Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nutrients ; 5(7): 2629-45, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23857172

ABSTRACT

Echium oil (EO), which is enriched in SDA (18:4 n-3), reduces plasma triglyceride (TG) concentrations in humans and mice. We compared mechanisms by which EO and fish oil (FO) reduce plasma TG concentrations in mildly hypertriglyceridemic male apoB100-only LDLrKO mice. Mice were fed one of three atherogenic diets containing 0.2% cholesterol and palm oil (PO; 20%), EO (10% EO + 10% PO), or FO (10% FO + 10% PO). Livers from PO- and EO-fed mice had similar TG and cholesteryl ester (CE) content, which was significantly higher than in FO-fed mice. Plasma TG secretion was reduced in FO vs. EO-fed mice. Plasma very low density lipoprotein (VLDL) particle size was ordered: PO (63 ± 4 nm) > EO (55 ± 3 nm) > FO (40 ± 2 nm). Post-heparin lipolytic activity was similar among groups, but TG hydrolysis by purified lipoprotein lipase was significantly greater for EO and FO VLDL compared to PO VLDL. Removal of VLDL tracer from plasma was marginally faster in EO vs. PO fed mice. Our results suggest that EO reduces plasma TG primarily through increased intravascular lipolysis of TG and VLDL clearance. Finally, EO may substitute for FO to reduce plasma TG concentrations, but not hepatic steatosis in this mouse model.


Subject(s)
Apolipoprotein B-100/blood , Echium/chemistry , Lipolysis/drug effects , Plant Oils/pharmacology , Receptors, LDL/blood , Triglycerides/blood , Animals , Cholesterol Esters/blood , Cholesterol, VLDL/blood , Fish Oils/pharmacology , Gene Expression , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Palm Oil , RNA/genetics , RNA/isolation & purification
2.
J Lipid Res ; 53(4): 686-95, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22279185

ABSTRACT

12/15 lipoxygenase (12/15LO) oxidizes polyunsaturated fatty acids (PUFAs) to form bioactive lipid mediators. The role of 12/15LO in atherosclerosis development remains controversial. We evaluated atherosclerosis development and lipid metabolism in 12/15LO-LDL receptor (LDLr) double knockout (DK) vs. LDLr knockout (SK) mice fed a PUFA-enriched diet to enhance production of 12/15LO products. Compared with SK controls, DK mice fed a PUFA-enriched diet had decreased plasma and liver lipid levels, hepatic lipogenic gene expression, VLDL secretion, and aortic atherosclerosis and increased VLDL turnover. Bone marrow transplantation and Kupffer cell ablation studies suggested both circulating leukocytes and Kupffer cells contributed to the lipid phenotype in 12/15LO-deficient mice. Conditioned medium from in vitro incubation of DK vs. SK macrophages reduced triglyceride secretion in McArdle 7777 hepatoma cells. Our results suggest that, in the context of dietary PUFA enrichment, macrophage 12/15LO expression adversely affects plasma and hepatic lipid metabolism, resulting in exacerbated atherosclerosis.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Atherosclerosis/pathology , Lipid Metabolism , Macrophages/enzymology , Animals , Bone Marrow Transplantation , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Culture Media, Conditioned , Diet, Atherogenic/adverse effects , Dietary Fats, Unsaturated/administration & dosage , Fatty Acids, Unsaturated/administration & dosage , Fatty Acids, Unsaturated/metabolism , Female , Gene Expression Regulation, Enzymologic , Hepatocytes/metabolism , Kupffer Cells/metabolism , Leukocytes/metabolism , Lipoproteins, VLDL/metabolism , Liver/drug effects , Liver/metabolism , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plaque, Atherosclerotic/pathology
3.
J Lipid Res ; 50(6): 1146-55, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19122238

ABSTRACT

The fetus has a high requirement for cholesterol and synthesizes cholesterol at elevated rates. Recent studies suggest that fetal cholesterol also can be obtained from exogenous sources. The purpose of the current study was to examine the transport of maternal cholesterol to the fetus and determine the mechanism responsible for any cholesterol-driven changes in transport. Studies were completed in pregnant hamsters with normal and elevated plasma cholesterol concentrations. Cholesterol feeding resulted in a 3.1-fold increase in the amount of LDL-cholesterol taken up by the fetus and a 2.4-fold increase in the amount of HDL-cholesterol taken up. LDL-cholesterol was transported to the fetus primarily by the placenta, and HDL-cholesterol was transported by the yolk sac and placenta. Several proteins associated with sterol transport and efflux, including those induced by activated liver X receptor, were expressed in hamster and human placentas: NPC1, NPC1L1, ABCA2, SCP-x, and ABCG1, but not ABCG8. NPC1L1 was the only protein increased in hypercholesterolemic placentas. Thus, increasing maternal lipoprotein-cholesterol concentrations can enhance transport of maternal cholesterol to the fetus, leading to 1) increased movement of cholesterol down a concentration gradient in the placenta, 2) increased lipoprotein secretion from the yolk sac (shown previously), and possibly 3) increased placental NPC1L1 expression.


Subject(s)
Cholesterol/blood , Cholesterol/metabolism , Fetus/metabolism , Maternal-Fetal Exchange/physiology , Animals , Biological Transport, Active , Cholesterol, Dietary/administration & dosage , Cholesterol, HDL/blood , Cholesterol, HDL/metabolism , Cholesterol, LDL/blood , Cholesterol, LDL/metabolism , Cricetinae , DNA-Binding Proteins/metabolism , Female , Humans , Infant, Newborn , Liver X Receptors , Male , Maternal-Fetal Exchange/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins , Mesocricetus , Orphan Nuclear Receptors , Placenta/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Smith-Lemli-Opitz Syndrome/blood , Smith-Lemli-Opitz Syndrome/metabolism , Yolk Sac/metabolism
4.
J Lipid Res ; 49(11): 2390-401, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18583707

ABSTRACT

We investigated the in vivo metabolic fate of pre-beta HDL particles in human apolipoprotein A-I transgenic (hA-I (Tg)) mice. Pre-beta HDL tracers were assembled by incubation of [(125)I]tyramine cellobiose-labeled apolipoprotein A-I (apoA-I) with HEK293 cells expressing ABCA1. Radiolabeled pre-beta HDLs of increasing size (pre-beta1, -2, -3, and -4 HDLs) were isolated by fast-protein liquid chromatography and injected into hA-I (Tg)-recipient mice, after which plasma decay, in vivo remodeling, and tissue uptake were monitored. Pre-beta2, -3, and -4 had similar plasma die-away rates, whereas pre-beta1 HDL was removed 7-fold more rapidly. Radiolabel recovered in liver and kidney 24 h after tracer injection suggested increased (P < 0.001) liver and decreased kidney catabolism as pre-beta HDL size increased. In plasma, pre-beta1 and -2 were rapidly (<5 min) remodeled into larger HDLs, whereas pre-beta3 and -4 were remodeled into smaller HDLs. Pre-beta HDLs were similarly remodeled in vitro with control or LCAT-immunodepleted plasma, but not when incubated with phospholipid transfer protein knockout plasma. Our results suggest that initial interaction of apoA-I with ABCA1 imparts a unique conformation that partially determines the in vivo metabolic fate of apoA-I, resulting in increased liver and decreased kidney catabolism as pre-beta HDL particle size increases.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/metabolism , High-Density Lipoproteins, Pre-beta/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Animals , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Conformation , Rats
5.
Arterioscler Thromb Vasc Biol ; 27(8): 1828-36, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17510466

ABSTRACT

OBJECTIVES: The aim of this study was to determine the role of ATP binding cassette transporter A1 (ABCA1) on generation of different-sized nascent HDLs. METHODS AND RESULTS: HEK293 cells stably-transfected with ABCA1 (HEK293-ABCA1) or non-transfected (control) cells were incubated with lipid free 125I-apoA-I for 24 hours. Incubation of apoA-I with HEK293-ABCA1 cells, but not control cells, led to the formation of heterogeneous-sized, pre-beta migrating nascent HDL subpopulations (pre-beta1 to -4) that varied in size (7.1 to 15.7 nm), lipid, and apoA-I content. Kinetic studies suggested that all subpopulations were formed simultaneously, with no evidence for a precursor-product relationship between smaller and larger-sized particles. When isolated nascent pre-beta HDLs (pre-beta1 to -4) were added back to HEK293-ABCA1 cells, their ability to bind to ABCA1 and efflux lipid was severely compromised. Heat-denaturation of pre-beta1 HDL resulted in partial recovery of ABCA1 binding, suggesting that initial interaction of apoA-I with ABCA1 results in a constrained conformation of apoA-I that decreases subsequent binding. CONCLUSIONS: Interaction of apoA-I with ABCA1 results in the simultaneous generation of pre-beta HDLs of discrete size and chemical composition. These nascent particles are poor substrates for subsequent lipidation by ABCA1 and presumably require additional non-ABCA1-mediated lipidation for further maturation.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/pharmacology , High-Density Lipoproteins, Pre-beta/metabolism , Lipoproteins, HDL/biosynthesis , ATP-Binding Cassette Transporters/genetics , Atherosclerosis/physiopathology , Binding, Competitive , Cells, Cultured , Cholesterol Ester Transfer Proteins , Culture Media , Humans , Probability , Sensitivity and Specificity , Transfection
6.
J Lipid Res ; 46(10): 2233-45, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16024913

ABSTRACT

Patients homozygous for Tangier disease have a near absence of plasma HDL as a result of mutations in ABCA1 and hypercatabolize normal HDL particles. To determine the relationship between ABCA1 expression and HDL catabolism, we investigated intravascular remodeling, plasma clearance, and organ-specific uptake of HDL in mice expressing the human apolipoprotein A-I (apoA-I) transgene in the Abca1 knockout background. Small HDL particles (7.5 nm), radiolabeled with (125)I-tyramine cellobiose, were injected into recipient mice to quantify plasma turnover and the organ uptake of tracer. Small HDL tracer was remodeled to 8.2 nm diameter particles within 5 min in human apolipoprotein A-I transgenic (hA-I(Tg)) mice (control) and knockout mice. Decay of tracer from plasma was 1.6-fold more rapid in knockout mice (P < 0.05) and kidney uptake was twice that of controls, with no difference in liver uptake. We also observed 2-fold greater hepatic expression of ABCA1 protein in hA-I(Tg) mice compared with nontransgenic mice, suggesting that overexpression of human apoA-I stabilized hepatic ABCA1 protein in vivo. We conclude that ABCA1 is not required for in vivo remodeling of small HDLs to larger HDL subfractions and that the hypercatabolism of normal HDL particles in knockout mice is attributable to a selective catabolism of HDL apoA-I by the kidney.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Kidney/metabolism , Lipoproteins, HDL/metabolism , ATP Binding Cassette Transporter 1 , Animals , Apolipoprotein A-I/genetics , Lipoproteins, HDL/blood , Liver/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Phosphatidylcholine-Sterol O-Acyltransferase/blood
7.
J Clin Invest ; 115(5): 1333-42, 2005 May.
Article in English | MEDLINE | ID: mdl-15841208

ABSTRACT

Patients with Tangier disease exhibit extremely low plasma HDL concentrations resulting from mutations in the ATP-binding cassette, sub-family A, member 1 (ABCA1) protein. ABCA1 controls the rate-limiting step in HDL particle assembly by mediating efflux of cholesterol and phospholipid from cells to lipid-free apoA-I, which forms nascent HDL particles. ABCA1 is widely expressed; however, the specific tissues involved in HDL biogenesis are unknown. To determine the role of the liver in HDL biogenesis, we generated mice with targeted deletion of the second nucleotide-binding domain of Abca1 in liver only (Abca1(-L/-L)). Abca1(-L/-L) mice had total plasma and HDL cholesterol concentrations that were 19% and 17% those of wild-type littermates, respectively. In vivo catabolism of HDL apoA-I from wild-type mice or human lipid-free apoA-I was 2-fold higher in Abca1(-L/-L) mice compared with controls due to a 2-fold increase in the catabolism of apoA-I by the kidney, with no change in liver catabolism. We conclude that in chow-fed mice, the liver is the single most important source of plasma HDL. Furthermore, hepatic, but not extrahepatic, Abca1 is critical in maintaining the circulation of mature HDL particles by direct lipidation of hepatic lipid-poor apoA-I, slowing its catabolism by the kidney and prolonging its plasma residence time.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Apolipoprotein A-I/metabolism , Kidney/metabolism , Lipoproteins, HDL/blood , Tangier Disease/genetics , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Gene Targeting , Genotype , Hepatocytes/metabolism , Humans , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Tangier Disease/metabolism
8.
J Lipid Res ; 45(4): 716-28, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14729861

ABSTRACT

We compared the in vivo metabolism of prebeta HDL particles isolated by anti-human apolipoprotein A-I (apoA-I) immunoaffinity chromatography (LpA-I) in human apoA-I transgenic (hA-I Tg) mice with that of lipid-free apoA-I (LFA-I) and small LpA-I. After injection, prebeta LpA-I were removed from plasma more rapidly than were LFA-I and small LpA-I. Prebeta LpA-I and LFA-I were preferentially degraded by kidney compared with liver; small LpA-I were preferentially degraded by the liver. Five minutes after tracer injection, 99% of LFA-I in plasma was found to be associated with medium-sized (8.6 nm) HDL, whereas only 37% of prebeta tracer remodeled to medium-sized HDL. Injection of prebeta LpA-I doses into C57Bl/6 recipients resulted in a slower plasma decay compared with hA-I Tg recipients and a greater proportion (>60%) of the prebeta radiolabel that was associated with medium-sized HDL. Prebeta LpA-I contained one to four molecules of phosphatidylcholine per molecule of apoA-I, whereas LFA-I contained less than one. We conclude that prebeta LpA-I has two metabolic fates in vivo, rapid removal from plasma and catabolism by kidney or remodeling to medium-sized HDL, which we hypothesize is determined by the amount of lipid associated with the prebeta particle and the particle's ability to bind to medium-sized HDL.


Subject(s)
Apolipoprotein A-I/metabolism , Lipoproteins, HDL/metabolism , Animals , Apolipoprotein A-I/genetics , High-Density Lipoproteins, Pre-beta , Humans , Kidney/metabolism , Kinetics , Lipoproteins, HDL/blood , Liver/metabolism , Mice , Mice, Transgenic , Particle Size , Phospholipids/analysis , Protein Binding
9.
Metabolism ; 51(8): 1053-60, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12145781

ABSTRACT

The effect of 6 months of endurance exercise training on plasma concentrations of lipoprotein (Lp)AI and LpAI:AII was determined in 39 sedentary subjects (17 men, 22 women, average age, 57 years) with abnormal cholesterol concentrations (total cholesterol [TC] > 200 mg/dL, or high-density lipoprotein-cholesterol [HDL-C] < 35 mg/dL). Following exercise training, plasma LpAI concentrations increased (+5.9 +/- 1.2 mg/dL; P <.001), but there was no change in total apolipoprotein (apo) A-I or LpAI:AII concentrations. The change in plasma LpAI concentration was positively correlated to changes in total HDL-C (r =.495, P =.001), the sum of HDL4-C(nmr) + HDL5-C(nmr) (r =.417, P =.008), and average HDL particle size (r =.415, P =.009), but not to changes in body composition or Vo2max. In the 8 subjects with the greatest change in LpAI concentration following training, the size distribution of LpAI and LpAI:AII particles in plasma also was measured before and after training. In these subjects, the size distribution of LpAI:AII particles did not change with training, but there was a significant increase (0.1 nm; P =.048) in the peak size of the "medium" (7.8 to 9.8 nm) LpAI particles after training. In 7 subjects who served as age- and weight-matched sedentary controls, plasma concentrations of total apo A-I, the LpAI and LpAI:AII subfractions, and plasma lipoprotein-lipids did not differ significantly between baseline and final testing. These data indicate that endurance exercise training increases the average size and plasma concentrations of LpAI, but not LpAI:AII, particles, which may represent possible enhancements of reverse cholesterol transport and may provide insight into the role that exercise plays in reducing cardiovascular disease risk.


Subject(s)
Exercise , Glycoproteins , Lipoprotein(a)/analogs & derivatives , Lipoprotein(a)/blood , Physical Endurance , Aged , Carrier Proteins/analysis , Cholesterol Ester Transfer Proteins , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Female , Humans , Male , Middle Aged , Particle Size , Sterol O-Acyltransferase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...