Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Arterioscler Thromb Vasc Biol ; 38(9): 2217-2224, 2018 09.
Article in English | MEDLINE | ID: mdl-30026269

ABSTRACT

Objective- Drug-eluting stents eluting canonical mTOR (mammalian target of rapamycin) inhibitors are widely used to treat coronary artery disease but accelerate the development of atherosclerosis within the stent (neoatherosclerosis)-a leading cause of late stent failure. We recently showed that canonical mTOR inhibitors bind FKBP12.6 (12.6-kDa FK506-binding protein 12), displace it from calcium release channels, resulting in activation of PKCα (protein kinase Cα) and dissociation of p-120-catenin (p120) from VE-CAD (vascular endothelial cadherin; promoting endothelial barrier dysfunction [EBD]). However, the relevance of these findings to drug-eluting stents remains unknown. Newer generation direct mTOR kinase inhibitors do not bind FKBP12.6 and offer the potential of improving endothelial barrier function while maintaining antirestenotic efficacy, but their actual effects are unknown. We examined the effects of 2 different pharmacological targeting strategies-canonical mTOR inhibitor everolimus and mTOR kinase inhibitors Torin-2-on EBD after stenting. Approach and Results- Using the rabbit model of stenting and a combination of Evans blue dye, confocal and scanning electron microscopy studies, everolimus-eluting stents resulted in long-term EBD compared with bare metal stents. EBD was mitigated by using stents that eluted mTOR kinase inhibitors (Torin-2-eluting stent). At 60 days after stent placement, everolimus-eluting stents demonstrated large areas of Evans blue dye staining and evidence of p120 VE-CAD dissociation consistent with EBD. These findings were absent in bare metal stents and significantly attenuated in Torin-2-eluting stent. As proof of concept of the role of EBD in neoatherosclerosis, 100 days after stenting, animals were fed an enriched cholesterol diet for an additional 30 days. Everolimus-eluting stents demonstrated significantly more macrophage infiltration (consistent with neoatherosclerosis) compared with both bare metal stents and Torin-2-eluting stent. Conclusions- Our results pinpoint interactions between FKBP12.6 and canonical mTOR inhibitors as a major cause of vascular permeability and neoatherosclerosis, which can be overcome by using mTOR kinase inhibitors. Our study suggests further refinement of molecular targeting of the mTOR complex may be a promising strategy (Graphic Abstract).


Subject(s)
Capillary Permeability/drug effects , Drug-Eluting Stents , Endothelium, Vascular/metabolism , Everolimus/pharmacology , Naphthyridines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Catenins/metabolism , Enzyme Activation , Everolimus/metabolism , Male , Models, Animal , Naphthyridines/metabolism , Proof of Concept Study , Protein Kinase C-alpha/metabolism , Rabbits , TOR Serine-Threonine Kinases/metabolism , Tacrolimus Binding Proteins/metabolism , Delta Catenin
3.
Interv Cardiol Clin ; 5(3): 321-329, 2016 07.
Article in English | MEDLINE | ID: mdl-28582030

ABSTRACT

Cardiovascular disease is a leading cause of death and disability worldwide. Current treatment strategies aimed at treating the symptoms and consequences of obstructive vascular disease have embraced both optimal medical therapy and catheter-based percutaneous coronary intervention with drug-eluting stents. Drug-eluting stents elute antiproliferative drugs inhibiting vascular smooth muscle cell proliferation, which occurs in response to injury and thus prevents restenosis. However, all drugs currently approved for use in drug-eluting stents do not discriminate between proliferating vascular smooth muscle cells and endothelial cells, thus delaying re-endothelialization and subsequent vascular healing.


Subject(s)
Coronary Restenosis/prevention & control , Drug-Eluting Stents , Endothelial Cells/drug effects , Muscle, Smooth, Vascular/drug effects , Endothelial Cells/physiology , Humans , Myocytes, Smooth Muscle , Paclitaxel , Stents
4.
Nat Commun ; 6: 7792, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26242746

ABSTRACT

Macrophages are an essential component of the immune response to ischaemic injury and play an important role in promoting inflammation and its resolution, which is necessary for tissue repair. The type I transmembrane glycoprotein CD163 is exclusively expressed on macrophages, where it acts as a receptor for haemoglobin:haptoglobin complexes. An extracellular portion of CD163 circulates in the blood as a soluble protein, for which no physiological function has so far been described. Here we show that during ischaemia, soluble CD163 functions as a decoy receptor for TWEAK, a secreted pro-inflammatory cytokine of the tumour necrosis factor family, to regulate TWEAK-induced activation of canonical nuclear factor-κB (NF-κB) and Notch signalling necessary for myogenic progenitor cell proliferation. Mice with deletion of CD163 have transiently elevated levels of TWEAK, which stimulate muscle satellite cell proliferation and tissue regeneration in their ischaemic and non-ischaemic limbs. These results reveal a role for soluble CD163 in regulating muscle regeneration after ischaemic injury.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Macrophages/physiology , Muscle, Skeletal/physiology , Receptors, Cell Surface/metabolism , Regeneration , Tumor Necrosis Factors/metabolism , Animals , Cytokine TWEAK , Male , Mice, Knockout , NF-kappa B/metabolism , Random Allocation , Receptors, Notch/metabolism , Reperfusion Injury
5.
Atherosclerosis ; 241(2): 692-700, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26125411

ABSTRACT

OBJECTIVES: Toll-like Receptor 4 (TLR4) is implicated in modulating inflammatory cytokines though its role in atherosclerosis remains uncertain. We have recently described a non-foam cell macrophage phenotype driven by ingestion of hemoglobin:haptoglobin complexes (HH), via the scavenger receptor CD163, characterized by reduced inflammatory cytokine production. In this study, we examined the role of iron metabolism in modulating TLR4 signaling in these cells. METHODS AND RESULTS: Areas in human atherosclerotic plaque with non-foam cell, CD163 positive macrophages demonstrated reduced expression of tumor necrosis factor alpha (TNF-α) and interferon-beta (INF-ß) compared to foam cells. Human macrophages differentiated in hemoglobin:haptoglobin (HH) complexes expressed the CD163 positive non-foam cell phenotype and demonstrated significantly less TNF-α and INF-ß compared to control macrophages when exposed to oxidized LDL (oxLDL) or lipopolysaccharide (LPS). LPS stimulated expression of TNF-α and INF-ß could be restored in HH macrophages by pretreatment with hepcidin, an endogenous suppressor of ferroportin1 (FPN), or by genetic suppression of FPN in macrophages derived from myeloid specific FPN knockout mice. LPS stimulated control macrophages demonstrated increase in TLR4 trafficking to lipid rafts; this response was suppressed in HH macrophages but was restored upon pretreatment with hepcidin. Using a pharmacologic hepcidin suppressor, we observed a decrease in cytokine expression and TLR4-lipid raft trafficking in LPS-stimulated in a murine macrophage model. CONCLUSION: TLR4 dependent macrophage signaling is controlled via hepcidin-ferroportin1 axis by influencing TLR4-lipid raft interactions. Pharmacologic manipulation of iron metabolism may represent a promising approach to limiting TLR4-mediated inflammatory responses.


Subject(s)
Cation Transport Proteins/metabolism , Hepcidins/chemistry , Macrophages/cytology , Plaque, Atherosclerotic/metabolism , Toll-Like Receptor 4/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Foam Cells/cytology , Haptoglobins/chemistry , Hemoglobins/chemistry , Humans , Inflammation , Iron/chemistry , Lipopolysaccharides/chemistry , Lipoproteins, LDL/chemistry , Macrophages/metabolism , Macrophages, Peritoneal/metabolism , Male , Membrane Microdomains/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Pharmacol Res ; 93: 22-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25533811

ABSTRACT

Coronary artery disease is a leading cause of death and disability worldwide with contemporary treatment strategies employing both optimal medical therapy and catheter based percutaneous coronary intervention (PCI) with drug eluting stents (DES). While DES have dramatically reduced restenosis rates, their use has been associated with an increased risk of late stent thrombosis and accelerated neointimal atherosclerosis (i.e. "neoatherosclerosis") both major contributors to late stent failure. The underlying substrate of late DES failure is likely related to vascular endothelial dysfunction such as poor endothelial regrowth and barrier function (i.e. "endothelial healing"). Initial concerns with 1st generation DES have lead to improvements in mechanical and biologic properties of current 2nd generation DES, which inhibit endothelial regrowth to a lesser extent, lessening late stent failure and resulting in an overall improved safety profile. Current guidelines recommend duration of at least one year of dual anti-platelet therapy with aspirin and a thienopyridine agent such as clopidogrel or prasugrel as sufficient to prevent late thrombotic complications. Recent studies, however, suggest a shorter duration of dual anti-platelet therapy may be equally as safe and efficacious in preventing stent thrombosis with newer generation DES. However, higher risk populations such as patients receiving 1st generation DES or those with increased risk for future ischemic events may benefit from a longer duration (i.e. 30 months) of DAPT to prevent major cardiovascular events with the caveat that such an approach may be associated with an increased risk for bleeding. This review examines the vascular responses to 1st and second generation DES and recent clinical trials examining DAPT duration.


Subject(s)
Drug-Eluting Stents , Platelet Aggregation Inhibitors/therapeutic use , Thrombosis/prevention & control , Animals , Atherosclerosis/etiology , Drug-Eluting Stents/adverse effects , Humans , TOR Serine-Threonine Kinases/antagonists & inhibitors
7.
Front Pharmacol ; 5: 195, 2014.
Article in English | MEDLINE | ID: mdl-25221512

ABSTRACT

Iron is an essential mineral needed for normal physiologic processes. While its function in oxygen transport and other important physiologic processes is well known, less is understood about its role in inflammatory diseases such as atherosclerosis. Existing paradigms suggest iron as a driver of atherosclerosis through its actions as a pro-oxidant capable of causing lipid oxidation and tissue damage. Recently we and others have identified hemoglobin (Hb) derived iron as an important factor in determining macrophage differentiation and function in areas of intraplaque hemorrhage within human atherosclerosis. Hb associated macrophages, M(Hb), are distinct from traditional macrophage foam cells because they do not contain large amounts of lipid or inflammatory cytokines, are characterized by high levels of expression of mannose receptor (CD206) and CD163 in addition to producing anti-inflammatory cytokines such as IL-10. Despite the well-known role of iron as an catalyst capable of producing lipid peroxidation through generation of reactive oxygen species (ROS) such as hydroxyl radical, we and others have shown that macrophages in areas of intraplaque hemorrhage demonstrate reduced intracellular iron and ROS which triggers production of anti-inflammatory cytokines as well as genes involved in cholesterol efflux. These data suggest that manipulation of macrophage iron itself may be a promising pharmacologic target for atherosclerosis prevention through its effects on macrophage inflammation and lipid metabolism. In this review we will summarize the current understanding of iron as it relates to plaque inflammation and discuss how further exploration of this subject may lead to new therapies for atherosclerosis.

8.
Circ Cardiovasc Interv ; 7(4): 526-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24915972

ABSTRACT

BACKGROUND: Preclinical evaluation of the vascular response of drug-eluting stents is limited especially in the setting of diabetes mellitus preventing the evaluation of changes in drug-eluting stent design and eluted drugs after clinical use. METHODS AND RESULTS: Cultured human aortic endothelial cells were used to assess the differences between sirolimus and its analog, everolimus, in the setting of hyperglycemia on various cellular functions necessary for endothelial recovery. A diabetic rabbit model of iliac artery stenting was used to compare histological and morphometric characteristics of the vascular response to everolimus-eluting, sirolimus-eluting, and bare metal stent placement. Under hyperglycemic conditions, sirolimus impaired human aortic endothelial cell barrier function, migration, and proliferation to a greater degree compared with everolimus. In our in vivo model of diabetes mellitus, endothelialization at 28 days was significantly lower and endothelial integrity was impaired in sirolimus-eluting stent compared with both everolimus-eluting and bare metal stents. Neointimal area, uncovered struts, and fibrin deposition were significantly higher in sirolimus-eluting compared with everolimus-eluting and bare metal stents. CONCLUSIONS: Use of everolimus-eluting stent results in improved vascular response in our preclinical models of diabetes mellitus.


Subject(s)
Aorta/pathology , Diabetes Mellitus/therapy , Drug-Eluting Stents/statistics & numerical data , Endothelial Cells/drug effects , Hyperglycemia/therapy , Animals , Cell Movement/drug effects , Cells, Cultured , Disease Models, Animal , Everolimus , Fibrin/metabolism , Humans , Iliac Artery/metabolism , Iliac Artery/pathology , Male , Neointima , Rabbits , Sirolimus/administration & dosage , Sirolimus/adverse effects , Sirolimus/analogs & derivatives
10.
Atherosclerosis ; 229(2): 385-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23880192

ABSTRACT

OBJECTIVES: Metformin impairs endothelialization of drug eluting stents (DES) due to convergent signaling at the mammalian target of rapamycin (mTOR) pathway. We assessed whether metformin will continue to adversely affect stent endothelialization despite design improvements in newer generation DES. METHODS: Rabbit iliac artery stenting with newer generation DES was performed followed by 14 days of either normal chow diet or one with metformin (100 mg/kg/day) added. Scanning electron microscopy was used to assess stent endothelialization after sacrifice. RESULTS: In the metformin-treated group there was significantly less endothelialization compared to the placebo-treated group. Paclitaxel-eluting stents in placebo-treated group had the greatest degree of endothelialization with significantly less in its metformin-treated counterpart and all-limus eluting stent groups. CONCLUSIONS: Metformin inhibited stent endothelialization in newer generation DES despite improvements in stent design. By impairing stent endothelialization, metformin may increase the risk for thrombotic complications after newer generation DES placement.


Subject(s)
Drug-Eluting Stents/adverse effects , Endothelium, Vascular/drug effects , Iliac Artery/drug effects , Iliac Artery/injuries , Metformin/pharmacology , Paclitaxel/pharmacology , Angioplasty, Balloon/adverse effects , Animals , Disease Models, Animal , Endothelium, Vascular/pathology , Everolimus , Hypoglycemic Agents/pharmacology , Iliac Artery/pathology , Immunosuppressive Agents/pharmacology , Male , Rabbits , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Thrombosis/pathology , Tubulin Modulators/pharmacology
11.
Arterioscler Thromb Vasc Biol ; 33(10): 2425-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23887639

ABSTRACT

OBJECTIVE: Sirolimus (SRL) is an immunosuppressant drug used to prevent rejection in organ transplantation and neointimal hyperplasia when delivered from drug-eluting stents. Major side effects of SRL include edema and local collection of intimal lipid deposits at drug-eluting stent sites, suggesting that SRL impairs endothelial barrier function (EBF). The aim of this study was to address the role of SRL on impaired EBF and the potential mechanisms involved. APPROACH AND RESULTS: Cultured human aortic endothelial cells (HAECs) and intact human and mouse endothelium was examined to determine the effect of SRL, which binds FKBP12.6 to inhibit the mammalian target of rapamycin, on EBF. EBF, measured by transendothelial electrical resistance, was impaired in HAECs when treated with SRL or small interfering RNA for FKBP12.6 and reversed when pretreated with ryanodine, a stabilizer of ryanodine receptor 2 intracellular calcium release channels. Intracellular calcium increased in HAECs treated with SRL and normalized with ryanodine pretreatment. SRL-treated HAECs demonstrated increases in protein kinase C-α phosphorylation, a calcium sensitive serine/threonine kinase important in vascular endothelial (VE) cadherin barrier function through its interaction with p120-catenin (p120). Immunostaining of HAECs, human coronary and mouse aortic endothelium treated with SRL showed disruption of p120-VE cadherin interaction treated with SRL. SRL impairment of HAEC EBF was reduced with protein kinase C-α small interfering RNA. Mice treated with SRL demonstrated increased vascular permeability by Evans blue albumin extravasation in the lungs, heart, and aorta. CONCLUSIONS: SRL-FKBP12.6 impairs EBF by activation of protein kinase C-α and downstream disruption of the p120-VE cadherin interaction in vascular endothelium. These data suggest this mechanism may be an important contributor of SRL side effects related to impaired EBF.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability/drug effects , Catenins/metabolism , Endothelial Cells/drug effects , Protein Kinase C-alpha/metabolism , Sirolimus/toxicity , Tacrolimus Binding Proteins/drug effects , Animals , Calcium Signaling/drug effects , Cells, Cultured , Endothelial Cells/enzymology , Enzyme Activation , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Binding , Protein Kinase C-alpha/genetics , RNA Interference , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/drug effects , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Time Factors , Transfection , Delta Catenin
12.
J Am Coll Cardiol ; 61(9): 971-80, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23449430

ABSTRACT

OBJECTIVES: This study sought to examine the effect of oral metformin (Mf) therapy on endothelialization in the setting of drug-eluting stents (DES). BACKGROUND: Mf is a commonly used therapy in diabetic patients receiving DES. Mf and locally eluted mammalian target of rapamycin (mTOR) inhibitors used in DES have convergent molecular signaling; however, the impact of this drug interaction on stent endothelialization is unknown. METHODS: We examined human endothelial aortic cells (HAECs) and a rabbit model of stenting to determine points on molecular convergence between these 2 agents and their impact on stent endothelialization. RESULTS: Western blotting of HAECs treated with Mf and the mTOR inhibitor sirolimus and 14-day rabbit iliacs treated with the combination of zotarolimus-eluting stents (ZES) and oral Mf demonstrated greater inhibition of S6 kinase (S6K), a downstream effector of mTOR complex 1, than either treatment alone. HAEC proliferation was significantly inhibited by Mf or sirolimus treatments alone and further reduced when they were combined. Knockdown of S6K via short interfering RNA in HAECs impaired cell proliferation via a cyclin D1-dependent mechanism, whereas its overexpression rescued the antiproliferative effects of both agents. Last, endothelialization and endothelial cell proliferation at 14 days were assessed in rabbits receiving ZES or bare-metal stents and Mf or placebo by scanning electron microscopy and bromodeoxyuridine/CD31 labeling, respectively. Both endpoints were inhibited by ZES treatment alone and were further reduced by the combination of Mf and ZES. CONCLUSIONS: Significant convergence of signaling occurs between Mf and locally delivered mTOR inhibitors at S6K. This further impairs endothelial recovery/proliferation via an S6K-dependent mechanism. Patients receiving Mf in combination with stents that elute mTOR inhibitors are potentially at increased risk of delayed endothelial healing and stent thrombosis.


Subject(s)
Cell Proliferation/drug effects , Drug-Eluting Stents , Endothelial Cells/drug effects , Metformin/adverse effects , Ribosomal Protein S6 Kinases/physiology , Sirolimus/antagonists & inhibitors , Administration, Oral , Animals , Aorta/cytology , Apoptosis , Cells, Cultured , Drug Interactions , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Humans , Iliac Artery , Metformin/administration & dosage , Microscopy, Electron, Scanning , Rabbits , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Sirolimus/analogs & derivatives , Sirolimus/pharmacology
13.
Am J Cardiol ; 111(3): 346-51, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23178050

ABSTRACT

Antidepressants might increase compliance with cardiovascular disease risk reduction interventions. However, antidepressants have been linked to deleterious metabolic effects. In the present multicenter study, we sought to determine whether patients who take antidepressants derive the expected benefits from cardiac rehabilitation in terms of improvements in multiple atherosclerotic risk factors. A cohort of 26,957 patients who had completed a baseline assessment before participating in an exercise-based cardiac rehabilitation program constituted the study population. The patients were stratified into 3 cohorts (i.e., nondepressed, depressed unmedicated, and depressed medicated) at baseline according to a self-reported history of depression and the current use of antidepressants. Risk factors were assessed at baseline and after ∼12 weeks of program participation. A self-reported history of depression was present at baseline in 5,172 patients (19.2%). Of these patients, 2,147 (41.5%) were taking antidepressants. Patients in the nondepressed cohort (49.4% completion) were more likely (p <0.001) to complete the exit assessment than patients in the depressed unmedicated (44.5% completion) or depressed medicated (43.5% completion) cohorts. Patients in all 3 cohorts who completed the exit assessment showed significant improvement in multiple risk factors. Moreover, the magnitude of improvement in blood pressure, serum lipids and lipoproteins, fasting glucose, weight, and body mass index was similar (p >0.05) in patients taking antidepressants and those who were not. In conclusion, our study is the first to show that antidepressants do not offset the average magnitude of improvement in multiple atherosclerotic risk factors that occurs with completion of a cardiac rehabilitation program.


Subject(s)
Antidepressive Agents/therapeutic use , Atherosclerosis/rehabilitation , Depression/drug therapy , Exercise Therapy/methods , Risk Assessment , Aged , Atherosclerosis/complications , Atherosclerosis/epidemiology , Depression/complications , Female , Humans , Male , Patient Compliance , Prevalence , Retrospective Studies , Risk Factors , Treatment Outcome
14.
J Am Coll Cardiol ; 59(2): 166-77, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22154776

ABSTRACT

OBJECTIVES: The purpose of this study was to examine selective macrophage differentiation occurring in areas of intraplaque hemorrhage in human atherosclerosis. BACKGROUND: Macrophage subsets are recognized in atherosclerosis, but the stimulus for and importance of differentiation programs remain unknown. METHODS: We used freshly isolated human monocytes, a rabbit model, and human atherosclerotic plaques to analyze macrophage differentiation in response to hemorrhage. RESULTS: Macrophages characterized by high expression of both mannose and CD163 receptors preferentially exist in atherosclerotic lesions at sites of intraplaque hemorrhage. These hemoglobin (Hb)-stimulated macrophages, M(Hb), are devoid of neutral lipids typical of foam cells. In vivo modeling of hemorrhage in the rabbit model demonstrated that sponges exposed to red cells showed an increase in mannose receptor-positive macrophages only when these cells contained Hb. Cultured human monocytes exposed to Hb:haptoglobin complexes, but not interleukin-4, expressed the M(Hb) phenotype and were characterized by their resistance to cholesterol loading and up-regulation of ATP-binding cassette (ABC) transporters. M(Hb) demonstrated increased ferroportin expression, reduced intracellular iron, and reactive oxygen species (ROS). Degradation of ferroportin using hepcidin increased ROS and inhibited ABCA1 expression and cholesterol efflux to apolipoprotein A-I, suggesting reduced ROS triggers these effects. Knockdown of liver X receptor alpha (LXRα) inhibited ABC transporter expression in M(Hb) and macrophages differentiated in the antioxidant superoxide dismutase. Last, LXRα luciferase reporter activity was increased in M(Hb) and significantly reduced by overnight treatment with hepcidin. Collectively, these data suggest that reduced ROS triggers LXRα activation and macrophage reverse cholesterol transport. CONCLUSIONS: Hb is a stimulus for macrophage differentiation in human atherosclerotic plaques. A decrease in macrophage intracellular iron plays an important role in this nonfoam cell phenotype by reducing ROS, which drives transcription of ABC transporters through activation of LXRα. Reduction of macrophage intracellular iron may be a promising avenue to increase macrophage reverse cholesterol transport.


Subject(s)
Atherosclerosis/immunology , Cell Differentiation , Hemoglobins/physiology , Hemorrhage/immunology , Macrophages/physiology , Plaque, Atherosclerotic/immunology , ATP-Binding Cassette Transporters/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Atherosclerosis/pathology , Down-Regulation , Humans , Interleukin-10/metabolism , Interleukin-4/metabolism , Iron/metabolism , Lectins, C-Type/metabolism , Liver X Receptors , Macrophages/cytology , Mannose Receptor , Mannose-Binding Lectins/metabolism , Monocytes/cytology , Monocytes/physiology , Neovascularization, Pathologic , Orphan Nuclear Receptors/metabolism , Plaque, Atherosclerotic/pathology , Rabbits , Reactive Oxygen Species/metabolism , Receptors, Cell Surface/metabolism , Receptors, Scavenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...