Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Arterioscler Thromb Vasc Biol ; 34(8): 1723-30, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24947528

ABSTRACT

OBJECTIVE: Perivascular adipose tissue (PVAT) expands during obesity, is highly inflamed, and correlates with coronary plaque burden and increased cardiovascular risk. We tested the hypothesis that PVAT contributes to the vascular response to wire injury and investigated the underlying mechanisms. APPROACH AND RESULTS: We transplanted thoracic aortic PVAT from donor mice fed a high-fat diet to the carotid arteries of recipient high-fat diet-fed low-density lipoprotein receptor knockout mice. Two weeks after transplantation, wire injury was performed, and animals were euthanized 2 weeks later. Immunohistochemistry was performed to quantify adventitial macrophage infiltration and neovascularization and neointimal lesion composition and size. Transplanted PVAT accelerated neointimal hyperplasia, adventitial macrophage infiltration, and adventitial angiogenesis. The majority of neointimal cells in PVAT-transplanted animals expressed α-smooth muscle actin, consistent with smooth muscle phenotype. Deletion of monocyte chemoattractant protein-1 in PVAT substantially attenuated the effects of fat transplantation on neointimal hyperplasia and adventitial angiogenesis, but not adventitial macrophage infiltration. Conditioned medium from perivascular adipocytes induced potent monocyte chemotaxis in vitro and angiogenic responses in cultured endothelial cells. CONCLUSIONS: These findings indicate that PVAT contributes to the vascular response to wire injury, in part through monocyte chemoattractant protein-1-dependent mechanisms.


Subject(s)
Adipose Tissue/transplantation , Carotid Artery Injuries/metabolism , Chemokine CCL2/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Actins/metabolism , Adipocytes/metabolism , Adipocytes/transplantation , Adipose Tissue/metabolism , Animals , Biomarkers/metabolism , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cells, Cultured , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemotaxis , Coculture Techniques , Culture Media, Conditioned/metabolism , Diet, High-Fat , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Hyperplasia , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Neovascularization, Pathologic , Phenotype , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Time Factors , Transendothelial and Transepithelial Migration
2.
J Am Heart Assoc ; 2(2): e000065, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23537804

ABSTRACT

BACKGROUND: Recruitment of macrophage precursors to the adventitia plays a key role in the pathogenesis of abdominal aortic aneurysms (AAAs), but molecular mechanisms remain undefined. The innate immune signaling molecule CD14 was reported to be upregulated in adventitial macrophages in a murine model of AAA and in monocytes cocultured with aortic adventitial fibroblasts (AoAf) in vitro, concurrent with increased interleukin-6 (IL-6) expression. We hypothesized that CD14 plays a crucial role in adventitial macrophage precursor recruitment early during AAA formation. METHODS AND RESULTS: CD14(-/-) mice were resistant to AAA formation induced by 2 different AAA induction models: aortic elastase infusion and systemic angiotensin II (AngII) infusion. CD14 gene deletion led to reduced aortic macrophage infiltration and diminished elastin degradation. Adventitial monocyte binding to AngII-infused aorta in vitro was dependent on CD14, and incubation of human acute monocytic leukemia cell line-1 (THP-1) monocytes with IL-6 or conditioned medium from perivascular adipose tissue (PVAT) upregulated CD14 expression. Conditioned medium from AoAf and PVAT induced CD14-dependent monocyte chemotaxis, which was potentiated by IL-6. CD14 expression in aorta and plasma CD14 levels were increased in AAA patients compared with controls. CONCLUSIONS: These findings link CD14 innate immune signaling via a novel IL-6 amplification loop to adventitial macrophage precursor recruitment in the pathogenesis of AAA.


Subject(s)
Aortic Aneurysm, Abdominal/immunology , Cell Movement/immunology , Interleukin-6/immunology , Lipopolysaccharide Receptors/immunology , Macrophages/immunology , Monocyte-Macrophage Precursor Cells/immunology , Adventitia/immunology , Animals , Cell Line, Tumor , Cell Migration Assays, Macrophage , Cells, Cultured , Disease Models, Animal , Humans , Immunity, Innate , Macrophages, Peritoneal , Mice , Mice, Transgenic , Signal Transduction/immunology
3.
J Inflamm (Lond) ; 8: 4, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21352551

ABSTRACT

BACKGROUND: Bacterial endotoxin, long recognized as a potent pro-inflammatory mediator in acute infectious processes, has more recently been identified as a risk factor for atherosclerosis and other cardiovascular diseases. When endotoxin enters the bloodstream, one of the first cells activated is the circulating monocyte, which exhibits a wide range of pro-inflammatory responses. METHODS: We studied the effect of low doses of E. coli LPS on IL-8 release and superoxide formation by freshly isolated human peripheral blood mononuclear cells (PBMC). RESULTS: IL-8 release was consistently detectable at 10 pg/ml of endotoxin, reaching a maximum at 1 ng/ml, and was exclusively produced by monocytes; the lymphocytes neither produced IL-8, nor affected monocyte IL-8 release. Superoxide production was detectable at 30 pg/ml of endotoxin, reaching a maximum at 3 ng/ml. Peak respiratory burst activity was seen at 15-20 min, and superoxide levels returned to baseline by 1 h. IL-8 release was dependent on both membrane-associated CD14 (mCD14) and Toll-like receptor 4 (TLR4. Superoxide production was dependent on the presence of LBP, but was not significantly affected by a blocking antibody to TLR4. Moreover, treatment with lovastatin inhibited LPS-dependent IL-8 release and superoxide production. CONCLUSIONS: These findings suggest that IL-8 release and the respiratory burst are regulated by distinct endotoxin-dependent signaling pathways in PBMC in low level of endotoxin exposure. Selectively modulating these pathways could lead to new approaches to treat chronic inflammatory diseases, such as atherosclerosis, while preserving the capacity of monocytes to respond to acute bacterial infections.

4.
Curr Opin Pharmacol ; 10(2): 191-6, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20060362

ABSTRACT

Crosstalk between cells in the blood vessel wall is vital to normal vascular function and is perturbed in diseases such as atherosclerosis and hypertension. Perivascular adipocytes reside at the adventitial border of blood vessels but until recently were virtually ignored in studies of vascular function. However, perivascular adipocytes have been demonstrated to be powerful endocrine cells capable of responding to metabolic cues and transducing signals to adjacent blood vessels. Accordingly, crosstalk between perivascular adipose tissue (PVAT) and blood vessels is now being intensely examined. Emerging evidence suggests that PVAT regulates vascular function through numerous mechanisms, but evidence to date suggests modulation of three key aspects that are the focus of this review: inflammation, vasoreactivity, and smooth muscle cell proliferation.


Subject(s)
Adipose Tissue/metabolism , Blood Vessels/metabolism , Muscle, Smooth, Vascular/physiology , Signal Transduction/physiology , Adipocytes/metabolism , Adipose Tissue/physiology , Animals , Blood Vessels/physiology , Cell Proliferation , Humans , Inflammation/metabolism , Inflammation/physiopathology , Models, Biological , Vasoconstriction/physiology , Vasodilation/physiology
5.
Circ Res ; 104(4): 541-9, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19122178

ABSTRACT

Adipose tissue depots originate from distinct precursor cells, are functionally diverse, and modulate disease processes in a depot-specific manner. However, the functional properties of perivascular adipocytes, and their influence on disease of the blood vessel wall, remain to be determined. We show that human coronary perivascular adipocytes exhibit a reduced state of adipocytic differentiation as compared with adipocytes derived from subcutaneous and visceral (perirenal) adipose depots. Secretion of antiinflammatory adiponectin is markedly reduced, whereas that of proinflammatory cytokines interleukin-6, interleukin-8, and monocyte chemoattractant protein-1, is markedly increased in perivascular adipocytes. These depot-specific differences in adipocyte function are demonstrable in both freshly isolated adipose tissues and in vitro-differentiated adipocytes. Murine aortic arch perivascular adipose tissues likewise express lower levels of adipocyte-associated genes as compared with subcutaneous and visceral adipose tissues. Moreover, 2 weeks of high-fat feeding caused further reductions in adipocyte-associated gene expression, while upregulating proinflammatory gene expression, in perivascular adipose tissues. These changes were observed in the absence of macrophage recruitment to the perivascular adipose depot. We conclude that perivascular adipocytes exhibit reduced differentiation and a heightened proinflammatory state, properties that are intrinsic to the adipocytes residing in this depot. Dysfunction of perivascular adipose tissue induced by fat feeding suggests that this unique adipose depot is capable of linking metabolic signals to inflammation in the blood vessel wall.


Subject(s)
Adipocytes/immunology , Adipogenesis , Connective Tissue/immunology , Dietary Fats/adverse effects , Inflammation Mediators/metabolism , Intra-Abdominal Fat/immunology , Subcutaneous Fat/immunology , Adipocytes/pathology , Adipogenesis/genetics , Adiponectin/metabolism , Adipose Tissue, Brown/immunology , Animals , Aorta, Thoracic/immunology , Atherosclerosis/immunology , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Shape , Cells, Cultured , Chemokine CCL2/metabolism , Connective Tissue/pathology , Coronary Vessels/immunology , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation , Humans , Inflammation/immunology , Interleukin-6/metabolism , Interleukin-8/metabolism , Intra-Abdominal Fat/pathology , Male , Mice , Mice, Inbred C57BL , Models, Animal , PPAR gamma/metabolism , Phenotype , Subcutaneous Fat/pathology , Time Factors
6.
Am J Physiol Heart Circ Physiol ; 294(5): H2053-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18359891

ABSTRACT

Surfactant protein D (SP-D) is a constituent of the innate immune system that plays a role in the host defense against lung pathogens and in modulating inflammatory responses. While SP-D has been detected in extrapulmonary tissues, little is known about its expression and function in the vasculature. Immunostaining of human coronary artery tissue sections demonstrated immunoreactive SP-D protein in smooth muscle cells (SMCs) and endothelial cells. SP-D was also detected in isolated human coronary artery SMCs (HCASMCs) by PCR and immunoblot analysis. Treatment of HCASMCs with endotoxin (LPS) stimulated the release of IL-8, a proinflammatory cytokine. This release was inhibited >70% by recombinant SP-D. Overexpression of SP-D by adenoviral-mediated gene transfer in HCASMCs inhibited both LPS- and TNF-alpha-induced IL-8 release. Overexpression of SP-D also enhanced uptake of Chlamydia pneumoniae elementary bodies into HCASMCs while attenuating IL-8 production induced by bacterial exposure. Both LPS and TNF-alpha increased SP-D mRNA levels by five- to eightfold in HCASMCs, suggesting that inflammatory mediators upregulate the expression of SP-D. In conclusion, SP-D is expressed in human coronary arteries and functions as an anti-inflammatory protein in HCASMCs. SP-D may also participate in the host defense against pathogens that invade the vascular wall.


Subject(s)
Immunity, Innate , Inflammation/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Cells, Cultured , Chlamydophila pneumoniae/metabolism , Chlamydophila pneumoniae/pathogenicity , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Humans , Inflammation/immunology , Inflammation/prevention & control , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/immunology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Phagocytosis , Pulmonary Surfactant-Associated Protein D/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Time Factors , Transduction, Genetic , Tumor Necrosis Factor-alpha/metabolism
7.
Peptides ; 29(1): 83-92, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18082911

ABSTRACT

Enkephalins are opioid peptides that are found at high levels in the brain and endocrine tissues. Studies have shown that enkephalins play an important role in behavior, pain, cardiac function, cellular growth, immunity, and ischemic tolerance. Our global hypothesis is that enkephalins are released from non-neuronal tissues in response to brief ischemia or exercise, and that this release contributes to cardioprotection. To identify tissues that could serve as potential sources of enkephalins, we used real-time PCR, Western blot analysis, ELISA, immunofluorescence microscopy, and ex vivo models of enkephalin release. We found widespread expression of preproenkephalin (pPENK) mRNA and production of the enkephalin precursor protein proenkephalin (PENK) in rat and mouse tissues, as well as in tissues and cells from humans and pigs. Immunofluorescence microscopy with anti-enkephalin antisera demonstrated immunoreactivity in rat tissues, including heart and skeletal muscle myocytes, intestinal and kidney epithelium, and intestinal smooth muscle cells. Finally, isolated tissue studies showed that heart, skeletal muscle, and intestine released enkephalins ex vivo. Together our studies indicate that multiple non-neuronal tissues produce PENK and release enkephalins. These data support the hypothesis that non-neuronal tissues could play a role in both local and systemic enkephalin-mediated effects.


Subject(s)
Enkephalins/metabolism , Gene Expression Profiling , Protein Precursors/genetics , Animals , Blotting, Western , Enkephalins/biosynthesis , Enkephalins/chemistry , Enkephalins/genetics , Enzyme-Linked Immunosorbent Assay , Epithelium/metabolism , Heart/physiology , Humans , Intestinal Mucosa/metabolism , Intestines/cytology , Kidney/cytology , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Middle Aged , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Protein Precursors/biosynthesis , Protein Precursors/chemistry , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Swine
8.
Am J Physiol Heart Circ Physiol ; 294(1): H402-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17951371

ABSTRACT

Exercise increases serum opioid levels and improves cardiovascular health. Here we tested the hypothesis that opioids contribute to the acute cardioprotective effects of exercise using a rat model of exercise-induced cardioprotection. For the standard protocol, rats were randomized to 4 days of treadmill training and 1 day of vigorous exercise (day 5), or to a sham exercise control group. On day 6, animals were killed, and global myocardial ischemic tolerance was assessed on a modified Langendorff apparatus. Twenty minutes of ischemia followed by 3 h of reperfusion resulted in a mean infarct size of 42 +/- 4% in hearts from sham exercise controls and 21 +/- 3% (P < 0.001) in the exercised group. The cardioprotective effects of exercise were gone by 5 days after the final exercise period. To determine the role of opioid receptors in exercise-induced cardioprotection, rats were exercised according to the standard protocol; however, just before exercise on days 4 and 5, rats were injected subcutaneously with 10 mg/kg of the opioid receptor antagonist naltrexone. Similar injections were performed in the sham exercise control group. Naltrexone had no significant effect on baseline myocardial ischemic tolerance in controls (infarct size 43 +/- 4%). In contrast, naltrexone treatment completely blocked the cardioprotective effect of exercise (infarct size 40 +/- 5%). Exercise was also associated with an early increase in myocardial mRNA levels for several opioid system genes and with sustained changes in a number of genes that regulate inflammation and apoptosis. These findings demonstrate that the acute cardioprotective effects of exercise are mediated, at least in part, through opioid receptor-dependent mechanisms that may include changes in gene expression.


Subject(s)
Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Opioid Peptides/metabolism , Physical Exertion , Protein Precursors/metabolism , Receptors, Opioid/metabolism , Animals , Apoptosis/genetics , Disease Models, Animal , Gene Expression , Inflammation/genetics , Inflammation/metabolism , Male , Myocardial Ischemia/complications , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Opioid Peptides/genetics , Protein Precursors/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid/genetics , Time Factors
9.
Curr Pharm Des ; 12(32): 4229-45, 2006.
Article in English | MEDLINE | ID: mdl-17100625

ABSTRACT

Cardiovascular disease ranks among the leading causes of morbidity and mortality in adult populations in the Western world. Significant progress in understanding the etiology of cardiovascular disease has come from recent recognition that chronic inflammation plays a key role in its development. The principal mediators of this inflammatory response, and the mechanisms by which they work, however, are incompletely understood. Moreover, the complex nature of the inflammatory response poses significant challenges to the development of effective and targeted treatments. Potentially promising targets to reduce inflammation in atherosclerosis include Toll-like receptor (TLR) pathways and anti-inflammatory factors that modulate TLR signaling. In this review, we outline studies that provide insight into the links between cardiovascular disease and inflammation, focusing on innate immunity and endotoxin/TLR4 signaling. We also discuss the contribution of specific host immune/inflammatory responses to atherogenesis, and describe cellular signaling pathways (lipopolysaccharide-binding protein [LBP], CD14, MD-2, TLR4, MyD88, and NF-kappaB, among others) that play key roles in innate immune signaling. Finally, we discuss the therapeutic potential of modulating these cellular signaling pathways as future strategies for the prevention and treatment of cardiovascular disease, including such approaches as specific targeting of the TLR4 signaling pathway, antibiotic therapy, drug classes with broad anti-inflammatory activity (statins, thiazolidinediones), and the potential of vaccine development. Because of the complexity of the links between low-level chronic infections, inflammation, and atherosclerosis, treatment and prevention of cardiovascular disease will likely require an integrated approach that utilizes a combination of these strategies to target the underlying inflammatory processes.


Subject(s)
Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Coronary Vessels/pathology , Endotoxins/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Cardiovascular Diseases/pathology , Coronary Vessels/metabolism , Humans , Toll-Like Receptor 4/immunology
10.
Pediatr Pulmonol ; 41(1): 23-34, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16267824

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors that play central roles in lipid and glucose homeostasis, cellular differentiation, and the immune/inflammatory response. Growing evidence indicates that changes in expression and activation of PPARs likely modulate conditions as diverse as diabetes, atherosclerosis, cancer, asthma, Parkinson's disease, and Alzheimer's disease. Activation of these receptors by natural or pharmacologic ligands leads to both gene-dependent and gene-independent effects that alter the expression of a wide array of proteins. In the lung, PPARs are expressed by alveolar macrophages, as well as by epithelial, endothelial, and smooth muscle cells. Studies both in vitro and in vivo suggest that PPAR ligands may have anti-inflammatory effects in asthma, pulmonary sarcoidosis, and pulmonary alveolar proteinosis, as well as antiproliferative and antiangiogenic effects in epithelial lung cancers. Further studies to understand the contribution of these receptors to health and disease will be important for determining whether they represent a promising target for therapeutic intervention.


Subject(s)
Lung Diseases/physiopathology , Lung Diseases/therapy , Peroxisome Proliferator-Activated Receptors/immunology , Peroxisome Proliferator-Activated Receptors/metabolism , Animals , Asthma/physiopathology , Asthma/therapy , Gene Expression Regulation , Humans , Inflammation Mediators/metabolism , Lipid Metabolism , Lung Neoplasms/physiopathology , Lung Neoplasms/therapy , Peroxisome Proliferator-Activated Receptors/genetics , Polymorphism, Genetic , Pulmonary Alveolar Proteinosis/physiopathology , Pulmonary Alveolar Proteinosis/therapy , Sarcoidosis, Pulmonary/physiopathology , Sarcoidosis, Pulmonary/therapy
11.
J Immunol ; 175(6): 4017-23, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16148150

ABSTRACT

Pseudomonas aeruginosa secretes numerous factors that alter host cell function and may contribute to disease pathogenesis. Among recognized virulence factors is the redox-active phenazine pyocyanin. We have recently demonstrated that the precursor for pyocyanin, phenazine-1-carboxylic acid (PCA), increases oxidant formation and alters gene expression in human airway epithelial cells. We report in this work that PCA and pyocyanin increase expression of ICAM-1 both in vivo and in vitro. Moreover, phenazines enhanced cytokine-dependent increases in IL-8 and ICAM-1. Antioxidant intervention studies indicated both similarities and differences between PCA and pyocyanin. The thiol antioxidant N-acetyl cysteine, extracellular catalase, and inducible NO synthase inhibitors inhibited ICAM-1 and IL-8 increases in response to both phenazines. However, pyocyanin was significantly more sensitive to N-acetylcysteine inhibition. Interestingly, hydroxyl radical scavengers inhibited the response to pyocyanin, but not to PCA. These studies suggest that P. aeruginosa phenazines coordinately up-regulate chemokines (IL-8) and adhesion molecules (ICAM-1) by mechanisms that are, at least in part, oxidant dependent. However, results indicate that the mechanisms by which PCA and pyocyanin exert their effects are not identical, and not all antioxidant interventions are equally effective in inhibiting phenazine-mediated proinflammatory effects.


Subject(s)
Intercellular Adhesion Molecule-1/biosynthesis , Interleukin-8/biosynthesis , Pyocyanine/pharmacology , Respiratory Mucosa/metabolism , Antioxidants/pharmacology , Cells, Cultured , Epithelial Cells , Free Radical Scavengers/pharmacology , Humans , Oxidation-Reduction , Phenazines/pharmacology , Pseudomonas aeruginosa/pathogenicity , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Up-Regulation/drug effects , Virulence Factors/pharmacology
12.
Timely Top Med Cardiovasc Dis ; 9: E1, 2005 Feb 04.
Article in English | MEDLINE | ID: mdl-15824760

ABSTRACT

Statins, a group of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, are widely used in clinical practice for their efficacy in producing significant reductions in plasma cholesterol and LDL cholesterol and in reducing morbidity and mortality from cardiovascular disease. However, several large clinical trials have suggested that the cholesterol-lowering effects of statins may not completely account for the reduced incidence of cardiovascular disease seen in patients receiving statin therapy. A number of recent reports have shown that statins may also have important antiinflammatory effects, in addition to their effects on plasma lipids. Since inflammation is closely linked to the production of reactive oxygen species (ROS), the molecular basis of the observed antiinflammatory effects of statins may relate to their ability block the production and/or activity of ROS. In this review, we will discuss both the inhibition of ROS generation by statins, through interference with NAD(P)H oxidase expression and activity, and the actions of statins that serve to blunt the damaging effects of these radicals, including effects on antioxidant enzymes, lipid peroxidation, LDL cholesterol oxidation and nitric oxide synthase. These antioxidant effects of statins likely contribute to their clinical efficacy in treating cardiovascular disease as well as other chronic conditions associated with increased oxidative stress in humans.


Subject(s)
Antioxidants , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Antioxidants/pharmacology , Cardiovascular Diseases/drug therapy , Cholesterol, LDL/blood , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipid Peroxidation/drug effects , NADPH Oxidases/metabolism , Nitric Oxide/therapeutic use , Nitric Oxide Synthase/metabolism , Reactive Oxygen Species/metabolism
13.
Arterioscler Thromb Vasc Biol ; 24(12): 2227-36, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15472123

ABSTRACT

Atherosclerosis is increasingly recognized as a chronic inflammatory disease. Although a variety of inflammatory markers (ie, C-reactive protein) have been associated with atherosclerosis and its consequences, it is important to identify principal mediators of the inflammatory responses. One potentially important source of vascular inflammation in atherosclerosis is bacterial endotoxin. Mutations in Toll-like receptor 4 (TLR-4), an integral component of the endotoxin signaling complex, are fairly common in the Caucasian population and have recently been associated with reduced incidence of atherosclerosis and other cardiovascular diseases in some studies. Moreover, epidemiological studies suggest that endotoxemia at levels as low as 50 pg/mL constitutes a strong risk factor for the development of atherosclerosis. Endotoxin concentrations in this range may be produced by a variety of common subclinical Gram-negative infections. In this article, we outline the main elements of the endotoxin signaling receptor complex that initiates proinflammatory signaling (lipopolysaccharide binding protein [LBP], CD14, TLR-4, and MD-2) and discuss how changes in expression of these molecules may affect proatherogenic responses in the vessel wall. We also describe some of the proinflammatory effects of endotoxin that may be relevant to atherosclerosis, and discuss how serum lipoproteins, especially high-density lipoprotein, may modulate endotoxin-induced inflammatory responses. Further, we discuss recent findings suggesting that the lipid-lowering statins may have an additional protective role in blocking at least some of these proinflammatory signaling pathways. Finally, we discuss species diversity with regard to endotoxin signaling that should be considered when extrapolating experimental data from animal models to humans.


Subject(s)
Arteriosclerosis/pathology , Endotoxins/physiology , Animals , Humans , Inflammation/metabolism
14.
J Immunol ; 173(2): 1336-43, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15240728

ABSTRACT

Low-level endotoxemia has been identified as a powerful risk factor for atherosclerosis. However, little is known about the mechanisms that regulate endotoxin responsiveness in vascular cells. We conducted experiments to compare the relative responses of human coronary artery endothelial cells (HCAEC) and smooth muscle cells (HCASMC) to very low levels of endotoxin, and to elucidate the mechanisms that regulate endotoxin responsiveness in vascular cells. Endotoxin (10-fold higher in magnitude at >10-fold lower threshold concentrations (10-30 pg/ml) compared with HCAEC. This remarkable sensitivity of HCASMC to very low endotoxin concentrations, comparable to that found in circulating monocytes, was not due to differential expression of TLR4, which was detected in HCAEC, HCASMC, and intact coronary arteries. Surprisingly, membrane-bound CD14 was detected in seven different lines of HCASMC, conferring responsiveness to endotoxin and to lipoteichoic acid, a product of Gram-positive bacteria, in these cells. These results suggest that the low levels of endotoxin associated with increased risk for atherosclerosis are sufficient to produce inflammatory responses in coronary artery cells. Because CD14 recognizes a diverse array of inflammatory mediators and functions as a pattern recognition molecule in inflammatory cells, expression of membrane-bound CD14 in HCASMC implies a potentially broader role for these cells in transducing innate immune responses in the vasculature.


Subject(s)
Endotoxins/pharmacology , Lipopolysaccharide Receptors/immunology , Myocytes, Smooth Muscle/immunology , Arteries/drug effects , Arteries/immunology , Chemokine CCL2/metabolism , Coronary Vessels/drug effects , Coronary Vessels/immunology , Humans , Interleukin-8/metabolism , Myocytes, Smooth Muscle/drug effects
15.
Circulation ; 109(18): 2221-6, 2004 May 11.
Article in English | MEDLINE | ID: mdl-15117840

ABSTRACT

BACKGROUND: Ghrelin is a novel growth hormone-releasing peptide that has been shown to improve cachexia in heart failure and cancer and to ameliorate the hemodynamic and metabolic disturbances in septic shock. Because cytokine-induced inflammation is critical in these pathological states and because the growth hormone secretagogue receptor has been identified in blood vessels, we examined whether ghrelin inhibits proinflammatory responses in human endothelial cells in vitro and after administration of endotoxin to rats in vivo. METHODS AND RESULTS: Human umbilical vein endothelial cells (HUVECs) were treated with or without tumor necrosis factor-alpha (TNF-alpha), and induction of proinflammatory cytokines and mononuclear cell adhesion were determined. Ghrelin (0.1 to 1000 ng/mL) inhibited both basal and TNF-alpha-induced cytokine release and mononuclear cell binding. Intravenous administration of ghrelin also inhibited endotoxin-induced proinflammatory cytokine production in rats in vivo. Ghrelin inhibited H2O2-induced cytokine release in HUVECs, suggesting that the peptide blocks redox-mediated cellular signaling. Moreover, ghrelin inhibited basal and TNF-alpha-induced activation of nuclear factor-kappaB. Des-acyl ghrelin had no effect on TNF-alpha-induced cytokine production in HUVECs, suggesting that the antiinflammatory effects of ghrelin require interaction with endothelial growth hormone secretagogue receptors. CONCLUSIONS: Ghrelin inhibits proinflammatory cytokine production, mononuclear cell binding, and nuclear factor-kappaB activation in human endothelial cells in vitro and endotoxin-induced cytokine production in vivo. These novel antiinflammatory actions of ghrelin suggest that the peptide could play a modulatory role in atherosclerosis, especially in obese patients, in whom ghrelin levels are reduced.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Endothelium, Vascular/drug effects , Peptide Hormones/pharmacology , Animals , Arteriosclerosis/metabolism , Cell Adhesion/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Depression, Chemical , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endotoxemia/metabolism , Gene Expression Regulation/drug effects , Genes, Reporter , Ghrelin , Humans , Hydrogen Peroxide/pharmacology , Interleukin-8/biosynthesis , Interleukin-8/genetics , NF-kappa B/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , U937 Cells/drug effects , U937 Cells/metabolism , Umbilical Veins
16.
Drugs Today (Barc) ; 40(12): 975-90, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15645009

ABSTRACT

Statins, a group of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, are widely used in clinical practice for their efficacy in producing significant reductions in plasma cholesterol and LDL cholesterol and in reducing morbidity and mortality from cardiovascular disease. However, several large clinical trials have suggested that the cholesterol-lowering effects of statins may not completely account for the reduced incidence of cardiovascular disease seen in patients receiving statin therapy. A number of recent reports have shown that statins may also have important antiinflammatory effects, in addition to their effects on plasma lipids. Since inflammation is closely linked to the production of reactive oxygen species (ROS), the molecular basis of the observed antiinflammatory effects of statins may relate to their ability block the production and/or activity of ROS. In this review, we will discuss both the inhibition of ROS generation by statins, through interference with NAD(P)H oxidase expression and activity, and the actions of statins that serve to blunt the damaging effects of these radicals, including effects on antioxidant enzymes, lipid peroxidation, LDL cholesterol oxidation and nitric oxide synthase. These antioxidant effects of statins likely contribute to their clinical efficacy in treating cardiovascular disease as well as other chronic conditions associated with increased oxidative stress in humans.


Subject(s)
Antioxidants/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Reactive Oxygen Species/adverse effects , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism
17.
Arterioscler Thromb Vasc Biol ; 23(9): 1576-82, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12816876

ABSTRACT

BACKGROUND: Low-level endotoxemia (ie, >or=50 pg/mL) in apparently healthy subjects was recently identified as a powerful, independent risk factor for atherosclerosis. METHODS AND RESULTS: We treated human saphenous veins (HSVs) with low levels of endotoxin. Release of the proinflammatory chemokines interleukin-8 (IL-8) and monocyte chemoattractant protein-1 (MCP-1) was measured by ELISA. Superoxide was determined by using the fluorescent probe dihydroethidium (HE), and monocyte binding was assessed with calcein-labeled U-937 cells. Three- to 4-fold increases in MCP-1 and IL-8 release were observed at endotoxin concentrations of 100 pg/mL; these increases were inhibited by the 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor atorvastatin. Studies in cultured endothelial cells suggest that the mechanism is related to inhibition of isoprenylation (ie, geranylgeranylation) rather than cholesterol formation. Endotoxin produced dose-dependent increases in HE fluorescence that were inhibited by the superoxide dismutase mimics Tiron and MnTBAP. Endotoxin potently induced U-937 cell binding to HSV; binding was inhibited by both Tiron and atorvastatin. Toll-like receptor-4 expression was detected in cultured HSV endothelial and smooth muscle cells and in intact HSV. CONCLUSIONS: Clinically relevant levels of endotoxin, as reported in ambulatory populations, have profound inflammatory effects on intact HSV. Inhibition of endotoxin-induced vascular inflammation might contribute to the beneficial effects of statins in treating atherosclerosis.


Subject(s)
Blood Vessels/physiology , Endotoxins/antagonists & inhibitors , Endotoxins/immunology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Blood Vessels/drug effects , Blood Vessels/metabolism , Cell Line, Tumor , Chemokine CCL2/metabolism , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/prevention & control , Interleukin-8/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Saphenous Vein/drug effects , Saphenous Vein/metabolism , Saphenous Vein/physiology , U937 Cells/metabolism
18.
Free Radic Biol Med ; 34(7): 937-46, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12654483

ABSTRACT

Oxidized lipids, such as 13-hydroperoxyoctadecadienoic acid (13-HPODE), have been implicated in the pathogenesis of atherosclerosis. 13-HPODE, a constituent of oxidized low-density lipoproteins, can induce cytotoxicity of vascular smooth muscle cells (SMC), which may facilitate plaque destabilization and/or rupture. 13-HPODE-induced cytotoxicity has been linked to oxidative stress, although the mechanisms by which this occurs are unknown. In the present study, we show that 13-HPODE and 9-HPODE (10-30 microM) increased superoxide (O2*-) production and induced cytotoxicity in SMC. The 13-HPODE-induced increase in O2*- was blocked by transfecting the cells with antisense oligonucleotides against p22phox, suggesting that the O2*- was produced by NAD(P)H oxidase. Similar concentrations of the corresponding HPODE reduction products, 13-hydroxyoctadecadienoic acid (13-HODE) and 9-HODE, neither increased O2*- production nor induced cytotoxicity, while 4-hydroxy nonenal (4-HNE), an unsaturated aldehyde lipid peroxidation product, induced cytotoxicity without increasing O2*- production. Treatment with superoxide dismutase or Tiron to scavenge O2*-, or transfection with p22phox antisense oligonucleotides to inhibit O2*- production, attenuated 13-HPODE-induced cytotoxicity, but not that induced by 4-HNE. These findings suggest that activation of NAD(P)H oxidase, and production of O2*-, play an important role in lipid hydroperoxide-induced smooth muscle cytotoxicity.


Subject(s)
Imidazoles , Lipid Peroxides/metabolism , Membrane Transport Proteins , NADPH Oxidases/metabolism , Animals , Arteriosclerosis , Cell Survival , Cells, Cultured , Enzyme Activation , Free Radicals , Linoleic Acids/pharmacology , Lipid Peroxides/pharmacology , Male , Microscopy, Confocal , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , NADPH Dehydrogenase/metabolism , Oligonucleotides, Antisense/pharmacology , Oxidants/metabolism , Oxidative Stress , Oxygen/metabolism , Phosphoproteins/metabolism , Pyrazines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Superoxides/metabolism , Transfection
19.
Am J Physiol Heart Circ Physiol ; 283(6): H2306-14, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12388281

ABSTRACT

Cytochrome P-450 epoxygenase-derived epoxyeicosatrienoic acids (EETs) play an important role in the regulation of vascular reactivity and function. Conversion to the corresponding dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolases is thought to be the major pathway of EET metabolism in mammalian vascular cells. However, when human coronary artery endothelial cells (HCEC) were incubated with (3)H-labeled 14,15-EET, chain-shortened epoxy fatty acids, rather than DHET, were the most abundant metabolites. After 4 h of incubation, 23% of the total radioactivity remaining in the medium was converted to 10,11-epoxy-hexadecadienoic acid (16:2), a product formed from 14,15-EET by two cycles of beta-oxidation, whereas only 15% was present as 14,15-DHET. Although abundantly present in the medium, 10,11-epoxy-16:2 was not detected in the cell lipids. Exogenously applied (3)H-labeled 10,11-epoxy-16:2 was neither metabolized nor retained in the cells, suggesting that 10,11-epoxy-16:2 is a major product of 14,15-EET metabolism in HCEC. 10,11-Epoxy-16:2 produced potent dilation in coronary microvessels. 10,11-Epoxy-16:2 also potently inhibited tumor necrosis factor-alpha-induced production of IL-8, a proinflammatory cytokine, by HCEC. These findings implicate beta-oxidation as a major pathway of 14,15-EET metabolism in HCEC and provide the first evidence that EET-derived chain-shortened epoxy fatty acids are biologically active.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/metabolism , Coronary Vessels/cytology , Endothelium, Vascular/metabolism , Epoxy Compounds/metabolism , Fatty Acids, Unsaturated/biosynthesis , Vasodilator Agents/metabolism , 8,11,14-Eicosatrienoic Acid/chemistry , Cell Line , Chromatography, Liquid , Coronary Vessels/drug effects , Coronary Vessels/physiology , Culture Media, Conditioned/chemistry , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/pharmacology , Humans , Interleukin-8/biosynthesis , Mass Spectrometry , Oxidation-Reduction , Tumor Necrosis Factor-alpha/pharmacology , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL