Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 44(6): 1379-1392, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38695167

ABSTRACT

BACKGROUND: Thromboembolic events secondary to rupture or erosion of advanced atherosclerotic lesions is the global leading cause of death. The most common and effective means to reduce these major adverse cardiovascular events, including myocardial infarction and stroke, is aggressive lipid lowering via a combination of drugs and dietary modifications. However, we know little regarding the effects of reducing dietary lipids on the composition and stability of advanced atherosclerotic lesions, the mechanisms that regulate these processes, and what therapeutic approaches might augment the benefits of lipid lowering. METHODS: Smooth muscle cell lineage-tracing Apoe-/- mice were fed a high-cholesterol Western diet for 18 weeks and then a zero-cholesterol standard laboratory diet for 12 weeks before treating them with an IL (interleukin)-1ß or control antibody for 8 weeks. We assessed lesion size and remodeling indices, as well as the cellular composition of aortic and brachiocephalic artery lesions, indices of plaque stability, overall plaque burden, and phenotypic transitions of smooth muscle cell and other lesion cells by smooth muscle cell lineage tracing combined with single-cell RNA sequencing, cytometry by time-of-flight, and immunostaining plus high-resolution confocal microscopic z-stack analysis. RESULTS: Lipid lowering by switching Apoe-/- mice from a Western diet to a standard laboratory diet reduced LDL cholesterol levels by 70% and resulted in multiple beneficial effects including reduced overall aortic plaque burden, as well as reduced intraplaque hemorrhage and necrotic core area. However, contrary to expectations, IL-1ß antibody treatment after diet-induced reductions in lipids resulted in multiple detrimental changes including increased plaque burden and brachiocephalic artery lesion size, as well as increasedintraplaque hemorrhage, necrotic core area, and senescence as compared with IgG control antibody-treated mice. Furthermore, IL-1ß antibody treatment upregulated neutrophil degranulation pathways but downregulated smooth muscle cell extracellular matrix pathways likely important for the protective fibrous cap. CONCLUSIONS: Taken together, IL-1ß appears to be required for the maintenance of standard laboratory diet-induced reductions in plaque burden and increases in multiple indices of plaque stability.


Subject(s)
Atherosclerosis , Disease Models, Animal , Interleukin-1beta , Mice, Knockout, ApoE , Myocytes, Smooth Muscle , Plaque, Atherosclerotic , Animals , Interleukin-1beta/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Atherosclerosis/metabolism , Atherosclerosis/genetics , Mice , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Male , Diet, Western , Mice, Inbred C57BL , Aorta/pathology , Aorta/metabolism , Aorta/drug effects , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Aortic Diseases/genetics , Aortic Diseases/metabolism , Diet, High-Fat , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Brachiocephalic Trunk/pathology , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/drug effects
2.
Arterioscler Thromb Vasc Biol ; 40(9): 2159-2170, 2020 09.
Article in English | MEDLINE | ID: mdl-32673527

ABSTRACT

OBJECTIVE: Evidence from preclinical and clinical studies has demonstrated that myocardial infarction promotes atherosclerosis progression. The impact of focal vascular inflammation on the progression and phenotype of remote atherosclerosis remains unknown. Approach and Results: We used a novel ApoE-/- knockout mouse model of sustained arterial inflammation, initiated by mechanical injury in the abdominal aorta. Using serial in vivo molecular MRI and ex vivo histology and flow cytometry, we demonstrate that focal arterial inflammation triggered by aortic injury, accelerates atherosclerosis in the remote brachiocephalic artery. The brachiocephalic artery atheroma had distinct histological features including increased plaque size, plaque permeability, necrotic core to collagen ratio, infiltration of more inflammatory monocyte subsets, and reduced collagen content. We also found that arterial inflammation following focal vascular injury evoked a prolonged systemic inflammatory response manifested as a persistent increase in serum IL-6 (interleukin 6). Finally, we demonstrate that 2 therapeutic interventions-pravastatin and minocycline-had distinct anti-inflammatory effects at the plaque and systemic level. CONCLUSIONS: We show for the first time that focal arterial inflammation in response to vascular injury enhances systemic vascular inflammation, accelerates remote atheroma progression and induces plaques more inflamed, lipid-rich, and collagen-poor in the absence of ischemic myocardial injury. This inflammatory cascade is modulated by pravastatin and minocycline treatments, which have anti-inflammatory effects at both plaque and systemic levels that mitigate atheroma progression.


Subject(s)
Aortitis/complications , Atherosclerosis/etiology , Brachiocephalic Trunk/metabolism , Inflammation Mediators/blood , Plaque, Atherosclerotic , Animals , Anti-Inflammatory Agents/pharmacology , Aortitis/blood , Aortitis/pathology , Atherosclerosis/blood , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/pathology , Collagen/metabolism , Disease Models, Animal , Disease Progression , Interleukin-6/blood , Lipid Metabolism , Male , Mice, Knockout, ApoE , Minocycline/pharmacology , Necrosis , Pravastatin/pharmacology , Time Factors
3.
Diab Vasc Dis Res ; 15(4): 302-313, 2018 07.
Article in English | MEDLINE | ID: mdl-29499628

ABSTRACT

AIMS: Despite vast clinical experience linking diabetes and atherosclerosis, the molecular mechanisms leading to accelerated vascular damage are still unclear. Here, we investigated the effects of nuclear factor of activated T-cells inhibition on plaque burden in a novel mouse model of type 2 diabetes that better replicates human disease. METHODS & RESULTS: IGF-II/LDLR-/-ApoB100/100 mice were generated by crossbreeding low-density lipoprotein receptor-deficient mice that synthesize only apolipoprotein B100 (LDLR-/-ApoB100/100) with transgenic mice overexpressing insulin-like growth factor-II in pancreatic ß cells. Mice have mild hyperglycaemia and hyperinsulinaemia and develop complex atherosclerotic lesions. In vivo treatment with the nuclear factor of activated T-cells blocker A-285222 for 4 weeks reduced atherosclerotic plaque area and degree of stenosis in the brachiocephalic artery of IGF-II/LDLR-/-ApoB100/100 mice, as assessed non-invasively using ultrasound biomicroscopy prior and after treatment, and histologically after termination. Treatment had no impact on plaque composition (i.e. muscle, collagen, macrophages). The reduced plaque area could not be explained by effects of A-285222 on plasma glucose, insulin or lipids. Inhibition of nuclear factor of activated T-cells was associated with increased expression of atheroprotective NOX4 and of the anti-oxidant enzyme catalase in aortic vascular smooth muscle cells. CONCLUSION: Targeting the nuclear factor of activated T-cells signalling pathway may be an attractive approach for the treatment of diabetic macrovascular complications.


Subject(s)
Apolipoproteins B/deficiency , Atherosclerosis/prevention & control , Brachiocephalic Trunk/drug effects , Insulin-Like Growth Factor II/deficiency , NFATC Transcription Factors/antagonists & inhibitors , Plaque, Atherosclerotic , Pyrazoles/pharmacology , Receptors, LDL/deficiency , Animals , Apolipoprotein B-100 , Apolipoproteins B/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Catalase/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Female , Genetic Predisposition to Disease , Insulin-Like Growth Factor II/genetics , Male , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4/metabolism , NFATC Transcription Factors/metabolism , Oxidative Stress/drug effects , Phenotype , Receptors, LDL/genetics , Signal Transduction
4.
Pharmacol Rep ; 69(1): 112-118, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27915184

ABSTRACT

BACKGROUND: Cholesterol-dependent and independent mechanisms were proposed to explain anti-atherosclerotic action of statins in humans. However, their effects in murine models of atherosclerosis have not been consistently demonstrated. Here, we studied the effects of pravastatin on atherosclerosis in ApoE/LDLR-/- mice fed a control and atherogenic diet. METHODS: ApoE/LDLR-/- mice were fed a control (CHOW) or an atherogenic (Low Carbohydrate High Protein, LCHP) diet. Two doses of pravastatin (40mg/kg and 100mg/kg) were used. The anti-atherosclerotic effects of pravastatin in en face aorta, cross-sections of aortic roots and brachiocephalic artery (BCA) were analysed. The lipid profile was determined. Fourier Transform Infrared Spectroscopy followed by Fuzzy C-Means (FCM) clustering was used for the quantitative assessment of plaque composition. RESULTS: Treatment with pravastatin (100mg/kg) decreased total and LDL cholesterol only in the LCHP group, but displayed a pronounced anti-atherosclerotic effect in BCA and abdominal aorta. The anti-atherosclerotic effect of pravastatin (100mg/kg) in BCA was associated with significant alterations of the chemical plaque composition, including a fall in cholesterol and cholesterol esters contents independently on total cholesterol and LDL concentration in plasma. CONCLUSIONS: Pravastatin at high (100mg/kg), but not low dose displayed a pronounced anti-atherosclerotic effect in ApoE/LDLR-/- mice fed a CHOW or LCHP diet that was remarkable in BCA, visible in en face aorta, whereas it was not observed in aortic roots, suggesting that previous inconsistencies might have been due to the various sites of atherosclerotic plaque analysis.


Subject(s)
Aorta/drug effects , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Brachiocephalic Trunk/drug effects , Pravastatin/therapeutic use , Receptors, LDL/deficiency , Animals , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Aorta/metabolism , Aorta/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Female , Mice , Mice, Knockout , Pravastatin/pharmacology , Treatment Outcome
5.
Arterioscler Thromb Vasc Biol ; 36(3): 491-500, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26821947

ABSTRACT

OBJECTIVE: Inflammatory monocytes/macrophages produce various proteinases, including matrix metalloproteinases, and degradation of the extracellular matrix by these activated proteinases weakens the mechanical strength of atherosclerotic plaques, which results in a rupture of the plaque. Peroxisome proliferator-activated receptor-γ induces a polarity shift of monocytes/macrophages toward less inflammatory phenotypes and has the potential to prevent atherosclerotic plaque ruptures. Therefore, we hypothesized that nanoparticle-mediated targeted delivery of the peroxisome proliferator-activated receptor-γ agonist pioglitazone into circulating monocytes could effectively inhibit plaque ruptures in a mouse model. APPROACH AND RESULTS: We prepared bioabsorbable poly(lactic-co-glycolic-acid) nanoparticles containing pioglitazone (pioglitazone-NPs). Intravenously administered poly(lactic-co-glycolic-acid) nanoparticles incorporated with fluorescein isothiocyanate were found in circulating monocytes and aortic macrophages by flow cytometric analysis. Weekly intravenous administration of pioglitazone-NPs (7 mg/kg per week) for 4 weeks decreased buried fibrous caps, a surrogate marker of plaque rupture, in the brachiocephalic arteries of ApoE(-/-) mice fed a high-fat diet and infused with angiotensin II. In contrast, administration of control-NPs or an equivalent dose of oral pioglitazone treatment produced no effects. Pioglitazone-NPs inhibited the activity of matrix metalloproteinases and cathepsins in the brachiocephalic arteries. Pioglitazone-NPs regulated inflammatory cytokine expression and also suppressed the expression of extracellular matrix metalloproteinase inducer in bone marrow-derived macrophages. CONCLUSIONS: Nanoparticle-mediated delivery of pioglitazone inhibited macrophage activation and atherosclerotic plaque ruptures in hyperlipidemic ApoE(-/-) mice. These results demonstrate a promising strategy with a favorable safety profile to prevent atherosclerotic plaque ruptures.


Subject(s)
Atherosclerosis/drug therapy , Cardiovascular Agents/pharmacology , Cell Differentiation/drug effects , Drug Carriers , Lactic Acid/chemistry , Macrophages, Peritoneal/drug effects , Monocytes/drug effects , Nanoparticles , Plaque, Atherosclerotic , Polyglycolic Acid/chemistry , Thiazolidinediones/pharmacology , Administration, Intravenous , Angiotensin II , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/chemically induced , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/chemistry , Cathepsins/metabolism , Cells, Cultured , Chemistry, Pharmaceutical , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Disease Progression , Inflammation Mediators/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Matrix Metalloproteinases/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Phenotype , Pioglitazone , Polylactic Acid-Polyglycolic Acid Copolymer , Rupture, Spontaneous , Thiazolidinediones/administration & dosage , Thiazolidinediones/chemistry
7.
Circ J ; 79(11): 2499-508, 2015.
Article in English | MEDLINE | ID: mdl-26346031

ABSTRACT

BACKGROUND: Atherosclerosis is a progressive inflammatory disease that can lead to sudden cardiac events by plaque rupture and subsequent thrombosis. Factor Xa (FXa) not only occupies a crucial position in the coagulation cascade responsible for thrombin generation, but also has pro-inflammatory effects. The hypothesis that Fondaparinux, the selective FXa inhibitor, attenuates plaque progression and promotes stability of atherosclerotic lesions was assessed. METHODS AND RESULTS: Fondaparinux (5 mg/kg body weight/day) or 0.9% saline was intraperitoneally administered for 4 weeks to apolipoprotein E-deficient mice (n=12 per group) with established atherosclerotic lesions in the innominate arteries. Fondaparinux did not remarkably decrease the progression of atherosclerosis development in apolipoprotein E-deficient mice, but increased the thickness of fibrous cap (P=0.049) and decreased the ratio of necrotic core (P=0.001) significantly. Moreover, Fondaparinux reduced the staining against Mac-2 (P=0.017), α-SMA (P=0.002), protease-activated receptor (PAR)-1 (P=0.001), PAR-2 (P=0.003), CD-31 (P=0.024), MMP-9 (P=0.000), MMP-13(P=0.011), VCAM-1 (P=0.041) and the mRNA expression of inflammatory mediators (P<0.05) significantly, such as interleukin (IL)-6, MCP-1, IFN-γ, TNF-α, IL-10 and Egr-1. CONCLUSIONS: Fondaparinux, the selective FXa inhibitor, can promote the stability of atherosclerotic lesions in apolipoprotein E-deficient mice, possibly through inhibiting expression of the inflammatory mediators in plaque and reduced synthesis of MMP-9 and MMP-13.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Brachiocephalic Trunk/drug effects , Factor Xa Inhibitors/pharmacology , Polysaccharides/pharmacology , Animals , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Blood Coagulation/drug effects , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Fibrosis , Fondaparinux , Inflammation Mediators/metabolism , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice, Knockout , Necrosis , Plaque, Atherosclerotic , Time Factors
8.
J Nutr Health Aging ; 19(7): 710-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26193853

ABSTRACT

INTRODUCTION: Low Carbohydrate High Protein diet represents a popular strategy to achieve weight loss. OBJECTIVE: The aim of this study was to characterize effects of low carbohydrate, high protein diet (LCHP) on atherosclerotic plaque development in brachiocephalic artery (BCA) in apoE/LDLR-/- mice and to elucidate mechanisms of proatherogenic effects of LCHP diet. MATERIALS AND METHODS: Atherosclerosis plaques in brachiocephalic artery (BCA) as well as in aortic roots, lipoprotein profile, inflammation biomarkers, expression of SREBP-1 in the liver as well as mortality were analyzed in Control diet (AIN-93G) or LCHP (Low Carbohydrate High Protein) diet fed mice. RESULTS: Area of atherosclerotic plaques in aortic roots or BCA from LCHP diet fed mice was substantially increased as compared to mice fed control diet and was characterized by increased lipids and cholesterol contents (ORO staining, FT-IR analysis), increased macrophage infiltration (MOMA-2) and activity of MMPs (zymography). Pro-atherogenic phenotype of LCHP fed apoE/LDLR-/- mice was associated with increased plasma total cholesterol concentration, and in LDL and VLDL fractions, increased TG contents in VLDL, and a modest increase in plasma urea. LCHP diet increased SCD-1 index, activated SREBP-1 transcription factor in the liver and triggered acute phase response as evidence by an increased plasma concentration of haptoglobin, CRP or AGP. Finally, in long-term experiment survival of apoE/LDLR-/- mice fed LCHP diet was substantially reduced as compared to their counterparts fed control diet suggesting overall detrimental effects of LCHP diet on health. CONCLUSIONS: The pro-atherogenic effect of LCHP diet in apoE/LDLR-/- mice is associated with profound increase in LDL and VLDL cholesterol, VLDL triglicerides, liver SREBP-1 upregulation, and systemic inflammation.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/chemically induced , Diet, Atherogenic/adverse effects , Diet, Carbohydrate-Restricted/adverse effects , Dietary Proteins/pharmacology , Receptors, LDL/genetics , Acute-Phase Reaction/chemically induced , Animals , Aorta/drug effects , Aorta/pathology , Apolipoproteins E/deficiency , Atherosclerosis/blood , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/pathology , Cholesterol, LDL/blood , Cholesterol, VLDL/blood , Dietary Proteins/administration & dosage , Female , Inflammation/chemically induced , Liver/drug effects , Liver/metabolism , Macrophages/drug effects , Male , Mice , Mice, Knockout , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/chemically induced , Receptors, LDL/deficiency , Sterol Regulatory Element Binding Protein 1/metabolism , Survival Analysis , Triglycerides/blood , Urea/blood
9.
Atherosclerosis ; 241(2): 297-304, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26062989

ABSTRACT

OBJECTIVE: Angiopoietin-2 (Ang-2) blocking agents are currently undergoing clinical trials for use in cancer treatment. Ang-2 has also been associated with rupture-prone atherosclerotic plaques in humans, suggesting a role for Ang-2 in plaque stability. Despite the availability of Ang-2 blocking agents, their clinical use is still lacking. Our aim was to establish if Ang-2 has a role in atheroma development and in the transition of subclinical to clinically relevant atherosclerosis. We investigated the effect of antibody-mediated Ang-2 blockage on atherogenesis after in a mouse model of atherosclerosis. METHODS: Hypercholesterolemic (low-density lipoprotein receptor(-/-) apolipoprotein B(100/100)) mice were subjected to high-cholesterol diet for eight weeks, one group with and one group without Ang-2 blocking antibody treatment during weeks 4-8.To enhance plaque development, a peri-adventitial collar was placed around the carotid arteries at the start of antibody treatment. Aortic root, carotid arteries and brachiocephalic arteries were analyzed to evaluate the effect of Ang-2 blockage on atherosclerotic plaque size and stable plaque characteristics. RESULTS: Anti-Ang-2 treatment reduced the size of fatty streaks in the brachiocephalic artery (-72%, p < 0.05). In addition, antibody-mediated Ang-2 blockage reduced plasma triglycerides (-27%, p < 0.05). In contrast, Ang-2 blockage did not have any effect on the size or composition (collagen content, macrophage percentage, adventitial microvessel density) of pre-existing plaques in the aortic root or collar-induced plaques in the carotid artery. CONCLUSIONS: Ang-2 blockage was beneficial as it decreased fatty streak formation and plasma triglyceride levels, but had no adverse effect on pre-existing atherosclerosis in hypercholesterolemic mice.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Antibodies/pharmacology , Atherosclerosis/prevention & control , Brachiocephalic Trunk/drug effects , Plaque, Atherosclerotic , Angiopoietin-2/immunology , Angiopoietin-2/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoprotein B-100/deficiency , Apolipoprotein B-100/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Brachiocephalic Trunk/immunology , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/blood , Carotid Artery Diseases/genetics , Carotid Artery Diseases/immunology , Carotid Artery Diseases/pathology , Carotid Artery Diseases/prevention & control , Cholesterol, Dietary/blood , Disease Models, Animal , Hypercholesterolemia/blood , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Male , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors , Triglycerides/blood
10.
Ross Fiziol Zh Im I M Sechenova ; 100(3): 328-38, 2014 Mar.
Article in Russian | MEDLINE | ID: mdl-25464733

ABSTRACT

In acute experiments in anesthetized cats myocardial left ventricular ischemia caused the decreasing of the arterial pressure, cardiac output, superior and inferior vena cava flow and venous return. The diminishing of the superior vena cava flow was caused by the decreasing of the cardiac output, increasing of the vascular resistance and decreasing of the blood flow in the region of the brachiocephalica artery. The inferior vena cava flow decreased following the diminishing of the cardiac output and abdominal aorta blood flow, while vascular resistance in this region did not change. In acute experiments in anesthetized rabbits following myocardial ischemia after the blockade of N-cholinoreceptors the superior and inferior vena cava flow decreased in the same level as in control animals. Following myocardial ischemia after the blockade of α-adrenoreceptors the superior and inferior vena cava flow decreased more significant, than in control animals.


Subject(s)
Hemodynamics/drug effects , Myocardial Ischemia/physiopathology , Vena Cava, Inferior/physiopathology , Vena Cava, Superior/physiopathology , Adrenergic alpha-Antagonists/pharmacology , Animals , Arterial Pressure/drug effects , Blood Pressure/drug effects , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/physiopathology , Cardiac Output/drug effects , Cats , Cholinergic Antagonists/pharmacology , Myocardial Ischemia/drug therapy , Rabbits , Receptors, Adrenergic, alpha/metabolism , Receptors, Cholinergic/metabolism , Vascular Resistance/drug effects , Vena Cava, Inferior/drug effects , Vena Cava, Superior/drug effects
11.
Physiol Genomics ; 46(14): 523-32, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24824211

ABSTRACT

Estradiol (E2) is a well-known modulator of fetal neuroendocrine activity and has been proposed as a critical endocrine signal readying the fetus for birth and postnatal life. To investigate the modulatory role of E2 on fetal stress responsiveness and the response of the fetal brain to asphyxic stress, we subjected chronically catheterized fetal sheep to a transient (10 min) brachiocephalic artery occlusion (BCO) or sham occlusion. Half of the fetuses received subcutaneous pellets that increased plasma E2 concentrations within the physiological range. Hypothalamic mRNA was analyzed using the Agilent 8x15k ovine array (019921), processed and annotated as previously reported by our laboratory. Analysis of the data by ANOVA revealed that E2 differentially regulated (DR) 561 genes, and BCO DR 894 genes compared with control and E2+BCO DR 1,153 genes compared with BCO alone (all P < 0.05). E2 upregulated epigenetic pathways and downregulated local steroid biosynthesis but did not significantly involve genes known to directly respond to the estrogen receptor. Brachiocephalic occlusion upregulated kinase pathways as well as genes associated with lymphocyte infiltration into the brain and downregulated neuropeptide synthesis. E2 upregulated immune- and apoptosis-related pathways after BCO and reduced kinase and epigenetic pathway responses to the BCO. Responses to BCO are different from responses to hypoxic hypoxia suggesting that mechanisms of responses to these two forms of brain hypoxia are distinct. We conclude that cerebral ischemia caused by BCO might stimulate lymphocyte infiltration into the brain and that this response appears to be modified by estradiol.


Subject(s)
Brachiocephalic Trunk/drug effects , Estradiol/pharmacology , Fetus/drug effects , Hypothalamus/drug effects , Transcriptome/drug effects , Transcriptome/genetics , Animals , Brachiocephalic Trunk/metabolism , Brain/drug effects , Brain/metabolism , Brain Ischemia/genetics , Brain Ischemia/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Fetus/metabolism , Hypothalamus/metabolism , Hypoxia/embryology , Hypoxia/genetics , Lymphocytes/drug effects , Neuropeptides/genetics , Neuropeptides/metabolism , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Sheep/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
12.
J Am Heart Assoc ; 3(2): e000492, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24755147

ABSTRACT

BACKGROUND: Bisphenol A (BPA) is a base chemical used extensively in many consumer products. BPA has recently been associated with increased risk of cardiovascular disease (CVD) in multiple large-scale human population studies, but the underlying mechanisms remain elusive. We previously reported that BPA activates the pregnane X receptor (PXR), which acts as a xenobiotic sensor to regulate xenobiotic metabolism and has pro-atherogenic effects in animal models upon activation. Interestingly, BPA is a potent agonist of human PXR but does not activate mouse or rat PXR signaling, which confounds the use of rodent models to evaluate mechanisms of BPA-mediated CVD risk. This study aimed to investigate the atherogenic mechanism of BPA using a PXR-humanized mouse model. METHODS AND RESULTS: A PXR-humanized ApoE deficient (huPXR•ApoE(-/-)) mouse line was generated that respond to human PXR ligands and feeding studies were performed to determine the effects of BPA exposure on atherosclerosis development. Exposure to BPA significantly increased atherosclerotic lesion area in the aortic root and brachiocephalic artery of huPXR•ApoE(-/-) mice by 104% (P<0.001) and 120% (P<0.05), respectively. By contrast, BPA did not affect atherosclerosis development in the control littermates without human PXR. BPA exposure did not affect plasma lipid levels but increased CD36 expression and lipid accumulation in macrophages of huPXR•ApoE(-/-) mice. CONCLUSION: These findings identify a molecular mechanism that could link BPA exposure to increased risk of CVD in exposed individuals. PXR is therefore a relevant target for future risk assessment of BPA and related environmental chemicals in humans.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/chemically induced , Benzhydryl Compounds/toxicity , Environmental Pollutants/toxicity , Phenols/toxicity , Receptors, Steroid/agonists , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , CD36 Antigens/metabolism , Humans , Lipids/blood , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pregnane X Receptor , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Risk Assessment , Risk Factors
13.
Circulation ; 129(8): 896-906, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24305567

ABSTRACT

BACKGROUND: Preventing atherosclerotic plaque destabilization and rupture is the most reasonable therapeutic strategy for acute myocardial infarction. Therefore, we tested the hypotheses that (1) inflammatory monocytes play a causative role in plaque destabilization and rupture and (2) the nanoparticle-mediated delivery of pitavastatin into circulating inflammatory monocytes inhibits plaque destabilization and rupture. METHODS AND RESULTS: We used a model of plaque destabilization and rupture in the brachiocephalic arteries of apolipoprotein E-deficient (ApoE(-/-)) mice fed a high-fat diet and infused with angiotensin II. The adoptive transfer of CCR2(+/+)Ly-6C(high) inflammatory macrophages, but not CCR2(-/-) leukocytes, accelerated plaque destabilization associated with increased serum monocyte chemoattractant protein-1 (MCP-1), monocyte-colony stimulating factor, and matrix metalloproteinase-9. We prepared poly(lactic-co-glycolic) acid nanoparticles that were incorporated by Ly-6G(-)CD11b(+) monocytes and delivered into atherosclerotic plaques after intravenous administration. Intravenous treatment with pitavastatin-incorporated nanoparticles, but not with control nanoparticles or pitavastatin alone, inhibited plaque destabilization and rupture associated with decreased monocyte infiltration and gelatinase activity in the plaque. Pitavastatin-incorporated nanoparticles inhibited MCP-1-induced monocyte chemotaxis and the secretion of MCP-1 and matrix metalloproteinase-9 from cultured macrophages. Furthermore, the nanoparticle-mediated anti-MCP-1 gene therapy reduced the incidence of plaque destabilization and rupture. CONCLUSIONS: The recruitment of inflammatory monocytes is critical in the pathogenesis of plaque destabilization and rupture, and nanoparticle-mediated pitavastatin delivery is a promising therapeutic strategy to inhibit plaque destabilization and rupture by regulating MCP-1/CCR2-dependent monocyte recruitment in this model.


Subject(s)
Drug Delivery Systems/methods , Monocytes/drug effects , Nanoparticles/metabolism , Plaque, Atherosclerotic/drug therapy , Quinolines/pharmacokinetics , Adoptive Transfer , Animals , Apolipoproteins E/genetics , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/immunology , Brachiocephalic Trunk/pathology , Cell Movement/drug effects , Cell Movement/immunology , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/metabolism , Disease Models, Animal , Genetic Therapy/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Nanoparticles/therapeutic use , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Receptors, CCR2/genetics
14.
World J Pediatr Congenit Heart Surg ; 4(4): 373-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24327630

ABSTRACT

BACKGROUND: Indirect clinical evidence suggests that coronary blood flow (CBF) is altered in patients palliated with systemic-to-pulmonary artery shunts (SPSs). The addition of epinephrine may exert additional effects. METHODS: A total of 11 newborn piglets underwent placement of a 3.5- to 4-mm graft between the innominate artery and the pulmonary artery. Doppler probes measured flow continuously in the aorta (aortic flow [AoF]), pulmonary artery and left coronary artery at baseline (SPS closed), SPS open, and during epinephrine administration (SPS closed and open). Each animal served as its own control. Systolic and diastolic CBF, resistance (coronary vascular resistance index [CVRI]), and myocardial oxygen supply demand ratio were calculated. RESULTS: Opening the SPS increased AoF and decreased systolic and diastolic pressure from baseline, with and without the presence of epinephrine. The CBF and CVRI decreased on opening the SPS in the presence of epinephrine. The decrease occurred only in diastole and was proportional to pulmonary-to-systemic flow ratio (Qp/Qs). Epinephrine infusion itself reduced CVRI with SPS closed, but there was little further decrease on opening SPS. Myocardial oxygen supply-demand ratio decreased on opening SPS at baseline and with epinephrine. CONCLUSIONS: This study suggests that SPS decreases CBF, especially in the presence of a higher Qp/Qs and epinephrine. The mechanism is largely due to the decrease in diastolic pressure and the inability of the coronary arteries to compensate with vasodilation.


Subject(s)
Brachiocephalic Trunk/surgery , Coronary Circulation/drug effects , Epinephrine/pharmacology , Heart Defects, Congenital/surgery , Pulmonary Artery/surgery , Pulmonary Circulation/drug effects , Vasoconstriction/drug effects , Anastomosis, Surgical/methods , Animals , Animals, Newborn , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/physiopathology , Coronary Circulation/physiology , Disease Models, Animal , Heart Defects, Congenital/physiopathology , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Circulation/physiology , Swine , Vasoconstrictor Agents/pharmacology
15.
Part Fibre Toxicol ; 10: 61, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24330719

ABSTRACT

OBJECTIVE: Diesel exhaust particulate (DEP), a major component of urban air pollution, has been linked to atherogenesis and precipitation of myocardial infarction. We hypothesized that DEP exposure would increase and destabilise atherosclerotic lesions in apolipoprotein E deficient (ApoE-/-) mice. METHODS: ApoE-/- mice were fed a 'Western diet' (8 weeks) to induce 'complex' atherosclerotic plaques, with parallel experiments in normal chow fed wild-type mice. During the last 4 weeks of feeding, mice received twice weekly instillation (oropharyngeal aspiration) of 35 µL DEP (1 mg/mL, SRM-2975) or vehicle (saline). Atherosclerotic burden was assessed by en-face staining of the thoracic aorta and histological examination of the brachiocephalic artery. RESULTS: Brachiocephalic atherosclerotic plaques were larger in ApoE-/- mice treated with DEP (59 ± 10%) than in controls (32 ± 7%; P = 0.017). In addition, DEP-treated mice had more plaques per section of artery (2.4 ± 0.2 vs 1.8 ± 0.2; P = 0.048) and buried fibrous layers (1.2 ± 0.2 vs 0.4 ± 0.1; P = 0.028). These changes were associated with lung inflammation and increased antioxidant gene expression in the liver, but not with changes in endothelial function, plasma lipids or systemic inflammation. CONCLUSIONS: Increased atherosclerosis is caused by the particulate component of diesel exhaust producing advanced plaques with a potentially more vulnerable phenotype. These results are consistent with the suggestion that removal of the particulate component would reduce the adverse cardiovascular effects of diesel exhaust.


Subject(s)
Oxidative Stress/drug effects , Particulate Matter/toxicity , Plaque, Atherosclerotic/chemically induced , Plaque, Atherosclerotic/pathology , Vehicle Emissions/toxicity , Airway Remodeling/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Apolipoproteins E/genetics , Blood Coagulation/drug effects , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , C-Reactive Protein/analysis , Disease Models, Animal , Fibrinogen/analysis , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oropharynx/metabolism , Particulate Matter/pharmacokinetics , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology
16.
Eur J Pharmacol ; 697(1-3): 106-16, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23085268

ABSTRACT

Hydrogen sulfide (H(2)S) is a novel gaseous mediator that plays important roles in atherosclerosis. The present study investigated the effect of a novel H(2)S-releasing aspirin, ACS14 (2-acetyloxybenzoic acid 4-(3-thioxo-3H-1,2-dithiol-5-yl)phenyl ester), on atherosclerotic plaques in fat-fed apoE(-/-) mice and the underlying mechanism with respect to CX3C chemokine receptor 1 (CX3CR1) in macrophages. Mouse macrophage cell line RAW264.7 or mouse peritoneal macrophages were preincubated with aspirin (50, 100 or 200µM), ACS14 (50, 100 or 200µM) or vehicle for 6h, and then stimulated with interferon (IFN)-γ (500U/ml) or lipopolysaccharide (LPS; 10µg/ml) for 12h. ACS14, but not aspirin, dose-dependently inhibited IFN-γ or LPS-induced CX3CR1 expression and CX3CR1-mediated chemotaxis in macrophages. The inhibitory effect of ACS14 on CX3CR1 expression was abolished by pretreatment with GW9662, a selective peroxisome proliferator-activated receptor (PPAR)-γ antagonist, suggesting that suppression of macrophage CX3CR1 expression by ACS14 is PPAR-γ dependent. Eight-week-old male apoE(-/-) mice received intraperitoneal ACS14 (15 or 30µmol/kg/day) or aspirin (15 or 30µmol/kg/day) 4 weeks after fat feeding. Twelve weeks after ACS14 or aspirin treatment, mice were sacrificed to evaluate the extent of atherosclerosis and CX3CR1 expression in brachiocephalic artery (BCA). We found that ACS14, but not aspirin, significantly downregulated CX3CR1 expression in atherosclerotic plaques. ACS14 considerably impeded the formation and development of atherosclerosis as compared to a molar equivalent dose of aspirin. These data indicate that ACS14 may prevent the progression of atherosclerosis by downregulating macrophage CX3CR1 expression via a PPAR-γ-dependent mechanism.


Subject(s)
Apolipoproteins E/deficiency , Aspirin/analogs & derivatives , Atherosclerosis/drug therapy , Brachiocephalic Trunk/drug effects , Disulfides/pharmacology , Hydrogen Sulfide/metabolism , Macrophages/drug effects , Animals , Apolipoproteins E/genetics , Aspirin/administration & dosage , Aspirin/metabolism , Aspirin/pharmacology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brachiocephalic Trunk/immunology , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , CX3C Chemokine Receptor 1 , Cell Line , Chemotaxis/drug effects , Disease Models, Animal , Disulfides/administration & dosage , Disulfides/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Immunohistochemistry , Injections, Intraperitoneal , Interferon-gamma/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/drug effects , PPAR gamma/metabolism , Plaque, Atherosclerotic , Receptors, Chemokine/drug effects , Receptors, Chemokine/metabolism , Time Factors
17.
Atherosclerosis ; 222(2): 337-43, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22446027

ABSTRACT

OBJECTIVE: Proliferation signal inhibitors/mTOR-inhibitors have been shown to reduce de novo development of hypercholesterolemic atherosclerosis in animal models. However, their effect on pre-existing atherosclerosis has not yet been studied. METHODS AND RESULTS: Feeding LDL-R-KO mice a high cholesterol diet for 12 weeks resulted in formation of moderate fibroatheroma (induction phase). Sixty mice received either everolimus (1 or 5 mg/kg) or no everolimus for further 12 weeks (treatment phase). Everolimus significantly enhanced hypercholesterolemia (plasma cholesterol +45%, p<0.001). Atherosclerosis progressed obstructively in treated and non-treated mice. Everolimus (5 mg/kg) tended to reduced progression in aortic root lesions (0.28±0.02 vs. 0.33±0.03 mm(2), p=ns) and brachiocephalic lesions (0.044±0.006 vs. 0.066±0.012 mm(2), p=ns) but without significance. Everolimus (5mg/kg) resulted in an arrest of CD68 positive plaque area (p=0.03) and nearly halved CD68 fraction (p=0.05) in aortic root lesions but not in brachiocephalic lesions. Taken together, despite a trend to reduced progression and inflammatory cell content there was less conclusive net effect of everolimus treatment than expected. CONCLUSION: A higher potential of everolimus in the treatment of atherosclerosis might be obscured by its concomitant hypercholesterolemia. Considering stronger effects in previous studies we suggest that everolimus might exert more potent anti-atherogenic properties in earlier stages of atherogenesis than in advanced atherosclerosis.


Subject(s)
Aorta/drug effects , Atherosclerosis/drug therapy , Brachiocephalic Trunk/drug effects , Hypercholesterolemia/complications , Protein Kinase Inhibitors/pharmacology , Receptors, LDL/deficiency , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aorta/enzymology , Aorta/pathology , Atherosclerosis/blood , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Biomarkers/blood , Brachiocephalic Trunk/enzymology , Brachiocephalic Trunk/pathology , Cholesterol/blood , Disease Models, Animal , Everolimus , Hypercholesterolemia/blood , Hypercholesterolemia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Time Factors
18.
Cardiovasc Drugs Ther ; 26(1): 23-30, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22038096

ABSTRACT

PURPOSE: Simvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, has antioxidant and anti-inflammatory properties that are independent of lipid-lowering abilities. This experiment was carried out to explore the effects of simvastatin on apoptosis in vulnerable atherosclerotic plaques of apoE-deficient mice. METHODS AND RESULTS: Eight weeks-old apoE(-/-) mice were fed a Western-type diet. Vulnerable atherosclerotic lesions were formed in the branchiocephalic artery at the age of 30-weeks, before simvastatin administration for 8 weeks. Simvastatin did neither affect the levels of plasma glucose and lipids, nor the size of atherosclerotic lesions. Analysis of plaque composition showed that simvastatin decreased the area of lipid core and increased the amounts of macrophages and smooth muscle cells in atherosclerotic plaques of apoE(-/-) mice. In addition, simvastatin down-regulated the expression of vascular cell adhesion molecule-1 (VCAM-1) by both inhibition of nuclear factor kappa B (NF-кB) activation and suppression of the expression of the receptor for advanced glycation end products (RAGE). Moreover, we found that simvastatin administration led to reduced TUNEL-positive cells in the aortic root lesions, accompanied by up-regulation of Bcl-2 and Bcl-xL expression, and decreased P(53) expression as shown by Western blot. CONCLUSION: In the present study, we show novel data to suggest that simvastatin could suppress apoptosis in vulnerable atherosclerotic plaques of apoE(-/-) mice by regulating the expression of apoptosis-related proteins, such as p(53), Bcl-2 and Bcl-xL.


Subject(s)
Apoptosis/drug effects , Atherosclerosis/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Plaque, Atherosclerotic/metabolism , Simvastatin/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Blood Glucose/analysis , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipids/blood , Male , Mice , Mice, Knockout , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Simvastatin/therapeutic use , Tumor Suppressor Protein p53/metabolism , bcl-X Protein/metabolism
19.
Int J Cardiovasc Imaging ; 28(5): 1181-91, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21688134

ABSTRACT

We have applied an imaging system of phase-contrast X-ray CT to the detection of atherosclerotic plaque components by means of the differences of tissue mass densities. In this study, we investigated the effect of the anti-platelet therapies, widely used for secondly prevention of cardiovascular events, on plaque stability and examined whether this novel technique could detect the changes of plaque components under the therapy. Apolipoprotein E-deficient mice were fed on high-cholesterol diet alone and either with 0.1% cilostazol or clopidogrel for 10 weeks. We assessed atherosclerotic lesion volumes and components at brachiocephalic artery by the phase-contrast X-ray CT imaging and histochemistry. The phase-contrast X-ray CT imaging could reveal that cilostazol and clopidogrel significantly decreased atherosclerotic lesion volumes at brachiocephalic artery (31.2% reduction in cilostazol group and 37.4% reduction in clopidogrel group), compared with control group. In addition, the mass densities calculated by this method revealed the anti-platelet treatment increased stable plaque areas including high collagen content, but decreased unstable plaque areas including lipid and macrophage content. These findings were confirmed by histological analyses. Real-time PCR analyses indicated that anti-platelets inhibited gene expressions of cytokines and adhesion molecules, such as IFNγ and ICAM-1. Anti-platelet therapies had a beneficial effect on plaque stability maybe due to anti-inflammatory actions. Phase-contrast X-ray CT imaging could quantify the plaque volume and qualify the plaque components affected by anti-platelet therapies. This novel phase-contrast X-ray CT imaging system could be a plausible method to detect the unstable plaque non-invasively in the future.


Subject(s)
Atherosclerosis/diagnostic imaging , Atherosclerosis/drug therapy , Brachiocephalic Trunk/drug effects , Brachiocephalic Trunk/diagnostic imaging , Platelet Aggregation Inhibitors/pharmacology , Tetrazoles/pharmacology , Ticlopidine/analogs & derivatives , Tomography, X-Ray Computed , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Body Weight , Brachiocephalic Trunk/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cholesterol, Dietary , Cilostazol , Clopidogrel , Collagen/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Immunohistochemistry , Inflammation Mediators/metabolism , Lipids/blood , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Plaque, Atherosclerotic , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ticlopidine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...