Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Atherosclerosis ; 319: 79-85, 2021 02.
Article in English | MEDLINE | ID: mdl-33494008

ABSTRACT

BACKGROUND AND AIMS: There is extensive evidence from bone marrow transplantation studies that hematopoietic ATP binding cassette A1 (Abca1) is atheroprotective in low-density lipoprotein receptor (Ldlr) deficient mice. In contrast, studies using lysosyme M promoter-driven deletion of Abca1 in Ldlr deficient mice failed to show similar effects. It was hypothesized that the discrepancy between these studies might be due to the presence of Ldlr in bone marrow-derived cells in the transplantation model. In this study, we aim to determine the contribution of Ldlr to the atheroprotective effect of hematopoietic Abca1 in the murine bone marrow transplantation model. METHODS: Wild-type, Ldlr-/-, Abca1-/-, and Abca1-/-Ldlr-/- bone marrow was transplanted into hypercholesterolemic Ldlr-/- mice. RESULTS: Bone marrow Lldr deficiency did not influence the effects of Abca1 on macrophage cholesterol efflux, foam cell formation, monocytosis or plasma cholesterol. Ldlr deficiency did reduce circulating and peritoneal lymphocyte counts, albeit only in animals lacking Abca1 in bone marrow-derived cells. Importantly, the effects of Abca1 deficiency on atherosclerosis susceptibility were unaltered by the presence or absence of Ldlr. Bone marrow Ldlr deficiency did lead to marginally but consistently decreased atherosclerosis, regardless of Abca1 deficiency. Thus, Ldlr expression on bone marrow-derived cells does, to a minimal extent, influence atherosclerotic lesion development, albeit independent of Abca1. CONCLUSIONS: This study provides novel insight into the relative impact of Ldlr and Abca1 in bone marrow-derived cells on macrophage foam cell formation and atherosclerosis development in vivo. We have shown that Ldlr and Abca1 differentially and independently influence atherosclerosis development in a murine bone marrow transplantation model of atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Atherosclerosis , Bone Marrow , ATP-Binding Cassette Transporters/genetics , Animals , Atherosclerosis/genetics , Bone Marrow/metabolism , Cholesterol , Lipoproteins, LDL/metabolism , Mice , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/metabolism
2.
Biochem Pharmacol ; 86(11): 1594-602, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24095721

ABSTRACT

While numerous studies have aimed to develop strategies to inhibit the development and progression of atherosclerosis, recent attention has focussed on the regression of pre-existing atherosclerotic plaques. As important regulator of total body cholesterol homeostasis, the liver X receptor (LXR) could possibly be an important target to induce regression. Here, we describe the effect of LXR activation by the synthetic agonist T0901317 on lesion regression in different mouse models with early fatty streak lesions or advanced collagen-rich lesions. Although T0901317 caused a dramatic increase in plasma (V)LDL levels in low-density lipoprotein (LDL) receptor knockout mice, no further increase in lesion size was observed, which points to beneficial LXR activity in the vascular wall. In normolipidemic C57BL/6 mice with cholate diet-induced atherosclerotic lesions, T0901317 treatment improved plasma lipoprotein levels and induced lesion regression (-43%, p<0.05). Apolipoprotein E (APOE) reconstitution in APOE knockout mice by means of bone marrow transplantation dramatically improved plasma lipoprotein profiles and resulted in a marked regression of initial (-45%, p<0.001) and advanced lesions (-23%, p<0.01). Atherosclerosis regression was associated with a decrease in the absolute macrophage content (-84%, p<0.001). T0901317 supplementation further decreased the size of early (-71%, p<0.001 vs baseline; -48%, p<0.01 vs chow diet alone) and more advanced atherosclerotic lesions (-36%, p<0.001 and -17%, p=0.06 respectively). In conclusion, our study highlights the potential of LXR agonist T0901317 to stimulate removal of macrophages from atherosclerotic lesions ultimately leading to a highly significant plaque regression of both early and advanced atherosclerotic lesions.


Subject(s)
Hydrocarbons, Fluorinated/therapeutic use , Macrophages/drug effects , Orphan Nuclear Receptors/agonists , Plaque, Atherosclerotic/drug therapy , Sulfonamides/therapeutic use , Animals , Apolipoproteins E/genetics , Bone Marrow Transplantation , Cell Count , Cholesterol, VLDL/blood , Diet , Disease Models, Animal , Female , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/pharmacology , Liver X Receptors , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/pathology , Receptors, LDL/genetics , Severity of Illness Index , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Triglycerides/blood
3.
Atherosclerosis ; 223(2): 332-41, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22748276

ABSTRACT

OBJECTIVE: The ABCA2 transporter shares high structural homology to ABCA1, which is crucial for the removal of excess cholesterol from macrophages and, by extension, in atherosclerosis. It has been suggested that ABCA2 sequesters cholesterol inside the lysosomes, however, little is known of the macrophage-specific role of ABCA2 in regulating lipid homeostasis in vivo and in modulating susceptibility to atherosclerosis. METHODS: Chimeras with dysfunctional macrophage ABCA2 were generated by transplantation of bone marrow from ABCA2 knockout (KO) mice into irradiated LDL receptor (LDLr) KO mice. RESULTS: Interestingly, lack of ABCA2 in macrophages resulted in a diminished lesion size in the aortic root (-24.5%) and descending thoracic aorta (-36.6%) associated with a 3-fold increase in apoptotic cells, as measured by both caspase 3 and TUNEL. Upon oxidized LDL exposure, macrophages from wildtype (WT) transplanted animals developed filipin-positive droplets in lysosomal-like compartments, corresponding to free cholesterol (FC) accumulation. In contrast, ABCA2-deficient macrophages displayed an abnormal diffuse distribution of FC over peripheral regions. The accumulation of neutral sterols in lipid droplets was increased in ABCA2-deficient macrophages, but primarily in cytoplasmic clusters and not in lysosomes. Importantly, apoptosis of oxLDL loaded macrophages lacking ABCA2 was increased 2.7-fold, probably as a consequence of the broad cellular distribution of FC. CONCLUSIONS: Lack of functional ABCA2 generates abnormalities in intracellular lipid distribution/trafficking in macrophages consistent with its lysosomal sequestering role, leading to an increased susceptibility to apoptosis in response to oxidized lipids and reduced atherosclerotic lesion development.


Subject(s)
ATP-Binding Cassette Transporters/deficiency , Aorta/metabolism , Aortic Diseases/prevention & control , Apoptosis , Atherosclerosis/prevention & control , Cholesterol/metabolism , Macrophages/metabolism , Receptors, LDL/deficiency , ATP-Binding Cassette Transporters/genetics , Animals , Aorta/pathology , Aortic Diseases/etiology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow Transplantation , Caspase 3/metabolism , Cholesterol/blood , Disease Models, Animal , Filipin/metabolism , Foam Cells/metabolism , Foam Cells/pathology , Homeostasis , In Situ Nick-End Labeling , Lipoproteins, LDL/metabolism , Lysosomes/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Receptors, LDL/genetics , Time Factors , Transplantation Chimera , Whole-Body Irradiation
4.
PLoS One ; 7(3): e30984, 2012.
Article in English | MEDLINE | ID: mdl-22403608

ABSTRACT

ABCA7, a close relative of ABCA1 which facilitates cholesterol efflux to lipid-poor apoproteins, has been implicated in macrophage lipid efflux and clearance of apoptotic cells in in vitro studies. In the current study, we investigated the in vivo effects of macrophage ABCA7 deficiency on lipid metabolism and atherosclerosis. Chimeras with dysfunctional ABCA7 in macrophages and other blood cells were generated by transplantation of bone marrow from ABCA7 knockout (KO) mice into irradiated low-density lipoprotein receptor (LDLr) KO mice. Unexpectedly, macrophage ABCA7 deficiency did not significantly affect atherosclerosis susceptibility of LDLr KO mice after 10 weeks Western-type diet feeding. However, ABCA7 deficiency was associated with 2-fold (p<0.05) higher macrophage ABCA1 mRNA expression levels. Combined disruption of ABCA1 and ABCA7 in bone-marrow-derived cells increased atherosclerotic lesion development (1.5-fold (p>0.05) as compared to wild type transplanted mice. However, single deletion of ABCA1 had a similar effect (1.8-fold, p<0.05). Macrophage foam cell accumulation in the peritoneal cavity was reduced in ABCA1/ABCA7 dKO transplanted animals as compared to single ABCA1 KO transplanted mice, which was associated with increased ABCG1 expression. Interestingly, spleens of ABCA1/ABCA7 double KO transplanted mice were significantly larger as compared to the other 3 groups and showed massive macrophage lipid accumulation, a reduction in CD3+ T-cells, and increased expression of key regulators of erythropoiesis. In conclusion, deletion of ABCA7 in bone marrow-derived cells does not affect atherogenesis in the arterial wall neither in the absence or presence of ABCA1. Interestingly, combined deletion of bone marrow ABCA1 and ABCA7 causes severe splenomegaly associated with cellular lipid accumulation, a reduction in splenic CD3+ T cells, and induced markers of erythropoeisis. Our data indicate that ABCA7 may play a role in T cell proliferation and erythropoeisis in spleen.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Gene Knockout Techniques , Lipid Metabolism/genetics , Macrophages/metabolism , ATP Binding Cassette Transporter 1 , Animals , Atherosclerosis/blood , Atherosclerosis/surgery , Bone Marrow Transplantation , Foam Cells/metabolism , Foam Cells/pathology , Gene Expression Regulation/genetics , Lipids/blood , Macrophages/pathology , Male , Mice , Receptors, LDL/deficiency , Receptors, LDL/genetics , Up-Regulation/genetics
5.
Atherosclerosis ; 218(2): 314-22, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21840001

ABSTRACT

High-density lipoprotein (HDL) mediated reverse cholesterol transport (RCT) is regarded to be crucial for prevention of foam cell formation and atherosclerosis. ABC-transporter A1 (ABCA1) and scavenger receptor BI (SR-BI) are involved in the biogenesis of HDL and the selective delivery of HDL cholesterol to the liver, respectively. In the present study, we phenotypically characterized mice lacking these two proteins essential for HDL metabolism. ABCA1×SR-BI double knockout (dKO) mice showed severe hypocholesterolemia mainly due to HDL loss, despite a 90% reduction of HDL cholesterol uptake by liver. VLDL production was increased in dKO mice. However, non-HDL cholesterol levels were reduced, probably due to enhanced clearance via LRP1. Hepatobiliary cholesterol transport and fecal sterol excretion were not impaired in dKO mice. In contrast, the macrophage RCT in dKO mice was markedly impaired as compared to WT mice, associated with the accumulation of macrophage foam cells in the lung and Peyer's patches. Strikingly, no atherosclerotic lesion formation was observed in dKO mice. In conclusion, both ABCA1 and SR-BI are essential for maintaining a properly functioning HDL-mediated macrophage RCT, while the potential anti-atherosclerotic functions of ABCA1 and SR-BI are not evident in dKO mice due to the absence of pro-atherogenic lipoproteins.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Atherosclerosis/metabolism , CD36 Antigens/metabolism , Dyslipidemias/metabolism , Foam Cells/metabolism , ATP Binding Cassette Transporter 1 , Animals , Cholesterol/metabolism , Female , Foam Cells/cytology , Lipid Metabolism , Macrophages/cytology , Male , Mice , Mice, Knockout , Time Factors
6.
Am J Pathol ; 179(3): 1522-32, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21741939

ABSTRACT

Reducing the concentration of circulating lipids leads to decreased cardiovascular morbidity and mortality, but the dynamic remodeling that established atherosclerotic lesions undergo upon lipid lowering is poorly understood. Early or advanced lesions in the aortic root were induced by feeding LDL receptor knockout mice a high-fat, high-cholesterol Western-type diet for 5 or 9 weeks, respectively. In the first week after switching to a chow diet, plasma total cholesterol levels dropped 70%, but both early and advanced lesions increased in size. Early lesions grew because of an increase in smooth muscle cells; advanced lesions had an enlargement of absolute macrophage area. From 1 to 3 weeks after the diet switch, plasma total cholesterol levels were completely normalized, but the size of early lesions remained stable; however, advanced lesions became smaller due to a reduction of the absolute macrophage area. From 3 to 6 weeks, both early and advanced lesions progressed further, as a result of expansion of the absolute collagen and necrotic core area. In contrast, early lesions became proinflammatory, as evidenced by the increased infiltration of neutrophils and increased oxidative stress, probably caused by the activation of mast cells in the adventitia. Thus, the severity of atherosclerotic lesions affects their dynamic response to lipid lowering, indicating the importance of establishing stage-specific therapeutic protocols for the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/blood , Cholesterol/metabolism , Receptors, LDL/metabolism , Animals , Aorta, Thoracic/pathology , Aortic Diseases/pathology , Atherosclerosis/pathology , Cholesterol, Dietary/administration & dosage , Connective Tissue/pathology , Coronary Artery Disease/pathology , Diet, Atherogenic , Mice , Mice, Knockout , Monocytes , Neutrophils , Oxidative Stress
7.
Biochim Biophys Acta ; 1811(2): 104-10, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21126601

ABSTRACT

Gemfibrozil and fenofibrate, two of the fibrates most used in clinical practice, raise HDL cholesterol (HDLc) and are thought to reduce the risk of atherosclerotic cardiovascular disease. These drugs act as PPARα agonists and upregulate the expression of genes crucial in reverse cholesterol transport (RCT). In the present study, we determined the effects of these two fibrates on RCT from macrophages to feces in vivo in human apoA-I transgenic (hApoA-ITg) mice. [(3)H]cholesterol-labeled mouse macrophages were injected intraperitoneally into hApoA-ITg mice treated with intragastric doses of fenofibrate, gemfibrozil or a vehicle solution for 17days, and radioactivity was determined in plasma, liver and feces. Fenofibrate, but not gemfibrozil, enhanced [(3)H]cholesterol flux to plasma and feces of female hApoA-ITg mice. Fenofibrate significantly increased plasma HDLc, HDL phospholipids, hApoA-I levels and phospholipid transfer protein activity, whereas these parameters were not altered by gemfibrozil treatment. Unlike gemfibrozil, fenofibrate also induced the generation of larger HDL particles, which were more enriched in cholesteryl esters, together with higher potential to generate preß-HDL formation and caused a significant increase in [(3)H]cholesterol efflux to plasma. Our findings demonstrate that fenofibrate promotes RCT from macrophages to feces in vivo and, thus, highlight a differential action of this fibrate on HDL.


Subject(s)
Cholesterol/metabolism , Feces/chemistry , Fenofibrate/pharmacology , Gemfibrozil/pharmacology , Hypolipidemic Agents/pharmacology , Macrophages/drug effects , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Biological Transport , Cell Line , Female , Humans , Lipid Metabolism , Liver/chemistry , Liver/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Random Allocation
8.
Circ Res ; 107(12): e20-31, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21071707

ABSTRACT

RATIONALE: macrophages cannot limit the uptake of lipids and rely on cholesterol efflux mechanisms for maintaining cellular cholesterol homeostasis. Important mediators of macrophage cholesterol efflux are ATP-binding cassette transporter 1 (ABCA1), which mediates the efflux of cholesterol to lipid-poor apolipoprotein AI, and scavenger receptor class B type I (SR-BI), which promotes efflux to mature high-density lipoprotein. OBJECTIVE: the aim of the present study was to increase the insight into the putative synergistic roles of ABCA1 and SR-BI in foam cell formation and atherosclerosis. METHODS AND RESULTS: low-density lipoprotein receptor knockout (LDLr KO) mice were transplanted with bone marrow from ABCA1/SR-BI double knockout mice, the respective single knockouts, or wild-type littermates. Serum cholesterol levels were lower in ABCA1/SR-BI double knockout transplanted animals, as compared to the single knockout and wild-type transplanted animals on Western-type diet. Despite the lower serum cholesterol levels, massive foam cell formation was found in macrophages from spleen and the peritoneal cavity. Interestingly, ABCA1/SR-BI double knockout transplanted animals also showed a major increase in proinflammatory KC (murine interleukin-8) and interleukin-12p40 levels in the circulation. Furthermore, after 10 weeks of Western-type diet feeding, atherosclerotic lesion development in the aortic root was more extensive in the LDLr KO mice reconstituted with ABCA1/SR-BI double knockout bone marrow. CONCLUSIONS: deletion of ABCA1 and SR-BI in bone marrow-derived cells enhances in vivo macrophage foam cell formation and atherosclerotic lesion development in LDLr KO mice on Western diet, indicating that under high dietary lipid conditions, both macrophage ABCA1 and SR-BI contribute significantly to cholesterol homeostasis in the macrophage in vivo and are essential for reducing the risk for atherosclerosis.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Atherosclerosis/etiology , Atherosclerosis/pathology , Foam Cells/pathology , Gene Deletion , Scavenger Receptors, Class B/genetics , ATP Binding Cassette Transporter 1 , Animals , Atherosclerosis/genetics , Bone Marrow/pathology , Bone Marrow Transplantation , Cholesterol/blood , Homeostasis , Inflammation/etiology , Inflammation/genetics , Lipid Metabolism , Lipoproteins, LDL/genetics , Macrophages/pathology , Mice , Mice, Knockout
9.
Biochem Biophys Res Commun ; 395(3): 387-94, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20382126

ABSTRACT

OBJECTIVES: To determine the role of macrophage ATP-binding cassette transporter A5 (ABCA5) in cellular cholesterol homeostasis and atherosclerotic lesion development. METHODS AND RESULTS: Chimeras with dysfunctional macrophage ABCA5 (ABCA5(-M/-M)) were generated by transplantation of bone marrow from ABCA5 knockout (ABCA5(-/-)) mice into irradiated LDLr(-/-) mice. In vitro, bone marrow-derived macrophages from ABCA5(-M/-M) chimeras exhibited a 29% (P<0.001) decrease in cholesterol efflux to HDL, whereas a 21% (P=0.07) increase in cholesterol efflux to apoA-I was observed. Interestingly, expression of ABCA1, but not ABCG1, was up-regulated in absence of functional ABCA5 in macrophages. To induce atherosclerosis, the transplanted LDLr(-/-) mice were fed a high-cholesterol Western-type diet (WTD) for 6, 10, or 18weeks, allowing analysis of effects on initial as well as advanced lesion development. Atherosclerosis development was not affected in male ABCA5(-M/-M) chimeras after 6, 10, and 18weeks WTD feeding. However, female ABCA5(-M/-M) chimeras did develop significantly (P<0.05) larger aortic root lesions as compared with female controls after 6 and 10weeks WTD feeding. CONCLUSIONS: ABCA5 influences macrophage cholesterol efflux, and selective disruption of ABCA5 in macrophages leads to increased atherosclerotic lesion development in female LDLr(-/-) mice.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Atherosclerosis/metabolism , Cholesterol/metabolism , Macrophages/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Cholesterol/genetics , Female , Lipids/blood , Male , Mice , Mice, Knockout , Receptors, LDL/genetics
10.
Clín. investig. arterioscler. (Ed. impr.) ; 21(5): 232-239, sept.-oct. 2009. ilus, tab
Article in Spanish | IBECS | ID: ibc-84479

ABSTRACT

Introducción. La expresión de PTEC humana enratones transgénicos reduce el colesterol de las lipoproteínas de alta densidad (cHDL) y aumenta la susceptibilidad a la arteriosclerosis. Por otro lado, el gemfibrozilo, uno de los fibratos más utilizados en clínica, y la rosiglitazona aumentan el cHDL y reducen la susceptibilidad a la arteriosclerosis en modelos murinos. El objetivo principal de este estudio es evaluar el efecto de la expresión de la PTEC humana, el gemfibrozilo y la rosiglitazona en el transporte inverso de colesterol(TIC) específico de macrófagos. Materiales y método. Para determinar el TIC específico de macrófagos se aplicó inyección intraperitoneal de macrófagos P388D1 marcados con 3H-colesterol en ratones controles (C57BL/6) y ratones transgénicos de PTEC humana. Este mismo proceso se realizó en ratones transgénicos de apoA-I humana (h) que recibieron una dosis diaria por vía oral de gemfibrozilo (625 mg/kg) orosiglitazona (10 mg/kg) durante 17 días y se los comparó con los que recibieron la solución vehículo. A las 48 h, se sacrificó a los animales y se determinó el 3H-colesterol en plasma, hígado y heces. Resultados. Los ratones transgénicos de PTEC humana presentaron una disminución significativa de cHDL en plasma respecto a los ratones controles. El gemfibrozilo incrementó el colesterol total y el cHDL en los ratones transgénicos deapoA-Ih. Sin embargo, ni la expresión de PTEC humana ni los tratamientos farmacológicos alteraron la excreción fecal de colesterol y ácidos biliares. No se encontraron diferencias significativas en el TIC de 3H-colesterol desde los macrófagos P388D1 a heces en los diferentes grupos experimentales. Conclusiones. Ni la expresión de PTEC humana en ratones transgénicos ni el tratamiento congemfibrozilo o rosiglitazona en ratones transgénicos de apoA-Ih modifican el transporte inverso de colesterol especifico de macrófagos in vivo (AU)


Background. Human CETP expression in micereduces cholesterol from high density lipoprotein HDLc) and increases atherosclerosis susceptibility. On the other hand, gemfibrozil, one of the most used fibrates in clinical practice, and rosiglitazone, increase HDLc and reduce the atherosclerotic susceptibility in mouse models of atherosclerosis. The main aim of this study was to evaluate the effect of human CETP expression, and the effect of gemfibrozil or rosiglitazone treatment on one of the most important HDL anti-atherogenic properties, the macrophage-specific reverse cholesterol transport (RCT).Materials and method. To determinate the macrophage-specific RCT, [3H]cholesterol-labelledP388D1 macrophages were injected intraperitoneally into control mice (C57BL/6) and into human CETP transgenic mice. We performed the same procedure in human apoA-I transgenic mice treated during 17 days with an oral daily gavage dose of gemfibrozil (625 mg/kg),rosiglitazone (10 mg/kg) or vehicle solution. At 48h, the mice were euthanized and [3H]cholesterol in plasma, liver and faeces were measured. Results. Human CETP transgenic mice showed decreased HDLc compared to control mice. Human apoA-I transgenic mice showed an increase in total cholesterol and HDLc when treated with gemfibrozil, but not with rosiglitazone. Neither the human CETP activity, nor either of the two pharmacological treatments altered faecal cholesterol or bile acid excretion. Furthermore, the[3H]tracer detected in liver, faecal cholesterol and bile acid was not significantly different in any of the animal groups. Conclusions. Neither human CETP expression intransgenic mice, nor treatment with gemfibrozil orrosiglitazone in human apoA-I transgenic micemodified macrophage-specific reverse cholesterol transport (AU)


Subject(s)
Animals , Mice , Gemfibrozil/pharmacokinetics , Cholesterol Ester Transfer Proteins/pharmacokinetics , Lipoproteins, HDL , Arteriosclerosis/prevention & control , Clofibric Acid/pharmacokinetics , Atherosclerosis/physiopathology , Macrophages/metabolism , Feces
11.
Curr Atheroscler Rep ; 11(5): 391-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19664384

ABSTRACT

A number of studies have raised the possibility of circulating plant sterols being a risk factor in the pathogenesis of atherosclerosis. Evidence in support of this hypothesis comes mainly from observations in sitosterolemic patients, who hyperabsorb plant sterols and suffer premature atherosclerosis. Accordingly, the atherogenicity of plant sterols of dietary origin is currently under debate, in view of the widespread use of cholesterol-lowering functional foods enriched with these compounds. Although some reports have suggested the vascular perils of small increases in plasma plant sterol concentrations, other prospective and large population-based studies have indicated otherwise. Further, the potential risk of plant sterol-enriched foods may be counterbalanced by the notable reduction in plasma cholesterol. This review summarizes the current evidence on the possible impact of plant sterols as a risk factor for atherosclerosis.


Subject(s)
Atherosclerosis , Hypercholesterolemia/drug therapy , Phytosterols/pharmacokinetics , Atherosclerosis/blood , Atherosclerosis/chemically induced , Atherosclerosis/epidemiology , Dose-Response Relationship, Drug , Humans , Hypercholesterolemia/blood , Incidence , Phytosterols/adverse effects , Prognosis , Risk Factors
12.
Atherosclerosis ; 203(1): 18-31, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18692849

ABSTRACT

Plant sterols and stanols (phytosterols/phytostanols) are known to reduce serum low-density lipoprotein (LDL)-cholesterol level, and food products containing these plant compounds are widely used as a therapeutic dietary option to reduce plasma cholesterol and atherosclerotic risk. The cholesterol-lowering action of phytosterols/phytostanols is thought to occur, at least in part, through competition with dietary and biliary cholesterol for intestinal absorption in mixed micelles. However, recent evidence suggests that phytosterols/phytostanols may regulate proteins implicated in cholesterol metabolism both in enterocytes and hepatocytes. Important advances in the understanding of intestinal sterol absorption have provided potential molecular targets of phytosterols. An increased activity of ATP-binding cassette transporter A1 (ABCA1) and ABCG5/G8 heterodimer has been proposed as a mechanism underlying the hypocholesterolaemic effect of phytosterols. Conclusive studies using ABCA1 and ABCG5/G8-deficient mice have demonstrated that the phytosterol-mediated inhibition of intestinal cholesterol absorption is independent of these ATP-binding cassette (ABC) transporters. Other reports have proposed a phytosterol/phytostanol action on cholesterol esterification and lipoprotein assembly, cholesterol synthesis and apolipoprotein (apo) B100-containing lipoprotein removal. The accumulation of phytosterols in ABCG5/G8-deficient mice, which develop features of human sitosterolaemia, disrupts cholesterol homeostasis by affecting sterol regulatory element-binding protein (SREBP)-2 processing and liver X receptor (LXR) regulatory pathways. This article reviews the progress to date in studying these effects of phytosterols/phytostanols and the molecular mechanisms involved.


Subject(s)
Cholesterol/metabolism , Phytosterols/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP-Binding Cassette Transporters/chemistry , Absorption , Animals , Cholesterol, LDL/metabolism , Dimerization , Homeostasis , Humans , Lipoproteins/chemistry , Mice , Models, Biological , Models, Chemical , Sitosterols/metabolism
13.
J Lipid Res ; 49(9): 1904-11, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18509196

ABSTRACT

Liver X receptor (LXR) agonists increase both total fecal sterol excretion and macrophage-specific reverse cholesterol transport (RCT) in vivo. In this study, we assessed the effects of ABCG5/G8 deficiency as well as those of LXR agonist-induction of RCT from macrophages to feces in vivo. A [(3)H]cholesterol-labeled macrophage cell line was injected intraperitoneally into ABCG5/G8-deficient (G5/G8(-/-)), heterozygous (G5G8(+/-)), and wild-type G5/G8(+/+) mice. G5/G8(-/-)mice presented increased radiolabeled HDL-bound [(3)H]cholesterol 24 h after the label injection. However, the magnitude of macrophage-derived [(3)H]cholesterol in liver and feces did not differ between groups. A separate experiment was conducted in G5G8(+/+) and G5G8(-/-) mice treated with or without the LXR agonist T0901317. Treatment with T0901317 increased liver ABCG5/G8 expression, which was associated with a 2-fold increase in macrophage-derived [(3)H]cholesterol in feces of G5/G8(+/+) mice. However, T0901317 treatment had no effect on fecal [(3)H]cholesterol excretion in G5G8(-/-) mice. Additionally, LXR activation stimulated the fecal excretion of labeled cholesterol after an intravenous injection of HDL-[(3)H]cholesteryl oleate in G5/G8(+/+) mice, but failed to enhance fecal [(3)H]cholesterol in G5/G8(-/-) mice. Our data provide direct in vivo evidence of the crucial role of ABCG5 and ABCG8 in LXR-mediated induction of macrophage-specific RCT.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cholesterol/metabolism , DNA-Binding Proteins/physiology , Feces/chemistry , Lipoproteins/physiology , Macrophages/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , Animals , Biological Transport , Female , Lipoproteins/deficiency , Liver X Receptors , Male , Mice , Orphan Nuclear Receptors
14.
Atherosclerosis ; 196(2): 505-13, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17588582

ABSTRACT

CETP inhibition increases HDL cholesterol levels and presumably could contribute to human atheroprotection via increasing macrophage-specific reverse cholesterol transport (RCT) and antioxidant properties of HDL. However, the impact of CETP activity variation on these two antiatherogenic functions of HDL remain unknown. In this study, we assessed the effects of overexpressing CETP in transgenic (Tg) mice on macrophage-specific RCT and HDL ability to protect against LDL oxidative modification. [(3)H]cholesterol-labeled macrophages were injected intraperitoneally into mice maintained on a chow diet or an atherogenic diet, after which the appearance of [(3)H]cholesterol in plasma, liver and feces over 48 h was determined. The degree of protection of oxidative modification of LDL coincubated with HDL was evaluated by measuring relative electrophoretic mobility and dichlorofluorescein fluorescence. CETP-Tg mice presented decreased radiolabeled HDL-bound [(3)H]cholesterol 24 and 48 h after the label injection. However, the magnitude of macrophage-derived [(3)H]cholesterol in liver and feces did not differ between CETP-Tg and control mice on either diet. Similar results were found when [(3)H]cholesterol-labeled endogenous peritoneal macrophages were injected into the CETP-Tg and control mice. Further, the injection of endogenous macrophages from CETP-Tg mice did not alter macrophage RCT in control mice. HDL from CETP-Tg and control mice protected LDL from oxidative modification similarly, and paraoxonase 1, platelet activated factor acetyl-hydrolase and lecithin-cholesterol acyl transferase activities of transgenic mice did not differ from those of control mice. In conclusion, CETP overexpression in transgenic mice does not affect RCT from macrophages to feces in vivo or the protection conferred by HDL against LDL oxidative modification.


Subject(s)
Antioxidants/physiology , Atherosclerosis/prevention & control , Cholesterol Ester Transfer Proteins/genetics , Cholesterol Ester Transfer Proteins/physiology , Cholesterol/pharmacokinetics , Lipoproteins, HDL/physiology , Lipoproteins, LDL/physiology , Macrophages/metabolism , Animals , Cells, Cultured , Cholesterol, Dietary , Humans , Macaca fascicularis , Mice , Mice, Transgenic
15.
Exp Mol Pathol ; 83(3): 361-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17920586

ABSTRACT

We describe the effect of MCP-1 deficiency in mice rendered hyperlipemic by the concomitant ablation of the LDL receptor. The MCP-1(-/-)LDLr(-/-) mice in comparison with LDLr(-/-) mice showed a decreased lipoprotein clearance, derangements in free fatty acids delivery and less glucose tolerance when fed a regular chow, and they showed a partial resistance to alterations in glucose and lipid metabolism induced by dietary fat and cholesterol. They also were less prone to the development of diet-induced obesity. Our results suggest that the role of MCP-1 in metabolism is relevant and that, although new hidden complexities are evident, the function of MCP-1/CCL2 extends far beyond the monocyte chemoattractant effect. Therefore, the regulatory mechanisms influenced by MCP-1 should be fully ascertained to understand the metabolic consequences of inflammation and before considering MCP-1 as a therapeutic target.


Subject(s)
Chemokine CCL2/metabolism , Glucose/metabolism , Lipid Metabolism , Receptors, LDL/metabolism , Animals , Chemokine CCL2/genetics , Cholesterol/blood , Diet , Glucose Tolerance Test , Insulin/metabolism , Liver/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/genetics , Triglycerides/blood
16.
Proteomics ; 7(15): 2659-66, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17610203

ABSTRACT

The molecular mechanisms involved in the phytosterol-induced decrease in intestinal cholesterol absorption remain unclear. Further, other biological properties such as immunomodulatory activity and protection against cancer have also been ascribed to these plant compounds. To gain insight into the mechanisms underlying phytosterol actions, we conducted a proteomic study in the intestinal mucosa of phytosterol-fed apolipoprotein E-deficient hypercholesterolemic (apoE-/-) mice. With respect to control-fed apoE-/- mice, nine differentially expressed proteins were identified in whole-enterocyte homogenates using 2-D DIGE and MALDI-TOF MS. These proteins are involved in plasma membrane stabilization, cytoskeleton assembly network, and cholesterol metabolism. Four of these proteins were selected for further study since they showed the highest abundance change or had a potential functional relationship with known effects of phytosterols. Annexin A2 (ANXA2) and beta-actin decrease and annexin A4 (ANXA4) and annexin A5 (ANXA5) increase were confirmed by Western blot analysis. Intestinal gene expression of ANXA2 and A5 and beta-actin was reduced, whereas that of ANXA4 was unchanged. The main results were retested in normocholesterolemic C57BL/6J mice. ANXA4 and ANXA5 protein upregulation and ANXA2 and beta-actin downregulation were reproduced in these animals. However, no changes in gene expression were found in C57BL/6J mice in either of the four proteins selected. ANXA2, A4, and A5 and beta-actin are proteins of special interest given their pleiotropic functions that include cholesterol-ester transport from caveolae, apoptosis, and anti-inflammatory properties. Therefore, the protein expression changes identified in this study might be involved in the biological effects of phytosterols.


Subject(s)
Cholesterol, Dietary/metabolism , Intestinal Mucosa/metabolism , Phytosterols/administration & dosage , Proteins/metabolism , Proteome/analysis , Amino Acid Sequence , Animals , Apolipoproteins E/deficiency , Cholesterol/blood , Cholesterol/metabolism , Gene Expression , Intestine, Small/metabolism , Isoelectric Point , Lipoproteins/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Phytosterols/pharmacology , Proteins/chemistry , Proteins/genetics , Proteomics/methods , Time Factors
17.
Life Sci ; 80(21): 1951-6, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17382351

ABSTRACT

Although most studies have focused on the cholesterol-lowering activity of phytosterols, other biological actions have been ascribed to these plant sterol compounds, one of which is a potential immune modulatory effect. To gain insight into this issue, we used a mouse model of acute, aseptic inflammation induced by a single subcutaneous turpentine injection. Hypercholesterolemic apolipoprotein E-deficient (apoE(-/-)) mice, fed with or without a 2% phytosterol supplement, were treated with turpentine or saline and euthanized 48 h later. No differences were observed in spleen lymphocyte subsets between phytosterol- and control-fed apoE(-/-) mice. However, cultured spleen lymphocytes of apoE(-/-) mice fed with phytosterols and treated with turpentine showed increased IL-2 and IFN-gamma secretion (T-helper type1, Th1 lymphocyte cytokines) compared with turpentine-treated, control-fed animals. In contrast, there was no change in Th2 cytokines IL-4 and IL-10. Phytosterols also inhibit intestinal cholesterol absorption in wild-type C57BL/6J mice but, in this case, without decreasing plasma cholesterol. Spleen lymphocytes of turpentine-treated C57BL/6J mice fed with phytosterols also showed increased IL-2 production, but IFN-gamma, IL-4 and IL-10 production was unchanged. The Th1/Th2 ratio was significantly increased both in phytosterol-fed apoE(-/-) and C57BL/6J mice. We conclude that phytosterols modulate the T-helper immune response in vivo, in part independently of their hypocholesterolemic effect in a setting of acute, aseptic inflammation. Further study of phytosterol effects on immune-based diseases characterized by an exacerbated Th2 response is thus of interest.


Subject(s)
Immunity, Cellular/drug effects , Inflammation/drug therapy , Inflammation/immunology , Phytosterols/therapeutic use , Th1 Cells/drug effects , Analysis of Variance , Animals , Apolipoproteins E/genetics , Cholesterol/blood , Dietary Supplements , Flow Cytometry , Fluorescent Antibody Technique , Inflammation/chemically induced , Interferon-gamma/metabolism , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phytosterols/pharmacology , Th1 Cells/metabolism , Turpentine/toxicity
19.
Clín. investig. arterioscler. (Ed. impr.) ; 18(5): 182-192, sept. 2006. ilus, tab
Article in Es | IBECS | ID: ibc-048712

ABSTRACT

Introducción. La función de la apolipoproteína A-II (apo A-II) en el metabolismo lipoproteico y su relación con la arteriosclerosis es poco conocida. Los estudios realizados en humanos y ratones modificados genéticamente han demostrado un efecto directo de la apo A-II en el metabolismo de los triglicéridos y los ácidos grasos libres (AGL) y la sensibilidad a la insulina. El objetivo principal de este estudio es la identificación de proteínas diferencialmente expresadas en el hígado de ratones transgénicos de apo A-II humana (h) y su potencial relación con el metabolismo de los triglicéridos y la glucosa. Material y métodos. Se realizaron estudios metabólicos de las lipoproteínas ricas en triglicéridos, la betaoxidación hepática y la prueba de tolerancia a la glucosa en ratones transgénicos de apo A-IIh y en controles en situación de ayunas y tras una carga oral de aceite de oliva. Los cambios en el perfil de expresión proteica se analizaron mediante el análisis comparativo de geles bidimensionales y la identificación de proteínas mediante espectrometría de masas MALDI-TOF. Resultados. Los ratones transgénicos de apo A-IIh presentaban un incremento del colesterol y los triglicéridos de las lipoproteínas que contienen apo B, hipertrigliceridemia aumentada tras la carga oral de ácido oleico, así como un aclaramiento acelerado de la glucosa tras la prueba de sobrecarga de glucosa. Estos cambios estaban asociados a una reducción en el catabolismo de las lipoproteínas ricas en triglicéridos y la tasa de betaoxidación hepática, pero sin cambios significativos en la actividad de la lipoproteína lipasa. De las más de 1.000 manchas resueltas en el rango pH 3 a 10, se identificaron 55 alteraciones significativas en los ratones transgénicos en comparación con los ratones controles, 16 de las cuales estaban relacionadas directamente con el metabolismo de los AGL y los carbohidratos. Conclusiones. La sobreexpresión apo A-IIh en ratones transgénicos induce hipertrigliceridemia posprandial debido, al menos en parte, a un defecto en el catabolismo de las lipoproteínas ricas en triglicéridos. La aproximación proteómica nos ha permitido detectar y caracterizar diferencias en el proteoma hepático de los ratones transgénicos de apo A-IIh y establecer proteínas potencialmente involucradas en el metabolismo de los AGL. Se requieren más estudios bioquímicos y moleculares para investigar el significado funcional de los cambios encontrados (AU)


Introduction. The role of apolipoprotein A-II (apo A-II) in lipoprotein metabolism and its relationship with atherosclerosis is poorly understood. Several studies both in humans and genetically modified mice have shown a direct effect of apo A-II on triglyceride and free fatty acid (FFA) metabolism and on insulin sensitivity. The aim of this study was to identify the proteins differentially expressed in the livers of human apo A-II transgenic mice and their potential relationship with triglyceride and glucose metabolism. Matherial and methods. Metabolic studies of triglyceride-rich lipoproteins, liver beta-oxidation and the glucose tolerance test were conducted in C57BL/6 control mice and human apo A-II transgenic mice in both fasting and postprandial states. The changes in protein expression patterns were determined by analysis of two-dimensional polyacrylamide gel electrophoresis while protein identification was performed by mass spectrometry (MALDI-TOF). Results. Human apo A-II transgenic mice showed an increase in cholesterol and triglycerides from apo B-containing lipoproteins, a higher response after oral fat test with oleic acid and higher glucose clearance after the glucose test. These changes were associated with a reduction in the clearance of triglyceride-rich lipoproteins and in the rate of liver beta-oxidation, but there were no significant changes in lipoprotein lipase activity. Within the pH 3-10 range, 55 out of more than 1,000 spots were identified to be significantly altered in human apo A-II transgenic mice compared with control mice, and 16 of these spots were directly related with FFA and carbohydrate metabolism. Conclusions. Overexpression of human apo A-II in transgenic mice induces postprandial hypertriglyceridemia. This finding is at least partly due to reduced catabolism of triglyceride-rich lipoproteins. We have been able to detect and characterize differences in the liver proteome of human apo A-II transgenic mice and to identify proteins potentially related to FFA metabolism. Further biochemical and molecular studies are required to investigate the functional significance of the changes found (AU)


Subject(s)
Animals , Mice , Apolipoprotein A-II/metabolism , Triglycerides/metabolism , Glucose/metabolism , Proteome/metabolism , Liver/metabolism , Animals, Genetically Modified , Electrophoresis, Gel, Two-Dimensional , Gas Chromatography-Mass Spectrometry
20.
Cytokine ; 34(3-4): 224-32, 2006 May.
Article in English | MEDLINE | ID: mdl-16815711

ABSTRACT

There are increasing evidences showing that inflammation participates in atherosclerosis. Therefore, the therapeutic use of anti-inflammatory agents should be considered. We have induced chronic, aseptic inflammation upon the injection of turpentine and tested the effect of dexamethasone on lipoprotein metabolism and, consequently, atherosclerosis in apolipoprotein E-deficient mice. Aseptic inflammation caused a significant decrease in hyperlipidemia. Treatment with dexamethasone elicited the opposite effect increasing hyperlipidemia through mechanisms related to the increase in the synthesis of triglyceride-rich lipoproteins. Changes in plasma lipids correlated with those observed in the size of atherosclerotic lesions. Our data suggest the presence of a common mechanism present in both observations and which is probably related to the cytokine secretion. Among the candidates, we chose to test the effect of interleukin-6 because it is involved in both processes, atherosclerosis and inflammation, and its expression is efficiently repressed by corticosteroids. The injection of recombinant interleukin-6 in our mice elicited the same effects observed in our model of inflammation. We conclude that manipulation of inflammation-related mechanisms modulates lipid homeostasis and development of atherosclerotic plaque in rodents.


Subject(s)
Apolipoproteins E/deficiency , Hyperlipidemias/pathology , Interleukin-6/metabolism , ATP Binding Cassette Transporter, Subfamily B/analysis , Animals , Apolipoproteins E/genetics , Atherosclerosis/chemically induced , Atherosclerosis/pathology , Blood Glucose , Body Weight , Hyperlipidemias/chemically induced , Inflammation/chemically induced , Inflammation/pathology , Lipids/analysis , Lipids/blood , Liver/chemistry , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Turpentine , ATP-Binding Cassette Sub-Family B Member 4
SELECTION OF CITATIONS
SEARCH DETAIL
...