Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Arterioscler Thromb Vasc Biol ; 35(2): 309-11, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25524774

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the role of cluster of differentiation 43 (CD43), an integral membrane glycoprotein with both proadhesive and antiadhesive activities, in atherosclerosis. APPROACH AND RESULTS: Low-density lipoprotein receptor-deficient mice were lethally irradiated and reconstituted with either bone marrow from CD43(-/-) mice or from wild-type controls. We found that mice lacking the CD43 on their leukocytes had significantly less severe atherosclerosis and that, contrary to our expectation, macrophage infiltration into the vessel wall was not affected by the lack of CD43 in the leukocytes. However, we found that CD43 mediates cholesterol homeostasis in macrophages by facilitating cholesterol efflux. This resulted in a significant reduction in storage of cholesterol in the aorta of mice lacking CD43 in the leukocytes. CONCLUSIONS: CD43 may be an important mediator of macrophage lipid homeostasis, thereby affecting macrophage foam cell formation and ultimately atherosclerotic plaque development.


Subject(s)
Aorta/metabolism , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Leukocytes/metabolism , Leukosialin/metabolism , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Cholesterol/metabolism , Disease Models, Animal , Down-Regulation , Leukocytes/immunology , Leukosialin/genetics , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors
2.
Am J Pathol ; 183(1): 313-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23685110

ABSTRACT

Chitinase 1 (CHIT1) is secreted by activated macrophages. Chitinase activity is raised in atherosclerotic patient sera and is present in atherosclerotic plaque. However, the role of CHIT1 in atherosclerosis is unknown. Preliminary studies of atherosclerosis in cynomolgous monkeys revealed CHIT1 to be closely correlated with areas of macrophage infiltration. Thus, we investigated the effects of a chitinase inhibitor, allosamidin, on macrophage function in vitro and on atherosclerotic development in vivo. In RAW264.7 cells, allosamidin elevated monocyte chemoattractant protein 1 and tumor necrosis factor alpha expression, and increased activator protein 1 and nuclear factor-κB transcriptional activity. Although inducible nitric oxide synthase, IL-6, and IL-1ß expression were increased, Arg1 expression was decreased by chitinase inhibition, suggesting that suppression of CHIT1 activity polarizes macrophages into a M1 phenotype. Allosamidin decreased scavenger receptor AI, CD36, ABCA1, and ABCG1 expression which led to suppression of cholesterol uptake and apolipoprotein AI-mediated cholesterol efflux in macrophages. These effects were confirmed with CHIT1 siRNA transfection and CHIT1 plasmid transfection experiments in primary macrophages. Apolipoprotein E-deficient hyperlipidemic mice treated for 6 weeks with constant administration of allosamidin and fed an atherogenic diet showed aggravated atherosclerotic lesion formation. These data suggest that CHIT1 exerts protective effects against atherosclerosis by suppressing inflammatory responses and polarizing macrophages toward an M2 phenotype, and promoting lipid uptake and cholesterol efflux in macrophages.


Subject(s)
Acetylglucosamine/analogs & derivatives , Atherosclerosis/enzymology , Chitinases/antagonists & inhibitors , Enzyme Inhibitors/adverse effects , Macrophages/enzymology , Trisaccharides/adverse effects , Acetylglucosamine/adverse effects , Animals , Atherosclerosis/chemically induced , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Biomarkers/metabolism , Cell Line , Chitinases/metabolism , Cytokines/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
3.
Clin Sci (Lond) ; 126(9): 671-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24256203

ABSTRACT

In the present study we sought to determine the effect of CoCl2, an inhibitor of PHD (prolyl hydroxylase domain protein), on the development of AAA (abdominal aortic aneurysm). AAA was induced in C57BL/6 mice by periaortic application of CaCl2 (AAA group). NaCl (0.9%)-treated mice were used as a sham control (SHAM group). Mice were treated with 0.05% CoCl2 in the drinking water (AAA/CoCl2 group). At 1 and 6 weeks after the operation, aortic tissue was excised for further examination. After 6 weeks of CaCl2 treatment, aortic diameter and macrophage infiltration into the aortic adventitia were increased in the AAA group compared with the SHAM group. Treatment with CoCl2 reduced the aneurysmal size and macrophage infiltration compared with the AAA group. Aortic expression of inflammatory cytokines and MCP-1 (monocyte chemoattractant protein-1) and the activities of MMP-9 (matrix metalloproteinase-9) and MMP-2 were enhanced in the AAA group and attenuated in the AAA/CoCl2 group. Expression of cytokines and the activities of MMPs were already increased after 1 week of CaCl2 treatment, but were suppressed by CoCl2 treatment in association with reduced NF-κB (nuclear factor κB) phosphorylation. Treatment with CoCl2 in mice prevented the development of CaCl2-induced AAA in association with reduced inflammation and ECM (extracellular matrix) disruption. The results of the present study suggest that PHD plays a critical role in the development of AAA and that there is a therapeutic potential for PHD inhibitors in the prevention of AAA development.


Subject(s)
Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/prevention & control , Aortitis/prevention & control , Cobalt/pharmacology , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Animals , Aorta, Abdominal/enzymology , Aorta, Abdominal/immunology , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/enzymology , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Aortitis/chemically induced , Aortitis/enzymology , Aortitis/immunology , Aortitis/pathology , Calcium Chloride , Catalase/metabolism , Cytokines/metabolism , Disease Models, Animal , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/immunology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Time Factors , Transcription Factor RelA/metabolism
4.
Nat Med ; 13(6): 719-24, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17546038

ABSTRACT

Mast cells contribute importantly to allergic and innate immune responses by releasing various preformed and newly synthesized mediators. Previous studies have shown mast cell accumulation in human atherosclerotic lesions. This report establishes the direct participation of mast cells in atherogenesis in low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice. Atheromata from compound mutant Ldlr(-/-) Kit(W-sh)(/W-sh) mice showed decreased lesion size, lipid deposition, T-cell and macrophage numbers, cell proliferation and apoptosis, but increased collagen content and fibrous cap development. In vivo, adoptive transfer of syngeneic wild-type or tumor necrosis factor (TNF)-alpha-deficient mast cells restored atherogenesis to Ldlr(-/-)Kit(W-sh/W-sh) mice. Notably, neither interleukin (IL)-6- nor interferon (IFN)-gamma-deficient mast cells did so, indicating that the inhibition of atherogenesis in Ldlr(-/-)Kit(W-sh/W-sh) mice resulted from the absence of mast cells and mast cell-derived IL-6 and IFN-gamma. Compared with wild-type or TNF-alpha-deficient mast cells, those lacking IL-6 or IFN-gamma did not induce expression of proatherogenic cysteine proteinase cathepsins from vascular cells in vitro or affect cathepsin and matrix metalloproteinase activities in atherosclerotic lesions, implying that mast cell-derived IL-6 and IFN-gamma promote atherogenesis by augmenting the expression of matrix-degrading proteases. These observations establish direct participation of mast cells and mast cell-derived IL-6 and IFN-gamma in mouse atherogenesis and provide new mechanistic insight into the pathogenesis of this common disease.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/pathology , Cytokines/metabolism , Inflammation Mediators/physiology , Mast Cells/immunology , Mast Cells/metabolism , Animals , Atherosclerosis/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
5.
FASEB J ; 24(8): 2869-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20354139

ABSTRACT

In atherogenesis, macrophage foam cell formation is modulated by pathways involving both the uptake and efflux of cholesterol. We recently showed that interleukin-10 (IL-10) modulates lipid metabolism by enhancing both uptake and efflux of cholesterol in macrophages. However, the mechanistic details of these properties in vivo have been unclear. Thus, the purpose of this study was to determine whether expression of IL-10 in macrophages would alter susceptibility to atherosclerosis and whether IL-10 exerts its antiatherosclerotic properties by modulating lipid metabolism in macrophages. We utilized a macrophage-specific retroviral vector that allows long-term in vivo expression of IL-10 in macrophages through transplantation of retrovirally transduced bone marrow cells (BMCs). IL-10 expressed by macrophages derived from transduced BMCs inhibited atherosclerosis in LDLR(-/-) mice by reducing cholesteryl ester accumulation in atherosclerotic sites. Experiments with primary macrophages indicated that macrophage source of IL-10 stimulated both the uptake (by up-regulating scavenger receptors) and efflux of cholesterol (by activating the PPARgamma-LXR-ABCA1/ABCG1 pathway), thereby reducing inflammation and apoptosis in atherosclerosis. These findings indicate that BMC-transduced macrophage IL-10 production can act as a strong antiatherogenic agent, and they highlight a novel antiatherosclerotic therapy using a simple, yet effective, stem cell transduction system that facilitates long-term expression of IL-10 in macrophages.


Subject(s)
Atherosclerosis/drug therapy , Hyperlipidemias/complications , Interleukin-10/genetics , Macrophages/metabolism , Animals , Apoptosis/drug effects , Atherosclerosis/pathology , Genetic Therapy , Inflammation , Interleukin-10/biosynthesis , Interleukin-10/pharmacology , Lipid Metabolism/drug effects , Mice , Transduction, Genetic
6.
J Biol Chem ; 284(47): 32950-8, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19776020

ABSTRACT

Foam cell formation is a hallmark event during atherosclerosis. The current paradigm is that lipid uptake by scavenger receptor in macrophages initiates the chronic proinflammatory cascade and necrosis core formation that characterize atherosclerosis. We report here that a cytokine considered to be anti-atherogenic, interleukin-10 (IL10), promotes cholesterol uptake from modified lipoproteins in macrophages and its transformation into foam cells by increasing the expression of scavenger receptor CD36 and scavenger receptor A. Although uptake of modified lipoproteins is considered proatherogenic, we found that IL10 also increases cholesterol efflux from macrophages to protect against toxicity of free cholesterol accumulation in the cell. This process was PPARgamma-dependent and was mediated through up-regulation of ABCA1 (ATP-binding cassette transporter A1) protein expression. Importantly, expression of inflammatory molecules, such as tumor necrosis factor-alpha, intercellular adhesion molecule-1, and MMP9 as well as apoptosis were dramatically suppressed in lipid-laden foam cells treated with IL10. The notion that IL10 can mediate both the uptake of cholesterol from modified lipoproteins and the efflux of stored cholesterol suggests that the process of foam cell formation is not necessarily detrimental as long as mechanisms of cholesterol efflux and transfer to an exogenous acceptor are functioning robustly. Our results present a comprehensive antiatherogenic role of IL10 in macrophages, including enhanced disposal of harmful lipoproteins, inhibition of inflammatory molecules, and reduced apoptosis.


Subject(s)
Atherosclerosis/metabolism , Cholesterol/metabolism , Interleukin-10/metabolism , Macrophages/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , CD36 Antigens/metabolism , Caspase 3/metabolism , Cell Line , Cytokines/metabolism , Flow Cytometry , Lipids/chemistry , Lipoproteins/chemistry , Mice , Up-Regulation
7.
Front Physiol ; 11: 714, 2020.
Article in English | MEDLINE | ID: mdl-32655419

ABSTRACT

OBJECTIVE: To determine whether overexpression of the chitin degrading enzyme, chitotriosidase (CHIT1), modulates macrophage function and ameliorates atherosclerosis. APPROACH AND RESULTS: Using a mouse model that conditionally overexpresses CHIT1 in macrophages (CHIT1-Tg) crossbred with the Ldlr -/- mouse provided us with a means to investigate the effects of CHIT1 overexpression in the context of atherosclerosis. In vitro, CHIT1 overexpression by murine macrophages enhanced protein expression of IL-4, IL-8, and G-CSF by BMDM upon stimulation with a combination of lipopolysaccharide (LPS) and interferon-γ (IFN-γ). Phosphorylation of ERK1/2 and Akt was also down regulated when exposed to the same inflammatory stimuli. Hyperlipidemic, Ldlr -/--CHIT1-Tg (CHIT1-OE) mice were fed a high-fat diet for 12 weeks in order to study CHIT1 overexpression in atherosclerosis. Although plaque size and lesion area were not affected by CHIT1 overexpression in vivo, the content of hyaluronic acid (HA) and collagen within atherosclerotic plaques of CHIT1-OE mice was significantly greater. Localization of both ECM components was markedly different between groups. CONCLUSIONS: These data demonstrate that CHIT1 alters cytokine expression and signaling pathways of classically activated macrophages. In vivo, CHIT1 modifies ECM distribution and content in atherosclerotic plaques, both of which are important therapeutic targets.

8.
FASEB J ; 22(10): 3561-70, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18556458

ABSTRACT

Rho kinases (ROCKs) are serine-threonine protein kinases that regulate the actin cytoskeleton. Recent studies suggest that ROCKs also play an important role in cardiovascular disease. However, the isoform- and tissue-specific role of ROCKs in mediating this process is unknown. Using homologous recombination, we generated mutant mice harboring alleles with homozygous deletion of ROCK1 (ROCK1(-/-)). Most ROCK1(-/-) mice die perinatally. However, a few ROCK1(-/-) mice survive to adulthood, are phenotypically normal, and have no apparent compensatory changes in ROCK2. Using these ROCK1(-/-) mice, we show that ROCK1 in bone marrow-derived macrophages is critical to the development of atherosclerosis, in part, by mediating foam cell formation and macrophage chemotaxis. Lipid accumulation and atherosclerotic lesions were reduced in atherosclerosis-prone LDLR(-/-) mice, whose bone marrows have been replaced with bone marrows derived from ROCK1(-/-) mice. Bone marrow-derived ROCK1-deficient macrophages exhibited impaired chemotaxis to monocyte chemotactic protein-1 and showed reduced ability to take up lipids and to develop into foam cells when exposed to modified low-density lipoprotein. These findings indicate that ROCK1 in bone marrow-derived cells is a critical mediator of atherogenesis and suggest that macrophage ROCK1 may be an important therapeutic target for vascular inflammation and atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Chemotaxis , Macrophages/immunology , rho-Associated Kinases/genetics , Animals , Atherosclerosis/enzymology , Lipoproteins, LDL/metabolism , Macrophages/enzymology , Mice , Mice, Mutant Strains , Receptors, LDL/genetics
9.
Circulation ; 115(15): 2065-75, 2007 Apr 17.
Article in English | MEDLINE | ID: mdl-17404153

ABSTRACT

BACKGROUND: Remodeling of the arterial extracellular matrix participates importantly in atherogenesis and plaque complication. Increased expression of the elastinolytic and collagenolytic enzyme cathepsin L (Cat L) in human atherosclerotic lesions suggests its participation in these processes, a hypothesis tested here in mice. METHODS AND RESULTS: We generated Cat L and low-density lipoprotein receptor (LDLr) double-deficient (LDLr-/- Cat L-/-) mice by crossbreeding Cat L-null (Cat L-/-) and LDLr-deficient (LDLr-/-) mice. After 12 and 26 weeks of a Western diet, LDLr-/- Cat L-/- mice had significantly smaller atherosclerotic lesions and lipid cores compared with littermate control LDLr-/- Cat L+/- and LDLr-/- Cat L+/+ mice. In addition, lesions from the compound mutant mice showed significantly reduced levels of collagen, medial elastin degradation, CD4+ T cells, macrophages, and smooth muscle cells. Mechanistic studies showed that Cat L contributes to the degradation of extracellular matrix elastin and collagen by aortic smooth muscle cells. Smooth muscle cells from LDLr-/- Cat L-/- mice or those treated with a Cat L-selective inhibitor demonstrated significantly less degradation of elastin and collagen and delayed transmigration through elastin in vitro. Cat L deficiency also significantly impaired monocyte and T-lymphocyte transmigration through a collagen matrix in vitro, suggesting that blood-borne leukocyte penetration through the arterial basement membrane requires Cat L. Cysteine protease active site labeling demonstrated that Cat L deficiency did not affect the activity of other atherosclerosis-associated cathepsins in aortic smooth muscle cells and monocytes. CONCLUSIONS: Cat L directly participates in atherosclerosis by degrading elastin and collagen and regulates blood-borne leukocyte transmigration and lesion progression.


Subject(s)
Atherosclerosis/enzymology , Cathepsins/deficiency , Cysteine Endopeptidases/deficiency , Diet, Atherogenic , Receptors, LDL/deficiency , Animals , Aorta, Thoracic/pathology , Atherosclerosis/chemically induced , Atherosclerosis/genetics , Cathepsin L , Cathepsins/genetics , Cells, Cultured , Chemotaxis/genetics , Cysteine Endopeptidases/genetics , Disease Progression , Endothelial Cells/cytology , Lipids/blood , Male , Mice , Mice, Knockout , Monocytes/cytology , Muscle, Smooth, Vascular/cytology , Receptors, LDL/genetics
10.
Circ Res ; 94(9): 1203-10, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15059935

ABSTRACT

Activated monocytes are present in the arterial walls of hypertensive patients and animals. Monocyte chemoattractant protein-1 (MCP-1), which controls monocyte function through its receptor (CCR2), is implicated in hypertensive inflammatory changes in the arterial wall. The role of CCR2 expression on monocytes in hypertension-induced vascular remodeling, however, has not been addressed. We hypothesized that CCR2 on monocytes is critical in hypertension-induced vascular inflammation and remodeling. Hypertension was induced by infusion of angiotensin II (Ang II) into wild-type mice, CCR2-deficient (CCR2-/-) mice, and bone marrow-transferred mice with a leukocyte-selective CCR2 deficiency (BMT-CCR2-/-). In wild-type mice, Ang II increased CCR2 intensity in circulating monocytes, which was prevented by an Ang II type-1 (AT1) receptor blocker or blunted in AT1 receptor-deficient mice. Enhanced CCR2 intensity on monocytes was observed in hypertensive patients and rats, and was reduced by treatment with the Ang II receptor blocker, supporting the clinical relevance of the observation in mice. In CCR2-/- and BMT-CCR2-/- mice, Ang II-induced vascular inflammation and vascular remodeling (aortic wall thickening and fibrosis) were blunted as compared with control mice. In contrast, Ang II-induced left ventricular hypertrophy developed in CCR2-/- and BMT-CCR2-/- mice. The present study suggests that CCR2 expression in monocytes has a critical role in vascular inflammation and remodeling in Ang II-induced hypertension, and possibly in other forms of hypertension.


Subject(s)
Hypertension/metabolism , Monocytes/physiology , Receptors, Chemokine/physiology , Angiotensin II/toxicity , Angiotensin II Type 1 Receptor Blockers , Animals , Aorta/drug effects , Aorta/metabolism , Bone Marrow Transplantation , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemotaxis, Leukocyte/physiology , Enzyme Inhibitors/pharmacology , Humans , Hypertension/chemically induced , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Imidazoles/pharmacology , Inflammation/metabolism , Infusion Pumps, Implantable , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Olmesartan Medoxomil , Pilot Projects , Radiation Chimera , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 1/deficiency , Receptor, Angiotensin, Type 1/genetics , Receptors, CCR2 , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Recombinant Fusion Proteins/physiology , Superoxide Dismutase/genetics , Tetrazoles/pharmacology , Up-Regulation/drug effects
11.
Atherosclerosis ; 244: 113-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26613536

ABSTRACT

OBJECTIVE: Activation of vagal nerve suppresses inflammatory responses through activation of α7 nicotinic acetylcholine receptor (nAchR). We sought to determine whether AR-R17779, a selective agonist of α7nAchR, affects the development of abdominal aortic aneurysm (AAA). METHODS AND RESULTS: AAA was induced by topical application of calcium chloride (CaCl2) to abdominal aorta (AAA group). NaCl (0.9%) was substituted for CaCl2 as a sham operation (SHAM group). AR-R17779 was administered in drinking water (AAA/AR-R group). One and 6 weeks after the operation, aortic tissue was excised for histological and molecular analyses. Aortic diameter and macrophage infiltration into the aortic adventitia were increased in AAA group compared with SHAM group at 6 weeks. Treatment with AR-R17779 reduced the diameter of the aorta and macrophage infiltration compared with AAA group. Wavy morphology of the elastic lamellae was lost in AAA group while it was preserved in AAA/AR-R group. Expression of inflammatory cytokines and matrix metalloproteinase (MMP) activities were enhanced in AAA group, which was suppressed in AAA/AR-R group. AR-R17779 treatment suppressed CaCl2-induced expression of cytokines, activities of MMPs and NF-κB activation at 1 week when aortic dilatation had not developed. CONCLUSION: Treatment with AR-R17779 prevented the enlargement of abdominal aorta induced by CaCl2 in association with reduced inflammation and extracellular matrix disruption. These findings suggest therapeutic potential of α7nAchR activation for prevention of AAA development.


Subject(s)
Aortic Aneurysm, Abdominal/prevention & control , Bridged-Ring Compounds/pharmacology , Spiro Compounds/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Blotting, Western , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Disease Progression , Gene Expression Regulation/drug effects , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred C57BL , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Circulation ; 105(9): 1110-5, 2002 Mar 05.
Article in English | MEDLINE | ID: mdl-11877364

ABSTRACT

BACKGROUND: It remains unclear whether vascular endothelial growth factor (VEGF) is a proarteriosclerotic or an antiarteriosclerotic factor. We recently reported that long-term inhibition of nitric oxide by administering Nomega-nitro-L-arginine methyl ester (L-NAME) induces coronary vascular inflammation and arteriosclerosis. METHODS AND RESULTS: We used this animal model to investigate the role of VEGF in arteriosclerosis. We blocked VEGF activity in vivo by transfecting with plasmid DNA encoding the murine soluble FLT-1 (sFLT-1) gene into thigh muscle. Soluble FLT-1 can suppress VEGF activity both by sequestering VEGF and by functioning as a dominant-negative inhibitor of VEGF receptors. We observed vascular inflammation associated with increased VEGF expression within 3 days of L-NAME administration, which was prevented by pretreatment with ACE inhibitor, angiotensin II receptor antagonist, or neutralizing monocyte chemoattractant protein-1 antibody. The sFLT-1 gene transfer attenuated the early vascular inflammation and prevented late arteriosclerosis. The sFLT-1 gene transfer also inhibited increased expression of monocyte chemoattractant protein-1 and transforming growth factor-beta, indicating creation of a positive feedback loop to cause arteriosclerosis. CONCLUSIONS: VEGF is necessary in the development of arteriosclerosis by mediating monocyte recruitment and activation in this model.


Subject(s)
Arteriosclerosis/metabolism , Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Monocytes/metabolism , Nitric Oxide/antagonists & inhibitors , Proto-Oncogene Proteins/administration & dosage , Receptor Protein-Tyrosine Kinases/administration & dosage , Animals , Arteriosclerosis/etiology , Arteriosclerosis/immunology , Arteriosclerosis/pathology , Biological Assay , Blood Pressure/drug effects , Cell Division/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemotaxis, Leukocyte/drug effects , Coronary Vessels/drug effects , Coronary Vessels/immunology , Coronary Vessels/pathology , Disease Models, Animal , Endothelial Growth Factors/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Gene Expression/drug effects , Gene Transfer, Horizontal , Genetic Vectors/administration & dosage , Genetic Vectors/biosynthesis , Genetic Vectors/genetics , Inflammation/immunology , Inflammation/pathology , Injections, Intramuscular , Lymphokines/antagonists & inhibitors , Male , Mice , Monocytes/immunology , Monocytes/pathology , NG-Nitroarginine Methyl Ester/administration & dosage , Neovascularization, Physiologic/drug effects , Nitric Oxide/blood , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Inbred WKY , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Time , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
13.
Circulation ; 109(20): 2454-61, 2004 May 25.
Article in English | MEDLINE | ID: mdl-15148275

ABSTRACT

BACKGROUND: Stromal cell-derived factor-1alpha (SDF-1alpha) is implicated as a chemokine for endothelial progenitor cells (EPCs). We therefore hypothesized that SDF-1alpha gene transfer would induce therapeutic neovascularization in vivo by functioning as a chemokine of EPC. METHODS AND RESULTS: To examine SDF-1alpha-induced mobilization of EPC, we used bone marrow-transplanted mice whose blood cells ubiquitously express beta-galactosidase (LacZ). We produced unilateral hindlimb ischemia in the mice and transfected them with plasmid DNA encoding SDF-1alpha or empty plasmids into the ischemic muscles. SDF-1alpha gene transfer mobilized EPCs into the peripheral blood, augmented recovery of blood perfusion to the ischemic limb, and increased capillary density associated with partial incorporation of LacZ-positive cells into the capillaries of the ischemic limb, suggesting that SDF-1alpha induced vasculogenesis and angiogenesis. SDF-1alpha gene transfer did not affect ischemia-induced expression of vascular endothelial growth factor (VEGF) but did enhance Akt and endothelial nitric oxide synthase (eNOS) activity. Blockade of VEGF or NOS prevented all such SDF-1alpha-induced effects. CONCLUSIONS: SDF-1alpha gene transfer enhanced ischemia-induced vasculogenesis and angiogenesis in vivo through a VEGF/eNOS-related pathway. This strategy might become a novel chemokine therapy for next generation therapeutic neovascularization.


Subject(s)
Chemokines, CXC/genetics , Genetic Therapy , Ischemia/therapy , Neovascularization, Physiologic , Nitric Oxide Synthase/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Capillaries/growth & development , Chemokine CXCL12 , Chemotaxis , Endothelium, Vascular/cytology , Hindlimb/blood supply , Ischemia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction , Stem Cells/physiology , Transfection
14.
Circulation ; 110(16): 2444-52, 2004 Oct 19.
Article in English | MEDLINE | ID: mdl-15477409

ABSTRACT

BACKGROUND: Therapeutic angiogenesis by delivery of vascular endothelial growth factor (VEGF) has attracted attention. However, the role and function of VEGF in experimental restenosis (neointimal formation) after vascular intraluminal injury have not been addressed. METHODS AND RESULTS: We report herein that blockade of VEGF by soluble VEGF receptor 1 (sFlt-1) gene transfer attenuated neointimal formation after intraluminal injury in rabbits, rats, and mice. sFlt-1 gene transfer markedly attenuated the early vascular inflammation and proliferation and later neointimal formation. sFlt-1 gene transfer also inhibited increased expression of inflammatory factors such as monocyte chemoattractant protein-1 and VEGF. Intravascular VEGF gene transfer enhanced angiogenesis in the adventitia but did not reduce neointimal formation. CONCLUSIONS: Increased expression and activity of VEGF are essential in the development of experimental restenosis after intraluminal injury by recruiting monocyte-lineage cells.


Subject(s)
Carotid Artery Injuries/therapy , Femoral Artery/injuries , Genetic Therapy , Monocytes/pathology , Proteins/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Wound Healing/physiology , Adenoviridae/genetics , Animals , Bone Marrow Transplantation , Carotid Artery Injuries/pathology , Catheterization/adverse effects , Cell Division , Cell Lineage , Constriction, Pathologic , Endothelium, Vascular/physiology , Extracellular Matrix Proteins , Femoral Artery/pathology , Gene Expression Regulation/drug effects , Genetic Vectors/pharmacology , Genetic Vectors/therapeutic use , Hyperplasia , Inflammation/prevention & control , Male , Mice , Mice, Transgenic , Myosin Heavy Chains , Neovascularization, Physiologic , Nonmuscle Myosin Type IIB , Proteins/genetics , Rabbits , Rats , Rats, Inbred WKY , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Receptors, Vascular Endothelial Growth Factor/genetics , Recombinant Fusion Proteins/physiology , Recurrence , Regeneration , Solubility , Transduction, Genetic , Transfection , Tunica Intima/pathology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/physiology
15.
Circulation ; 106(21): 2700-6, 2002 Nov 19.
Article in English | MEDLINE | ID: mdl-12438296

ABSTRACT

BACKGROUND: Monocyte infiltration into the arterial wall and its activation is the central event in atherogenesis. Thus, monocyte chemoattractant protein-1 (MCP-1) might be a novel therapeutic target against atherogenesis. We and others recently reported that blockade or abrogation of the MCP-1 pathway attenuates the initiation of atheroma formation in hypercholesterolemic mice. It remains unclear, however, whether blockade of MCP-1 can limit progression or destabilization of established lesions. METHODS AND RESULTS: We report here that blockade of MCP-1 by transfecting an N-terminal deletion mutant of the MCP-1 gene limited progression of preexisting atherosclerotic lesions in the aortic root in hypercholesterolemic mice. In addition, blockade of MCP-1 changed the lesion composition into a more stable phenotype, ie, containing fewer macrophages and lymphocytes, less lipid, and more smooth muscle cells and collagen. This strategy decreased expression of CD40 and the CD40 ligand in the atherosclerotic plaque and normalized the increased chemokine (RANTES and MCP-1) and cytokine (tumor necrosis factor alpha, interleukin-6, interleukin-1beta, and transforming growth factor beta(1)) gene expression. These data suggest that MCP-1 is a central mediator in the progression and destabilization of established atheroma. CONCLUSIONS: The results of the present study suggest that the inflammatory responses mediated by MCP-1 are important in atherosclerosis and its complications.


Subject(s)
Apolipoproteins E/deficiency , Arteriosclerosis/therapy , Chemokine CCL2/antagonists & inhibitors , Genetic Therapy/methods , Animals , Apolipoproteins E/genetics , Arteriosclerosis/genetics , Arteriosclerosis/pathology , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokines/genetics , Chemokines/metabolism , Collagenases/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Gene Transfer Techniques , Hypercholesterolemia/genetics , Matrix Metalloproteinase 13 , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Receptors, CCR2 , Receptors, Chemokine/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , T-Lymphocytes/pathology , Treatment Outcome , Up-Regulation/drug effects
16.
FASEB J ; 16(13): 1838-40, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12223446

ABSTRACT

Prevention of restenosis after coronary intervention is a major clinical challenge, which highlights the need of new therapeutic options. Vascular injury may involve inflammatory responses that accelerate the recruitment and activation of monocytes through the activation of chemotactic factors, including monocyte chemoattractant protein-1 (MCP-1). However, there is no definitive evidence supporting the role of MCP-1 in restenosis. We recently devised a new strategy for anti-MCP-1 gene therapy by transfecting an N-terminal deletion mutant of the MCP-1 gene into skeletal muscles. We demonstrate here that this strategy suppressed monocyte infiltration/activation in the injured site and markedly inhibited restenotic changes (neointimal hyperplasia) after balloon injury of the carotid artery in rats and monkeys. This strategy also suppressed the local production of MCP-1 and inflammatory cytokines. Therefore, monocyte infiltration and activation mediated by MCP-1 are essential in the development of restenotic changes after balloon injury. This strategy may be a useful form of gene therapy against human restenosis.


Subject(s)
Carotid Artery Injuries/prevention & control , Chemokine CCL2/genetics , Tunica Intima/pathology , Actins/analysis , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/etiology , Catheterization/adverse effects , Chemokine CCL2/blood , Chemokine CCL2/metabolism , Chemokines/metabolism , Cytokines/metabolism , Hyperplasia , Immunohistochemistry , Macaca fascicularis , Male , Muscle, Smooth/chemistry , Mutation , Plasmids/genetics , Proliferating Cell Nuclear Antigen/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred WKY , Receptors, CCR2 , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Time Factors , Transfection , Tunica Intima/metabolism , von Willebrand Factor/analysis
17.
Arterioscler Thromb Vasc Biol ; 24(10): 1886-90, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15284091

ABSTRACT

OBJECTIVE: Accelerated coronary arteriosclerosis remains a major problem in the long-term survival of cardiac transplant recipients. However, the pathogenesis of graft vasculopathy is poorly understood, and there is no effective therapy. Transplant arteriosclerosis is characterized by early mononuclear cell attachment on the transplanted vessel followed by development of concentric neointimal hyperplasia. Early and persistent expression of monocyte chemoattractant protein-1 (MCP-1) in cardiac allografts has been implicated for the pathogenesis of transplant arteriosclerosis. METHODS AND RESULTS: We investigated whether anti-MCP-1 gene therapy can inhibit the development of intima hyperplasia in a mouse model of cardiac transplantation. Either the dominant-negative form of MCP-1 (7ND) or control vector was transfected into the skeletal muscles of B10.D2 mice. Cardiac allografts from DBA/2 mice were transplanted heterotopically into B10.D2 mice. 7ND gene transfer was associated with a significant reduction of the number of mononuclear cells accumulating in the lumen of the graft coronary arteries at 1 week and an attenuation of the development of the lesion at 8 weeks (intima/media ratio 0.79+/-0.05 versus 0.48+/-0.04). CONCLUSIONS: The MCP-1/chemokine receptor 2 (CCR2) signaling pathway plays a critical role in the pathogenesis of graft vasculopathy. This new anti-MCP-1 gene therapy might be useful to treat graft vascular disease.


Subject(s)
Arteriosclerosis/prevention & control , Chemokine CCL2/genetics , Genetic Therapy/methods , Animals , Chemokine CCL2/biosynthesis , Coronary Vessels/physiology , Disease Models, Animal , Gene Transfer Techniques , Genes, Dominant/physiology , Heart Transplantation/methods , Hyperplasia/prevention & control , Inflammation/genetics , Inflammation/pathology , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred DBA , Mice, Inbred Strains , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Receptors, CCR2 , Receptors, Chemokine/metabolism , Signal Transduction/genetics , Transplantation, Heterotopic , Transplantation, Homologous , Tunica Intima/pathology
18.
Arterioscler Thromb Vasc Biol ; 24(8): 1522-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15178560

ABSTRACT

OBJECTIVE: Anti-atherosclerotic effects of statins might be mediated partly by pleiotropic cholesterol-lowering independent mechanisms. We used nonhuman primates and examined whether treatment with pravastatin or antimonocyte chemoattractant protein-1 (MCP-1) therapy can induce regression and stabilization of established atherosclerotic lesions through cholesterol-lowering independent mechanisms. METHODS AND RESULTS: Advanced atherosclerosis was induced in the abdominal aorta and the common iliac artery of cynomolgus monkeys by undergoing balloon injury and giving atherogenic diet for 6 months. At 6 months, the diet was changed to normal chow, and the animals were allocated to 4 treatment groups: control vehicle group and other groups treated with pravastatin (1 or 10 mg/kg) or with mutant MCP-1 gene transfection for additional 6 months. Each compound was treated instead of the atherogenic diet, and cholesterol contents in pravastatin-treated groups were adjusted to equalize plasma cholesterol level among groups. Pravastatin reduced neointimal formation in the aorta, but not in the common iliac artery. Pravastatin reduced intimal macrophage area and other markers of plaque destabilization in the common iliac artery. Equivalent inhibitory effects were observed in animals that received mutant MCP-1 gene transfection. No serious side effects were noted by 2 therapeutic modalities. CONCLUSIONS: This study demonstrated cholesterol-lowering independent regression and stabilization of established atherosclerotic lesions by pravastatin and by anti-MCP-1 therapy in nonhuman primates. An anti-inflammatory mechanism may be involved in the beneficial effects of pravastatin.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aortic Diseases/drug therapy , Arteriosclerosis/drug therapy , Chemokine CCL2/genetics , Genetic Therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pravastatin/pharmacology , Angiotensin II/blood , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Aorta, Abdominal/injuries , Aorta, Abdominal/pathology , Aortic Diseases/blood , Arteriosclerosis/blood , Arteriosclerosis/etiology , Catheterization , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/chemistry , Chemokine CCL2/immunology , Chemotaxis , Cholesterol/blood , Cytokines/blood , DNA, Complementary/genetics , Diet, Atherogenic , Drug Evaluation, Preclinical , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/blood , Hypercholesterolemia/complications , Hypercholesterolemia/drug therapy , Iliac Artery/injuries , Iliac Artery/pathology , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Immunoglobulin M/biosynthesis , Immunoglobulin M/immunology , Inflammation , Macaca fascicularis , Male , Peptidyl-Dipeptidase A/blood , Phenotype , Pravastatin/therapeutic use , Random Allocation , Renin/blood , Transfection
19.
Arterioscler Thromb Vasc Biol ; 24(3): 534-9, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14739122

ABSTRACT

OBJECTIVE: Chronic inflammatory processes might be involved in the progression and destabilization of atherosclerotic plaques. Therefore, identification of the mechanism underlying arterial inflammatory function might lead to the development of novel therapeutic strategies. Angiotensin II (AngII) is implicated in atherogenesis by activating the vascular inflammation system, mainly through monocyte chemotaxis. Therefore, we hypothesized that AngII increases plaque size and promotes destabilization of established atheromas by activating the monocyte chemoattractant protein-1 (MCP-1) pathway. METHODS AND RESULTS: We report here that 4-week infusion of AngII not only increased plaque size but also induced a destabilization phenotype (ie, increased macrophages and lipids and decreased collagen and smooth muscle cells) of pre-existing atherosclerotic lesions of hypercholesterolemic mice. AngII also enhanced the gene expression of inflammatory cytokines (TNFalpha, IL-6, etc.) and chemokines (MCP-1, CCR2, etc). Blockade of MCP-1, by transfecting the deletion mutant of the human MCP-1 gene into the skeletal muscles, limited AngII-induced progression and destabilization of established atherosclerotic lesions and suppressed the induction of proinflammatory genes. CONCLUSIONS: These data suggest that MCP-1 functions as a central inflammatory mediator in the AngII-induced progression and changes in plaque composition of established atheroma.


Subject(s)
Angiotensin II/toxicity , Arteriosclerosis/metabolism , Chemokine CCL2/physiology , Hyperlipoproteinemia Type II/complications , Inflammation Mediators/physiology , Angiotensin II/administration & dosage , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aortic Diseases/etiology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/etiology , Arteriosclerosis/pathology , Chemokines/biosynthesis , Chemokines/genetics , Cytokines/biosynthesis , Cytokines/genetics , Disease Progression , Gene Expression Regulation/drug effects , Gene Targeting , Humans , Imidazoles/pharmacology , Inflammation , Injections, Intramuscular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Olmesartan Medoxomil , Random Allocation , Recombinant Fusion Proteins/physiology , Sequence Deletion , Single-Blind Method , Tetrazoles/pharmacology
20.
PLoS One ; 10(6): e0128546, 2015.
Article in English | MEDLINE | ID: mdl-26061913

ABSTRACT

BACKGROUND: Clinical trials have shown that treatment of patients with type 2 diabetes with pioglitazone, a peroxisome proliferator-activated receptor (PPAR)γ agonist, reduces cardiovascular events. However, the effect of PPARγ agonists on endoplasmic reticulum (ER) stress that plays an important role in the progression of atherosclerosis has not been determined. We sought to determine the effect of PPARγ agonists on ER stress induced by palmitate, the most abundant saturated fatty acid in the serum. METHODS AND RESULTS: Protein expression of ER stress marker was evaluated by Western blot analysis and stearoyl-CoA desaturase1 (SCD-1) mRNA expression was evaluated by qRT-PCR. Macrophage apoptosis was detected by flowcytometry. Pioglitazone and rosiglitazone reduced palmitate-induced phosphorylation of PERK, a marker of ER stress, in RAW264.7, a murine macrophage cell line. Pioglitazone also suppressed palmitate-induced apoptosis in association with inhibition of CHOP expression, JNK phosphorylation and cleavage of caspase-3. These effects of pioglitazone were reversed by GW9662, a PPARγ antagonist, indicating that PPARγ is involved in this process. PPARγ agonists increased expression of SCD-1 that introduces a double bond on the acyl chain of long-chain fatty acid. 4-(2-Chlorophenoxy)-N-(3-(3-methylcarbamoyl)phenyl)piperidine-1-carboxamide, an inhibitor of SCD-1, abolished the anti-ER stress and anti-apoptotic effects of pioglitazone. These results suggest that PPARγ agonists attenuate palmitate-induced ER stress and apoptosis through SCD-1 induction. Up-regulation of SCD-1 may contribute to the reduction of cardiovascular events by treatment with PPARγ agonists.


Subject(s)
Macrophages/drug effects , PPAR gamma/agonists , Palmitates/toxicity , Stearoyl-CoA Desaturase/genetics , Thiazolidinediones/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Macrophages/metabolism , Mice , Pioglitazone , Rosiglitazone , Stearoyl-CoA Desaturase/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL