Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 276(49): 45729-39, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11577090

ABSTRACT

Macrophage-derived foam cells in atherosclerotic lesions are generally thought to play a major role in the pathology of the disease. Because macrophages play a central role in the inflammatory response, and the atherosclerotic lesion has features associated with chronic inflammatory settings, we investigated foam cell inflammatory potential. THP-1-derived macrophages were treated with oxidized low density lipoprotein (OxLDL) for 3 days to lipid load the macrophages and establish a foam cell-like phenotype. The cells were then activated by treatment with lipopolysaccharide (LPS), and RNA was harvested at 0, 1, and 6 h after LPS addition. RNA from treated and control cells was hybridized to microarrays containing approximately 16,000 human cDNAs. Genes that exhibited a 4-fold or greater increase or decrease at either 1 or 6 h after LPS treatment were counted as LPS-responsive genes. Employing these criteria, 127 LPS-responsive genes were identified. Prior treatment of THP-1 macrophages with OxLDL affected the expression of 57 of these 127 genes. Among these 57 genes was a group of chemokine, cytokine, and signal transduction genes with pronounced expression changes. OxLDL pretreatment resulted in a significant perturbation of LPS-induced NF kappa B activation. Furthermore, some of the OxLDL effects appear to be mediated by the nuclear receptors retinoid X receptor and peroxisomal proliferator-activated receptor gamma because pretreatment of THP-1 macrophages with ligands for these receptors, followed by LPS treatment, recapitulates the OxLDL plus LPS results for several of the most significantly modulated genes.


Subject(s)
Lipoproteins, LDL/physiology , Macrophages/physiology , Transcription, Genetic/physiology , Base Sequence , Cell Line , Cholesterol/metabolism , DNA Primers , Humans , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Signal Transduction/physiology
2.
J Lipid Res ; 42(8): 1173-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11483617

ABSTRACT

It is widely believed that HDL functions to transport cholesterol from peripheral cells to the liver by reverse cholesterol transport, a pathway that may protect against atherosclerosis by clearing excess cholesterol from arterial cells. A cellular ATP-binding cassette transporter (ABC) called ABCA1 mediates the first step of reverse cholesterol transport: the transfer of cellular cholesterol and phospholipids to lipid-poor apolipoproteins. Mutations in ABCA1 cause Tangier disease (TD), a severe HDL deficiency syndrome characterized by accumulation of cholesterol in tissue macrophages and prevalent atherosclerosis. Studies of TD heterozygotes revealed that ABCA1 activity is a major determinant of plasma HDL levels and susceptibility to CVD. Drugs that induce ABCA1 in mice increase clearance of cholesterol from tissues and inhibit intestinal absorption of dietary cholesterol. Multiple factors related to lipid metabolism and other processes modulate expression and tissue distribution of ABCA1.Therefore, as the primary gatekeeper for eliminating tissue cholesterol, ABCA1 has a major impact on cellular and whole body cholesterol metabolism and is likely to play an important role in protecting against cardiovascular disease.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Cholesterol/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Biological Transport , Cardiovascular Diseases/genetics , Gene Expression Regulation , Humans , Lipoproteins, HDL/blood , Mutation , Tangier Disease/genetics
4.
J Biol Chem ; 276(25): 22209-14, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11301336

ABSTRACT

Lp(a) concentrations vary considerably among individuals and are primarily determined by the apo(a) gene locus. We have previously shown that mean plasma Lp(a) levels in the chimpanzee are significantly higher than those observed in humans (Doucet, C., Huby, T., Chapman, J., and Thillet, J. (1994) J. Lipid Res 35, 263-270). To evaluate the possibility that this difference may result from a high level of expression of chimpanzee apo(a), we cloned and sequenced 1.4 kilobase (kb) of the 5'-flanking region of the gene and compared promoter activity to that of its human counterpart. Sequence analysis revealed 98% homology between chimpanzee and human apo(a) 5' sequences; among the differences observed, two involved polymorphic sites associated with Lp(a) levels in humans. The TTTTA repeat located 1.3 kb 5' of the apo(a) gene, present in a variable number of copies (n = 5-12) in humans, is uniquely present as four copies in the chimpanzee sequence. The second position concerns the +93 C>T polymorphism that creates an additional ATG start codon in the human apo(a) gene, thereby impairing translation efficiency. In chimpanzee, this position did not appear polymorphic, and a base difference at position +94 precluded the presence of an additional ATG. In transient transfection assays, the chimpanzee apo(a) promoter exhibited a 5-fold elevation in transcriptional activity as compared with its human counterpart. This marked difference in activity was maintained with either 1.4 kb of 5' sequence or the minimal promoter region -98 to +141 of the human and chimpanzee apo(a) genes. Using point mutational analyses, nucleotides present at positions -3, -2, and +8 (relative to the start site of transcription) were found to be essential for the high transcription efficiency of the chimpanzee apo(a) promoter. High transcriptional activity of the chimpanzee apo(a) gene may therefore represent a key factor in the elevated plasma Lp(a) levels characteristic of this non-human primate.


Subject(s)
Apolipoproteins/genetics , Lipoprotein(a)/genetics , Promoter Regions, Genetic , Transcription, Genetic/genetics , Animals , Apolipoproteins/physiology , Apoprotein(a) , Base Sequence , Cell Line , Electrophoresis, Polyacrylamide Gel , Humans , Lipoprotein(a)/physiology , Molecular Sequence Data , Mutagenesis, Site-Directed , Pan troglodytes , Sequence Homology, Nucleic Acid
5.
Arterioscler Thromb Vasc Biol ; 21(3): 378-85, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231917

ABSTRACT

The present study examines the expression of ATP-binding cassette transporter 1 (ABC1) mRNA in normal and atherosclerotic tissues by using in situ hybridization in an effort to better understand the function of this cholesterol transport protein. Samples of normal baboon tissues as well as human normal and atherosclerotic aortas were hybridized with (35)S-labeled ABC1 sense and antisense riboprobes. Widespread expression of ABC1 was observed generally in tissues containing inflammatory cells and lymphocytes. Other noninflammatory cells that were also sites of ABC1 synthesis included the ductal cells of the kidney medulla, Leydig cells in the testis, and glial cells in the baboon cerebellum. Although normal veins and arteries did not express ABC1 mRNA, it was found to be upregulated in the setting of atherosclerosis, where widespread expression was found in macrophages within atherosclerotic lesions. These results are consistent with the proposed role of ABC1 in cholesterol transport in inflammatory cells. The specific upregulation of ABC1 mRNA in the setting of atherosclerosis probably reflects the response of leukocytes to cholesterol loading. However, the presence of ABC1 in ductal cells of the kidney medulla and in the small intestine suggest a more general role for this protein in cholesterol transport in other cell types.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Arteriosclerosis/genetics , Glycoproteins/genetics , ATP Binding Cassette Transporter 1 , Aorta/metabolism , Aorta/pathology , Arteriosclerosis/pathology , Gene Expression , Humans , In Situ Hybridization , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Distribution
6.
J Biol Chem ; 276(5): 3158-66, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11073951

ABSTRACT

Efflux of excess cellular cholesterol mediated by lipid-poor apolipoproteins occurs by an active mechanism distinct from passive diffusion and is controlled by the ATP-binding cassette transporter ABCA1. Here we examined whether ABCA1-mediated lipid efflux involves the selective removal of lipids associated with membrane rafts, plasma membrane domains enriched in cholesterol and sphingomyelin. ABCA1 was not associated with cholesterol and sphingolipid-rich membrane raft domains based on detergent solubility and lack of colocalization with marker proteins associated with raft domains. Lipid efflux to apoA-I was accounted for by decreases in cellular lipids not associated with cholesterol/sphingomyelin-rich membranes. Treating cells with filipin, to disrupt raft structure, or with sphingomyelinase, to digest plasma membrane sphingomyelin, did not impair apoA-I-mediated cholesterol or phosphatidylcholine efflux. In contrast, efflux of cholesterol to high density lipoproteins (HDL) or plasma was partially accounted for by depletion of cholesterol from membrane rafts. Additionally, HDL-mediated cholesterol efflux was partially inhibited by filipin and sphingomyelinase treatment. Apo-A-I-mediated cholesterol efflux was absent from fibroblasts with nonfunctional ABCA1 (Tangier disease cells), despite near normal amounts of cholesterol associated with raft domains and normal abilities of plasma and HDL to deplete cholesterol from these domains. Thus, the involvement of membrane rafts in cholesterol efflux applies to lipidated HDL particles but not to lipid-free apoA-I. We conclude that cholesterol and sphingomyelin-rich membrane rafts do not provide lipid for efflux promoted by apolipoproteins through the ABCA1-mediated lipid secretory pathway and that ABCA1 is not associated with these domains.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Membrane Lipids/metabolism , Membrane Microdomains/metabolism , ATP Binding Cassette Transporter 1 , Anti-Bacterial Agents/pharmacology , Biological Transport , Cells, Cultured , Cholesterol/metabolism , Detergents/chemistry , Fibroblasts/metabolism , Filipin/pharmacology , Humans , Octoxynol/chemistry , Sphingomyelin Phosphodiesterase/pharmacology , Tangier Disease/pathology
7.
Arterioscler Thromb Vasc Biol ; 20(10): 2184-91, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11031202

ABSTRACT

Vascular injury induces extensive alteration to the extracellular matrix (ECM). These changes contribute to lesion formation and promote cell migration and proliferation. To elucidate ECM response to arterial injury, we used real-time polymerase chain reaction monitoring to quantitate the expression levels of 81 genes involved in the synthesis and breakdown of ECM as well as receptors and signaling proteins that communicate and respond to ECM molecules. The temporal regulation of gene expression in the carotid was measured at 1, 3, 5, 7, 9, 14, and 28 days postinjury. Among the 68 genes that showed detectable expression by our method, 47 (69%) were significantly induced or repressed over time, confirming the extensive ECM gene response in this model. More ECM-related genes (31) were regulated at day 1 than at any other time point, and the number of regulated genes decreased over time. However, 14 of the genes were still induced or repressed at day 28, indicating that return to preinjury expression patterns did not occur and no new steady state was achieved over 28 days. In spite of the large number of changes in gene expression, only a small number of expression patterns was observed, suggesting that ECM-related genes could potentially be coregulated.


Subject(s)
Carotid Artery Injuries/genetics , Animals , Carotid Arteries/metabolism , Carotid Artery Injuries/physiopathology , Cluster Analysis , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Gene Expression Profiling/methods , Gene Expression Regulation , Male , RNA, Messenger/analysis , RNA, Messenger/isolation & purification , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
8.
J Biol Chem ; 275(48): 37324-32, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-10973959

ABSTRACT

We conducted large scale gene expression analysis of the response of macrophages to exposure to oxidized low density lipoprotein (Ox-LDL). Much of the vessel wall lesion of atherosclerosis is composed of macrophages that have become engorged with cholesterol. These resulting "foam cells" contribute to the progression of vascular disease through several pathways. As a potential model of foam cell formation, we treated THP-1 cells with 12-O-tetradecanoylphorbol 13-acetate to differentiate them into a macrophage-like phenotype and subsequently treated them with oxidized low density lipoprotein for various time periods. RNA from Ox-LDL treated and time-matched control untreated cells was hybridized to microarrays containing 9808 human genes. 268 genes were found to be at least 2-fold regulated at one or more time points. These regulation patterns were classified into seven clusters of expression profiles. The data is discussed in terms of the overall pattern of gene expression, the thematic classification of the responding genes, and the clustering of functional groups in distinct expression patterns. The magnitude and the temporal patterns of gene expression identified known and novel molecular components of the cellular response that are implicated in the growth, survival, migratory, inflammatory, and matrix remodeling activity of vessel wall macrophages. In particular, the role of nuclear receptors in mediating the gene expression modulation by Ox-LDL is highlighted.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Cell Line , Humans , Oligonucleotide Array Sequence Analysis , Transcription, Genetic
9.
Biochem Biophys Res Commun ; 274(3): 794-802, 2000 Aug 11.
Article in English | MEDLINE | ID: mdl-10924356

ABSTRACT

ATP-binding cassette transporter 1 (ABC1) mediates the active efflux of cholesterol from cells to apolipoproteins. To study the mechanisms of regulation of ABC1 gene expression, RAW 264.7 macrophages were transiently transfected with ABC1 promoter-luciferase reporter gene-fusion constructs. Transcription from a 1.64 kb fragment was induced by cholesterol loading but was not responsive to cAMP. Treatment of the cells with 9-cis retinoic acid or 20(S)-hydroxycholesterol, ligands for the nuclear receptors LXR and RXR, resulted in a marked induction of luciferase expression. The responsible control element was mapped to an imperfect direct repeat of the nuclear receptor half-site TGACCT separated by four bases (DR-4) that binds LXR/RXR heterodimers. Endogenous ABC1 gene expression in RAW cells and apolipoprotein A-I mediated cholesterol efflux were also upregulated by both receptor ligands. These findings raise the possibility that ligands that activate the LXR-RXR heterodimer may be useful for the therapeutic modulation of the ABC1 pathway.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Apolipoprotein A-I/physiology , Cholesterol/metabolism , Glycoproteins/physiology , Macrophages/physiology , Receptors, Cytoplasmic and Nuclear/physiology , ATP Binding Cassette Transporter 1 , Animals , Base Sequence , Biological Transport/physiology , Cell Line , DNA-Binding Proteins , Genes, Reporter , Ligands , Liver X Receptors , Luciferases/genetics , Mice , Molecular Sequence Data , Orphan Nuclear Receptors , Promoter Regions, Genetic , Signal Transduction/physiology , Transfection
10.
J Biol Chem ; 275(44): 34508-11, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10918070

ABSTRACT

Lipid-poor high density lipoprotein apolipoproteins remove cholesterol and phospholipids from cells by an active secretory pathway controlled by an ABC transporter called ABCA1. This pathway is induced by cholesterol and cAMP analogs in a cell-specific manner. Here we provide evidence that increased plasma membrane ABCA1 accounts for the enhanced apolipoprotein-mediated lipid secretion from macrophages induced by cAMP analogs. Treatment of RAW264 macrophages with 8-bromo-cAMP caused parallel increases in apoA-I-mediated cholesterol efflux, ABCA1 mRNA and protein levels, incorporation of ABCA1 into the plasma membrane, and binding of apoA-I to cell-surface ABCA1. All of these parameters declined to near base-line values within 6 h after removal of 8-bromo-cAMP, indicating that ABCA1 is highly unstable and is degraded rapidly in the absence of inducer. Thus, ABCA1 is likely to be the cAMP-inducible apolipoprotein receptor that promotes removal of cholesterol and phospholipids from macrophages.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/metabolism , Cholesterol/metabolism , Cyclic AMP/metabolism , Macrophages/metabolism , Receptors, Lipoprotein/metabolism , ATP Binding Cassette Transporter 1 , Animals , Cell Line , Mice , Protein Binding , Receptors, Lipoprotein/biosynthesis
11.
Endothelium ; 7(4): 253-64, 2000.
Article in English | MEDLINE | ID: mdl-11201523

ABSTRACT

Elevated plasma level of lipoprotein(a) (Lp(a)) is a well established risk factor for premature atherosclerosis and coronary artery disease. Recent studies showed impaired endothelium-dependent vasodilatation in humans with elevated plasma Lp(a). However, these human studies could not determine whether (1) elevated Lp(a) levels alone are the cause of endothelial dysfunction (these patients had multiple risk factors), and (2) native or oxidatively modified Lp(a) contributes to endothelial dysfunction (no measurements of native/oxidized Lp(a) ratio was reported in humans). In order to test whether apo(a) (an essential component of Lp(a) which is required for binding to endothelial cells) and native Lp(a) cause endothelial dysfunction, in the present study we tested endothelium-dependent vasorelaxation in aortic rings isolated from control and transgenic male mice either expressing the human apo(a) gene (TgA) or both the human apo(a) and human apo B100 genes (TgL). The TgA mice had plasma apo(a) levels of 8.8 +/- 1.2 mg/dl (n=6) and the double transgenic TgL mice had plasma Lp(a) levels of 15.3 +/- 1.4 mg/dl (n=8). Isolated aortic rings with and without endothelium were mounted in organ chambers and contracted with U46619 (10(-8) M) in the presence of ibuprofen (10(-5) M). Acetylcholine caused concentration-dependent (10(-9)-10(-5) M) relaxation, which could be prevented by endothelium removal and by NG-L-nitro-arginine (10(-4) M). Basal and acetylcholine-stimulated endothelium-dependent relaxation and endothelium-independent relaxation to nitroglycerin (10(-6) M) were not significantly different in aortic rings isolated from control and TgA or TgL mice. Twenty-four hour incubation of aortic rings isolated from control mice with recombinant human apo(a) or native Lp(a) (up to 300 microg/ml) caused no impairment of endothelium-dependent relaxations. In contrast, incubation with oxidized Lp(a) (50 microg/ml) or oxidized LDL (250 microg/ml) caused significant suppression of acetylcholine-induced endothelium-dependent vasorelaxation. These results show for the first time that elevated plasma levels of apo(a) and Lp(a) do not cause endothelial dysfunction in transgenic mice.


Subject(s)
Aorta/physiopathology , Apolipoproteins/physiology , Endothelium, Vascular/physiopathology , Lipoprotein(a)/physiology , Vasodilation , Animals , Aorta/metabolism , Apolipoprotein B-100 , Apolipoproteins/genetics , Apolipoproteins B/genetics , Apolipoproteins B/physiology , Apoprotein(a) , Humans , In Vitro Techniques , Lipoprotein(a)/genetics , Lipoproteins, LDL/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide/metabolism
12.
Atherosclerosis ; 153(2): 349-54, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11164423

ABSTRACT

The aortic root from 21 LPA transgenic mice and 18 control litter mates on cholesterol enriched chow were studied histologically for the presence of atherosclerotic lesions. Serial sections were cut and the total area of the lesions was measured by use of computerised image analysis. Lipid staining lesions were found in 17 aortas of the transgenic mice and were five times more common than in the controls. Foam cell lesions were the only type of lesion in 12 of the aortas from transgenic animals, while five animals had developed fibrofatty lesions. Immunostaining revealed monocytes/macrophages on the endothelial surface, and in the subendothelial space of foam cell lesions. In fibrofatty lesions, spindle shaped cells formed a cap around the lipid core. This study supports the view that transgenic mice expressing human apolipoprotein (a) on a high fat and cholesterol diet, are more susceptible to aortic lesions than control mice and develop early atherosclerotic lesions comparable to lesions in man. Aminoguanidin in the drinking water had no effect on the aortic lesions, but lesion size was significantly, negatively correlated with plasma glucose concentration.


Subject(s)
Arteriosclerosis/genetics , Arteriosclerosis/pathology , Lipoprotein(a)/genetics , Animals , Arteriosclerosis/metabolism , Cholesterol, Dietary/administration & dosage , Gene Expression Regulation , Humans , Lipoprotein(a)/metabolism , Mice , Mice, Transgenic
13.
J Clin Invest ; 104(8): R25-31, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10525055

ABSTRACT

The ABC1 transporter was identified as the defect in Tangier disease by a combined strategy of gene expression microarray analysis, genetic mapping, and biochemical studies. Patients with Tangier disease have a defect in cellular cholesterol removal, which results in near zero plasma levels of HDL and in massive tissue deposition of cholesteryl esters. Blocking the expression or activity of ABC1 reduces apolipoprotein-mediated lipid efflux from cultured cells, and increasing expression of ABC1 enhances it. ABC1 expression is induced by cholesterol loading and cAMP treatment and is reduced upon subsequent cholesterol removal by apolipoproteins. The protein is incorporated into the plasma membrane in proportion to its level of expression. Different mutations were detected in the ABC1 gene of 3 unrelated patients. Thus, ABC1 has the properties of a key protein in the cellular lipid removal pathway, as emphasized by the consequences of its defect in patients with Tangier disease.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Apolipoproteins/physiology , Glycoproteins/physiology , Lipid Metabolism , Tangier Disease/genetics , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Adolescent , Animals , Cells, Cultured , Gene Expression Regulation , Glycoproteins/genetics , Humans , Male , Mice , Mutation , Tangier Disease/complications
14.
Blood ; 94(6): 2007-19, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10477730

ABSTRACT

Retention of lipoproteins within the vasculature is a central event in the pathogenesis of atherosclerosis. However, the signals that mediate this process are only partially understood. Prompted by putative links between inflammation and atherosclerosis, we previously reported that alpha-defensins released by neutrophils are present in human atherosclerotic lesions and promote the binding of lipoprotein(a) [Lp(a)] to vascular cells without a concomitant increase in degradation. We have now tested the hypothesis that this accumulation results from the propensity of defensin to form stable complexes with Lp(a) that divert the lipoprotein from its normal cellular degradative pathways to the extracellular matrix (ECM). In accord with this hypothesis, defensin stimulated the binding of Lp(a) to vascular matrices approximately 40-fold and binding of the reactants to the matrix was essentially irreversible. Defensin formed stable, multivalent complexes with Lp(a) and with its components, apoprotein (a) and low-density lipoprotein (LDL), as assessed by optical biosensor analysis, gel filtration, and immunoelectron microscopy. Binding of defensin/Lp(a) complexes to matrix was inhibited (>90%) by heparin and by antibodies to fibronectin (>70%), but not by antibodies to vitronectin or thrombospondin. Defensin increased the binding of Lp(a) (10 nmol/L) to purified fibronectin more than 30-fold. Whereas defensin and Lp(a) readily traversed the endothelial cell membranes individually, defensin/Lp(a) complexes lodged on the cell surface. These studies demonstrate that alpha-defensins released from activated or senescent neutrophils stimulate the binding of an atherogenic lipoprotein to the ECM of endothelial cells, a process that may contribute to lipoprotein accumulation in atherosclerotic lesions.


Subject(s)
Endothelium, Vascular/physiology , Extracellular Matrix/physiology , Lipoprotein(a)/blood , Proteins/physiology , Apolipoproteins/blood , Apoprotein(a) , Biosensing Techniques , Blood Proteins/physiology , Blood Proteins/ultrastructure , Cells, Cultured , Defensins , Extracellular Matrix/ultrastructure , Humans , Kinetics , Lipoprotein(a)/ultrastructure , Lipoproteins, LDL/blood , Microscopy, Immunoelectron , Protein Binding , Proteins/ultrastructure , Umbilical Veins
15.
J Biol Chem ; 274(22): 15569-74, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10336452

ABSTRACT

Elevated plasma levels of the lipoprotein particle Lp(a) are a major risk factor for cardiovascular disease. Lp(a) plasma levels are determined by the level of expression of its characteristic protein component, apo(a). Apo(a) expression is modulated by several hormones, of which estrogens are the best known. The chromosomal region responsible for estrogen response was identified within an apo(a) enhancer located at approximately 26 kilobases from the apo(a) promoter. Although the estrogen-responsive unit contains a potential estrogen response element, binding of estrogen receptor-alpha to DNA was not necessary. The receptor, activated by bound estradiol, interacts through its transactivation domains with a transcription factor necessary for the function of the enhancer, preventing its binding to DNA.


Subject(s)
Apolipoproteins A/genetics , Estrogens/pharmacology , Base Sequence , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Estradiol/metabolism , Estrogen Receptor alpha , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Molecular Sequence Data , Plasminogen/genetics , Promoter Regions, Genetic , Protein Binding , Receptors, Estrogen/genetics , Regulatory Sequences, Nucleic Acid , Transcriptional Activation/genetics , Transfection , Tumor Cells, Cultured
16.
Biochem Biophys Res Commun ; 255(3): 639-44, 1999 Feb 24.
Article in English | MEDLINE | ID: mdl-10049763

ABSTRACT

The study of human lipoprotein (a) [Lp(a)] has been hampered due to the lack of appropriate animal models since apolipoprotein (a) [apo(a)] is found only in primates and humans. In addition, human apo(a) in transgenic mice can not bind to murine apoB to form Lp(a) particles. In this study, we generated three independent transgenic rabbits expressing human apo(a) in their plasma at 1.8-4.5 mg/dl. In the plasma of transgenic rabbits, unlike the plasma of transgenic mice, about 80% of the apo(a) was covalently associated with rabbit apo-B and was contained in the fractions with density 1.02-1.10 g/ml, indicating the formation of Lp(a). These results suggest that transgenic rabbits expressing human apo(a) exhibit efficient assembly of Lp(a) and can be used as an animal model for the study of human Lp(a).


Subject(s)
Apolipoproteins B/metabolism , Lipoprotein(a)/genetics , Animals , Animals, Genetically Modified , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Female , Humans , Immunohistochemistry , In Situ Hybridization , Lipoprotein(a)/metabolism , Lipoproteins/metabolism , Liver/metabolism , Male , RNA, Messenger/genetics , Rabbits
17.
Exp Mol Pathol ; 65(2): 53-63, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9828147

ABSTRACT

Apolipoprotein(a) [apo(a)] contains a kringle domain(IV) homologous to that of angiostatin, a natural angiogenic inhibitor. Because of this structural similarity we suspected that apo(a) could be an inhibitor of angiogenesis. The possible role of apo(a) in microvascular proliferation was studied in an in vivo quantitative model, the disc angiogenesis system (DAS) and compared to angiostatin. Apo(a) and other test compounds were placed in the center of a polyvinyl alcohol foam disc that was implanted subcutaneously in mice. After 14 days, the disc was removed and vascular growth into the disc was measured. Apo(a) did not affect spontaneous vessel growth into the disc, while angiostatin suppressed this growth and basic fibroblast growth factor (bFGF) increased it. Additionally, apo(a) did not modify the vascular growth induced by bFGF. Transgenic mice expressing the human apo(a) gene were used to study the systemic effect of apo(a): neither an increase nor a decrease in vascular growth was detected. Our results suggest that apo(a) is unlikely to play a significant role in the control of angiogenesis. Furthermore, our experiments confirm the inhibitory effect of angiostatin not only on induced angiogenesis but also on baseline, spontaneous angiogenesis.


Subject(s)
Apolipoproteins A/pharmacology , Neovascularization, Pathologic/prevention & control , Peptide Fragments/pharmacology , Plasminogen/pharmacology , Angiostatins , Animals , Apolipoproteins A/genetics , Capillaries/drug effects , Capillaries/pathology , Connective Tissue/blood supply , Connective Tissue/drug effects , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/drug effects , Gene Deletion , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/pathology , Skin/blood supply
18.
Proc Natl Acad Sci U S A ; 95(21): 12591-5, 1998 Oct 13.
Article in English | MEDLINE | ID: mdl-9770530

ABSTRACT

To test directly whether fibrin(ogen) is a key binding site for apolipoprotein(a) [apo(a)] in vessel walls, apo(a) transgenic mice and fibrinogen knockout mice were crossed to generate fibrin(ogen)-deficient apo(a) transgenic mice and control mice. In the vessel wall of apo(a) transgenic mice, fibrin(ogen) deposition was found to be essentially colocalized with focal apo(a) deposition and fatty-streak type atherosclerotic lesions. Fibrinogen deficiency in apo(a) transgenic mice decreased the average accumulation of apo(a) in vessel walls by 78% and the average lesion (fatty streak type) development by 81%. Fibrinogen deficiency in wild-type mice did not significantly reduce lesion development. Our results suggest that fibrin(ogen) provides one of the major sites to which apo(a) binds to the vessel wall and participates in the generation of atherosclerosis.


Subject(s)
Afibrinogenemia/metabolism , Aorta/metabolism , Apolipoproteins/metabolism , Arteriosclerosis/metabolism , Lipoprotein(a) , Animals , Apolipoproteins/genetics , Apoprotein(a) , Mice , Mice, Inbred C57BL , Mice, Transgenic
19.
Arterioscler Thromb Vasc Biol ; 17(9): 1741-5, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9327772

ABSTRACT

Plasma levels of lipoprotein(a) (Lp(a)), are regulated by the synthetic rate of apolipoprotein(a) (apo(a)), a major protein component of this atherogenic lipoprotein. Exogenously administered sex steroid hormones are potent regulators of plasma Lp(a) concentrations. We utilized a recently developed apo(a) yeast artificial chromosome (YAC) transgenic mouse model to study the effects of ovariectomy, estrus cycle, and exogenous administration of ethinyl-estradiol, the partial estrogen receptor agonist, tamoxifen, and progesterone on circulating apo(a) plasma levels. Analysis of liver RNA revealed that estrogen and tamoxifen exerts their plasma apo(a) lowering effect at the level of apo(a) mRNA. This action of estrogen and tamoxifen may contribute to their antiatherosclerotic and cardiovascular protective effect.


Subject(s)
Apolipoproteins/genetics , Estradiol Congeners/pharmacology , Estrogen Antagonists/pharmacology , Estrus/physiology , Ethinyl Estradiol/pharmacology , Gene Expression/drug effects , Lipoprotein(a) , Progesterone/pharmacology , Tamoxifen/pharmacology , Animals , Apolipoproteins/blood , Apoprotein(a) , Drug Combinations , Female , Gene Expression/physiology , Liver/metabolism , Mice , Mice, Transgenic/genetics , Ovariectomy , RNA, Messenger/metabolism
20.
Proc Natl Acad Sci U S A ; 94(22): 11992-7, 1997 Oct 28.
Article in English | MEDLINE | ID: mdl-9342350

ABSTRACT

Apolipoprotein(a) [apo(a)] is the distinguishing protein component of lipoprotein(a), a major inherited risk factor for atherosclerosis. Human apo(a) is homologous to plasminogen. It contains from 15 to 50 repeated domains closely related to plasminogen kringle four, plus single kringle five-like and inactive protease-like domains. This expressed gene is confined to a subset of primates. Although most mammals lack apo(a), hedgehogs produce an apo(a)-like protein composed of highly repeated copies of a plasminogen kringle three-like domain, with complete absence of protease domain sequences. Both human and hedgehog apo(a)-like proteins form covalently linked lipoprotein particles that can bind to fibrin and other substrates shared with plasminogen. DNA sequence comparisons and phylogenetic analysis indicate that the human type of apo(a) evolved from a duplicated plasminogen gene during recent primate evolution. In contrast, the kringle three-based type of apo(a) evolved from an independent duplication of the plasminogen gene approximately 80 million years ago. In a type of convergent evolution, the plasminogen gene has been independently remodeled twice during mammalian evolution to produce similar forms of apo(a) in two widely divergent groups of species.


Subject(s)
Apolipoproteins A/genetics , Evolution, Molecular , Haplorhini/genetics , Hedgehogs/genetics , Macropodidae/genetics , Plasminogen/genetics , Animals , Cattle , Hominidae/genetics , Humans , Kringles , Macaca mulatta/genetics , Mice , Models, Genetic , Molecular Sequence Data , Multigene Family
SELECTION OF CITATIONS
SEARCH DETAIL
...