Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nature ; 535(7611): 303-7, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27383786

ABSTRACT

Cellular mechanisms that mediate steatohepatitis, an increasingly prevalent condition in the Western world for which no therapies are available, are poorly understood. Despite the fact that its synthetic agonists induce fatty liver, the liver X receptor (LXR) transcription factor remains a target of interest because of its anti-atherogenic, cholesterol removal, and anti-inflammatory activities. Here we show that tetratricopeptide repeat domain protein 39B (Ttc39b, C9orf52) (T39), a high-density lipoprotein gene discovered in human genome-wide association studies, promotes the ubiquitination and degradation of LXR. Chow-fed mice lacking T39 (T39(-/-)) display increased high-density lipoprotein cholesterol levels associated with increased enterocyte ATP-binding cassette transporter A1 (Abca1) expression and increased LXR protein without change in LXR messenger RNA. When challenged with a high fat/high cholesterol/bile salt diet, T39(-/-) mice or mice with hepatocyte-specific T39 deficiency show increased hepatic LXR protein and target gene expression, and unexpectedly protection from steatohepatitis and death. Mice fed a Western-type diet and lacking low-density lipoprotein receptor (Ldlr(-/-)T39(-/-)) show decreased fatty liver, increased high-density lipoprotein, decreased low-density lipoprotein, and reduced atherosclerosis. In addition to increasing hepatic Abcg5/8 expression and limiting dietary cholesterol absorption, T39 deficiency inhibits hepatic sterol regulatory element-binding protein 1 (SREBP-1, ADD1) processing. This is explained by an increase in microsomal phospholipids containing polyunsaturated fatty acids, linked to an LXRα-dependent increase in expression of enzymes mediating phosphatidylcholine biosynthesis and incorporation of polyunsaturated fatty acids into phospholipids. The preservation of endogenous LXR protein activates a beneficial profile of gene expression that promotes cholesterol removal and inhibits lipogenesis. T39 inhibition could be an effective strategy for reducing both steatohepatitis and atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Fatty Liver/genetics , Lipoproteins, HDL/deficiency , Lipoproteins, HDL/genetics , Orphan Nuclear Receptors/metabolism , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/metabolism , Animals , Atherosclerosis/prevention & control , Atherosclerosis/therapy , Bile Acids and Salts/metabolism , Cholesterol, Dietary/metabolism , Cholesterol, HDL/metabolism , Diet, High-Fat , Fatty Acids, Unsaturated/metabolism , Fatty Liver/prevention & control , Fatty Liver/therapy , Female , Gene Expression Regulation , Hepatocytes/metabolism , Ligands , Lipogenesis/genetics , Lipoproteins/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Liver X Receptors , Male , Mice , Orphan Nuclear Receptors/genetics , Phosphatidylcholines/biosynthesis , Phosphatidylcholines/metabolism , Protein Stability , Proteolysis , Receptors, LDL/deficiency , Receptors, LDL/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Ubiquitination
2.
Arterioscler Thromb Vasc Biol ; 38(7): 1454-1467, 2018 07.
Article in English | MEDLINE | ID: mdl-29748333

ABSTRACT

OBJECTIVE: Reverse cholesterol transport (RCT) is a major mechanism by which HDL (high-density lipoprotein) protects against atherosclerosis. Endothelial lipase (EL) reportedly reduces HDL levels, which, in theory, would increase atherosclerosis. However, it remains unclear whether EL affects RCT in vivo. APPROACH AND RESULTS: Adenoviral vectors expressing EL or luciferase were intravenously injected into mice, and a macrophage RCT assay was performed. As expected, hepatic EL overexpression markedly reduced HDL levels. In parallel, plasma 3H-cholesterol counts from the EL-expressing mice decreased by 85% compared with control. Surprisingly, there was no difference in fecal 3H-cholesterol excretion between the groups. Kinetic studies revealed increased catabolism/hepatic uptake of 3HDL-cholesteryl ether, resulting in no change in fecal HDL-cholesteryl ester excretion in the mice. To explore underlying mechanisms for the preservation of RCT despite low HDL levels in the EL-expressing mice, we investigated the effects of hepatic SR-BI (scavenger receptor class B type I) knockdown. RCT assay revealed that knockdown of SR-BI alone reduced fecal excretion of macrophage-derived 3H-cholesterol. Interestingly, hepatic EL overexpression under SR-BI inhibition further attenuated fecal tracer counts as compared with control. Finally, we observed that EL overexpression enhanced in vivo RCT under pharmacological inhibition of hepatic ABCA1 (ATP-binding cassette transporter A1) by probucol. CONCLUSIONS: Hepatic EL expression compensates for reduced macrophage-derived cholesterol efflux to plasma because of low HDL levels by promoting cholesterol excretion to bile/feces via an SR-BI pathway, maintaining overall RCT in vivo. In contrast, EL-modified HDL might negatively regulate RCT via hepatic ABCA1. Despite extreme hypoalphalipoproteinemia, RCT is maintained in EL-expressing mice via SR-BI/ABCA1-dependent pathways.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cholesterol, HDL/blood , Lipase/biosynthesis , Liver/enzymology , Macrophages, Peritoneal/metabolism , Scavenger Receptors, Class B/metabolism , Adenoviridae/genetics , Animals , Enzyme Induction , Gene Transfer Techniques , Genetic Vectors , Hep G2 Cells , Humans , Lipase/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , RAW 264.7 Cells , RNA Interference , Scavenger Receptors, Class B/genetics
3.
Arterioscler Thromb Vasc Biol ; 36(4): 591-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26848156

ABSTRACT

OBJECTIVE: Oxidized products of probucol, spiroquinone and diphenoquinone, were shown to increase cell cholesterol release and plasma high-density lipoprotein (HDL) by inhibiting degradation of ATP-binding cassette transporter A1. We investigated whether these compounds enhance reverse cholesterol transport in mice. APPROACH AND RESULTS: Spiroquinone and diphenoquinone increased ATP-binding cassette transporter A1 protein (2.8- and 2.6-fold, respectively, P<0.01) and apolipoprotein A-I-mediated cholesterol release (1.4- and 1.4-fold, P<0.01 and P<0.05, respectively) in RAW264.7 cells. However, diphenoquinone, but not spiroquinone, enhanced cholesterol efflux to HDL (+12%, P<0.05), whereas both increased ATP-binding cassette transporter G1 protein, by 1.8- and 1.6-fold, respectively. When given orally to mice, both compounds significantly increased plasma HDL-cholesterol, by 19% and 20%, respectively (P<0.05), accompanied by an increase in hepatic and macrophage ATP-binding cassette transporter A1 but not ATP-binding cassette transporter G1. We next evaluated in vivo reverse cholesterol transport by injecting RAW264.7 cells labeled with (3)H-cholesterol intraperitoneally into mice. Both spiroquinone and diphenoquinone increased fecal excretion of the macrophage-derived (3)H-tracer, by 25% and 28% (P<0.01 and P<0.05), respectively. spiroquinone/diphenoquinone did not affect fecal excretion of HDL-derived (3)H-cholesterol, implying that macrophage-to-plasma was the most important step in spiroquinone/diphenoquinone-mediated promotion of in vivo reverse cholesterol transport. Finally, spiroquinone significantly reduced aortic atherosclerosis in apolipoprotein E null mice when compared with the vehicle. CONCLUSIONS: Spiroquinone and diphenoquinone increase functional ATP-binding cassette transporter A1 in both the macrophages and the liver, elevate plasma HDL-cholesterol, and promote overall reverse cholesterol transport in vivo. These compounds are promising as therapeutic reagents against atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1/drug effects , Androstadienes/pharmacology , Anticholesteremic Agents/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Cholesterol/blood , Hypercholesterolemia/drug therapy , Macrophages/drug effects , Probucol/pharmacology , Quinones/pharmacology , ATP Binding Cassette Transporter 1/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoprotein A-I/blood , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Biological Transport , Cholesterol, HDL/blood , Disease Models, Animal , Feces/chemistry , Hypercholesterolemia/blood , Hypercholesterolemia/genetics , Liver/drug effects , Liver/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Plaque, Atherosclerotic , RAW 264.7 Cells , Time Factors
4.
Biochim Biophys Acta ; 1841(9): 1247-55, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24989153

ABSTRACT

Reverse cholesterol transport (RCT) is pivotal in the return of excess cholesterol from peripheral tissues to the liver for excretion in bile and eventually feces. RCT from macrophages is a critical anti-atherogenicity mechanism of HDL. As the cholesterol absorption inhibitor ezetimibe promoted RCT in mice, which lack cholesterol ester transfer protein (CETP), we investigated its effects in hamsters, which have CETP. A high-cholesterol diet (HC) increased cholesterol levels throughout lipoprotein fractions and ezetimibe markedly reduced VLDL/LDL cholesterol levels under both normal chow (NC) and HC. However, ezetimibe did not affect and reduced HDL-cholesterol levels under NC and HC, respectively. Intraperitoneal injection of (3)H-cholesterol pre-labeled macrophages in an in vivo RCT assay increased tracer accumulation in the liver but reduced it in bile under HC, and these changes were completely cancelled by ezetimibe. Under both NC and HC, ezetimibe reduced tracer levels in the liver but increased them in feces, indicating promotion of RCT in vivo. We performed a RCT assay using hamsters subjected to bile duct ligation (BDL) to clarify whether a transintestinal cholesterol efflux (TICE) pathway contributes to ezetimibe's enhancement of RCT. BDL markedly inhibited macrophage-derived (3)H-cholesterol excretion to feces and cancelled ezetimibe's stimulatory effect on RCT, suggesting that biliary cholesterol excretion is a major contributor in RCT promotion by ezetimibe but the contribution of the TICE pathway is minimal. In conclusions, ezetimibe exerts an additive anti-atherogenic property by enhancing RCT in hamsters. Our findings suggest that this property is independent of the TICE pathway.


Subject(s)
Anticholesteremic Agents/pharmacology , Azetidines/pharmacology , Bile/drug effects , Cholesterol, HDL/metabolism , Liver/drug effects , Macrophages/drug effects , Animals , Bile/metabolism , Bile Ducts/surgery , Biological Transport/drug effects , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol, LDL/metabolism , Cholesterol, VLDL/metabolism , Cricetinae , Diet , Ezetimibe , Feces/chemistry , Liver/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Tritium
5.
Arterioscler Thromb Vasc Biol ; 34(6): 1171-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24675665

ABSTRACT

OBJECTIVE: Low-density lipoprotein receptor (LDLR) is degraded by inducible degrader of LDLR (Idol) and protein convertase subtilisin/kexin type 9 (PCSK9), thereby regulating circulating LDL levels. However, it remains unclear whether, and if so how, these LDLR degraders affect each other. We therefore investigated effects of liver-specific expression of Idol on LDL/PCSK9 metabolism in mice and hamsters. APPROACH AND RESULTS: Injection of adenoviral vector expressing Idol (Ad-Idol) induced a liver-specific reduction in LDLR expression which, in turn, increased very-low-density lipoprotein/LDL cholesterol levels in wild-type mice because of delayed LDL catabolism. Interestingly, hepatic Idol overexpression markedly increased plasma PCSK9 levels. In LDLR-deficient mice, plasma PCSK9 levels were already elevated at baseline and unchanged by Idol overexpression, which was comparable with the observation for Ad-Idol-injected wild-type mice, indicating that Idol-induced PCSK9 elevation depended on LDLR. In wild-type mice, but not in LDLR-deficient mice, Ad-Idol enhanced hepatic PCSK9 expression, with activation of sterol regulatory element-binding protein 2 and subsequently increased expression of its target genes. Supporting in vivo findings, Idol transactivated PCSK9/LDLR in sterol regulatory element-binding protein 2/LDLR-dependent manners in vitro. Furthermore, an in vivo kinetic study using (125)I-labeled PCSK9 revealed delayed clearance of circulating PCSK9, which could be another mechanism. Finally, to extend these findings into cholesteryl ester transfer protein-expressing animals, we repeated the above in vivo experiments in hamsters and obtained similar results. CONCLUSIONS: A vicious cycle in LDLR degradation might be generated by PCSK9 induced by hepatic Idol overexpression via dual mechanisms: sterol regulatory element-binding protein 2/LDLR. Furthermore, these effects would be independent of cholesteryl ester transfer protein expression.


Subject(s)
Liver/metabolism , Proprotein Convertases/blood , Receptors, LDL/physiology , Serine Endopeptidases/blood , Signal Transduction , Sterol Regulatory Element Binding Protein 2/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Cholesterol Ester Transfer Proteins/physiology , Cricetinae , Hep G2 Cells , Humans , Lipoproteins, LDL/metabolism , Liver X Receptors , Mesocricetus , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/physiology , Proprotein Convertase 9 , Proprotein Convertases/physiology , Serine Endopeptidases/physiology
6.
Biochim Biophys Acta ; 1831(8): 1402-11, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23747827

ABSTRACT

Stearoyl-coenzyme A desaturase 1 (SCD1) is the rate-limiting enzyme in the synthesis of monounsaturated fatty acids. However, the impact of SCD1 on atherosclerosis remains unclear. The aim of this study was to determine whether SCD1 affects macrophage reverse cholesterol transport (RCT) in mice. Compared to the control, adenoviral-mediated SCD1 overexpression in RAW264.7 macrophages increased cholesterol efflux to HDL, but not to apoA-I, without clear changes in ABCA1, ABCG1 and SR-BI expressions. While knockdown of ABCG1 and SR-BI did not affect the SCD1-induced cholesterol efflux to HDL, SCD1-overexpressing macrophages promoted the formation of both normal- and large-sized HDL in media, accompanying increased apolipoprotein A-I levels in HDL fractions. Transformation to larger particles of HDL was independently confirmed by nuclear magnetic resonance-based lipoprotein analysis. Interestingly, media transfer assays revealed that HDL generated by SCD1 had enhanced cholesterol efflux potential, indicating that SCD1 transformed HDL to a more anti-atherogenic phenotype. To study macrophage RCT in vivo, (3)H-cholesterol-labeled RAW264.7 cells overexpressing SCD1 or the control were intraperitoneally injected into mice. Supporting the in vitro data, injection of SCD1-macrophages resulted in significant increases in (3)H-tracer in plasma, liver, and feces compared to the control. Moreover, there was a shift towards larger particles in the (3)H-tracer distribution of HDL fractions obtained from the mice. In conclusion, macrophage-specific SCD1 overexpression promotes overall RCT through increased cholesterol efflux to HDL, suggesting that macrophage SCD1 achieves an anti-atherogenic effect by enhancing RCT.


Subject(s)
Cholesterol/metabolism , Gene Expression Regulation, Enzymologic , Lipoproteins, HDL/metabolism , Macrophages/enzymology , Stearoyl-CoA Desaturase/biosynthesis , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/therapy , Biological Transport, Active/genetics , Cell Line , Cholesterol/genetics , Humans , Lipoproteins/genetics , Lipoproteins/metabolism , Lipoproteins, HDL/genetics , Macrophages/pathology , Mice , Stearoyl-CoA Desaturase/genetics
7.
J Clin Biochem Nutr ; 55(1): 32-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25120277

ABSTRACT

Reverse cholesterol transport (RCT) is a mechanism critical to the anti-atherogenic property of HDL. Although citrulline contributes to the amelioration of atherosclerosis via endothelial nitric oxide production, it remains unclear whether it affects RCT. This study was undertaken to clarify the effects of citrulline on expressions of specific transporters such as ATP binding cassette transporters (ABC)A1 and ABCG1, and the cholesterol efflux from macrophages to apolipoprotein (apo) A-I or HDL in vitro and ex vivo. Citrulline increased ABCA1 and ABCG1 mRNA and protein levels in THP-1 macrophages, translating into enhanced apoA-I- and HDL-mediated cholesterol efflux. In the human crossover study, 8 healthy male volunteers (age 30-49 years) consumed either 3.2 g/day citrulline or placebo for 1 week. Citrulline consumption brought about significant increases in plasma levels of citrulline and arginine. Supporting the in vitro data, monocyte-derived macrophages (MDM) differentiated under autologous post-citrulline sera demonstrated enhancement of both apoA-I- and HDL-mediated cholesterol efflux through increased ABCA1 and ABCG1 expressions, compared to MDM differentiated under pre-citrulline sera. However, the placebo did not modulate these parameters. Therefore, in addition to improving endothelium function, citrulline might have an anti-atherogenic property by increasing RCT of HDL.

8.
Biochim Biophys Acta ; 1821(4): 561-72, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22353356

ABSTRACT

ABC transporter G1 (ABCG1) plays a pivotal role in HDL-mediated cholesterol efflux and atherogenesis. We investigated whether, and how, retinoic acid receptors (RARs) regulate ABCG1 expression in macrophages. All-trans retinoic acid (ATRA), an RAR ligand, increased ABCG1 protein levels and apoA-I/HDL-mediated cholesterol efflux from the macrophages. Both ATRA and other RAR agonists, TTNPB and Am580, increased major transcripts driven by promoter B upstream of exon 5, though minor transcripts driven by promoter A upstream of exon 1 were only increased by ATRA. The stimulatory effects of ATRA on ABCG1 expression were completely abolished in the presence of RAR/RXR antagonists but were only partially canceled in the presence of an LXR antagonist. Adenovirus with overexpressed oxysterol sulfotransferase abolished the LXR pathway, as previously reported, and ATRA-responsiveness in ABCA1/ABCG1 expressions were respectively attenuated by 38 and 22% compared to the control virus. Promoter assays revealed that ABCG1 levels were regulated more by promoter B than promoter A, and ATRA activated promoter B in a liver X receptor-responsive element (LXRE)-dependent manner. Further, LXRE-B in intron 7, but not LXRE-A in intron 5, enhanced ATRA responsiveness under overexpression of all RAR isoforms-RARα/ß/γ. In contrast, the activation of promoter B by TTNPB depended on LXRE-B and RARα, but not on RARß/γ. Finally, chromatin immunoprecipitation and gel-shift assays revealed a specific and direct repeat 4-dependent binding of RARα to LXRE-B. In conclusion, RAR ligands increase ABCA1/G1 expression and apoA-I/HDL-mediated cholesterol efflux from macrophages, and modulate ABCG1 promoter activity via LXRE-dependent mechanisms.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Macrophages/drug effects , Receptors, Retinoic Acid/agonists , Tretinoin/pharmacology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Animals , Benzoates/pharmacology , Biological Transport/drug effects , Blotting, Western , COS Cells , Cell Line , Chlorocebus aethiops , Cholesterol/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Liver X Receptors , Macrophages/cytology , Macrophages/metabolism , Models, Genetic , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Response Elements/genetics , Retinoid X Receptors/agonists , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Retinoids/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tetrahydronaphthalenes/pharmacology
9.
Int J Food Sci Nutr ; 64(4): 407-14, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23317012

ABSTRACT

Endothelium-bound extracellular superoxide dismutase (eEC-SOD), a major antioxidative enzyme in the vasculature, is involved in anti-atherogenesis by inhibiting low-density lipoprotein (LDL) oxidation. The objective was to investigate whether the polyphenol-rich juar tea had beneficial effects on LDL oxidation and eEC-SOD levels in patients with metabolic syndrome (MetS). A total of 20 men with MetS participated in a randomized cross-over trial, comparing consumption of five cups/day of juar tea with that of a polyphenol-poor tea, barley tea, for 4 weeks. Although there was no change in LDL oxidizability after consumption of either tea, juar tea significantly increased eEC-SOD levels by 16% (p < 0.05), whereas barley tea significantly decreased levels by 15% (p < 0.05). It is noteworthy that the changes in eEC-SOD were positively associated with those in LDL oxidizability after tea consumption (r(2) = 0.11, p < 0.05). Tea polyphenols may provide anti-atherosclerotic effects by inhibiting LDL oxidation through EC-SOD bound to the endothelium.


Subject(s)
Camellia sinensis/chemistry , Endothelium, Vascular/drug effects , Lipoproteins, LDL/metabolism , Metabolic Syndrome/drug therapy , Phytotherapy , Polyphenols/therapeutic use , Superoxide Dismutase/metabolism , Adult , Antioxidants/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Endothelium, Vascular/metabolism , Hordeum , Humans , Male , Metabolic Syndrome/metabolism , Oxidation-Reduction , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Polyphenols/pharmacology , Tea/chemistry
10.
Circ Res ; 106(4): 779-87, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20075335

ABSTRACT

RATIONALE: Association of habitual coffee consumption with coronary heart disease morbidity and mortality has not been established. We hypothesized that coffee may enhance reverse cholesterol transport (RCT) as the antiatherogenic properties of high-density lipoprotein (HDL). OBJECTIVE: This study was to investigate whether the phenolic acids of coffee and coffee regulates RCT from macrophages in vitro, ex vivo and in vivo. METHODS AND RESULTS: Caffeic acid and ferulic acid, the major phenolic acids of coffee, enhanced cholesterol efflux from THP-1 macrophages mediated by HDL, but not apoA-I. Furthermore, these phenolic acids increased both the mRNA and protein levels of ATP-binding cassette transporter (ABC)G1 and scavenger receptor class B type I (SR-BI), but not ABCA1. Eight healthy volunteers were recruited for the ex vivo study, and blood samples were taken before and 30 minutes after consumption of coffee or water in a crossover study. The mRNA as well as protein levels of ABCG1, SR-BI, and cholesterol efflux by HDL were increased in the macrophages differentiated under autologous sera obtained after coffee consumption compared to baseline sera. Finally, effects of coffee and phenolic acid on in vivo RCT were assessed by intraperitoneally injecting [(3)H]cholesterol-labeled acetyl low-density lipoprotein-loaded RAW264.7 cells into mice, then monitoring appearance of (3)H tracer in plasma, liver, and feces. Supporting in vitro and ex vivo data, ferulic acid was found to significantly increase the levels of (3)H tracer in feces. CONCLUSIONS: Coffee intake might have an antiatherogenic property by increasing ABCG1 and SR-BI expression and enhancing HDL-mediated cholesterol efflux from the macrophages via its plasma phenolic acids.


Subject(s)
Beverages , Caffeic Acids/pharmacology , Cholesterol/metabolism , Coffee , Coumaric Acids/pharmacology , Lipoproteins, HDL/metabolism , Macrophages/drug effects , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adult , Animals , Apolipoprotein A-I/metabolism , Bile/metabolism , Biological Transport , Caffeic Acids/blood , Cell Line , Cholesterol/blood , Coronary Disease/metabolism , Coronary Disease/prevention & control , Coumaric Acids/blood , Cross-Over Studies , Dose-Response Relationship, Drug , Feces/chemistry , Female , Genes, Reporter , Humans , Lipoproteins, HDL/blood , Lipoproteins, LDL/metabolism , Liver/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA, Messenger/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Time Factors , Transfection , Up-Regulation
11.
Arterioscler Thromb Vasc Biol ; 31(9): 1980-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21817095

ABSTRACT

OBJECTIVE: ATP-binding cassette transporter A1 (ABCA1) and ABCG1 are key molecules in an initial step of reverse cholesterol transport (RCT), a major antiatherogenic property of high-density lipoprotein (HDL). The ubiquitin-proteasome system (UPS) mediates nonlysosomal pathways for protein degradation and is known to be involved in atherosclerosis. However, little is known about the effects of the UPS on these molecules and overall RCT. We therefore investigated whether UPS inhibition affects ABCA1/G1 expression in macrophages and RCT in vitro and in vivo. METHODS AND RESULTS: Various proteasome inhibitors increased ABCA1/G1 expression in macrophages, translating into enhanced apolipoprotein A-I- and HDL-mediated cholesterol efflux from macrophages. ABCA1 and ABCG1 were found to undergo polyubiquitination in the macrophages and HEK293 cells overexpressing these proteins, and pulse-chase analysis revealed that proteasome inhibitors inhibited ABCA1/G1 protein degradation. In in vivo experiments, the proteasome inhibitor bortezomib increased ABCA1/G1 protein levels in mouse peritoneal macrophages, and RCT assays showed that it significantly increased the fecal (54% increase compared with saline) and plasma (23%) appearances of the tracer derived from intraperitoneally injected (3)H-cholesterol-labeled macrophages. CONCLUSIONS: The present study provided evidence that the UPS is involved in ABCA1/G1 degradation, thereby affecting RCT in vivo. Therefore, specific inhibition of the UPS pathway might lead to a novel HDL therapy that enhances RCT.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cholesterol/metabolism , Lipoproteins/physiology , Macrophages/metabolism , Proteasome Endopeptidase Complex/physiology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/analysis , Animals , Apolipoprotein A-I/physiology , Boronic Acids/pharmacology , Bortezomib , Cells, Cultured , Hep G2 Cells , Humans , Lipoproteins/analysis , Lipoproteins, HDL/physiology , Mice , Phosphorylation , Proteasome Inhibitors , Pyrazines/pharmacology , Ubiquitin-Protein Ligases/physiology , Ubiquitination
12.
J Atheroscler Thromb ; 29(5): 775-784, 2022 May 01.
Article in English | MEDLINE | ID: mdl-33952812

ABSTRACT

AIMS: Inflammation is involved in various processes of atherosclerosis development. Serum C-reactive protein (CRP) levels, a predictor for cardiovascular risk, are reportedly reduced by statins. However, several studies have demonstrated that CRP is a bystander during atherogenesis. While S100A12 has been focused on as an inflammatory molecule, it remains unclear whether statins affect circulating S100A12 levels. Here, we investigated whether atorvastatin treatment affected S100A12 and which biomarkers were correlated with changes in arterial inflammation. METHODS: We performed a prospective, randomized open-labeled trial on whether atorvastatin affected arterial (carotid and thoracic aorta) inflammation using 18fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG-PET/CT) and inflammatory markers. Thirty-one statin-naïve patients with carotid atherosclerotic plaques were randomized to either a group receiving dietary management (n=15) or one receiving atorvastatin (10mg/day, n=16) for 12weeks. 18F-FDG-PET/CT and flow-mediated vasodilation (FMD) were performed, the latter to evaluate endothelial function. RESULTS: Atorvastatin, but not the diet-only treatment, significantly reduced LDL-cholesterol (LDL-C, -43%), serum CRP (-37%) and S100A12 levels (-28%) and improved FMD (+38%). 18F-FDG-PET/CT demonstrated that atorvastatin, but not the diet-only treatment, significantly reduced accumulation of 18F-FDG in the carotid artery and thoracic aorta. A multivariate analysis revealed that reduction in CRP, S100A12, LDL-C, oxidized-LDL, and increase in FMD were significantly associated with reduced arterial inflammation in the thoracic aorta, but not in the carotid artery. CONCLUSIONS: Atorvastatin treatment reduced S100A12/CRP levels, and the changes in these circulating markers mirrored the improvement in arterial inflammation. Our observations suggest that S100A12 may be an emerging therapeutic target for atherosclerosis.


Subject(s)
Arteritis , Atherosclerosis , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Plaque, Atherosclerotic , Atherosclerosis/metabolism , Atorvastatin/therapeutic use , Biomarkers , Carotid Arteries/diagnostic imaging , Carotid Arteries/metabolism , Cholesterol, LDL/metabolism , Fluorodeoxyglucose F18 , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/drug therapy , Plaque, Atherosclerotic/drug therapy , Positron Emission Tomography Computed Tomography/methods , Prospective Studies , S100A12 Protein
13.
J Atheroscler Thromb ; 14(3): 133-41, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17587765

ABSTRACT

AIM: The ATP binding cassette transporters A1 and G1 (ABCA1/G1) and scavenger receptor class B type I (SR-BI) are key molecules in cholesterol efflux and atherogenesis. These genes are regulated by peroxisome proliferator-activated receptor gamma (PPARgamma) and liver X receptor (LXR). Telmisartan is an angiotensin type 1 receptor blocker which has been reported to act as a ligand for PPARgamma. We investigated whether PPARgamma-activating ARBs affect the expression of these genes and cholesterol efflux from macrophages. METHODS AND RESULTS: Telmisartan increased ABCA1, ABCG1 and SR-BI mRNA levels in THP-1 macrophages in a dose- and time-dependent fashion. It also increased their protein levels and enhanced apoA-I- and HDL-mediated cholesterol efflux from macrophages. The knockdown of PPARgamma by siRNA abolished the telmisartan-induced expression of these genes. The knockdown of LXRalpha resulted in the complete and partial abolishment of telmisartan-induced ABCA1 and ABCG1 expression, respectively. We also demonstrated that telmisartan-induced SR-BI expression was dependent on the PPARgamma pathway but not on the LXRalpha pathway. A luciferase assay using an ABCA1 promoter construct showed that telmisartan activated ABCA1 transcription, which was abolished if the LXR binding element was mutated, indicating that increased ABCA1 transcription by telmisartan is LXR-dependent. CONCLUSION: Our results showed that telmisartan enhanced both apoA-I- and HDL-mediated cholesterol efflux from macrophages by increasing ABCA1, ABCG1 and SR-BI expression via PPARgamma-dependent and LXR-dependent/independent pathways.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Benzimidazoles/pharmacology , Benzoates/pharmacology , Cholesterol/metabolism , Gene Expression Regulation , Macrophages/drug effects , PPAR gamma/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Lipoproteins, HDL/metabolism , Liver X Receptors , Macrophages/metabolism , Orphan Nuclear Receptors , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , RNA, Small Interfering/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Telmisartan
14.
Intern Med ; 56(6): 687-689, 2017.
Article in English | MEDLINE | ID: mdl-28321071

ABSTRACT

Massive hemoptysis is a medical emergency and needs immediate treatment. It occurs in a wide variety of pulmonary diseases and typically originates from the bronchial arteries. We herein report a very rare case of a patient bleeding from a right phrenic artery-to-pulmonary artery fistula accompanied with focal bronchiectasis in the right middle lobe of the lung. In this case, multi-detector computed tomography was useful for clarifying the etiology and the abnormal anastomosis and facilitated effective angiographic embolization.


Subject(s)
Arterio-Arterial Fistula/complications , Bronchial Arteries/pathology , Hemoptysis/etiology , Lung/pathology , Pulmonary Artery/pathology , Aged, 80 and over , Angiography , Arterio-Arterial Fistula/therapy , Embolization, Therapeutic/methods , Female , Hemoptysis/therapy , Humans , Tomography, X-Ray Computed/adverse effects
15.
Atherosclerosis ; 232(1): 31-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24401214

ABSTRACT

OBJECTIVE: Although previous randomized clinical trials established a basis for lipid guidelines worldwide, they employed fixed doses of statins throughout trials (fire-and-forget approach). In the real clinical setting, however, statin doses are titrated to achieve target low-density lipoprotein cholesterol (LDL-C) levels (treat-to-target approach). The major objective was to investigate whether intensive lipid-lowering therapy using the treat-to-target approach yielded greater regression of aortic plaques. METHODS: We therefore performed a prospective, randomized trial comparing the effects of standard (achieve LDL-C levels recommended by the Japanese guidelines) and intensive (achieve 30% lower LDL-C levels than standard) rosuvastatin therapy for 1 year in 60 hypercholesterolemic patients with a primary endpoint of aortic atherosclerotic plaques evaluated by non-invasive magnetic resonance imaging (MRI). RESULTS: Average doses were 2.9 ± 3.1 and 6.5 ± 5.1 mg/day for standard (n = 29) and intensive therapy group (n = 31), respectively. Although both therapies significantly reduced LDL-C and high-sensitivity C-reactive protein (hsCRP) levels, LDL-C reduction was significantly greater in the intensive group (-46 vs. -34%). MRI study showed that thoracic aortic plaques were significantly regressed in both groups, with greater regression of thoracic plaque in the intensive group (-9.1 vs. -3.2%, p = 0.01). Multivariate analyses revealed that thoracic plaque regression was significantly correlated with hsCRP reduction, but not with changes in serum lipids, endothelial function, or doses of rosuvastatin. CONCLUSION: Intensive statin therapy with titration targeting lower LDL-C levels resulted in greater thoracic aortic plaque regression compared to standard therapy, which was correlated with hsCRP reduction, suggesting that intensive statin therapy could provide better clinical outcomes.


Subject(s)
C-Reactive Protein/metabolism , Cholesterol, LDL/blood , Fluorobenzenes/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Aged , Anthropometry , Aorta/pathology , Atherosclerosis , Endothelium, Vascular/metabolism , Female , Guidelines as Topic , Humans , Lipids/blood , Magnetic Resonance Imaging , Male , Middle Aged , Prospective Studies , Rosuvastatin Calcium , Treatment Outcome
16.
J Am Heart Assoc ; 2(1): e003277, 2013 Jan 28.
Article in English | MEDLINE | ID: mdl-23525426

ABSTRACT

BACKGROUND: Endothelial dysfunction is an independent predictor for cardiovascular events in patients with type 2 diabetes (T2DM). Glucagon like peptide-1 (GLP-1) reportedly exerts vasodilatory actions, and inhibitors of dipeptidyl peptidase-4 (DPP-4), an enzyme-degrading GLP-1, are widely used to treat T2DM. We therefore hypothesized that DPP-4 inhibitors (DPP-4Is) improve endothelial function in T2DM patients and performed 2 prospective, randomized crossover trials to compare the DPP-4I sitagliptin and an α-glucosidase inhibitor, voglibose (in study 1) and the DPP-4Is sitagliptin and alogliptin (in study 2). METHODS AND RESULTS: In study 1, 24 men with T2DM (46±5 years) were randomized to sitagliptin or voglibose for 6 weeks without washout periods. Surprisingly, sitagliptin significantly reduced flow-mediated vasodilatation (FMD; -51% compared with baseline, P<0.05) of the brachial artery despite improved diabetic status. In contrast, voglibose did not affect FMD. To confirm this result and determine whether it is a class effect, we conducted another trial (study 2) to compare sitagliptin and alogliptin in 42 T2DM patients (66±8 years) for 6 weeks with 4-week washout periods. Both DPP-4Is improved glycemic control but significantly attenuated FMD (7.2/4.3%, P<0.001, before/after sitagliptin; 7.0/4.8%, P<0.001, before/after alogliptin, respectively). Interestingly, FMD reduction was less evident in subjects who were on statins or whose LDL cholesterol levels were reduced by them, but this was not correlated with parameters including DPP-4 activity and GLP-1 levels or diabetic parameters. CONCLUSIONS: Our 2 independent trials demonstrated that DPP-4 inhibition attenuated endothelial function as evaluated by FMD in T2DM patients. This unexpected unfavorable effect may be a class effect of DPP-4Is. CLINICAL TRIAL REGISTRATION: URL: http://center.umin.ac.jp, Unique Identifiers: UMIN000005682 (sitagliptin versus voglibose) and UMIN000005681 (sitagliptin versus alogliptin).


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Endothelium, Vascular/drug effects , Inositol/analogs & derivatives , Piperidines/adverse effects , Pyrazines/adverse effects , Triazoles/adverse effects , Uracil/analogs & derivatives , Vasodilation/drug effects , Adult , Aged , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Cholesterol, LDL/blood , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/physiopathology , Endothelium, Vascular/physiopathology , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inositol/adverse effects , Japan , Linear Models , Male , Middle Aged , Prospective Studies , Regional Blood Flow , Sitagliptin Phosphate , Time Factors , Treatment Outcome , Uracil/adverse effects
17.
J Nutr Sci Vitaminol (Tokyo) ; 58(2): 96-104, 2012.
Article in English | MEDLINE | ID: mdl-22790567

ABSTRACT

ATP-binding cassette transporters (ABC) A1 and G1 are key molecules in cholesterol efflux from macrophages, which is an initial step of reverse cholesterol transport (RCT), a major anti-atherogenic property of high-density lipoprotein (HDL). Astaxanthin is one of the naturally occurring carotenoids responsible for the pink-red pigmentation in a variety of living organisms. Although astaxanthin is known to be a strong antioxidant, it remains unclear through what mechanism of action it affects cholesterol homeostasis in macrophages. We therefore investigated the effects of astaxanthin on cholesterol efflux and ABCA1/G1 expressions in macrophages. Astaxanthin enhanced both apolipoprotein (apo) A-I- and HDL-mediated cholesterol efflux from RAW264.7 cells. In supporting these enhanced cholesterol efflux mechanisms, astaxanthin promoted ABCA1/G1 expression in various macrophages. In contrast, peroxisome proliferator-activated receptor γ, liver X receptor (LXR) α and LXRß levels remained unchanged by astaxanthin. An experiment using actinomycin D demonstrated that astaxanthin transcriptionally induced ABCA1/G1 expression, and oxysterol depletion caused by overexpression of cholesterol sulfotransferase further revealed that these inductions in ABCA1/G1 were independent of LXR-mediated pathways. Finally, we performed luciferase assays using human ABCA1/G1 promoter-reporter constructs to reveal that astaxanthin activated both promoters irrespective of the presence or absence of LXR-responsive elements, indicating LXR-independence of these activations. In conclusion, astaxanthin increased ABCA1/G1 expression, thereby enhancing apoA-I/HDL-mediated cholesterol efflux from the macrophages in an LXR-independent manner. In addition to the anti-oxidative properties, the potential cardioprotective properties of astaxanthin might therefore be associated with an enhanced anti-atherogenic function of HDL.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cholesterol/metabolism , Gene Expression/drug effects , Lipoproteins/genetics , Macrophages/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/physiology , Animals , Antioxidants , Apolipoprotein A-I/drug effects , Apolipoprotein A-I/physiology , Cardiotonic Agents , Cell Line , Lipoproteins/physiology , Lipoproteins, HDL/drug effects , Lipoproteins, HDL/physiology , Liver X Receptors , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/physiology , Xanthophylls/pharmacology
18.
Cardiovasc Res ; 95(2): 251-9, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22396501

ABSTRACT

AIMS: Endothelin-1 (ET-1) contributes to the pathogenesis of cardiovascular diseases with multiple properties such as vasoconstriction. Human ET-1 gene expression is up-regulated by the transcription factor hypoxia-inducible factor-1 (HIF-1) through hypoxia response element (HRE). Although previous studies suggested that 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) alter HIF-1-related gene expression, it remained unclear whether statins modulate HIF-1-mediated ET-1 expression. Therefore, we investigated the effect of fluvastatin on hypoxia-induced human ET-1 expression in vascular smooth muscle cells (VSMC). METHODS AND RESULTS: Hypoxia (1% O(2)), compared with the normoxic condition (21% O(2)), significantly induced the expression of preproET-1 mRNA, ET-1 protein, and ET-1 secretion in VSMC. Hypoxia induced a 2.3-fold increase in HRE-dependent ET-1 reporter gene activation. Under concentrations of 1 µmol/L or greater, fluvastatin attenuated the hypoxia-induced ET-1 gene expression through the accelerated ubiquitin/proteasome-dependent degradation of HIF-1α, thus consequently attenuating HIF-1α binding to the HRE of the ET-1 gene. These inhibitory effects of fluvastatin were cancelled by concomitant treatment with mevalonate, farnesyl pyrophosphate, or geranylgeranyl pyrophosphate, but not squalene. CONCLUSION: The present study suggests that fluvastatin attenuates HIF-1-dependent ET-1 gene expression in conjunction with the stimulation of HIF-1α ubiquitin/proteasome-dependent degradation via isoprenoid-dependent mechanisms.


Subject(s)
Endothelin-1/metabolism , Fatty Acids, Monounsaturated/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Indoles/pharmacology , Myocytes, Smooth Muscle/metabolism , Cells, Cultured , Fluvastatin , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Proteolysis/drug effects , Transcription Factors
19.
J Atheroscler Thromb ; 18(6): 513-30, 2011.
Article in English | MEDLINE | ID: mdl-21636950

ABSTRACT

AIM: Reverse cholesterol transport (RCT) is a critical mechanism for the anti-atherogenic property of HDL. The inhibitory effect of the sulfonylurea agent (SUA) glibenclamide on ATP binding-cassette transporter (ABC) A1 may decrease HDL function but it remains unclear whether it attenuates RCT in vivo. We therefore investigated how the SUAs glibenclamide and glimepiride affected the functionality of ABCA1/ABCG1 and scavenger receptor class B type I (SR-BI) expression in macrophages in vitro and overall RCT in vivo. METHODS: RAW264.7, HEK293 and BHK-21 cells were used for in vitro studies. To investigate RCT in vivo, 3H-cholesterol-labeled and acetyl LDL-loaded RAW264.7 cells were injected into mice. RESULTS: High dose (500µM) of glibenclamide inhibited ABCA1 function and apolipoprotein A-I (apoA-I)-mediated cholesterol efflux, and attenuated ABCA1 expression. Although glimepiride maintained apoA-I-mediated cholesterol efflux from RAW264.7 cells, like glibenclamide, it inhibited ABCA1-mediated cholesterol efflux from transfected HEK293 cells. Similarly, the SUAs inhibited SR-BI-mediated cholesterol efflux from transfected BHK-21 cells. High doses of SUAs increased ABCG1 expression in RAW264.7 cells, promoting HDL-mediated cholesterol efflux in an ABCG1-independent manner. Low doses (0.1-100 µM) of SUAs did not affect cholesterol efflux from macrophages despite dose-dependent increases in ABCA1/G1 expression. Furthermore, they did not change RCT or plasma lipid levels in mice. CONCLUSION: High doses of SUAs inhibited the functionality of ABCA1/SR-BI, but not ABCG1. At lower doses, they had no unfavorable effects on cholesterol efflux or overall RCT in vivo. These results indicate that SUAs do not have adverse effects on atherosclerosis contrary to previous findings for glibenclamide.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol/metabolism , Glyburide/pharmacology , Hypoglycemic Agents/pharmacology , Scavenger Receptors, Class B/metabolism , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Animals , Biological Transport , Blotting, Western , Cells, Cultured , Cricetinae , HEK293 Cells , Humans , In Vitro Techniques , Liver/cytology , Liver/drug effects , Liver/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Scavenger Receptors, Class B/antagonists & inhibitors , Scavenger Receptors, Class B/genetics
20.
Atherosclerosis ; 219(1): 141-50, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21862012

ABSTRACT

OBJECTIVE: Pioglitazone, a peroxisome proliferator-activated receptor γ (PPARγ) agonist, reportedly reduces cardiovascular events in diabetic patients. ATP cassette binding transporters (ABC) A1 and G1 are pivotal molecules for cholesterol efflux (ChE) from macrophages and high density-lipoprotein biogenesis, and the A1 transporter is regulated by a PPARγ-liver receptor X (LXR) pathway. Also, pioglitazone induces ABCG1 expression, though the exact mechanism remains unclear. We therefore investigated the effects of pioglitazone on ABCA1/G1 expression in vitro and ex vivo. METHODS: The effects of pioglitazone on ChE and ABCA1/G1 expressions in macrophages were assessed. Then, mRNA was quantified in macrophages when PPARγ/LXR inhibition by siRNA or overexpression of oxysterol sulfotransferase was performed. ABCA1/G1 promoter activity with mutated LXR-responsive elements was also measured. As an ex vivo study, 15 type 2 diabetic patients were administered pioglitazone or placebo, and ChE assays and protein expressions were determined using macrophages cultured with the corresponding sera. RESULTS: Pioglitazone increased LXRα/ABCA1/G1 expressions, which enhanced ChE from macrophages. Inhibition of PPARγ/LXR pathways revealed that LXR was primarily involved in pioglitazone's transactivation of ABCA1 but only partially involved for ABCG1. Promoter assays showed that ABCG1 was regulated more by the promoter in intron 4 than that upstream of exon 1 but both promoters were responsive to LXR activation. Sera obtained after pioglitazone treatment promoted ChE and ABCA1/G1 expressions in macrophages. CONCLUSION: Pioglitazone enhanced ChE from macrophages by increasing ABCA1/G1 in LXR-dependent and -independent manners. Our comparable in vitro and ex vivo results shed new light on pioglitazone's novel anti-atherogenic property.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Cholesterol/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/pharmacology , Macrophages/physiology , Orphan Nuclear Receptors/biosynthesis , PPAR gamma/metabolism , Thiazolidinediones/pharmacology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Cells, Cultured , HEK293 Cells , Humans , Liver X Receptors , Macrophages/drug effects , Pioglitazone , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL