Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Assay Drug Dev Technol ; 11(8): 501-7, 2013 Oct.
Article En | MEDLINE | ID: mdl-24116940

Apolipoprotein A-II (ApoA-II) is the second most abundant protein constituent of high-density lipoprotein (HDL). The physiologic role of ApoA-II is poorly defined. ApoA-II may inhibit lecithin:cholesterol acyltransferase and cholesteryl-ester-transfer protein activities, but may increase the hepatic lipase activity. ApoA-II may also inhibit the hepatic cholesteryl uptake from HDL probably through the scavenger receptor class B type I depending pathway. Interpretation of data from transgenic and knockout mice of genes involved in lipoprotein metabolism has been often complicated as clinical implications because of species difference. So it is important to obtain human ApoA-II for further studies about its functions. In our studies, Pichia pastoris expression system was first used to express a high-level secreted recombinant human ApoA-II (rhApoA-II). We have cloned the cDNA encoding human ApoA-II and achieved its high-level secreting expression with a yield of 65 mg/L of yeast culture and the purification process was effective and easy to handle. The purified rhApoA-II can be used to further study its biological activities.


Apolipoprotein A-II/biosynthesis , Pichia/metabolism , Recombinant Proteins/biosynthesis , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Blotting, Western , Cloning, Molecular , Culture Media , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Fermentation , Genetic Vectors , Humans , Indicators and Reagents , Liposomes/chemistry , Methanol/metabolism , Promoter Regions, Genetic/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
2.
J Clin Endocrinol Metab ; 97(9): E1658-66, 2012 Sep.
Article En | MEDLINE | ID: mdl-22745238

CONTEXT: Cellular cholesterol efflux is a key step in reverse cholesterol transport and may depend on the metabolism of apolipoprotein (apo) B-100, apoA-I, and apoA-II. OBJECTIVE: We examined the associations between cholesterol efflux and plasma concentrations and kinetics of very low-density lipoprotein (VLDL), intermediate-density lipoprotein (IDL), and low-density lipoprotein (LDL)-apoB-100, high-density lipoprotein (HDL)-apoA-I, and HDL-apoA-II in men. DESIGN, SUBJECTS, AND METHODS: Thirty men were recruited from the community with a wide range of body mass index. The capacity of plasma and HDL to efflux cholesterol was measured ex vivo. Apolipoprotein kinetics were measured using stable isotope techniques and multicompartmental modeling. RESULTS: Cholesterol efflux to whole plasma was correlated with plasma levels of cholesterol, triglyceride, apoB-100, insulin, cholesteryl ester transfer protein, and lecithin-cholesterol acyltransferase, body mass index and waist circumference (P < 0.05 in all). Cholesterol efflux was inversely correlated with the fractional catabolic rate (FCR) of VLDL (r = -0.728), IDL (r = -0.662), and LDL-apoB-100 (r = -0.479) but positively correlated with the FCR (r = 0.438) and production rate (r = 0.468) of HDL-apoA-II. In multiple regression analysis, the concentration and FCR of VLDL-apoB-100 (ß-coefficient = 0.708 and -0.518, respectively) and IDL-apoB-100 (ß-coefficient = 0.354 and -0.447, respectively) were independent predictors of cholesterol efflux. The association of cholesterol efflux with apoB-100 metabolism was diminished after removal of apoB-100-containing lipoproteins from plasma prior to efflux. All associations, except for cholesteryl ester transfer protein, were lost when cholesterol efflux to isolated HDL was tested. CONCLUSIONS: The plasma concentration and kinetics of apoB-100-containing lipoproteins are significant predictors of the capacity of whole plasma to effect cellular cholesterol efflux.


Apolipoprotein A-II/biosynthesis , Apolipoprotein B-100/biosynthesis , Cholesterol/metabolism , Adult , Aged , Anthropometry , Apolipoprotein A-I/biosynthesis , Blood Chemical Analysis , Cholesterol, HDL/metabolism , Cholesterol, LDL/blood , Cholesterol, VLDL/blood , Humans , Kinetics , Male , Middle Aged , Models, Statistical , Regression Analysis
3.
Biochemistry ; 48(33): 8070-6, 2009 Aug 25.
Article En | MEDLINE | ID: mdl-19618959

Human plasma HDL are the target of streptococcal serum opacity factor (SOF), a virulence factor that clouds human plasma. Recombinant (r) SOF transfers cholesteryl esters (CE) from approximately 400,000 HDL particles to a CE-rich microemulsion (CERM), forms a cholesterol-poor HDL-like particle (neo HDL), and releases lipid-free (LF) apo A-I. Whereas the rSOF reaction requires labile apo A-I, the modulation effects of other apos are not known. We compared the products and rates of the rSOF reaction against human HDL and HDL from mice overexpressing apos A-I and A-II. Kinetic studies showed that the reactivity of various HDL species is apo-specific. LpA-I reacts faster than LpA-I/A-II. Adding apos A-I and A-II inhibited the SOF reaction, an effect that was more profound for apo A-II. The rate of SOF-mediated CERM formation was slower against HDL from mice expressing human apos A-I and A-II than against WT mice HDL and slowest against HDL from apo A-II overexpressing mice. The lower reactivity of SOF against HDL containing human apos is due to the higher hydropathy of human apo A-I, particularly its C-terminus relative to mouse apo A-I, and the higher lipophilicity of human apo A-II. The SOF-catalyzed reaction is the first to target HDL rather than its transporters and receptors in a way that enhances reverse cholesterol transport (RCT). Thus, effects of apos on the SOF reaction are highly relevant. Our studies show that the "humanized" apo A-I-expressing mouse is a good animal model for studies of rSOF effects on RCT in vivo.


Apolipoprotein A-II/physiology , Apolipoprotein A-I/physiology , Lipoproteins, HDL/antagonists & inhibitors , Peptide Hydrolases/blood , Streptococcus pyogenes/physiology , Animals , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-II/genetics , Apolipoprotein A-II/metabolism , Catalysis , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Cholesterol Ester Transfer Proteins/chemistry , Cholesterol Ester Transfer Proteins/physiology , Cholesterol Esters/metabolism , Emulsions , Humans , Lipoproteins, HDL/blood , Mice , Mice, Transgenic , Nephelometry and Turbidimetry , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Peptide Fragments/physiology , Peptide Hydrolases/genetics , Peptide Hydrolases/physiology , Protein Binding , Recombinant Proteins/blood , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Streptococcus pyogenes/chemistry
4.
Clin Chim Acta ; 398(1-2): 48-52, 2008 Dec.
Article En | MEDLINE | ID: mdl-18727923

BACKGROUND: The discovery of molecular biomarkers is crucial to the diagnosis of head and neck squamous cell cancer (HNSCC). METHODS: Proteins from pre-surgery serum samples of patients with HNSCC and healthy individuals were analyzed by 2-dimensional gel electrophoresis (2-DE) using a 17 cm-long immobilized pH gradient gel strip (large gel). The differentially expressed protein spots were detected by statistical analysis. Because 2 haptoglobin (Hp) alpha chains were found to be differentially expressed, the genotypic distribution of Hp alpha chains in patients and healthy individuals was assayed by polymerase chain reaction. The protein expression levels of Hp alpha chains in individuals carrying different Hp alleles were analyzed by 2-DE with a small gel. RESULTS: Two isoforms of haptoglobin alpha2 chain (Hp alpha2) in patients' sera were found from 2-DE analysis to be up-regulated, while the isoforms of haptoglobin alpha1 chain (Hp alpha1) were significantly down-regulated. Apolipoprotein AII and 2 isoforms of apolipoprotein CII were also differentially expressed in the sera of patients with HNSCC. The Hp alpha2 chain was significantly up-regulated in the patients carrying at least one haptoglobin 2 allele, according to the spot intensities from scanned images of small-gel 2-DE. CONCLUSIONS: The expression pattern of seven differentially expressed polypeptides and the up-regulation of Hp alpha2 in individuals with the Hp 2 allele are potential biomarkers.


Haptoglobins/metabolism , Head and Neck Neoplasms/diagnosis , Neoplasms, Squamous Cell/diagnosis , Adolescent , Aged , Alleles , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-II/genetics , Apolipoproteins C/biosynthesis , Apolipoproteins C/genetics , Biomarkers , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Female , Genomics , Haptoglobins/analysis , Haptoglobins/genetics , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/genetics , Humans , Hydrogen-Ion Concentration , Isoelectric Focusing , Isomerism , Male , Middle Aged , Neoplasms, Squamous Cell/blood , Neoplasms, Squamous Cell/genetics , Proteome/genetics , Reverse Transcriptase Polymerase Chain Reaction , Silver Staining , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Young Adult
5.
Metabolism ; 57(7): 973-9, 2008 Jul.
Article En | MEDLINE | ID: mdl-18555840

Hydroxymethylglutaryl-coenzyme A reductase inhibitors (statins) are extensively used to regulate dyslipidemia and to reduce atherosclerotic cardiovascular disease. In addition to effectively lowering cholesterol and low-density lipoprotein levels, rosuvastatin and certain other statins can also increase plasma high-density lipoprotein (HDL) cholesterol modestly. However, the mechanism of action of rosuvastatin on HDL metabolic processes is not understood. Using cultured human hepatoblastoma cells (Hep G2) as an in vitro model system, we assessed the effect of rosuvastatin on apolipoprotein (apo) A-I and apo A-II (the major proteins of HDL) synthesis and HDL catabolic processes. Rosuvastatin dose-dependently increased messenger RNA expression and de novo synthesis of apo A-I but not apo A-II. Rosuvastatin selectively increased the synthesis of HDL particles containing only apo A-I (LP A-I) but not particles containing both apo A-I and A-II (LP A-I + A-II). The HDL(3)-protein or HDL(3)-cholesterol ester uptake by Hep G2 cells was not affected by rosuvastatin. The apo A-I-containing particles secreted by rosuvastatin-treated Hep G2 significantly increased cholesterol efflux from fibroblasts. The data indicate that rosuvastatin increases hepatic apo A-I but not apo A-II messenger RNA transcription, thereby selectively increasing the synthesis of functionally active apo A-I-containing HDL particles, which mediate cholesterol efflux from peripheral tissues. We suggest that this mechanism of action of rosuvastatin to increase apo A-I production without apo A-I/HDL removal may result in increased apo A-I turnover that results in accelerated reverse cholesterol transport.


Apolipoprotein A-II/biosynthesis , Apolipoprotein A-I/biosynthesis , Fluorobenzenes/pharmacology , Hepatocytes/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Cell Line, Tumor , Cholesterol, HDL/metabolism , Fibroblasts/metabolism , Hepatocytes/drug effects , Humans , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Rosuvastatin Calcium , Stimulation, Chemical , Transcription, Genetic
6.
Lab Invest ; 87(7): 633-43, 2007 Jul.
Article En | MEDLINE | ID: mdl-17468778

In mice, apolipoprotein A-II (apoA-II) self-associates to form amyloid fibrils (AApoAII) in an age-associated manner. We postulated that the two most important factors in apoA-II amyloidosis are the Apoa2(c) allele, which codes for the amyloidogenic protein APOA2C (Gln5, Ala38) and transmission of amyloid fibrils. To characterize further the contribution of the Apoa2(c) allele to amyloidogenesis and improve detection of amyloidogenic materials, we established transgenic mice that overexpress APOA2C protein under the cytomegalovirus (CMV) immediate early gene (CMV-IE) enhancer/chicken beta promoter. Compared to transgene negative (Tg(-/-)) mice that express apoA-II protein mainly in the liver, mice homozygous (Tg(+/+)) and heterozygous (Tg(+/-)) for the transgene express a high level of apoA-II protein in many tissues. They also have higher plasma concentrations of apoA-II, higher ratios of ApoA-II/apolipoprotein A-I (ApoA-I) and higher concentrations of high-density lipoprotein (HDL) cholesterol. Following injection of AApoAII fibrils into Tg(+/+) mice, amyloid deposition was observed in the testis, liver, kidney, heart, lungs, spleen, tongue, stomach and intestine but not in the brain. In Tg(+/+) mice, but not in Tg(-/-) mice, amyloid deposition was induced by injection of less than 10(-8) mug AApoAII fibrils. Furthermore, deposition in Tg(+/+) mice occurred more rapidly and to a greater extent than in Tg(-/-) mice. These studies indicate that increased levels of APOA2C protein lead to earlier and greater amyloid deposition and enhanced sensitivity to the transmission of amyloid fibrils in transgenic mice. This transgenic mouse model should prove valuable for studies of amyloidosis.


Amyloid/metabolism , Amyloid/ultrastructure , Amyloidosis/metabolism , Apolipoprotein A-II/biosynthesis , Neurofibrils/ultrastructure , Amyloidosis/pathology , Animals , Apolipoprotein A-II/ultrastructure , Cloning, Molecular , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Inbred Strains , Mice, Transgenic , Microscopy, Electron, Transmission , Neurofibrils/pathology , Protein Biosynthesis , Tissue Distribution
7.
Am J Physiol Regul Integr Comp Physiol ; 293(1): R70-7, 2007 Jul.
Article En | MEDLINE | ID: mdl-17363680

This study investigated the effect of clofibrate treatment on expression of target genes of peroxisome proliferator-activated receptor (PPAR)-alpha and various genes of the lipid metabolism in liver and adipose tissue of pigs. An experiment with 18 pigs was performed in which pigs were fed either a control diet or the same diet supplemented with 5 g clofibrate/kg for 28 days. Pigs treated with clofibrate had heavier livers, moderately increased mRNA concentrations of various PPAR-alpha target genes in liver and adipose tissue, a higher concentration of 3-hydroxybutyrate, and markedly lower concentrations of triglycerides and cholesterol in plasma and lipoproteins than control pigs (P < 0.05). mRNA concentrations of sterol regulatory element-binding proteins (SREBP)-1 and -2, insulin-induced genes (Insig)-1 and Insig-2, and the SREBP target genes acetyl-CoA carboxylase, 3-methyl-3-hydroxyglutaryl-CoA reductase, and low-density lipoprotein receptor in liver and adipose tissue and mRNA concentrations of apolipoproteins A-I, A-II, and C-III in the liver were not different between both groups of pigs. In conclusion, this study shows that clofibrate treatment activates PPAR-alpha in liver and adipose tissue and has a strong hypotriglyceridemic and hypocholesterolemic effect in pigs. The finding that mRNA concentrations of some proteins responsible for the hypolipidemic action of fibrates in humans were not altered suggests that there were certain differences in the mode of action compared with humans. It is also shown that PPAR-alpha activation by clofibrate does not affect hepatic expression of SREBP target genes involved in synthesis of triglycerides and cholesterol homeostasis in liver and adipose tissue of pigs.


Adipose Tissue/metabolism , Anticholesteremic Agents/pharmacology , Clofibrate/pharmacology , Liver/metabolism , PPAR alpha/biosynthesis , PPAR alpha/genetics , Sterol Regulatory Element Binding Proteins/genetics , 3-Hydroxybutyric Acid/blood , Adipose Tissue/drug effects , Animals , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-II/genetics , Apolipoprotein C-III/biosynthesis , Apolipoprotein C-III/genetics , Body Weight/drug effects , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cholesterol/metabolism , Eating/drug effects , Lipoproteins/blood , Liver/drug effects , Organ Size/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Swine , Triglycerides/metabolism , Up-Regulation/drug effects
8.
J Lipid Res ; 45(12): 2377-87, 2004 Dec.
Article En | MEDLINE | ID: mdl-15466364

We previously demonstrated that transgenic mice overexpressing mouse apolipoprotein A-II (apoA-II) exhibit several traits associated with the insulin resistance (IR) syndrome, including increased atherosclerosis, hypertriglyceridemia, obesity, and IR. The skeletal muscle appeared to be the insulin-resistant tissue in the apoA-II transgenic mice. We now demonstrate a decrease in FA oxidation in skeletal muscle of apoA-II transgenic mice, consistent with reports that decreased skeletal muscle FA oxidation is associated with increased skeletal muscle triglyceride accumulation, skeletal muscle IR, and obesity. The decrease in FA oxidation is not due to decreased carnitine palmitoyltransferase 1 activity, because oxidation of palmitate and octanoate were similarly decreased. Quantitative RT-PCR analysis of gene expression demonstrated that the decrease in FA oxidation may be explained by a decrease in medium chain acyl-CoA dehydrogenase. We previously demonstrated that HDLs from apoA-II transgenic mice exhibit reduced binding to CD36, a scavenger receptor involved in FA metabolism. However, studies of combined apoA-II transgenic and CD36 knockout mice suggest that the major effects of apoA-II are independent of CD36. Rosiglitazone treatment significantly ameliorated IR in the apoA-II transgenic mice, suggesting that the underlying mechanisms of IR in this animal model may share common features with certain types of human IR.


Apolipoprotein A-II/genetics , Insulin Resistance/physiology , Animals , Apolipoprotein A-II/biosynthesis , Female , Gene Dosage , Glycogen/metabolism , Heterozygote , Homozygote , Insulin Resistance/genetics , Liver/metabolism , Male , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism
9.
Circ Res ; 95(8): 789-97, 2004 Oct 15.
Article En | MEDLINE | ID: mdl-15388641

Apolipoprotein A-II (apoA-II), the second major high-density lipoprotein (HDL) apolipoprotein, has been linked to familial combined hyperlipidemia. Human apoA-II transgenic mice constitute an animal model for this proatherogenic disease. We studied the ability of human apoA-II transgenic mice HDL to protect against oxidative modification of apoB-containing lipoproteins. When challenged with an atherogenic diet, antigens related to low-density lipoprotein (LDL) oxidation were markedly increased in the aorta of 11.1 transgenic mice (high human apoA-II expressor). HDL from control mice and 11.1 transgenic mice were coincubated with autologous very LDL (VLDL) or LDL, or with human LDL under oxidative conditions. The degree of oxidative modification of apoB lipoproteins was then evaluated by measuring relative electrophoretic mobility, dichlorofluorescein fluorescence, 9- and 13-hydroxyoctadecadienoic acid content, and conjugated diene kinetics. In all these different approaches, and in contrast to control mice, HDL from 11.1 transgenic mice failed to protect LDL from oxidative modification. A decreased content of apoA-I, paraoxonase (PON1), and platelet-activated factor acetyl-hydrolase activities was found in HDL of 11.1 transgenic mice. Liver gene expression of these HDL-associated proteins did not differ from that of control mice. In contrast, incubation of isolated human apoA-II with control mouse plasma at 37 degrees C decreased PON1 activity and displaced the enzyme from HDL. Thus, overexpression of human apoA-II in mice impairs the ability of HDL to protect apoB-containing lipoproteins from oxidation. Further, the displacement of PON1 by apoA-II could explain in part why PON1 is mostly found in HDL particles with apoA-I and without apoA-II, as well as the poor antiatherogenic properties of apoA-II-rich HDL.


Apolipoprotein A-II/physiology , Aryldialkylphosphatase/metabolism , Dinoprost/analogs & derivatives , Hyperlipoproteinemia Type II/metabolism , Lipoproteins, HDL/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Aorta/metabolism , Aortic Diseases/etiology , Aortic Diseases/metabolism , Apolipoprotein A-I/metabolism , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-II/genetics , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Cholesterol, HDL/blood , Diet, Atherogenic , Dinoprost/blood , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Hyperlipoproteinemia Type II/genetics , Lipoproteins, LDL/blood , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/metabolism , Liver/metabolism , Male , Mice , Mice, Transgenic , Oxidation-Reduction , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/physiology , Thiobarbituric Acid Reactive Substances/analysis
10.
J Lipid Res ; 43(10): 1734-42, 2002 Oct.
Article En | MEDLINE | ID: mdl-12364558

To ascertain the mechanisms underlying the hypoalphalipoproteinemia present in mice overexpressing human apolipoprotein A-II (apoA-II) (line 11.1), radiolabeled HDL or apoA-I were injected into mice. Fractional catabolic rate of [(3)H]cholesteryl oleoyl ether HDL ([(3)H]HDL) was 2-fold increased in 11.1 transgenic mice compared with control mice and this was concomitant with increased radioactivity in liver, gonads, and adrenals. However, scavenger receptor class B, type I (SR-BI) was increased only in adrenals. [(3)H]HDL of 11.1 transgenic mice presented greater binding but decreased uptake compared with control mice when Chinese hamster ovary cells transfected with SR-BI were used, thereby pointing to unknown but SR-BI-independent mechanisms as being responsible for the increased (3)H-radioactivity seen in liver and gonads. Synthesis rate (SR) of plasma [(3)H]HDL was 2-fold decreased in 11.1 transgenic mice. Mouse (125)I-apoA-I was 2-fold more rapidly catabolized (mainly by the kidney) in transgenic mice. Mouse apoA-I displacement from HDL by the addition of isolated human apoA-II was reproduced ex vivo; thus, this mechanism may be involved in the increased renal catabolism of apoA-I. ApoA-I SR was 2-fold decreased in 11.1 transgenic mice and this was concomitant with a 2.3-fold decrease in hepatic apoA-I mRNA abundance. Our findings show that multiple mechanisms are involved in the HDL deficiency presented by mice overexpressing human apoA-II.


Apolipoprotein A-II/biosynthesis , Cholesterol, HDL/deficiency , Actins/biosynthesis , Animals , Apolipoprotein A-I/metabolism , Apolipoprotein A-II/metabolism , Binding, Competitive , CD36 Antigens/biosynthesis , CHO Cells/metabolism , Cholesterol, HDL/analysis , Cholesterol, HDL/metabolism , Cricetinae , Female , Humans , Kinetics , Liver/metabolism , Mice , Mice, Transgenic , Organ Specificity , RNA, Messenger/biosynthesis , Radioligand Assay , Transfection , Tritium
11.
J Histochem Cytochem ; 49(6): 739-48, 2001 Jun.
Article En | MEDLINE | ID: mdl-11373320

Apolipoprotein A-II (apoA-II), an apolipoprotein in serum high-density lipoprotein, is a precursor of mouse senile amyloid fibrils. The liver has been considered to be the primary site of synthesis. However, we performed nonradioactive in situ hybridization analysis in tissue sections from young and old amyloidogenic (R1.P1-Apoa2C) and amyloid-resistant (SAMR1) mice and revealed that other tissues in addition to the liver synthesize apoA-II. We found a strong hybridization signal in the basal cells of the squamous epithelium and the chief cells of the fundic gland in the stomach, the crypt cells and a small portion of the absorptive epithelial cells in the small intestine, the basal cells of the tongue mucosa, and the basal cells of the epidermis and hair follicles in the skin in both mouse strains. Expression of apoA-II mRNA in those tissues was also examined by RT-PCR analysis. Immunolocalization of apoA-II protein also indicated the cellular localization of apoA-II. ApoA-II transcription was not observed in the heart. Amyloid deposition was observed around the cells expressing apoA-II mRNA in the old R1.P1-Apoa2C mice. These results demonstrate that the apoA-II mRNA is transcribed and translated in various extrahepatic tissues and suggest a possible contribution of apoA-II synthesized in these tissues to amyloid deposition.


Aging/physiology , Amyloidosis/etiology , Apolipoprotein A-II/isolation & purification , Animals , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-II/genetics , Gastric Mucosa/metabolism , Immunohistochemistry , In Situ Hybridization , Intestinal Mucosa/metabolism , Intestines/chemistry , Liver/chemistry , Liver/metabolism , Mice , RNA, Messenger/isolation & purification , Skin/chemistry , Skin/metabolism , Stomach/chemistry , Tongue/chemistry , Tongue/metabolism
12.
J Biol Chem ; 276(7): 4634-9, 2001 Feb 16.
Article En | MEDLINE | ID: mdl-11050100

Similar to fibrate hypolipidemic drugs, long chain polyunsaturated fatty acids contained in fish oil are activators of peroxisome proliferator-activated receptor alpha (PPARalpha). The goal of this study was to assess the contribution of PPARalpha in mediating the effect of fish oil on plasma lipid, lipoprotein, and apolipoprotein levels. To this end, PPARalpha-deficient mice and wild-type littermates were fed isocaloric fish oil or coconut oil diets, the content of which varied reciprocally between 0, 3, 7, and 10% for 1 week. In both wild-type and PPARalpha-deficient mice, fish oil feeding was associated with a dose-dependent decrease in triglycerides, cholesterol, and phospholipids associated with lower levels of very low density lipoprotein (VLDL) triglycerides and high density lipoprotein (HDL) cholesterol. The lowering of triglycerides and VLDL triglycerides was associated with a significant decrease of plasma apoC-III in both genotypes. Fish oil treatment did not influence hepatic apoC-III mRNA levels in either genotype indicating that apoC-III is not under transcriptional control by fish oil. The lowering of HDL cholesterol observed in both genotypes was associated with reduced plasma apoA-II without changes in liver apoA-II mRNA levels. In contrast, plasma apoA-I and liver apoA-I mRNA levels were decreased in wild-type but not in PPARalpha-deficient mice after fish oil feeding indicating that PPARalpha contributes to the effect of fish oil on apoA-I gene expression. In conclusion, PPARalpha is not rate-limiting for fish oil to exert its triglyceride- and HDL-lowering action. Furthermore, PPARalpha mediates, at least partly, the decrease of apoA-I after fish oil treatment, whereas apoC-III and apoA-II levels are affected in a PPARalpha-independent manner. Altogether, these results show major molecular differences in action between fibrates and fish oil providing a molecular rationale for combination treatment with these compounds.


Fish Oils/pharmacology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Acyl-CoA Oxidase , Animals , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/blood , Apolipoprotein A-I/genetics , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-II/blood , Apolipoprotein A-II/genetics , Apolipoprotein C-III , Apolipoproteins C/biosynthesis , Apolipoproteins C/blood , Apolipoproteins C/genetics , Cholesterol/blood , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/genetics , Dose-Response Relationship, Drug , Fenofibrate/pharmacology , Liver/enzymology , Liver/metabolism , Mice , Mice, Knockout , Oxidoreductases/biosynthesis , Oxidoreductases/genetics , Phospholipids/blood , RNA, Messenger/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Triglycerides/blood
13.
Biochim Biophys Acta ; 1488(3): 233-44, 2000 Nov 15.
Article En | MEDLINE | ID: mdl-11082533

We investigated the mechanisms that lead to combined hyperlipidemia in transgenic mice that overexpress human apolipoprotein (apo) A-II (line 11.1). The 11.1 transgenic mice develop pronounced hypertriglyceridemia, and a moderate increase in free fatty acid (FFA) and plasma cholesterol, especially when fed a high-fat/high-cholesterol diet. Post-heparin plasma lipoprotein lipase and hepatic lipase activities (using artificial or natural autologous substrates), the decay of plasma triglycerides with fasting, and the fractional catabolic rate of the radiolabeled VLDL-triglyceride (both fasting and postprandial) were similar in 11. 1 transgenic mice and in control mice. In contrast, a 2.5-fold increase in hepatic VLDL-triglyceride production was observed in 11. 1 transgenic mice in a period of 2 h in which blood lipolysis was inhibited. This increased synthesis of hepatic VLDL-triglyceride used preformed FFA rather than FFA of de novo hepatic synthesis. The 11.1 transgenic mice also presented reduced epididymal/parametrial white adipose tissue weight (1.5-fold), increased rate of epididymal/parametrial hormone-sensitive lipase-mediated lipolysis (1.2-fold) and an increase in cholesterol and, especially, in triglyceride liver content, suggesting an enhanced mobilization of fat as the source of preformed FFA reaching the liver. Increased plasma FFA was reverted by insulin, demonstrating that 11.1 transgenic mice are not insulin resistant. We conclude that the overexpression of human apoA-II in transgenic mice induces combined hyperlipidemia through an increase in VLDL production. These mice will be useful in the study of molecular mechanisms that regulate the overproduction of VLDL, a situation of major pathophysiological interest since it is the basic mechanism underlying familial combined hyperlipidemia.


Apolipoprotein A-II/genetics , Dietary Fats/administration & dosage , Hyperlipidemia, Familial Combined/genetics , Lipoproteins, VLDL/biosynthesis , Animals , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-II/blood , Blood Glucose , Cholesterol, Dietary/administration & dosage , Fatty Acids, Nonesterified/blood , Female , Food Deprivation , Gene Expression Regulation , Glucose Tolerance Test , Humans , Hyperlipidemia, Familial Combined/blood , Insulin/blood , Insulin Resistance , Lipolysis , Lipoproteins, VLDL/blood , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Transgenic , Time Factors , Triglycerides/blood
14.
Int J Tissue React ; 22(2-3): 67-78, 2000.
Article En | MEDLINE | ID: mdl-10937356

Our understanding of the in vivo metabolic functions of apoA-I and A-II has greatly advanced with the use of transgenic mice, but the physiological role of apoA-IV remains elusive. Both apoA-I and A-II are necessary for the structural stability of high-density lipoprotein (HDL). Structural differences exist between human and mouse A apoproteins because: i) human cholesterol ester transfer protein, lecithin cholesterol acyl transferase and phospholipid transfer protein interact better with human apoA-I; ii) human apoA-I and A-II, alone or in combination, form polydisperse instead of monodisperse HDL particles. Human apoA-II overexpression has highlighted its inhibitory effect on lipoprotein lipase and hepatic lipase, resulting in hypertriglyceridemia and concomitantly decreased HDL and apoA-I. After long-term challenge with an atherogenic diet, mice are less protected against lesion formation by human apoA-II, mouse apoA-II being overtly proatherogenic. On the other hand, human apoA-I confers great protection against lesion formation and causes reduction of preexisting lesions. Human apoA-IV is also protective, although the mechanisms by which this protection is achieved remain to be determined.


Apolipoprotein A-II/biosynthesis , Apolipoprotein A-I/biosynthesis , Apolipoproteins A/biosynthesis , Cholesterol/metabolism , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-II/genetics , Apolipoprotein A-II/physiology , Apolipoproteins A/genetics , Arteriosclerosis , Biological Transport, Active , Disease Susceptibility , Gene Expression Profiling , Homeostasis , Humans , Lipoproteins, HDL/blood , Mice , Mice, Knockout , Mice, Transgenic
15.
Circulation ; 102(1): 21-7, 2000 Jul 04.
Article En | MEDLINE | ID: mdl-10880410

BACKGROUND: Coronary heart disease patients with low high-density lipoprotein cholesterol (HDL-C) levels, high triglyceride levels, or both are at an increased risk of cardiovascular events, but the clinical impact of raising HDL-C or decreasing triglycerides remains to be confirmed. METHODS AND RESULTS: In a double-blind trial, 3090 patients with a previous myocardial infarction or stable angina, total cholesterol of 180 to 250 mg/dL, HDL-C < or =45 mg/dL, triglycerides < or =300 mg/dL, and low-density lipoprotein cholesterol < or =180 mg/dL were randomized to receive either 400 mg of bezafibrate per day or a placebo; they were followed for a mean of 6.2 years. The primary end point was fatal or nonfatal myocardial infarction or sudden death. Bezafibrate increased HDL-C by 18% and reduced triglycerides by 21%. The frequency of the primary end point was 13. 6% on bezafibrate versus 15.0% on placebo (P=0.26). After 6.2 years, the reduction in the cumulative probability of the primary end point was 7.3%, (P=0.24). In a post hoc analysis in the subgroup with high baseline triglycerides (> or =200 mg/dL), the reduction in the cumulative probability of the primary end point by bezafibrate was 39.5% (P=0.02). Total and noncardiac mortality rates were similar, and adverse events and cancer were equally distributed. CONCLUSIONS: Bezafibrate was safe and effective in elevating HDL-C levels and lowering triglycerides. An overall trend in a reduction of the incidence of primary end points was observed. The reduction in the primary end point in patients with high baseline triglycerides (> or =200 mg/dL) requires further confirmation.


Bezafibrate/administration & dosage , Cholesterol, HDL/blood , Coronary Disease/drug therapy , Coronary Disease/prevention & control , Hypolipidemic Agents/administration & dosage , Triglycerides/blood , Aged , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-II/biosynthesis , Apolipoprotein C-III , Apolipoproteins C/biosynthesis , Bezafibrate/adverse effects , Coronary Disease/blood , Coronary Disease/mortality , Female , Fibrinogen/metabolism , Follow-Up Studies , Humans , Hypolipidemic Agents/adverse effects , Lipoprotein Lipase/metabolism , Male , Middle Aged , Survival Analysis , Treatment Outcome
16.
Atherosclerosis ; 149(1): 43-9, 2000 Mar.
Article En | MEDLINE | ID: mdl-10704613

Although the role of multiple humoral agents (such as plasma albumin, glucose, hormones etc.) are implicated in lipoprotein metabolism, the mechanism of action of these agents on various steps of the synthesis and secretion of lipoproteins and apolipoproteins (protein moieties of lipoproteins) are not completely understood. Specifically, the hepatocellular mechanisms of the effect of albumin and fatty acids on apolipoprotein (apo) AI and AII [major proteins of high density lipoproteins (HDL)] synthesis and secretion are not known. Using human hepatoblastoma cells (Hep G2) as an in vitro model system, this study examined the effect of albumin and fatty acids on the synthesis, secretion, and the steady-state mRNA expression of apo AI and AII. The data indicated that the incubation of Hep G2 cells with albumin, dose-dependently, inhibited apo AI and AII accumulation (secretion) in the media, de novo synthesis, and the steady-state mRNA expression. Albumin did not alter total protein synthesis; thus the effect of albumin appeared to be specific for the synthesis and secretion of apo AI and apo AII. Free fatty acids (FFA) are transported by albumin and diseases characterized by enhanced FFA mobilization (e.g. diabetes mellitus) are associated with low HDL levels. Studies were therefore performed to examine the effect of albumin-bound-oleic acid on apo AI and apo AII production. The results showed that the albumin-oleate complex further increased the inhibitory effects of albumin on apo AI and apo AII production. These data suggest how HDL metabolism may be affected at the hepatocellular level by alterations in plasma albumin concentrations and/or fatty acid mobilization in clinical situations characterized by altered HDL levels.


Albumins/pharmacology , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-I/biosynthesis , Oleic Acid/pharmacology , Apolipoprotein A-I/drug effects , Apolipoprotein A-I/genetics , Apolipoprotein A-II/drug effects , Apolipoprotein A-II/genetics , Dose-Response Relationship, Drug , Drug Interactions , Hepatoblastoma/chemistry , Humans , Liver Neoplasms/chemistry , Probability , RNA, Messenger/analysis , Tumor Cells, Cultured
17.
J Soc Biol ; 193(1): 67-75, 1999.
Article Fr | MEDLINE | ID: mdl-10851558

Fibrates are old hypolipidemic drugs with pleitropic effects on lipid metabolism. Until, recently their intimate molecular mechanisms of action were mysterious. In the late 5 years, we have shown that the pharmacological effects of fibrates depend on their binding to "Peroxisome Proliferator Activated Receptor alpha" (PPAR alpha). The binding of fibrates to PPAR alpha induces the activation or the inhibition of multiple genes involved in lipid metabolism through the binding of the activated PPAR alpha to "Peroxisome Proliferator Response Element" (PPRE) located in the gene promoters. Fibrates reduce plasma triglyceride levels by altering the expression of numerous genes coding for proteins involved in fatty acid metabolism (fatty acid transport protein, acyl-CoA synthetase, etc.) and also by increasing the lipoprotein lipase synthesis and decreasing the apolipoprotein C-III synthesis. Fibrates increase HDL cholesterol levels by increasing apolipoprotein A-I and apolipoprotein A-II synthesis. Furthermore, we recently demonstrated that fibrates are potent anti-inflammatory molecules through an indirect modulation of the nuclear-factor-kappa B activity. Therefore, we suggest that fibrates inhibit atherosclerosis development not only by improving the plasma lipid profile but also by reducing inflammation in the vascular wall.


Anticholesteremic Agents/pharmacology , Gene Expression Regulation/physiology , Hypolipidemic Agents/pharmacology , Naphthalenes/pharmacology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/blood , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-II/blood , Cholesterol, HDL/blood , Gene Expression Regulation/drug effects , Humans , Triglycerides/blood
18.
Arterioscler Thromb Vasc Biol ; 18(6): 999-1006, 1998 Jun.
Article En | MEDLINE | ID: mdl-9633943

Estrogen therapy increases plasma HDL levels, which may reduce cardiovascular risk in postmenopausal women. The mechanism of action of estrogen in influencing various steps in hepatic HDL and apolipoprotein (apo) A-I synthesis and secretion are not fully understood. In this study, we have used the human hepatoblastoma cell line (Hep G2) as an in vitro model system to delineate the effect of estradiol on multiple regulatory steps involved in hepatic HDL metabolism. Incubation of Hep G2 cells with estradiol resulted in the following statistically significant findings: (1) increased accumulation of apoA-I in the medium without affecting uptake/removal of radiolabeled HDL-protein; (2) accelerated incorporation of [3H]leucine into apoA-I; (3) selective increase in [3H]leucine incorporation into lipoprotein (LP) A-I but not LP A-I+A-II HDL particles (HDL particles without and with apoA-II, respectively); (4) increased ability of apoA-I-containing particles to efflux cholesterol from fibroblasts; (5) stimulated steady state apoA-I but not apoA-II mRNA expression; and (6) increased newly transcribed apoA-I mRNA message without effect on apoA-I mRNA half-life. The data indicate that estradiol stimulates newly transcribed hepatic apoA-I mRNA, resulting in a selective increase in LP A-I, a subfraction of HDL that is associated with decreased atherosclerotic cardiovascular disease, especially in premenopausal women.


Apolipoprotein A-II/genetics , Apolipoprotein A-I/genetics , Estradiol/pharmacology , Lipoproteins, HDL , Pregnancy Proteins , Transcription, Genetic/drug effects , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-II/biosynthesis , Carrier Proteins/metabolism , Humans , RNA, Messenger , Racemases and Epimerases , Tumor Cells, Cultured
19.
Biosci Biotechnol Biochem ; 61(11): 1810-3, 1997 Nov.
Article En | MEDLINE | ID: mdl-9404059

Gene expression of apolipoproteins (apo) A-I, A-IV, and B, the predominant protein components of chylomicrons, was investigated in the residual ileum after a massive small bowel resection in rats. A Northern blot analysis showed that the apo A-IV mRNA level, but not the apo A-I and B mRNA levels, in the ileum was significantly higher in the resected rats than in the sham-operated rats 24 h and 2 wk post-surgery. RT-PCR coupled with a primer extension assay revealed that the apo B-48 mRNA/apo B-100 mRNA ratio, i.e., apo B mRNA editing, in the ileum was unchanged by the resection. It is thus concluded that, among the major intestinal apolipoproteins, apo A-IV is the only one whose gene expression is influenced by loss of the proximal intestine.


Adaptation, Physiological/physiology , Apolipoproteins/biosynthesis , Ileum/metabolism , Ileum/physiology , Animals , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , Apolipoprotein A-II/biosynthesis , Apolipoprotein A-II/genetics , Apolipoproteins/genetics , Apolipoproteins B/biosynthesis , Apolipoproteins B/genetics , Blotting, Northern , DNA, Complementary/metabolism , Gene Expression , Ileum/surgery , Male , Polymerase Chain Reaction , RNA/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar
20.
Am J Clin Nutr ; 64(5): 706-11, 1996 Nov.
Article En | MEDLINE | ID: mdl-8901789

Low plasma concentrations of high-density lipoprotein (HDL) are associated with increased risk of coronary heart disease. Several drugs that induce the microsomal cytochrome P-450-dependent enzyme system in liver and intestine, the sites of HDL apolipoprotein (apo) A-I and A-II synthesis, raise plasma HDL concentrations in humans. To test the hypothesis that phytochemicals with cytochrome P-450-inducing activity may also increase plasma HDL concentrations, two controlled dietary trials were undertaken in healthy nonsmoking males aged 20-28 y. One study examined the effect of replacing 300 g glucosinolate-free vegetables with 300 g Brussels sprouts/d for 3 wk. The other study examined the effects of 150 mg eugenol/d in capsule form, using a double-blind, placebo-controlled crossover design. There were no significant increases in plasma apo A-I, apo A-II, HDL cholesterol, or HDL phospholipids. These results suggest that dietary phytochemicals that induce members of the cytochrome P-450 system do not necessarily raise plasma HDL concentrations in humans, but do not exclude the possibility that some phytochemicals may have such an effect.


Cytochrome P-450 Enzyme System/metabolism , Glucosinolates/pharmacology , Lipoproteins, HDL/blood , Adult , Apolipoprotein A-II/biosynthesis , Brassica , Cross-Sectional Studies , Cytochrome P-450 Enzyme System/analysis , Double-Blind Method , Eugenol/pharmacology , Humans , Intestines/enzymology , Liver/enzymology , Male , Vegetables
...