Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
J Lipid Res ; 50(5): 880-4, 2009 May.
Article En | MEDLINE | ID: mdl-19141870

Apolipoprotein AV (apoAV) overexpression causes a decrease in plasma triglyceride (TG) levels, while deficiency of apoAV causes hypertriglyceridemia in both men and mice. However, contrary to what would be expected, plasma apoAV and TG levels in humans are positively correlated. To address this apparent paradox, we determined plasma apoAV levels in various mouse models with median TG levels ranging from 30 mg/dl in wild-type mice to 2089 mg/dl in glycosylphosphatidylinositol-anchored HDL binding protein 1-deficient mice. The data show that apoAV and TG levels are positively correlated in mice (r = +0.798, P < 0.001). In addition, we show that LPL gene transfer caused a simultaneous decrease in TG and apoAV in LPL-deficient mice. The combined data suggest that apoAV levels follow TG levels due to an intimate link between the apoAV molecule and TG-rich lipoproteins, comprising both secretion and removal of these lipoproteins. Taken together, the data suggest that higher plasma apoAV levels reflect an increased demand for plasma TG hydrolysis under normal physiological conditions.


Apolipoproteins A/blood , Triglycerides/blood , Animals , Apolipoprotein A-V , Humans , Lipoprotein Lipase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Diabetologia ; 51(6): 978-88, 2008 Jun.
Article En | MEDLINE | ID: mdl-18421434

AIMS/HYPOTHESIS: We previously used an integrative genetics approach to demonstrate that 5-lipoxygenase (5-LO) deficiency in mice (Alox5 (-/-)) protects against atherosclerosis despite increasing lipid levels and fat mass. In the present study, we sought to further examine the role of 5-LO in adiposity and pancreatic function. METHODS: Alox5 (-/-) and wild-type (WT) mice were characterised with respect to adiposity and glucose/insulin metabolism using in vivo and in vitro approaches. The role of ALOX5 in pancreatic function in human islets was assessed through short interfering RNA (siRNA) knockdown experiments. RESULTS: Beginning at 12 weeks of age, Alox5 (-/-) mice had significantly increased fat mass, plasma leptin levels and fasting glucose levels, but lower fasting insulin levels (p<0.05). Although Alox5 (-/-) mice did not exhibit insulin resistance, they had impaired insulin secretion in response to a bolus glucose injection. Histological analyses revealed that Alox5 (-/-) mice had increased islet area, beta cell nuclear size, and numbers of beta cells/mm(2) islet (p<0.05), indicative of both hyperplasia and hypertrophy. Basal and stimulated insulin secretion in isolated Alox5 (-/-) islets were significantly lower than in WT islets (p<0.05) and accompanied by a three- to fivefold decrease in the expression of the genes encoding insulin and pancreatic duodenal homeobox 1 (Pdx1). Direct perturbation of ALOX5 in isolated human islets with siRNA decreased insulin and PDX1 gene expression by 50% and insulin secretion by threefold (p<0.05). CONCLUSIONS/INTERPRETATION: These results provide strong evidence for pleiotropic metabolic effects of 5-LO on adiposity and pancreatic function and may have important implications for therapeutic strategies targeting this pathway for the treatment of cardiovascular disease.


Adipose Tissue/enzymology , Adipose Tissue/physiology , Carrier Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Pancreas/enzymology , 5-Lipoxygenase-Activating Proteins , Animals , Arachidonate 5-Lipoxygenase/deficiency , Arachidonate 5-Lipoxygenase/genetics , Blood Glucose/metabolism , Body Composition , Body Weight , Crosses, Genetic , Female , Glucose Tolerance Test , Humans , Insulin/blood , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA/genetics , RNA/isolation & purification , RNA Interference , RNA, Small Interfering/genetics , Sex Characteristics , Tissue Donors
4.
J Lipid Res ; 42(4): 563-70, 2001 Apr.
Article En | MEDLINE | ID: mdl-11290828

Studies with mice have revealed that increased expression of apolipoprotein A-II (apoA-II) results in elevations in high density lipoprotein (HDL), the formation of larger HDL, and the development of early atherosclerosis. We now show that the increased size of HDL results in part from an inhibition of the ability of hepatic lipase (HL) to hydrolyze phospholipids and triglycerides in the HDL and that the ratio of apoA-I to apoA-II determines HDL functional and antiatherogenic properties. HDL from apoA-II transgenic mice was relatively resistant to the action of HL in vitro. To test whether HL and apoA-II influence HDL size independently, combined apoA-II transgenic/HL knockout (HLko) mice were examined. These mice had HDL similar in size to apoA-II transgenic mice and HLko mice, suggesting that they do not increase HDL side by independent mechanisms. Overexpression of apoA-I from a transgene reversed many of the effects of apoA-II overexpression, including the ability of HDL to serve as a substrate for HL. Combined apoA-I/apoA-II transgenic mice exhibited significantly less atherosclerotic lesion formation than did apoA-II transgenic mice. These results were paralleled by the effects of the transgenes on the ability of HDL to protect against the proinflammatory effects of oxidized low density lipoprotein (LDL). Whereas nontransgenic HDL protected against oxidized LDL induction of adhesion molecules in endothelial cells, HDL from apoA-II transgenic mice was proinflammatory. HDL from combined apoA-I/apoA-II transgenic mice was equally as protective as HDL from nontransgenic mice. Our data suggest that as the ratio of apoA-II to apoA-I is increased, the HDL become larger because of inhibition of HL, and lose their antiatherogenic properties.


Apolipoprotein A-II/metabolism , Apolipoprotein A-I/metabolism , Arteriosclerosis/metabolism , Lipase/metabolism , Lipoproteins, HDL/metabolism , Animals , Aorta/pathology , Apolipoprotein A-I/genetics , Apolipoprotein A-II/genetics , Arteriosclerosis/etiology , Arteriosclerosis/pathology , Cell Adhesion/physiology , Cholesterol/blood , Diet, Atherogenic , Endothelium, Vascular/cytology , Humans , Lipids/blood , Lipoproteins, HDL/chemistry , Liver/enzymology , Male , Mice , Mice, Knockout , Mice, Transgenic , Monocytes/metabolism
5.
Arterioscler Thromb Vasc Biol ; 21(4): 481-8, 2001 Apr.
Article En | MEDLINE | ID: mdl-11304461

Oxidation of low density lipoprotein (LDL) phospholipids containing arachidonic acid at the sn-2 position occurs when a critical concentration of "seeding molecules" derived from the lipoxygenase pathway is reached in LDL. When this critical concentration is reached, the nonenzymatic oxidation of LDL phospholipids produces a series of biologically active, oxidized phospholipids that mediate the cellular events seen in the developing fatty streak. Normal high density lipoprotein (HDL) contains at least 4 enzymes as well as apolipoproteins that can prevent the formation of the LDL-derived oxidized phospholipids or inactivate them after they are formed. In the sense that normal HDL can prevent the formation of or inactivate these inflammatory LDL-derived oxidized phospholipids, normal HDL is anti-inflammatory. HDL from mice that are genetically predisposed to diet-induced atherosclerosis became proinflammatory when the mice are fed an atherogenic diet, injected with LDL-derived oxidized phospholipids, or infected with influenza A virus. Mice that were genetically engineered to be hyperlipidemic on a chow diet and patients with coronary atherosclerosis, despite normal lipid levels, also had proinflammatory HDL. It is proposed that LDL-derived oxidized phospholipids and HDL may be part of a system of nonspecific innate immunity and that the detection of proinflammatory HDL may be a useful marker of susceptibility to atherosclerosis.


Arteriosclerosis/metabolism , Inflammation/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Phospholipids/metabolism , Animals , Arteriosclerosis/diagnosis , Arteriosclerosis/physiopathology , Biomarkers , Coronary Artery Disease/diagnosis , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Diet, Atherogenic , Disease Models, Animal , Humans , Inflammation/physiopathology , Lipoproteins, HDL/physiology , Lipoproteins, LDL/physiology , Lipoxygenase/metabolism , Lipoxygenase/physiology , Mice , Oxidation-Reduction , Phospholipids/physiology
6.
J Biol Chem ; 276(26): 23304-11, 2001 Jun 29.
Article En | MEDLINE | ID: mdl-11323427

The finding that expression of a cholesterol 7alpha-hydroxylase (CYP7A1) transgene in cultured rat hepatoma cells caused a coordinate increase in lipogenesis and secretion of apoB-containing lipoproteins led to the hypothesis that hepatic production of apoB-containing lipoproteins may be linked to the expression of CYP7A1 (Wang, S.-L., Du, E., Martin, T. D., and Davis, R. A. (1997) J. Biol. Chem. 272, 19351-19358). To examine this hypothesis in vivo, a transgene encoding CYP7A1 driven by the constitutive liver-specific enhancer of the human apoE gene was expressed in C56BL/6 mice. The expression of CYP7A1 mRNA (20-fold), protein ( approximately 10-fold), and enzyme activity (5-fold) was markedly increased in transgenic mice compared with non-transgenic littermates. The bile acid pool of CYP7A1 transgenic mice was doubled mainly due to increased hydrophobic dihydroxy bile acids. In CYP7A1 transgenic mice, livers contained approximately 3-fold more sterol response element-binding protein-2 mRNA. Hepatic expression of mRNAs encoding lipogenic enzymes (i.e. fatty-acid synthase, acetyl-CoA carboxylase, stearoyl-CoA desaturase, squalene synthase, farnesyl-pyrophosphate synthase, 3-hydroxy-3-methylglutaryl-CoA reductase, and low density lipoprotein receptor) as well as microsomal triglyceride transfer protein were elevated approximately 3-5-fold in transgenic mice. CYP7A1 transgenic mice also displayed a >2-fold increase in hepatic production and secretion of triglyceride-rich apoB-containing lipoproteins. Despite the increased hepatic secretion of apoB-containing lipoproteins in CYP7A1 mice, plasma levels of triglycerides and cholesterol were not significantly increased. These data suggest that the 5-fold increased expression of the low density lipoprotein receptor displayed by the livers of CYP7A1 transgenic mice was sufficient to compensate for the 2-fold increase production of apoB-containing lipoproteins. These findings emphasize the important homeostatic role that CYP7A1 plays in balancing the anabolic lipoprotein assembly/secretion pathway with the cholesterol catabolic bile acid synthetic pathway.


Apolipoproteins B/biosynthesis , Cholesterol 7-alpha-Hydroxylase/physiology , Liver/enzymology , Animals , Apolipoprotein B-100 , Apolipoproteins B/blood , Apolipoproteins B/metabolism , Bile Acids and Salts/metabolism , CCAAT-Enhancer-Binding Proteins/biosynthesis , CCAAT-Enhancer-Binding Proteins/genetics , Carrier Proteins/metabolism , Cholesterol/metabolism , Cholesterol 7-alpha-Hydroxylase/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Hyperlipidemias/blood , Lipid Metabolism , Lipids/blood , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/biosynthesis , Receptors, LDL/biosynthesis , Receptors, LDL/genetics , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Taurochenodeoxycholic Acid/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics , Triglycerides/blood
7.
Diabetes ; 50(3): 643-51, 2001 Mar.
Article En | MEDLINE | ID: mdl-11246886

Apolipoprotein A-II (apoA-II) is the second most abundant protein in HDLs. Genetic studies in humans have provided evidence of linkage of the apoA-II gene locus to plasma free fatty acid (FFA) levels and to type 2 diabetes, and transgenic mice overexpressing mouse apoA-II have elevated levels of both FFA and triglycerides. We now show that apoA-II promotes insulin resistance and has diverse effects on fat homeostasis. ApoA-II transgenic mice have increased adipose mass and higher plasma leptin levels than C57BL/6J control mice. Fasting glucose levels were similar between apoA-II transgenic and control mice, but plasma insulin levels were elevated approximately twofold in the apoA-II transgenic mice. Compared with control mice, apoA-II transgenic mice exhibited a delay in plasma clearance of a glucose bolus. Adipose tissue isolated from fasted apoA-II transgenic mice exhibited a 50% decrease in triglyceride hydrolysis compared with adipose tissue from control mice. This is consistent with a normal response of adipose tissue to the increased insulin levels in the apoA-II transgenic mice and may partially explain the increased fat deposition. Skeletal muscle isolated from fasted apoA-II transgenic mice exhibited reduced uptake of 2-deoxyglucose compared with muscles isolated from control mice. Our observations indicate that a primary disturbance in lipoprotein metabolism can result in several traits associated with insulin resistance, consistent with the hypothesis that insulin resistance and type 2 diabetes can, under certain circumstances, be related primarily to altered lipid metabolism rather than glucose metabolism.


Adipose Tissue/anatomy & histology , Apolipoprotein A-II/physiology , Insulin Resistance/physiology , Lipoproteins, HDL/physiology , Adipose Tissue/metabolism , Animals , Apolipoprotein A-II/genetics , Blood Glucose/metabolism , Deoxyglucose/pharmacokinetics , Fasting/metabolism , Hydrolysis , Mice , Mice, Inbred C57BL , Mice, Transgenic/genetics , Muscle, Skeletal/metabolism , Reference Values , Triglycerides/metabolism
8.
Mamm Genome ; 12(3): 238-45, 2001 Mar.
Article En | MEDLINE | ID: mdl-11252174

Familial combined hyperlipidemia (FCHL) is a common genetic dyslipidemia predisposing to premature coronary heart disease (CHD). We previously identified a locus for FCHL on human Chromosome (Chr) 1q21-q23 in 31 Finnish FCHL families. We also mapped a gene for combined hyperlipidemia (Hyplip1) to a potentially orthologous region of mouse Chr 3 in the HcB-19/Dem mouse model of FCHL. The human FCHL locus was, however, originally mapped about 5 Mb telomeric to the synteny border, the centromeric part of which is homologous to mouse Chr 3 and the telomeric part to mouse Chr 1. To further localize the human Hyplip1 homolog and estimate its distance from the peak linkage markers, we fine-mapped the Hyplip1 locus and defined the borders of the region of conserved synteny between human and mouse. This involved establishing a physical map of a bacterial artificial chromosome (BAC) contig across the Hyplip1 locus and hybridizing a set of BACs to both human and mouse chromosomes by fluorescence in situ hybridization (FISH). We narrowed the location of the mouse Hyplip1 gene to a 1.5-cM region that is homologous only with human 1q21 and within approximately 5-10 Mb of the peak marker for linkage to FCHL. FCHL is a complex disorder and this distance may, thus, reflect the well-known problems hampering the mapping of complex disorders. Further studies identifying and sequencing the Hyplip1 gene will show whether the same gene predisposes to hyperlipidemia in human and mouse.


Hyperlipidemia, Familial Combined/genetics , Animals , Chromosome Mapping , Chromosomes, Artificial, Bacterial , Contig Mapping , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred BALB C , Microsatellite Repeats
9.
J Lipid Res ; 41(12): 1936-46, 2000 Dec.
Article En | MEDLINE | ID: mdl-11108726

Strain CAST/Ei (CAST) mice exhibit unusually low levels of high density lipoproteins (HDL) as compared with most other strains of mice, including C57BL/6J (B6). This appears to be due in part to a functional deficiency of lecithin:cholesterol acyltransferase (LCAT). LCAT mRNA expression in CAST mice is normal, but the mice exhibit several characteristics consistent with functional deficiency. First, the activity and mass of LCAT in plasma and in HDL of CAST mice were reduced significantly. Second, the HDL of CAST mice were relatively poor in phospholipids and cholesteryl esters, but rich in free cholesterol and apolipoprotein A-I (apoA-I). Third, the adrenals of CAST mice were depleted of cholesteryl esters, a phenotype similar to that observed in LCAT- and acyl-CoA:cholesterol acyltransferase-deficient mice. Fourth, in common with LCAT-deficient mice, CAST mice contained triglyceride-rich lipoproteins with "panhandle"-like protrusions. To examine the genetic bases of these differences, we studied HDL lipid levels in an intercross between strain CAST and the common laboratory strain B6 on a low fat, chow diet as well as a high fat, atherogenic diet. HDL levels exhibited complex inheritance, as 12 quantitative trait loci with significant or suggestive likelihood of observed data scores were identified. Several of the loci occurred over plausible candidate genes and these were investigated. The results indicate that the functional LCAT deficiency is unlikely to be due to variations of the LCAT gene. Our results suggest that novel genes are likely to be important in the control of HDL metabolism, and they provide evidence of genetic factors influencing the interaction of LCAT with HDL.


Cholesterol, HDL/blood , Adrenal Glands/metabolism , Animals , Apolipoprotein A-I/blood , Base Sequence , Crosses, Genetic , DNA Primers , Lipid Metabolism , Lod Score , Mice , Microscopy, Electron , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Quantitative Trait, Heritable , RNA, Messenger/genetics , Species Specificity
10.
J Lipid Res ; 41(4): 573-82, 2000 Apr.
Article En | MEDLINE | ID: mdl-10744778

Dietary cholesterol is known to raise total and low density lipoprotein cholesterol concentrations in humans and experimental animals, but the response among individuals varies greatly. Here we describe a mouse strain, C57BL/6ByJ (B6By), that is resistant to diet-induced hypercholesterolemia, in contrast to the phenotype seen in other common strains of mice including the closely related C57BL/6J (B6J) strain. Compared to B6J, B6By mice exhibit somewhat lower basal cholesterol levels on a chow diet, and show a relatively modest increase in absolute levels of total and LDL/VLDL cholesterol in response to an atherogenic diet containing 15% fat, 1.25% cholesterol, and 0.5% cholate. Correspondingly, B6By mice are also resistant to diet-induced aortic lesions, with less than 15% as many lesions as B6J. Food intake and cholesterol absorption are similar between B6By and B6J mice. To investigate the gene(s) underlying the resistant B6By phenotype, we performed genetic crosses with the unrelated mouse strain, A/J. A genome-wide scan revealed a locus, designated Diet1, on chromosome 2 near marker D2Mit117 showing highly significant linkage (lod = 9.6) between B6By alleles and hypo-response to diet. Examination of known genes in this region suggested that this locus represents a novel gene affecting plasma lipids and atherogenesis in response to diet.


Arteriosclerosis/genetics , Cholesterol, Dietary/metabolism , Cholesterol/blood , Hypercholesterolemia/genetics , Mice, Inbred Strains/genetics , Animals , Aorta/pathology , Cholesterol, LDL/blood , Cholesterol, VLDL/blood , Chromosome Mapping , Genetic Linkage , Genetic Predisposition to Disease , Intestinal Absorption , Mice
11.
J Biol Chem ; 275(23): 17527-35, 2000 Jun 09.
Article En | MEDLINE | ID: mdl-10748217

Serum paraoxonase (PON1), present on high density lipoprotein, may inhibit low density lipoprotein (LDL) oxidation and protect against atherosclerosis. We generated combined PON1 knockout (KO)/apolipoprotein E (apoE) KO and apoE KO control mice to compare atherogenesis and lipoprotein oxidation. Early lesions were examined in 3-month-old mice fed a chow diet, and advanced lesions were examined in 6-month-old mice fed a high fat diet. In both cases, the PON1 KO/apoE KO mice exhibited significantly more atherosclerosis (50-71% increase) than controls. We examined LDL oxidation and clearance in vivo by injecting human LDL into the mice and following its turnover. LDL clearance was faster in the double KO mice as compared with controls. There was a greater rate of accumulation of oxidized phospholipid epitopes and a greater accumulation of LDL-immunoglobulin complexes in the double KO mice than in controls. Furthermore, the amounts of three bioactive oxidized phospholipids were elevated in the endogenous intermediate density lipoprotein/LDL of double KO mice as compared with the controls. Finally, the expression of heme oxygenase-1, peroxisome proliferator-activated receptor gamma, and oxidized LDL receptors were elevated in the livers of double KO mice as compared with the controls. These data demonstrate that PON1 deficiency promotes LDL oxidation and atherogenesis in apoE KO mice.


Apolipoproteins E/blood , Arteriosclerosis/genetics , Esterases/blood , Lipoproteins, LDL/metabolism , Animals , Aortic Valve/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/pathology , Arteriosclerosis/physiopathology , Aryldialkylphosphatase , Autoantibodies/blood , Crosses, Genetic , Diet, Atherogenic , Dietary Fats , Esterases/deficiency , Esterases/genetics , Gene Expression Regulation , Humans , Lipoproteins, LDL/immunology , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout
12.
Arterioscler Thromb Vasc Biol ; 19(5): 1284-90, 1999 May.
Article En | MEDLINE | ID: mdl-10323781

Some observations have suggested that the extracellular group IIa phospholipase A2 (sPLA2), previously implicated in chronic inflammatory conditions such as arthritis, may contribute to atherosclerosis. We have examined this hypothesis by studying transgenic mice expressing the human enzyme. Compared with nontransgenic littermates, the transgenic mice exhibited dramatically increased atherosclerotic lesions when maintained on a high-fat, high-cholesterol diet. Surprisingly, the transgenic mice also exhibited significant atherosclerotic lesions when maintained on a low-fat chow diet. Immunohistochemical staining indicated that sPLA2 was present in the atherosclerotic lesions of the transgenic mice. On both chow and atherogenic diets, the transgenic mice exhibited decreased levels of HDLs and slightly increased levels of LDLs compared with nontransgenic littermates. These data indicate that group IIa sPLA2 may promote atherogenesis, in part, through its effects on lipoprotein levels. These data also provide a possible mechanism for the observation that there is an increased incidence of coronary artery disease in many chronic inflammatory diseases.


Arteriosclerosis/enzymology , Lipoproteins, LDL/biosynthesis , Phospholipases A/physiology , Animals , Arteriosclerosis/etiology , Arteriosclerosis/genetics , Aryldialkylphosphatase , Cholesterol, Dietary/toxicity , Diet, Atherogenic , Dietary Fats/toxicity , Esterases/deficiency , Female , Genetic Predisposition to Disease , Group II Phospholipases A2 , Humans , Lipids/blood , Lipoproteins, VLDL/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Phospholipases A/genetics , Phospholipases A2
13.
J Lipid Res ; 40(3): 397-404, 1999 Mar.
Article En | MEDLINE | ID: mdl-10064727

Foam cells formed from receptor-mediated uptake of lipoprotein cholesterol by macrophages in the arterial intima are critical in the initiation, progression, and stability of atherosclerotic lesions. Macrophages accumulate cholesterol when conditions favor esterification by acyl-CoA:cholesterol acyltransferase (ACAT) over cholesteryl-ester hydrolysis by a neutral cholesteryl-ester hydrolase, such as hormone-sensitive lipase (HSL), and subsequent cholesterol efflux mediated by extracellular acceptors. We recently made stable transfectants of a murine macrophage cell line, RAW 264.7, that overexpressed a rat HSL cDNA and had a 5-fold higher rate of cholesteryl-ester hydrolysis than control cells. The current study examined the effect of macrophage-specific HSL overexpression on susceptibility to diet-induced atherosclerosis in mice. A transgenic line overexpressing the rat HSL cDNA regulated with a macrophage-specific scavenger receptor promoter-enhancer was established by breeding with C57BL/6J mice. Transgenic peritoneal macrophages exhibited macrophage-specific 7-fold overexpression of HSL cholesterol esterase activity. Total plasma cholesterol levels in transgenic mice fed a chow diet were modestly elevated 16% compared to control littermates. After 14 weeks on a high-fat, high-cholesterol diet, total cholesterol increased 3-fold, with no difference between transgenics and controls. However, HSL overexpression resulted in thicker aortic fatty lesions that were 2.5-times larger in transgenic mice. HSL expression in the aortic lesions was shown by immunocytochemistry. Atherosclerosis was more advanced in transgenic mice exhibiting raised lesions involving the aortic wall, along with lipid accumulation in coronary arteries occurring only in transgenics. Thus, increasing cholesteryl-ester hydrolysis, without concomitantly decreasing ACAT activity or increasing cholesterol efflux, is not sufficient to protect against atherosclerosis. hormone-sensitive lipase overexpression in macrophages.


Arteriosclerosis/genetics , Macrophages, Peritoneal/enzymology , Membrane Proteins , Receptors, Lipoprotein , Sterol Esterase/genetics , Animals , Aorta/pathology , Arteriosclerosis/pathology , Cell Line , Cholesterol/blood , Cholesterol Esters/metabolism , Diet, Atherogenic , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation/genetics , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Rats , Receptors, Immunologic/genetics , Receptors, Scavenger , Scavenger Receptors, Class B , Transfection/genetics
14.
Nature ; 394(6690): 284-7, 1998 Jul 16.
Article En | MEDLINE | ID: mdl-9685159

Serum paraoxonase (PON1) is an esterase that is associated with high-density lipoproteins (HDLs) in the plasma; it is involved in the detoxification of organophosphate insecticides such as parathion and chlorpyrifos. PON1 may also confer protection against coronary artery disease by destroying pro-inflammatory oxidized lipids present in oxidized low-density lipoproteins (LDLs). To study the role of PON1 in vivo, we created PON1-knockout mice by gene targeting. Compared with their wild-type littermates, PON1-deficient mice were extremely sensitive to the toxic effects of chlorpyrifos oxon, the activated form of chlorpyrifos, and were more sensitive to chlorpyrifos itself. HDLs isolated from PON1-deficient mice were unable to prevent LDL oxidation in a co-cultured cell model of the artery wall, and both HDLs and LDLs isolated from PON1-knockout mice were more susceptible to oxidation by co-cultured cells than the lipoproteins from wild-type littermates. When fed on a high-fat, high-cholesterol diet, PON1-null mice were more susceptible to atherosclerosis than their wild-type littermates.


Arteriosclerosis/etiology , Chlorpyrifos/toxicity , Esterases/physiology , Insecticides/toxicity , Animals , Aorta/pathology , Arteriosclerosis/pathology , Aryldialkylphosphatase , Cells, Cultured , Chlorpyrifos/analogs & derivatives , Cholinesterase Inhibitors/toxicity , Coculture Techniques , Diet, Atherogenic , Esterases/blood , Esterases/deficiency , Esterases/genetics , Female , Humans , Inactivation, Metabolic , Lipid Peroxides/blood , Lipid Peroxides/metabolism , Lipoproteins, HDL/blood , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Restriction Mapping , Risk Factors
15.
Nat Genet ; 18(4): 374-7, 1998 Apr.
Article En | MEDLINE | ID: mdl-9537422

Familial combined hyperlipidaemia (FCHL) is a common, multifactorial disorder associated with elevated levels of plasma triglyceride, cholesterol, or both. A characteristic feature is increased secretion of very low density lipoproteins (VLDL) and apolipoprotein B (apoB). Although FCHL is the most common cause of premature coronary artery disease (CAD), accounting for over 10% of cases, its aetiology remains largely unknown. One powerful approach to the dissection of complex genetic traits involves the use of animal models. We have identified a mouse strain, HcB-19/Dem (HcB-19), which exhibits hypertriglyceridaemia, hypercholesterolaemia and elevated levels of plasma apoB. Like FCHL patients, HcB-19 mice also exhibit increased secretion of triglyceride-rich lipoproteins, and their hyperlipidaemia becomes progressively more severe with age. It is likely that the hyperlipidaemia results from a mutation of a novel gene that arose during development of strain HcB-19. We mapped the hyperlipidaemia gene (Hyplip1) to the distal portion of mouse chromosome 3. This region is syntenic to human chromosome 1q21-q23, which has recently been shown to harbour a gene associated with FCHL in families from a Finnish isolate.


Genes/genetics , Hyperlipidemias/genetics , Mice, Mutant Strains/genetics , Age Factors , Animals , Cholesterol/blood , Cholesterol, HDL/blood , Cholesterol, VLDL/blood , Chromosome Mapping , Chromosomes/genetics , Chromosomes, Human, Pair 1/genetics , Female , Genetic Linkage , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Microsatellite Repeats/genetics , Species Specificity , Triglycerides/blood
16.
J Lipid Res ; 38(11): 2232-9, 1997 Nov.
Article En | MEDLINE | ID: mdl-9392421

Lipoprotein metabolism is markedly altered during inflammation. The concentration of human secretory phospholipase A2 (sPLA2) can increase hundreds of fold in inflammatory fluids and in the circulation. It was detected in atherosclerotic lesions where many inflammatory genes are induced. As sPLA2 has been reported to act on lipoproteins as substrates, lipoprotein profiles in transgenic mice expressing sPLA2 were studied. HDL levels were markedly decreased in transgenic mice overexpressing sPLA2. HDL in the transgenics were smaller in size, with a significant decrease (11%) in phospholipid content compared to nontransgenic littermates. In sPLA2 transgenic mice and transgenic mice expressing both sPLA2 and human apolipoprotein B (apoB), the concentrations of apoB-containing lipoproteins were not altered. We conclude that sPLA2 alters HDL metabolism and could be responsible for the depressed levels of HDL that exist during chronic inflammatory diseases.


Lipoproteins, HDL/metabolism , Lipoproteins/metabolism , Phospholipases A/metabolism , Animals , Apolipoproteins B/blood , Apolipoproteins B/metabolism , Cholesterol/blood , Chromatography, Liquid , Female , Fibroblasts , Humans , Inflammation/metabolism , Lipids/blood , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Transgenic , Phospholipases A/genetics , Phospholipases A2 , Protein Binding
17.
J Clin Invest ; 100(2): 464-74, 1997 Jul 15.
Article En | MEDLINE | ID: mdl-9218525

Previous studies showed that transgenic mice overexpressing either apolipoprotein AI (apoAI) or apolipoprotein AII (apoAII), the major proteins of HDL, exhibited elevated levels of HDL cholesterol, but, whereas the apoAI-transgenic mice were protected against atherosclerosis, the apoAII-transgenic mice had increased lesion development. We now examine the basis for this striking functional heterogeneity. HDL from apoAI transgenics exhibited an enhanced ability to promote cholesterol efflux from macrophages, but HDL from apoAII transgenics and nontransgenics were not discernibly different in efflux studies. In contrast with HDL from nontransgenics and apoAI transgenics, HDL from the apoAII transgenics were unable to protect against LDL oxidation in a coculture model of the artery wall. Furthermore, HDL taken from apoAII-transgenic mice, but not HDL taken from either the apoAI transgenics or nontransgenic littermate controls, by itself stimulated lipid hydroperoxide formation in artery wall cells and induced monocyte transmigration, indicating that the apoAII-transgenic HDL were in fact proinflammatory. This loss in the ability of the apoAII-transgenic HDL to function as an antioxidant/antiinflammatory agent was associated with a decreased content of paraoxonase, an enzyme that protects against LDL oxidation. Reconstitution of the apoAII transgenic HDL with purified paraoxonase restored both paraoxonase activity and the ability to protect against LDL oxidation. We conclude that overexpression of apoAII converts HDL from an anti- to a proinflammatory particle and that paraoxonase plays a role in this transformation.


Apolipoprotein A-II/metabolism , Cholesterol/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Animals , Aorta/metabolism , Aorta/pathology , Apolipoprotein A-I/metabolism , Apolipoprotein A-II/genetics , Arteriosclerosis/etiology , Aryldialkylphosphatase , Coculture Techniques , Esterases/metabolism , Gene Expression Regulation , Lipid Peroxides/metabolism , Lipoproteins, HDL/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Phospholipases A/metabolism
18.
J Clin Invest ; 99(8): 1906-16, 1997 Apr 15.
Article En | MEDLINE | ID: mdl-9109435

Transgenic mouse lines carrying several copies of the mouse apo A-IV gene were produced. Lipoprotein composition and function, and aortic lesion development were examined. Apo A-IV levels in the plasma of transgenic mice were elevated threefold compared with nontransgenic littermates on a chow diet, and sixfold in mice fed an atherogenic diet. Plasma concentrations of total cholesterol, HDL cholesterol, triglycerides, and free fatty acids were similar in transgenic and control mice fed a chow diet. However, with the atherogenic diet, male transgenic mice exhibited significantly higher levels of plasma triglycerides (P < 0.05), total cholesterol (P < 0.01), HDL cholesterol (P < 0.0001), and free fatty acids (P < 0.05), and lower levels of unesterified cholesterol (P < 0.05), than nontransgenic littermates. Expression of the apo A-IV transgene had a protective effect against the formation of diet-induced aortic lesions, with transgenics exhibiting lesion scores of approximately 30% those seen in control mice. HDL-sized lipoproteins isolated from transgenic mice fed the atherogenic diet promoted cholesterol efflux from cholesterol-loaded human monocytes more efficiently than comparable lipoproteins from nontransgenic counterparts. Plasma from transgenics also exhibited higher endogenous cholesterol esterification rates. Taken together, these results suggest that apo A-IV levels influence the metabolism and antiatherogenic properties of HDL.


Aorta/pathology , Apolipoproteins A/genetics , Apolipoproteins A/metabolism , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Lipoproteins, HDL/blood , Animals , Arteriosclerosis/pathology , Cholesterol/metabolism , Cholesterol Esters/blood , Diet, Atherogenic , Female , Gene Expression , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
J Clin Invest ; 99(8): 2005-19, 1997 Apr 15.
Article En | MEDLINE | ID: mdl-9109446

We have examined the effects of mildly oxidized LDL and atherosclerosis on the levels of two proteins associated with HDL; apolipoprotein J (apoJ), and paraoxonase (PON). On an atherogenic diet, PON activity decreased by 52%, and apoJ levels increased 2.8-fold in fatty streak susceptible mice, C57BL/6J (BL/6), but not in fatty streak resistant mice, C3H/HeJ (C3H). Plasma PON activity was also significantly decreased, and apoJ levels were markedly increased in apolipoprotein E knockout mice on the chow diet, resulting in a 9.2-fold increase in the apoJ/PON ratio as compared to controls. Furthermore, a dramatic increase in the apoJ/PON ratio (over 100-fold) was observed in LDL receptor knockout mice when they were fed a 0.15%-cholesterol-enriched diet. Injection of mildly oxidized LDL (but not native LDL) into BL/6 mice (but not in C3H mice) on a chow diet resulted in a 59% decrease in PON activity (P < 0.01) and a 3.6-fold increase in apoJ levels (P < 0.01). When an acute phase reaction was induced in rabbits, or the rabbits were placed on an atherogenic diet, hepatic mRNA for apoJ was increased by 2.7-fold and 2.8-fold, respectively. Treatment of HepG2 cells in culture with mildly oxidized LDL (but not native LDL) resulted in reduced mRNA levels for PON (3.0-fold decrease) and increased mRNA levels for apoJ (2.0-fold increase). In normolipidemic patients with angiographically documented coronary artery disease who did not have diabetes and were not on lipid-lowering medication (n = 14), the total cholesterol/HDL cholesterol ratio was 3.1+/-0.9 as compared to 2.9+/-0.4 in the controls (n = 19). This difference was not statistically significant. In contrast, the apoJ/PON ratio was 3.0+/-0.4 in the patients compared to 0.72+/-0.2 in the controls (P < 0.009). In a subset of these normolipidemic patients (n = 5), the PON activity was low (48+/-6.6 versus 98+/-17 U/ml for controls; P < 0.009), despite similar normal HDL levels, and the HDL from these patients failed to protect against LDL oxidation in co-cultures of human artery wall cells. We conclude that: (a) mildly oxidized LDL can induce an increased apoJ/PON ratio, and (b) the apoJ/PON ratio may prove to be a better predictor of atherosclerosis than the total cholesterol/HDL cholesterol ratio.


Esterases/metabolism , Glycoproteins/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Molecular Chaperones , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Arteries/drug effects , Arteries/metabolism , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Aryldialkylphosphatase , Base Sequence , Cells, Cultured , Cholesterol/metabolism , Clusterin , Diet, Atherogenic , Esterases/genetics , Glycoproteins/genetics , Humans , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/chemistry , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Probes/genetics , Oxidation-Reduction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Receptors, LDL/genetics , Receptors, LDL/metabolism
20.
J Clin Invest ; 96(4): 1845-58, 1995 Oct.
Article En | MEDLINE | ID: mdl-7560076

A genetic cross was constructed from two parental inbred strains of mice, NZB/BINJ and SM/J, which differ markedly in their plasma lipoprotein levels. Plasma lipid and apolipoprotein values were measured in 184 F2 progeny on a normal chow diet and on an atherogenic diet. Genetic markers were typed at 126 loci spanning all chromosomes except the Y. Statistical analysis revealed significant linkage or suggestive linkage of lipoprotein levels with markers on a number of chromosomes. Chromosome 1 markers were linked to levels of total cholesterol (lod 5.9) and high density lipoprotein (HDL) cholesterol (lod 8.1), chromosome 5 markers were linked to levels of total cholesterol (lod 6.7) and HDL cholesterol (lod 5.6), and chromosome 7 markers were linked to levels of total plasma triglycerides (lod 5.1) and free fatty acids (lod 5.6). Plasma apoAII levels were linked to the apoAII gene (lod score 19.6) and were highly correlated with plasma HDL cholesterol levels (r = 0.63, P = 0.0001), indicating that apoAII expression influences HDL cholesterol levels. Molecular studies suggested that structural differences in the apoAII polypeptide of the two strains may contribute to differences in clearance of the protein.


Apolipoprotein A-II/genetics , Chromosome Mapping , Genetic Linkage , Lipoproteins/metabolism , Amino Acid Sequence , Animals , Apolipoproteins/metabolism , Base Sequence , Crosses, Genetic , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred NZB , Molecular Sequence Data , Rabbits
...