the transcription factor nuclear factor-kappa B (NF-κB) in aortic tissue. RESULTS: Treatment with the combination of olmesartan medoxomil and amlodipine besylate led to a significant reduction in atherosclerotic lesion size in ApoE(-/-) mice (olmesartan medoxomil/amlodipine besylate: 122,277±6,795 µm(2), number [n]=14; versus control: 177,502±10,814 µm(2), n=9; P<0.001). Treatment with amlodipine besylate (n=5) alone did not reach significance. However, a trend toward a decrease in lesion size in the amlodipine besylate-treated animals could be observed. In the histological analysis of atherosclerotic lesion composition, significantly thicker fibrous caps were found in treatment with amlodipine besylate (amlodipine: 5.12±0.26 µm, n=6; versus control: 3.98±0.18 µm, n=10; P<0.01). Furthermore, all sections revealed morphological signs of calcification, but no difference could be detected. Treatment with the combination of olmesartan medoxomil and amlodipine besylate showed no effect on lesion composition. Electrophoretic mobility shift assays of nuclear extracts demonstrated reduced activity of the transcription factor NF-κB when treated with olmesartan medoxomil, amlodipine besylate, or their combination, as compared to controls. CONCLUSION: Combined treatment with olmesartan medoxomil and amlodipine besylate attenuated atherosclerotic lesion progression, possibly due to anti-inflammatory mechanisms. Our data support the hypothesis that even in advanced atherosclerosis anti-inflammatory treatment, using angiotensin II type 1 receptor blockers and calcium channel antagonists of the dihydropyridine type can attenuate atherosclerotic lesion progression.


Subject(s)
Amlodipine Besylate, Olmesartan Medoxomil Drug Combination/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Amlodipine Besylate, Olmesartan Medoxomil Drug Combination/administration & dosage , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Atherosclerosis/pathology , Disease Models, Animal , Disease Progression , Electrophoretic Mobility Shift Assay , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism
5.
Circ Cardiovasc Interv ; 7(6): 806-12, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25351198

ABSTRACT

BACKGROUND: We aimed to test, for the first time, the feasibility of intracoronary delivery of an innovative, injectable bioabsorbable scaffold (IK-5001), to prevent or reverse adverse left ventricular remodeling and dysfunction in patients after ST-segment-elevation myocardial infarction. METHODS AND RESULTS: Patients (n=27) with moderate-to-large ST-segment-elevation myocardial infarctions, after successful revascularization, were enrolled. Two milliliters of IK-5001, a solution of 1% sodium alginate plus 0.3% calcium gluconate, was administered by selective injection through the infarct-related coronary artery within 7 days after myocardial infarction. IK-5001 is assumed to permeate the infarcted tissue, cross-linking into a hydrogel and forming a bioabsorbable cardiac scaffold. Coronary angiography, 3 minutes after injection, confirmed that the injection did not impair coronary flow and myocardial perfusion. Furthermore, IK-5001 deployment was not associated with additional myocardial injury or re-elevation of cardiac biomarkers. Clinical assessments, echocardiographic studies, 12-lead electrocardiograms, 24-hour Holter monitoring, blood tests, and completion of Minnesota Living with Heart Failure Questionnaires were repeated during follow-up visits at 30, 90, and 180 days after treatment. During a 6-month follow-up, these tests confirmed favorable tolerability of the procedure, without device-related adverse events, serious arrhythmias, blood test abnormalities, or death. Serial echocardiographic studies showed preservation of left ventricular indices and left ventricular ejection fraction. CONCLUSIONS: This first-in-man pilot study shows that intracoronary deployment of an IK-5001 scaffold is feasible and well tolerated. Our results have promoted the initiation of a multicenter, randomized controlled trial to confirm the safety and efficacy of this new approach in high-risk patients after ST-segment-elevation myocardial infarction. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01226563.


Subject(s)
Absorbable Implants , Alginates/administration & dosage , Calcium Gluconate/administration & dosage , Myocardial Infarction/therapy , Regenerative Medicine/methods , Tissue Engineering/methods , Tissue Scaffolds , Ventricular Dysfunction, Left/therapy , Ventricular Function, Left , Ventricular Remodeling , Absorbable Implants/adverse effects , Adult , Aged , Alginates/adverse effects , Belgium , Calcium Gluconate/adverse effects , Feasibility Studies , Female , Germany , Glucuronic Acid/administration & dosage , Glucuronic Acid/adverse effects , Heart Function Tests , Hexuronic Acids/administration & dosage , Hexuronic Acids/adverse effects , Humans , Hydrogels , Injections, Intra-Arterial , Male , Middle Aged , Myocardial Infarction/diagnosis , Myocardial Infarction/physiopathology , Pilot Projects , Predictive Value of Tests , Surveys and Questionnaires , Time Factors , Tissue Scaffolds/adverse effects , Treatment Outcome , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/physiopathology
6.
Eur J Med Res ; 18: 19, 2013 Jun 24.
Article in English | MEDLINE | ID: mdl-23800095

ABSTRACT

BACKGROUND: The cytokine growth differentiation factor-15 (GDF-15), a member of the TGF beta superfamily, has recently been discovered to play an important role in cardiovascular diseases. It is mostly expressed in macrophages of atherosclerotic lesions, but its impact on advanced atherosclerosis is still unknown. This study was performed to evaluate the effects of GDF-15 in an established mouse model of advanced atherosclerosis. METHODS: Thirty-eight LDL receptor deficient mice received a lethal body radiation. Half of the group was transplanted with bone marrow of GDF-15 deficient mice. Nineteen mice were transplanted with bone marrow from wild-type controls. After 24 weeks on an atherogenic diet, animals were euthanized and sections of the aortic sinus were prepared. Lesion size and lesion composition, as well as macrophage content,were evaluated. RESULTS: While demonstrating no difference in lesion size, LDL-receptor knockout mice transplanted with bone marrow from GDF-15 deficient mice showed enhanced macrophage accumulation and features of atherosclerotic plaque destabilization, such as thinning of fibrous caps. Immunostaining against intercellular adhesion molecule-1 further revealed an increased expression in mice receiving GDF-15-deficient bone marrow. CONCLUSIONS: This is the first study that demonstrates a protective role of GDF-15 in advanced atherosclerosis and macrophage accumulation, possibly due to the reduced expression of adhesion molecules.


Subject(s)
Atherosclerosis/drug therapy , Growth Differentiation Factor 15/metabolism , Macrophages/metabolism , Receptors, LDL/metabolism , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow Transplantation , Cell Adhesion/genetics , Cell Adhesion/radiation effects , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Growth Differentiation Factor 15/genetics , Humans , Lethal Dose 50 , Macrophages/pathology , Macrophages/radiation effects , Mice , Receptors, LDL/deficiency , Receptors, LDL/genetics , Whole-Body Irradiation
7.
Mol Med ; 18: 1375-86, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23114885

ABSTRACT

Psychosocial stress has been shown to be a contributing factor in the development of atherosclerosis. Although the underlying mechanisms have not been elucidated entirely, it has been shown previously that the transcription factor nuclear factor-κB (NF-κB) is an important component of stress-activated signaling pathway. In this study, we aimed to decipher the mechanisms of stress-induced NF-κB-mediated gene expression, using an in vitro and in vivo model of psychosocial stress. Induction of stress led to NF-κB-dependent expression of proinflammatory (tissue factor, intracellular adhesive molecule 1 [ICAM-1]) and protective genes (manganese superoxide dismutase [MnSOD]) via p50, p65 or cRel. Selective inhibition of the different subunits and the respective kinases showed that inhibition of cRel leads to the reduction of atherosclerotic lesions in apolipoprotein(-/-) (ApoE(-/-)) mice via suppression of proinflammatory gene expression. This observation may therefore provide a possible explanation for ineffectiveness of antioxidant therapies and suggests that selective targeting of cRel activation may provide a novel approach for the treatment of stress-related inflammatory vascular disease.


Subject(s)
Atherosclerosis/genetics , Gene Expression Regulation , NF-kappa B/metabolism , Protein Subunits/metabolism , Stress, Psychological/genetics , Thrombosis/genetics , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Atherosclerosis/complications , Atherosclerosis/psychology , Female , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , Norepinephrine , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-rel/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stress, Psychological/complications , Thrombosis/complications , Thrombosis/psychology
8.
JACC Cardiovasc Interv ; 4(8): 896-904, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21851905

ABSTRACT

OBJECTIVES: This study sought to demonstrate the noninferiority of endothelial progenitor cell capturing stents (ECS) relative to drug-eluting stents (DES) regarding target lesion failure (TLF) and the composite of cardiac death, myocardial infarction, and target lesion repeat revascularization within 1 year. BACKGROUND: A "pro-healing" approach for prevention of in-stent restenosis is theoretically favorable over the use of cytotoxic/cytostatic drugs released from DES to treat coronary artery disease. Promoting accelerated endothelialization of the stent, ECS have shown promising results in studies with patients carrying noncomplex lesions. METHODS: We undertook an international, clinical trial in 26 centers planning to randomize 1,300 patients with stable coronary artery disease and with a high risk of restenosis between treatment, with either ECS or DES. After a routine review with 50% of the patients enrolled, early cessation of the trial was recommended by the data and safety monitoring board when TLF in the ECS population was higher and treatment of new patients with an ECS would be unreasonable. RESULTS: At 1 year evaluating 304 patients receiving ECS and 318 receiving DES, TLF occurred in 17.4% of the ECS-treated patients and in 7.0% of the DES-treated patients (p = 0.98 for noninferiority). CONCLUSIONS: Within 1 year, inhibition of intimal hyperplasia by the ECS is not sufficiently strong to compete with DES in terms of restenosis prevention in patients/lesions with a high risk of restenosis. Furthermore, long-term follow-up is pivotal to fully appreciate the clinical value of ECS, including the effect on late intimal hyperplasia regression.


Subject(s)
Angioplasty, Balloon, Coronary/instrumentation , Coronary Artery Disease/therapy , Coronary Restenosis/prevention & control , Drug-Eluting Stents , Endothelial Cells/pathology , Stem Cells/pathology , Stents , Aged , Angioplasty, Balloon, Coronary/adverse effects , Angioplasty, Balloon, Coronary/mortality , Coronary Angiography , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/mortality , Coronary Restenosis/diagnostic imaging , Coronary Restenosis/etiology , Coronary Restenosis/mortality , Europe , Female , Humans , Hyperplasia , Kaplan-Meier Estimate , Male , Middle Aged , Myocardial Infarction/etiology , Myocardial Infarction/prevention & control , Prospective Studies , Prosthesis Design , Risk Assessment , Risk Factors , Single-Blind Method , Time Factors , Treatment Outcome
9.
Mediators Inflamm ; 2011: 432080, 2011.
Article in English | MEDLINE | ID: mdl-21772662

ABSTRACT

AIM: Thrombin not only plays a central role in thrombus formation and platelet activation, but also in induction of inflammatory processes. Activated factor X (FXa) is traditionally known as an important player in the coagulation cascade responsible for thrombin generation. We assessed the hypothesis that rivaroxaban, a direct FXa inhibitor, attenuates plaque progression and promotes stability of advanced atherosclerotic lesions in an in vivo model. METHODS AND RESULTS: Rivaroxaban (1 or 5 mg/kg body weight/day) or standard chow diet was administered for 26 weeks to apolipoprotein E-deficient mice (n = 20 per group) with already established atherosclerotic lesions. There was a nonsignificant reduction of lesion progression in the high-concentration group, compared to control mice. FXa inhibition with 5 mg Rivaroxaban/kg/day resulted in increased thickness of the protective fibrous caps (12.3 ± 3.8 µm versus 10.1 ± 2.7 µm; P < .05), as well as in fewer medial erosions and fewer lateral xanthomas, indicating plaque stabilizing properties. Real time-PCR from thoracic aortas revealed that rivaroxaban (5 mg/kg/day) treatment reduced mRNA expression of inflammatory mediators, such of IL-6, TNF-α, MCP-1, and Egr-1 (P < .05). CONCLUSIONS: Chronic administration of rivaroxaban does not affect lesion progression but downregulates expression of inflammatory mediators and promotes lesion stability in apolipoprotein E-deficient mice.


Subject(s)
Anticoagulants/therapeutic use , Apolipoproteins E/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Factor Xa Inhibitors , Morpholines/therapeutic use , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology , Thiophenes/therapeutic use , Animals , Apolipoproteins E/genetics , Female , Humans , Lipids/blood , Mice , Mice, Knockout , Random Allocation , Rivaroxaban
10.
Eur J Clin Invest ; 41(11): 1164-71, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21418204

ABSTRACT

BACKGROUND: The multiligand receptor for advanced glycation end products (RAGE) of the immunoglobulin superfamily is expressed on multiple cell types implicated in the inflammatory response in atherosclerosis. We sought to determine the role of bone marrow-derived RAGE in different stages of atherosclerotic development in apolipoprotein E-deficient mice (apoE(-/-)). METHODS: Seven- and 23-week-old apoE(-/-) mice (n = 40) were lethally irradiated and given bone marrow from RAGE null (RAGE(-/-)/apoE(-/-)) or RAGE-bearing (RAGE(+/+)/apoE(-/-)) mice to apoE(-/-) mice to generate double knockout bone marrow chimera (RAGE(-/-)/apoE(-/-bmc) and RAGE(+/+)/apoE(-/-bmc)-, respectively). After 16 weeks on a standard chow diet, mice were sacrificed and atherosclerotic lesion formation was evaluated. RESULTS: Plaques in the aortic root of the young mice showed no significant difference in maximum plaque size (217,470 ± 17,480 µm(2) for the RAGE(-/-) /apoE(-/-bmc) mice compared to 244,764 ± 45,840 µm(2)), whereas lesions in the brachiocephalic arteries of the older RAGE(-/-)/apoE(-/-bmc) mice had significantly smaller lesions (94,049 ± 13,0844 µm(2) vs. 145,570 ± 11,488 µm(2), P < 0.05) as well as reduced average necrotic core area (48,600 ± 9220 µm(2) compared to 89,502 ± 10,032 µm(2), P < 0.05) when compared to RAGE(+/+)/apoE(-/-bmc) mice. Reduced plaque size and more stable plaque morphology was associated with significant reduced expression of VCAM-1, ICAM-1 and MCP-1. Accumulation of the RAGE ligand HMGB-1 was also significantly reduced within the lesions of RAGE(-/-)/apoE(-/-bmc) mice. CONCLUSIONS: This study demonstrates that bone marrow-derived RAGE is an important factor in the progression of atherosclerotic plaques.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Bone Marrow Transplantation , Glycation End Products, Advanced/metabolism , Receptors, Immunologic/physiology , Animals , Atherosclerosis/metabolism , Bone Marrow/immunology , Chemokine CCL2/metabolism , Female , HMGB1 Protein/metabolism , Intercellular Adhesion Molecule-1/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor for Advanced Glycation End Products , Vascular Cell Adhesion Molecule-1/metabolism
11.
J Cardiovasc Pharmacol ; 57(4): 447-54, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21242806

ABSTRACT

Inflammation contributes to atherosclerotic plaque initiation and progression. Recent studies suggest that nicotinic acid has anti-inflammatory effects independent of its lipid-modifying capabilities. We assessed the hypothesis that administration of nicotinic acid to older apolipoprotein E (apoE)-deficient mice with established lesions will reduce lesion size and plaque inflammation independent of its lipid-modifying effects. Therefore nicotinic acid was administered to 27-week-old apo E-deficient mice exhibiting advanced atherosclerotic lesions within the innominate artery. After 27 weeks of treatment both animal groups had no significant changes in plasma lipid levels. Mice treated with nicotinic acid (n = 22) demonstrated a 30% reduction in total lesion area compared with controls (n = 20). Furthermore, they revealed a more stable plaque composition with an increase in fibrous cap thickness and a reduction in the size of the necrotic core. Immunohistochemistry demonstrated a reduced accumulation of macrophages and a reduced expression of vascular cell adhesion molecule-1 and tissue factor. Additionally, administration of nicotinic acid significantly reduced tumor necrosis factor alpha expression in the thoracic aorta as demonstrated by real-time PCR. In conclusion, these data suggest that long-term administration of nicotinic acid has anti-atherogenic and anti-inflammatory properties on advanced atherosclerotic lesions, which are independent of its lipid-modifying actions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Niacin/pharmacology , Plaque, Atherosclerotic/drug therapy , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Apolipoproteins E/genetics , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/pathology , Female , Inflammation/physiopathology , Lipids/blood , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Plaque, Atherosclerotic/pathology , Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/drug effects , Vascular Cell Adhesion Molecule-1/metabolism
12.
Basic Res Cardiol ; 106(1): 125-34, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21116822

ABSTRACT

A chronic (auto)immune response is the critical mechanism in atherosclerosis. Interleukin-17A is a pivotal effector cytokine, which modulates immune cell trafficking and initiates inflammation in (auto)immune and infectious diseases. However, expression of IL-17A in the context of human atherosclerosis has hardly been explored. Carotid artery plaques were collected from 79 patients undergoing endarterectomy. Patients were grouped according to their symptomatic status (TIA, stroke), plaque morphology and medication. Quantitative RT-PCR was used to analyze tissue inflammation and immunohistochemistry to assess cellular source of IL-17A expression and lesion morphology. Carotid plaques from patients with ischemic symptoms were characterized by a highly activated inflammatory milieu including accumulation of T cells (p = 0.04) and expression of IL-6 and VCAM1 (p = 0.02, 0.01). Expression of IL-17A and its positive regulators IL-21 and IL-23 was present in atherosclerotic lesions, significantly upregulated in atheromas of symptomatic patients (p = 0.005, 0.004, 0.03), and expression of IL-17A and IL-21 showed a strong correlation (p = 0.002, r = 0.52). The cellular sources of lesional IL-17A expression are T cells, macrophages, B cells and plasma cells. Vulnerable/ruptured (complicated) plaques were significantly associated with IL-17A expression levels (p = 0.003). In addition, IL-17A showed a marked negative correlation with the potent anti-inflammatory/atheroprotective cytokine IL-10 (p = 0.0006, r = -0.46). Furthermore, treatment with a HMG-CoA reductase inhibitor or acetylsalicylic acid showed reduced levels of IL-21, IL-23 and VCAM1 (all p < 0.05), but did not influence IL-17A. The association of IL-17A with ischemic symptoms and vulnerable plaque characteristics suggests that the pro-inflammatory cytokine IL-17A may contribute to atherosclerosis und plaque instability.


Subject(s)
Atherosclerosis/metabolism , Carotid Arteries/metabolism , Interleukin-17/metabolism , Plaque, Atherosclerotic/metabolism , Aged , Aged, 80 and over , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Carotid Arteries/pathology , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Ischemia/metabolism , Male , Osteopontin/metabolism , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology
13.
Mediators Inflamm ; 2010: 194896, 2010.
Article in English | MEDLINE | ID: mdl-20652055

ABSTRACT

INTRODUCTION: The Chinese extract Rhizoma coptidis is well known for its anti-inflammatory, antioxidative, antiviral, and antimicrobial activity. The exact mechanisms of action are not fully understood. METHODS: We examined the effect of the extract and its main compound, berberine, on LPS-induced inflammatory activity in a murine macrophage cell line. RAW 264.7 cells were stimulated with LPS and incubated with either Rhizoma coptidis extract or berberine. Activation of AP-1 and NFkappaB was analyzed in nuclear extracts, secretion of MCP-1/CCL2 was measured in supernatants. RESULTS: Incubation with Rhizoma coptidis and berberine strongly inhibited LPS-induced monocyte chemoattractant protein (MCP)-1 production in RAW cells. Activation of the transcription factors AP-1 and NFkappaB was inhibited by Rhizoma coptidis in a dose- and time-dependent fashion. CONCLUSIONS: Rhizoma coptidis extract inhibits LPS-induced MCP-1/CCL2 production in vitro via an AP-1 and NFkappaB-dependent pathway. Anti-inflammatory action of the extract is mediated mainly by its alkaloid compound berberine.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chemokine CCL2/biosynthesis , Drugs, Chinese Herbal/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Macrophages/drug effects , NF-kappa B/physiology , Transcription Factor AP-1/physiology , Animals , Berberine/pharmacology , Cells, Cultured , Coptis chinensis , Interleukin-12/biosynthesis , Interleukin-1beta/biosynthesis , Macrophages/metabolism , Mice , Nitric Oxide/biosynthesis , Reactive Oxygen Species/metabolism
14.
Am J Emerg Med ; 28(4): 477-84, 2010 May.
Article in English | MEDLINE | ID: mdl-20466229

ABSTRACT

INTRODUCTION: Even among health care professionals, resuscitation performance has been shown to be poor. So far, it remains unclear whether cardiac arrest staff with frequent practice in resuscitation requires training to adapt to the new International Liaison Committee on Resuscitation (ILCOR) guidelines of 2005. This study evaluated the need for basic life support training in nurses with emergency experience. METHODS AND RESULTS: Nurses (N = 24) recruited from an intensive care unit self-assessed their resuscitation skills and performed a cardiac arrest scenario using a manikin. After a theoretical instruction and hands-on training followed by feedback, participants once again performed a resuscitation scenario in addition to completing posttraining self-assessments. Participating nurses considered resuscitation skills training--in particular in adapting to the new ILCOR guidelines of 2005--to be important. Pretraining data revealed performance deficits even in this sample of emergency-experienced nursing staff. Training resulted in significant improvement in ventilation volume (P < .001), rate of compressions with correct depth (P < .031) and full release (P < .001), and a reduction in total hands-off time (P < .050). Objective data were mirrored in participants' self-assessed competencies. CONCLUSION: Results suggest that basic life support training based on the ILCOR guidelines of 2005 is necessary even in nurses with emergency experience. Training followed by the application of a feedback algorithm seems to improve short-term resuscitation performance and is well accepted by experienced nurses who work on an intensive care unit and who also comprise the inner-hospital cardiac arrest team.


Subject(s)
Cardiopulmonary Resuscitation/standards , Heart Arrest/therapy , Nursing Staff, Hospital/education , Practice Guidelines as Topic , Adult , Cardiopulmonary Resuscitation/education , Clinical Competence , Emergency Service, Hospital/standards , Female , Humans , Life Support Care , Male , Middle Aged , Needs Assessment , Workforce
15.
Cardiovasc Res ; 86(2): 321-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20110335

ABSTRACT

AIMS: Early growth response gene-1 (Egr-1) regulates the expression of genes important to cardiovascular disease. Within atherosclerotic lesions, Egr-1 is expressed in smooth muscle cells, endothelial cells, and macrophages. Since macrophages play a pivotal role in atherosclerotic lesion initiation and progression, this study investigated the effects of Egr-1 deficiency within bone marrow-derived cells on the development of atherosclerosis in a hyperlipidaemic mouse model. METHODS AND RESULTS: Bone marrow from Egr-1-deficient mice and wild-type controls was transplanted into lethally irradiated LDL receptor null mice. After 26 weeks on a high fat diet, atherosclerotic lesion size within the aortic sinus of recipients was evaluated. Mice receiving Egr-1-deficient bone marrow had significantly decreased lesion size compared with controls. Lesions of these mice contained fewer macrophages and had reduced expression of vascular cell adhesion molecule-1 (VCAM-1), tissue factor, as well as transforming growth factor receptor type II, which are target genes of Egr-1. These results were validated by in vitro analysis of Egr-1-deficient peritoneal macrophages which, after lipopolysaccharide stimulation, had decreased VCAM-1 and tissue factor mRNA expression compared with wild-type controls. CONCLUSION: This study demonstrates that bone marrow-derived Egr-1 promotes macrophage accumulation, atherosclerotic lesion development, and lesion complexity.


Subject(s)
Aorta, Thoracic/metabolism , Atherosclerosis/prevention & control , Bone Marrow Cells/metabolism , Early Growth Response Protein 1/deficiency , Hyperlipidemias/metabolism , Animals , Aorta, Thoracic/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow Transplantation , Disease Models, Animal , Early Growth Response Protein 1/genetics , Female , Gene Expression Regulation , Hyperlipidemias/complications , Hyperlipidemias/genetics , Hyperlipidemias/pathology , Lipids/blood , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Radiation Chimera , Receptor, Transforming Growth Factor-beta Type II , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, Transforming Growth Factor beta/metabolism , Thromboplastin/metabolism , Time Factors , Vascular Cell Adhesion Molecule-1/metabolism , Whole-Body Irradiation
16.
Basic Res Cardiol ; 105(2): 301-13, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19629561

ABSTRACT

Proinflammatory cytokines, including TNF family members, have been shown to play a critical role in cardiac remodeling. FGF-inducible 14-kDa protein (Fn14, TNFrsf12a or TWEAKR) is the smallest member of the TNF-receptor family. Currently, little is known about the functional role of Fn14 and its only known ligand TNF-like weak inducer of apoptosis (TWEAK) in the heart. We therefore evaluated the expression and regulation of Fn14 in cardiomyocytes and in experimental myocardial infarction. In order to study the regulation of Fn14, myocardial infarction was induced in CD-1 mice and neonatal rat cardiomyocytes were used for in vitro studies. TWEAK and Fn14 were markedly upregulated in the remodeling myocardium after experimental myocardial infarction in vivo. Likewise, fibroblast growth factor 1, norepinephrine and angiotensin II as well as mechanical stretch were able to strongly induce Fn14 expression in cardiomyocytes. This induction is mediated via the Rho/ROCK pathway, since the known inhibitors C3 exoenzyme for RhoA and Y27632 for ROCK prevented the upregulation of Fn14 in cardiomyocytes. Consistently, pretreatment of cardiomyocytes with siRNA against Rho A and ROCK also abolished Fn14 induction. Moreover, stimulation of cardiomyocytes with TWEAK promoted nuclear translocation of NF-kappaB and subsequent induction of NF-kappaB dependent genes such as RANTES and MCP-1. Conversely, when cells were pretreated with siRNA against Fn14, NF-kappaB activation by TWEAK was inhibited. We here provide the first evidence of a stress-induced regulation of the TWEAK/Fn14 axis in cardiomyocytes implying a role of the TWEAK/Fn14 pathway in cardiac remodeling.


Subject(s)
Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Cells, Cultured , Fibroblast Growth Factors/metabolism , Male , Mice , Myocardial Infarction/metabolism , Rats , Rats, Wistar , Signal Transduction , TWEAK Receptor , Up-Regulation
17.
J Immunol ; 183(12): 8167-75, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20007582

ABSTRACT

The importance of an (auto)immune response in atherogenesis is becoming increasingly well understood. IL-17A-expressing T cells modulate immune cell trafficking, initiating inflammation and cytokine production in (auto)immune diseases. In human carotid artery plaques, we previously showed the presence of IL-17A-producing T cells and IL-23; however, IL-17A effects on atherogenesis have not been studied. Aortic root sections from 8-wk-old apolipoprotein E-deficient mice fed a standard chow diet were examined after 12 wk for lesion area, plaque composition, cellular infiltration, cytokine expression, and apoptosis. The treatment group (n = 15) received anti-IL-17A Ab and the controls (n = 10) received irrelevant Abs. Inhibition of IL-17A markedly reduced atherosclerotic lesion area (p < 0.001), maximal stenosis (p < 0.001), and vulnerability of the lesion. IL-17A mAb-treated mice showed reduced cellular infiltration, down-regulation of activation markers on endothelium and immune cells (e.g., VCAM-1), and reduced cytokine/chemokine secretion (e.g., IL6, TNFalpha, CCL5). To investigate possible mechanisms, different atherogenic cell types (e.g., macrophages, dendritic cells, HUVECs, vascular smooth muscle cells) were stimulated with IL-17A in addition to TNF-alpha, IFN-gamma, or LPS to induce cellular activation or apoptosis in vitro. Stimulation with IL-17A induced proinflammatory changes in several atherogenic cell types and apoptotic cell death in murine cells. Functional blockade of IL-17A reduces atherosclerotic lesion development and decreases plaque vulnerability, cellular infiltration, and tissue activation in apolipoprotein E-deficient mice. The present data support a pathogenic role of IL-17A in the development of atherosclerosis by way of its widespread proinflammatory and proapoptotic effects on atherogenic cells.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/immunology , Atherosclerosis/pathology , Interleukin-17/antagonists & inhibitors , Interleukin-17/physiology , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Monoclonal/administration & dosage , Apolipoproteins E/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/physiology , Atherosclerosis/prevention & control , Cells, Cultured , Disease Models, Animal , Female , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Inflammation Mediators/physiology , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
Circulation ; 120(9): 774-84, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19687358

ABSTRACT

BACKGROUND: Clinical studies failed to provide clear evidence for a proatherogenic role of hypercoagulability. This is in contrast to the well-established detrimental role of hypercoagulability and thrombin during acute atherosclerotic complications. These seemingly opposing data suggest that hypercoagulability might exert both proatherogenic and antiatherogenic effects. We therefore investigated whether hypercoagulability mediates a beneficial effect during de novo atherogenesis. METHODS AND RESULTS: De novo atherogenesis was evaluated in 2 mouse models with hyperlipidemia and genetically imposed hypercoagulability (TM(Pro/Pro)ApoE(-/-) and FVL(Q/Q)ApoE(-/-) mice). In both mouse models, hypercoagulability resulted in larger plaques, but vascular stenosis was not enhanced secondary to positive vascular remodeling. Importantly, plaque stability was increased in hypercoagulable mice with less necrotic cores, more extracellular matrix, more smooth muscle cells, and fewer macrophages. Long-term anticoagulation reversed these changes. The reduced frequency of intraplaque macrophages in hypercoagulable mice is explained by an inhibitory role of thrombin and protease-activated receptor-1 on monocyte transendothelial migration in vitro. This is dependent on phospholipase-Cbeta, phosphoinositide 3-kinase, and nitric oxide signaling in monocytes but not in endothelial cells. CONCLUSIONS: Here, we show a new function of the coagulation system, averting stenosis and plaque destabilization during de novo atherogenesis. The in vivo and in vitro data establish that thrombin-induced signaling via protease-activated receptor-1, phospholipase-Cbeta, phosphoinositide 3-kinase, and nitric oxide in monocytes impairs monocyte transendothelial migration. This likely accounts for the reduced macrophage accumulation in plaques of hypercoagulable mice. Thus, in contrast to their role in unstable plaques or after vascular injury, hypercoagulability and thrombin convey a protective effect during de novo atherogenesis.


Subject(s)
Atherosclerosis/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C beta/metabolism , Receptor, PAR-1/metabolism , Thrombophilia/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Movement/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Hyperlipidemias/immunology , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Monocytes/cytology , Monocytes/metabolism , Nitric Oxide/metabolism , Signal Transduction/physiology , Thrombin/metabolism , Thrombophilia/immunology , Vasculitis/immunology , Vasculitis/metabolism , Vasculitis/pathology
19.
J Am Coll Cardiol ; 54(25): 2435-46, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20082935

ABSTRACT

OBJECTIVES: The extent of adverse myocardial remodeling contributes essentially to the prognosis after myocardial infarction (MI). In this study we investigated whether inhibition of "mammalian target of rapamycin" (mTOR) attenuates left ventricular (LV) remodeling after MI. BACKGROUND: Therapeutic strategies to inhibit remodeling are currently limited to inhibition of neurohumoral activation. The mTOR-dependent signaling mechanisms are centrally involved in remodeling processes and provide new therapeutic opportunities. METHODS: Everolimus (RAD) treatment was initiated on the day after or 3 days after induction of myocardial infarction (MI) in rats. RESULTS: After 28 days, RAD-treated animals had reduced post-MI remodeling, with improved LV function and smaller LV end-diastolic diameters (8.9 + or - 0.3 mm vs. 11.4 + or - 0.2 mm, p < 0.05), end-diastolic volumes (304 + or - 30 microl vs. 414 + or - 16 microl, p < 0.05), and cardiac myocyte size (-40% vs. vehicle, p < 0.05). Infarct size was significantly reduced compared with vehicle-treated animals. The mTOR inhibition increased autophagy and concomitantly decreased proteasome activity in the border zone of the infarcted myocardium. Measurement of autophagic flux demonstrated that RAD did not decrease autophagosome clearance. When RAD treatment was initiated 3 days after MI, adverse remodeling was still attenuated and increased autophagy was still present. Sustained improvement of LV function was observed 3 months after MI, even when RAD treatment was discontinued after 1 month. CONCLUSIONS: Inhibition of mTOR is a potential therapeutic strategy to limit infarct size and to attenuate adverse LV remodeling after MI.


Subject(s)
Immunosuppressive Agents/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Myocardial Infarction/physiopathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sirolimus/analogs & derivatives , Ventricular Remodeling/drug effects , Animals , Atrial Natriuretic Factor/metabolism , Autophagy/drug effects , Diastole/physiology , Echocardiography , Everolimus , Heart Ventricles/diagnostic imaging , Intracellular Signaling Peptides and Proteins/physiology , Male , Microtubule-Associated Proteins/physiology , Myocytes, Cardiac/pathology , NF-kappa B/drug effects , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 70-kDa/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Up-Regulation , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling/physiology
20.
Atherosclerosis ; 203(1): 105-11, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18691715

ABSTRACT

Hyperhomocysteinemia is an independent risk factor for atherosclerosis. Uptake of homocysteine induces oxidative stress in macrophages. Antioxidant response elements (AREs) are regulatory elements within promoters of genes, which protect cells against oxidative stress. The current study investigated whether homocysteine induces transcription of glutamate-cysteine ligase (Gcl), via ARE driven gene expression in mouse macrophages. Gcl is the rate-limiting enzyme in the synthesis of glutathione, an important endogenous antioxidant. Gcl is heterodimeric and the genes encoding the subunits of Gcl contain several AREs within their 5'-promoter regions. Treatment of mouse macrophages with d-/l-homocysteine (50microM) induced depletion of intracellular glutathione and a compensatory increase in Gcl activity. Electro mobiliy shift assays demonstrated increased binding of nuclear proteins to ARE-containing oligonucleotides. Real-time RT-PCR revealed increased mRNA-expression of the catalytic subunit of Gcl (Gclc) after treatment with homocysteine, and this occurred via increased transcription as demonstrated with luciferase promoter reporter constructs for Gclc. Additional site directed mutagenesis demonstrated that ARE4 plays a direct role in mediating induction of Gclc by homocysteine. Supershift analysis and Western blotting revealed that Nrf2 signalling is critical in homocysteine-induced activation of ARE4. Inhibition of MAP kinase activity reduced binding of nuclear proteins to the AREs, nuclear expression of Nrf2 and mRNA expression of Gclc. Western blotting demonstrated phosporylation of ERK1/2 in homocysteine treated macrophages. These data suggest that ARE-driven gene expression of Gclc via a MEK/Nrf2 pathway could help to protect macrophages from oxidative stress due to hyperhomocysteinemia.


Subject(s)
Antioxidants/metabolism , Gene Expression Regulation , Glutamate-Cysteine Ligase/metabolism , Homocysteine/metabolism , Macrophages/metabolism , Animals , Catalysis , Cell Line , Cell Nucleus/metabolism , Glutathione/metabolism , Mice , Models, Biological , Oxidative Stress , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...