Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 276(33): 30655-61, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11389145

ABSTRACT

Upon activation, platelets secrete a 120-kDa protein that competes for the binding and internalization of acetyl low density lipoproteins (AcLDL) by macrophages. From the amino-terminal amino acid sequence, amino acid composition, and immunoblot analysis, we identified the active factor in platelet secretion products as sAPP, an alpha-secretase cleavage product of the beta-amyloid precursor protein (APP), that contains a Kunitz-type protease inhibitor (KPI) domain. We showed that both sAPP751 (also called Nexin II) and sAPP695, which does not contain a KPI domain, are ligands for the class A scavenger receptor (SR-A). Chinese hamster ovary cells stably transfected to express the SR-A bound and internalized 4-fold more human platelet-derived sAPP than control cells. The binding and internalization of sAPP were inhibited by the SR-A antagonist fucoidin. In addition, sAPP competed as effectively as fucoidin for SR-A-mediated cell association and degradation of (125)I-AcLDL. To determine if the KPI domain is required for the binding of sAPP to the SR-A, APP751 and APP695 were expressed in Chinese hamster ovary cells, and sAPP751 and sAPP695 purified from the medium were tested for their binding to the SR-A. sAPP751 and sAPP695 were equally effective in competing for the cell association of (125)I-AcLDL by SR-A-expressing cells, demonstrating that the KPI domain is not essential for binding. We also found that sAPP751 is present in extracts of atherosclerotic lesions and that sAPP competes for the SR-A-mediated cell association of oxidized low density lipoprotein. Deletion mutagenesis indicated that a negatively charged region of APP (residues 191-264) contributes to binding to the SR-A. These results suggest that the SR-A contributes to the clearance of sAPP and that sAPP competes for the cell association of other SR-A ligands.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Blood Platelets/metabolism , Membrane Proteins , Proteoglycans/metabolism , Receptors, Immunologic/metabolism , Receptors, Lipoprotein , Amino Acid Sequence , Amyloid beta-Protein Precursor/chemistry , Animals , Humans , Ligands , Molecular Sequence Data , Rabbits , Receptors, Scavenger , Scavenger Receptors, Class A , Scavenger Receptors, Class B
2.
J Biol Chem ; 275(23): 17661-70, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10837497

ABSTRACT

Oxidative stress caused by phorbol esters or reactive oxygen up-regulates the class A scavenger receptor (SR-A) in human smooth muscle cells (SMC), which normally do not express this receptor. The increase in SR-A expression correlates with activation of the redox-sensitive transcription factors activating protein-1 c-Jun and CCAAT enhancer-binding protein beta. Here we show that coincubation of SMC with macrophages or oxidized low density lipoproteins (LDL) from macrophage-conditioned medium activates these same regulatory pathways and stimulates SR-A expression. The increased SR-A gene transcription induced by cell-oxidized LDL up-regulated SR-A mRNA and increased by 30-fold the uptake of acetyl LDL, a ligand for the SR-A. Copper-oxidized LDL also increased SR-A receptor expression. Oxidized LDL with a lipid peroxide level of 80-100 nmol/mg of LDL protein and an electrophoretic mobility approximately 1.5 times that of native LDL exhibited the greatest bioactivity. Inhibition of calcium flux suppressed SR-A induction by oxidized LDL. Conversely, calcium ionophore greatly enhanced SR-A up-regulation by oxidized LDL or other treatments that promote intracellular oxidative stress. This enhancement was dependent upon concurrent up-regulation of SMC cyclooxygenase-2 expression and activity and was blocked by the cyclooxygenase-2 inhibitors NS-398 and Resveratrol. In THP-1 cells, oxidized LDL induced monocyte-to-macrophage differentiation and increased SR-A expression. These findings support a role for mildly oxidized LDL in the redox regulation of macrophage differentiation and SR-A expression and suggest that increased vascular oxidative stress may contribute to the formation of both SMC and macrophage foam cells.


Subject(s)
Calcium/physiology , Gene Expression Regulation, Enzymologic/drug effects , Isoenzymes/genetics , Lipoproteins, LDL/pharmacology , Muscle, Smooth/physiology , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Immunologic/genetics , Transcription, Genetic/drug effects , Cell Differentiation , Cell Line , Cell Nucleus/metabolism , Culture Media, Conditioned , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Kinetics , Macrophages/cytology , Macrophages/drug effects , Membrane Proteins , Mitogen-Activated Protein Kinases/metabolism , Monocytes/cytology , Monocytes/drug effects , Nitrobenzenes/pharmacology , RNA, Messenger/genetics , Receptors, Scavenger , Resveratrol , Scavenger Receptors, Class A , Stilbenes/pharmacology , Sulfonamides/pharmacology , Transfection , Up-Regulation/drug effects
3.
Neuroscience ; 97(2): 207-10, 2000.
Article in English | MEDLINE | ID: mdl-10799751

ABSTRACT

Apolipoprotein E fulfills fundamental functions in lipid transport and neural tissue repair after injury.(6,8) Its three most common isoforms (E2, E3, and E4) are critical determinants of diverse human diseases, including major cardiovascular and neurodegenerative disorders.(8,14) Apolipoprotein E4 is associated with an increased risk for Alzheimer's disease(3,5) and poor clinical outcome after head injury or stroke.(11,16) The precise role of apolipoprotein E4 in these conditions remains unknown. To characterize the effects of human apolipoprotein E isoforms in vivo, we analysed transgenic Apoe knockout mice that express apolipoprotein E3 or E4 or both in the brain. Hemizygous and homozygous apolipoprotein E3 mice were protected against age-related and excitotoxin-induced neurodegeneration, whereas apolipoprotein E4 mice were not. Apolipoprotein E3/E4 bigenic mice were as susceptible to neurodegeneration as apolipoprotein E4 singly-transgenic mice. At eight months of age neurodegeneration was more severe in homozygous than in hemizygous apolipoprotein E4 mice consistent with a dose effect. Thus, apolipoprotein E4 is not only less neuroprotective than apolipoprotein E3 but also acts as a dominant negative factor that interferes with the beneficial function of apolipoprotein E3. The inhibition of this apolipoprotein E4 activity may be critical for the prevention and treatment of neurodegeneration in APOE varepsilon4 carriers.


Subject(s)
Apolipoproteins E/genetics , Brain/metabolism , Neurodegenerative Diseases/genetics , Alzheimer Disease/genetics , Animals , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/deficiency , Apolipoproteins E/physiology , Apolipoproteins E/toxicity , Axons/pathology , Brain/pathology , Dendrites/pathology , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/analysis , Neurodegenerative Diseases/pathology , Neuroprotective Agents , Presynaptic Terminals/pathology , Synaptophysin/analysis
5.
Am J Pathol ; 155(5): 1741-7, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10550330

ABSTRACT

The class A scavenger receptor (SR) is expressed on reactive microglia surrounding cerebral amyloid plaques in Alzheimer's disease (AD). Interactions between the SR and amyloid beta peptides (Abeta) in microglial cultures elicit phagocytosis of Abeta aggregates and release of neurotoxins. To assess the role of the SR in amyloid clearance and Abeta-associated neurodegeneration in vivo, we used the platelet-derived growth factor promoter to express human amyloid protein precursors (hAPPs) in neurons of transgenic mice. With increasing age, hAPP mice develop AD-like amyloid plaques. We bred heterozygous hAPP (hAPP(+/-)) mice that were wild type for SR (SR(+/+)) with SR knockout (SR(-/-)) mice. Crosses among the resulting hAPP(+/-)SR(+/-) offspring yielded hAPP(+/-) and hAPP(-/-) littermates that were SR(+/+) or SR(-/-). These second-generation mice were analyzed at 6 and 12 months of age for extent of cerebral amyloid deposition and loss of synaptophysin-immunoreactive presynaptic terminals. hAPP(-/-)SR(-/-) mice showed no lack of SR expression, plaque formation, or synaptic degeneration, indicating that lack of SR expression does not result in significant accumulation of endogenous amyloidogenic or neurotoxic factors. In hAPP(+/-) mice, ablation of SR expression did not alter number, extent, distribution, or age-dependent accumulation of plaques; nor did it affect synaptic degeneration. Our results do not support a critical pathogenic role for microglial SR expression in neurodegenerative alterations associated with cerebral beta amyloidosis.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Membrane Proteins , Plaque, Amyloid/pathology , Receptors, Immunologic/physiology , Receptors, Lipoprotein , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/physiology , Animals , Gene Expression Regulation/physiology , Humans , Mice , Mice, Transgenic , Receptors, Scavenger , Scavenger Receptors, Class A , Scavenger Receptors, Class B
6.
J Neurosci ; 19(12): 4867-80, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10366621

ABSTRACT

Apolipoprotein (apo) E isoforms are key determinants of susceptibility to Alzheimer's disease. The apoE4 isoform is the major known genetic risk factor for this disease and is also associated with poor outcome after acute head trauma or stroke. To test the hypothesis that apoE3, but not apoE4, protects against age-related and excitotoxin-induced neurodegeneration, we analyzed apoE knockout (Apoe-/-) mice expressing similar levels of human apoE3 or apoE4 in the brain under control of the neuron-specific enolase promoter. Neuronal apoE expression was widespread in the brains of these mice. Kainic acid-challenged wild-type or Apoe-/- mice had a significant loss of synaptophysin-positive presynaptic terminals and microtubule-associated protein 2-positive neuronal dendrites in the neocortex and hippocampus, and a disruption of neurofilament-positive axons in the hippocampus. Expression of apoE3, but not of apoE4, protected against this excitotoxin-induced neuronal damage. ApoE3, but not apoE4, also protected against the age-dependent neurodegeneration seen in Apoe-/- mice. These differences in the effects of apoE isoforms on neuronal integrity may relate to the increased risk of Alzheimer's disease and to the poor outcome after head trauma and stroke associated with apoE4 in humans.


Subject(s)
Apolipoproteins E/genetics , Brain Chemistry/genetics , Nerve Degeneration/genetics , Aging/metabolism , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Animals , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/analysis , Apolipoproteins E/cerebrospinal fluid , Dendrites/chemistry , Dendrites/metabolism , Enzyme-Linked Immunosorbent Assay , Excitatory Amino Acid Agonists , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Genotype , Humans , Kainic Acid , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/chemically induced , Neurotoxins/metabolism , Presynaptic Terminals/chemistry , Presynaptic Terminals/metabolism , RNA, Messenger/analysis , Synaptophysin/analysis
7.
Arterioscler Thromb Vasc Biol ; 18(9): 1440-9, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9743233

ABSTRACT

Reactive oxygen species generated by treatment of smooth muscle cells (SMCs) with either phorbol 12-myristate 13-acetate or with the combination of H2O2 and vanadate strongly induce expression of the class A scavenger receptor (SR-A) gene. In the current studies, cis-acting elements in the proximal 245 bp of the SR-A promoter were shown to direct luciferase reporter expression in response to oxidative stress in both SMCs and macrophages. A composite activating protein-1 (AP-1)/ets binding element located between -67 and -50 bp relative to the transcriptional start site is critical for macrophage SR-A activity. Mutation of either the AP-1 or the ets component of this site also prevented promoter activity in SMCs. Mutation of a second site located between -44 and -21 bp, which we have identified as a CCAAT/enhancer binding protein (C/EBP) element, reduced the inducible activity of the promoter in SMCs by 50%, suggesting that combinatorial interactions between these sites are necessary for optimal gene induction. Interactions between SMC nuclear extracts and the SR-A promoter were analyzed by electrophoretic mobility shift assay. c-Jun/AP-1 binding activity, specific for the -67- to -50-bp site, was induced in SMCs by the same conditions that increased SR-A expression. Moreover, phorbol 12-myristate 13-acetate, H2O2, or the combination of H2O2 and sodium orthovanadate (vanadate) activated c-Jun-activating kinase. The binding activity within SMC extracts specific for the C/EBP site was shown to be C/EBPbeta in SMCs. Taken together, these findings demonstrate that reactive oxygen species regulate the interactions between c-Jun/AP-1 and C/EBPbeta in the SR-A promoter. Furthermore, induction of oxidative stress in THP-1 cells, with a combination of 10 micromol/L vanadate and 100 micromol/L H2O2, induced macrophage differentiation, adhesion, and SR activity. These data suggest that vascular oxidative stress may contribute to the induction of SR-A expression and thereby promote the uptake of oxidatively modified low density lipoprotein by both macrophage and SMCs to produce foam cells in atherosclerotic lesions.


Subject(s)
DNA-Binding Proteins/physiology , JNK Mitogen-Activated Protein Kinases , Membrane Proteins , Mitogen-Activated Protein Kinase Kinases , Muscle, Smooth/metabolism , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-jun/physiology , Receptors, Immunologic/genetics , Receptors, Lipoprotein , Transcription Factor AP-1/metabolism , Transcription, Genetic , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins , Cell Line , DNA/genetics , DNA/metabolism , Gene Expression/drug effects , Humans , Hydrogen Peroxide/pharmacology , MAP Kinase Kinase 4 , Macrophages/metabolism , Mutation , Oxidative Stress , Promoter Regions, Genetic , Protein Kinases/metabolism , Rabbits , Receptors, Scavenger , Scavenger Receptors, Class A , Scavenger Receptors, Class B , Tetradecanoylphorbol Acetate/pharmacology
8.
Proc Natl Acad Sci U S A ; 95(18): 10914-9, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9724804

ABSTRACT

Apolipoprotein E (apoE) mediates the redistribution of lipids among cells and is expressed at highest levels in brain and liver. Human apoE exists in three major isoforms encoded by distinct alleles (epsilon2, epsilon3, and epsilon4). Compared with APOE epsilon2 and epsilon3, APOE epsilon4 increases the risk of cognitive impairments, lowers the age of onset of Alzheimer's disease (AD), and decreases the response to AD treatments. Besides age, inheritance of the APOE epsilon4 allele is the most important known risk factor for the development of sporadic AD, the most common form of this illness. Although numerous hypotheses have been advanced, it remains unclear how APOE epsilon4 might affect cognition and increase AD risk. To assess the effects of distinct human apoE isoforms on the brain, we have used the neuron-specific enolase (NSE) promoter to express human apoE3 or apoE4 at similar levels in neurons of transgenic mice lacking endogenous mouse apoE. Compared with NSE-apoE3 mice and wild-type controls, NSE-apoE4 mice showed impairments in learning a water maze task and in vertical exploratory behavior that increased with age and were seen primarily in females. These findings demonstrate that human apoE isoforms have differential effects on brain function in vivo and that the susceptibility to apoE4-induced deficits is critically influenced by age and gender. These results could be pertinent to cognitive impairments observed in human APOE epsilon4 carriers. NSE-apoE mice and similar models may facilitate the preclinical assessment of treatments for apoE-related cognitive deficits.


Subject(s)
Apolipoproteins E/physiology , Neurons/metabolism , Age Factors , Alleles , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Avoidance Learning , Exploratory Behavior , Female , Humans , Immunohistochemistry , Maze Learning , Mice , Mice, Knockout , Mice, Transgenic , Neurons/enzymology , Phosphopyruvate Hydratase/genetics , Sex Factors
10.
J Biol Chem ; 273(22): 13452-60, 1998 May 29.
Article in English | MEDLINE | ID: mdl-9593678

ABSTRACT

Isoform-specific effects of apolipoprotein E (apoE) on neurite outgrowth and the cytoskeleton are associated with higher intracellular levels of apoE3 than apoE4 in cultured neurons. The current studies, designed to determine the mechanism for the differential intracellular accumulation or retention of apoE, demonstrate that apoE3- and apoE4-containing beta-very low density lipoproteins (beta-VLDL) possess similar cell binding and internalization and delivery of cholesterol to the cells. However, as assessed by immunocytochemistry, analysis of extracted cellular proteins, or quantitation of 125I-apoE-enriched beta-VLDL, there was a 2-3-fold greater accumulation of apoE3 than apoE4 in Neuro-2a cells, fibroblasts, and hepatocytes (HepG2) after 1-2 h, and this differential was maintained for up to 48 h. ApoE2 also accumulated in Neuro-2a cells to a greater extent than apoE4. The differential effect was mediated by the apoE-enriched beta-VLDL and not by free apoE. Neither the low density lipoprotein receptor nor the low density lipoprotein receptor-related protein was responsible for the differential accumulation of apoE3 and apoE4, since cells deficient in either or both of these receptors also displayed the differential accumulation. The effect appears to be mediated primarily by cell surface heparan sulfate proteoglycans (HSPG). The retention of both apoE3 and apoE4 was markedly reduced, and the differential accumulation of apoE3 and apoE4 was eliminated both in mutant Chinese hamster ovary cells that did not express HSPG and in HSPG-expressing cells treated with heparinase. The data suggest that cell surface HSPG directly mediate the uptake of apoE-containing lipoproteins, that the differential accumulation/retention of apoE by cells is mediated via HSPG, and that there is a differential intracellular handling of the specific apoE isoforms.


Subject(s)
Apolipoproteins E/metabolism , Heparan Sulfate Proteoglycans/metabolism , Animals , CHO Cells , Cell Membrane/metabolism , Cricetinae , Endocytosis , Iodine Radioisotopes , Lipoproteins, VLDL/metabolism , Mice , Tumor Cells, Cultured
11.
Proc Natl Acad Sci U S A ; 95(13): 7585-90, 1998 Jun 23.
Article in English | MEDLINE | ID: mdl-9636193

ABSTRACT

Apolipoprotein E- (apoE) deficient (E-/-) mice develop severe hyperlipidemia and diffuse atherosclerosis. Low-dose expression of a human apoE3 transgene in macrophages of apoE-deficient mice (E-/-hTgE+/0), which results in about 5% of wild-type apoE plasma levels, did not correct hyperlipidemia but significantly reduced the extent of atherosclerotic lesions. To investigate the contribution of apoE to reverse cholesterol transport, we compared plasmas of wild-type (E+/+), E-/-, and E-/-hTgE+/0 mice for the appearance of apoE-containing lipoproteins by electrophoresis and their capacity to take up and esterify 3H-labeled cholesterol from radiolabeled fibroblasts or J774 macrophages. Wild-type plasma displayed lipoproteins containing apoE that were the size of high density lipoprotein and that had either electrophoretic alpha or gamma mobilities. Similar particles were also present in E-/-hTgE+/0 plasma. Depending on incubation time, E-/- plasma released 48-74% less 3H-labeled cholesterol from fibroblasts than E+/+ plasma, whereas cholesterol efflux into E-/-hTgE+/0 plasma was only 11-25% lower than into E+/+ plasma. E-/-hTgE+/0 plasma also released 10% more 3H-labeled cholesterol from radiolabeled J774 macrophages than E-/- plasma. E+/+ and E-/-hTgE+/0 plasma each esterified significantly more cell-derived 3H-labeled cholesterol than E-/- plasma. Moreover, E-/- plasma accumulated much smaller proportions of fibroblast-derived 3H-labeled cholesterol in fractions with electrophoretic gamma and alpha mobility than E+/+ and E-/-hTgE+/0 plasma. Thus, low-dose expression of apoE in macrophages nearly restored the cholesterol efflux capacity of apoE-deficient plasma through the formation of apoE-containing particles, which efficiently take up cell-derived cholesterol, and through the increase of cholesterol esterification activity. Thus, macrophage-derived apoE may protect against atherosclerosis by increasing cholesterol efflux from arterial wall cells.


Subject(s)
Apolipoproteins E/genetics , Cholesterol/blood , Gene Expression , Macrophages/metabolism , Animals , Apolipoproteins E/blood , Cells, Cultured , Cholesterol Esters/blood , Electrophoresis, Agar Gel , Electrophoresis, Gel, Two-Dimensional , Fibroblasts/metabolism , Humans , Lipids/blood , Lipoproteins/blood , Mice , Mice, Inbred C57BL , Transgenes
12.
J Lipid Res ; 38(11): 2207-15, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9392418

ABSTRACT

Peritoneal macrophages are easily isolated by lavage, suggesting that they are either nonadherent or weakly adherent in situ. Cultured macrophages express class A scavenger receptors (SCR), which mediate Ca2+-independent adhesion in vitro. We examined fresh peritoneal macrophages from mice and from women with endometriosis to determine whether the adherence of these cells was associated with increased expression of class A SCR. Fresh human macrophages were not immunoreactive to SCR antibodies; however, SCR immunoreactivity increased with time in culture. Fresh mouse and human macrophages took up minimal amounts of 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine (DiI)-acetyl-low density lipoproteins (Ac-LDL), a class A SCR ligand. Murine macrophages in culture for 24-72 h internalized four times more Ac-LDL than fresh cells. Cells cultured for 2 days incorporated 3.2 times more [14C] oleate than freshly isolated cells (55.7 +/- 7.9 versus 17.6 +/- 3.0 nmol/mg cell protein). In contrast to SCR activity, mouse macrophage SCR mRNA expression was similar in freshly isolated macrophages and those cultured for 3 days. These results suggest that peritoneal macrophages express only low levels of SCR activity in situ and that posttranscriptional regulation after isolation leads to an increase in SCR activity that correlates with adherence of the macrophages in vitro.


Subject(s)
Macrophages, Peritoneal/metabolism , Membrane Proteins , Receptors, Immunologic/metabolism , Receptors, Lipoprotein , Animals , Blotting, Northern , Carbocyanines/metabolism , Cell Adhesion/physiology , Cells, Cultured , Cycloheximide/pharmacology , Female , Fluorescent Dyes/metabolism , Humans , Lipoproteins, LDL/metabolism , Macrophages, Peritoneal/cytology , Mice , Microscopy, Fluorescence , Oleic Acid/metabolism , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/analysis , Receptors, Immunologic/biosynthesis , Receptors, Scavenger , Scavenger Receptors, Class A , Scavenger Receptors, Class B , Time Factors
13.
Arterioscler Thromb Vasc Biol ; 17(5): 969-78, 1997 May.
Article in English | MEDLINE | ID: mdl-9157963

ABSTRACT

Phorbol esters increase scavenger-receptor mRNA expression and receptor activity in smooth muscle cells (SMCs). Our present results demonstrate that activation of protein kinase C (PKC) mediates this increase in receptor expression. This conclusion is based on the findings that (1) phorbol esters induced translocation of PKC-alpha from the cytosol to the membrane fraction; (2) PKC inhibitors blocked the effect of phorbol esters on receptor expression; (3) diacylglycerol, a physiological PKC agonist, enhanced scavenger-receptor activity; and (4) in cotransfected human SMCs, constitutively active PKC-alpha stimulated the expression of a reporter gene under control of the scavenger-receptor promoter. Phorbol ester treatment of SMCs increased intracellular reactive oxygen, and the increase in receptor activity was reduced 30% by the antioxidant N-acetyl cysteine (NAC), suggesting a role for reactive oxygen in phorbol ester-mediated receptor regulation. Furthermore, direct treatment of SMCs with reactive oxygen species increased scavenger-receptor activity. In rabbit SMCs, 100 micromol/L H2O2 alone slightly increased scavenger-receptor mRNA and protein expression. In combination, 100 micromol/L H2O2 and 10 micromol/L vanadate, which promotes formation of OH and enhances the inhibition of protein tyrosine phosphatase by H2O2, increased scavenger-receptor mRNA expression 25-fold in rabbit SMCs and 8-fold in human SMCs. NAC reduced the effect of H2O2 and vanadate by 93%. The increase in SMC scavenger-receptor expression occurs at the level of gene transcription. Receptor mRNA half-life was unchanged after treatment with either phorbol esters or reactive oxygen (approximately 14.5 hours), and induction by phorbol esters increased SMC scavenger-receptor mRNA transcription, as determined by nuclear run-on assay. Multiple cytokines and growth factors that contribute to the generation of reactive oxygen species are present in atherosclerotic lesions. These factors may all contribute to the upregulation of SMC scavenger-receptor activity and therefore to the formation of smooth muscle foam cells.


Subject(s)
Gene Expression Regulation , Membrane Proteins , Muscle, Smooth, Vascular/metabolism , Oxidative Stress , Protein Kinase C/physiology , Receptors, Immunologic/genetics , Receptors, Lipoprotein , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Luciferases/genetics , Protein Kinase C/antagonists & inhibitors , RNA, Messenger/biosynthesis , Rabbits , Reactive Oxygen Species , Receptors, Scavenger , Recombinant Fusion Proteins , Scavenger Receptors, Class B , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Up-Regulation
14.
Brain Res ; 749(1): 135-8, 1997 Feb 21.
Article in English | MEDLINE | ID: mdl-9070638

ABSTRACT

Apolipoprotein E4 (ApoE4) increases the risk of late-onset Alzheimer's disease (AD). It binds tightly to beta-amyloid protein (A beta), which is known to activate the classical complement pathway in vitro. Since complement activation is a possible mechanism for promoting inflammation in AD, we tested, utilizing ELISA techniques, whether the various isoforms of ApoE could influence A beta complement activation, or could themselves activate the pathway. A beta applied alone to ELISA plate wells at concentrations of 100-500 ng showed a linear increase in ability to activate serum complement, but all the ApoE isoproteins were inactive. When 200 or 430 ng of A beta were plated and then exposed to solutions of 100-200 ng of ApoE2, ApoE3, ApoE4 or bovine serum albumin (BSA), only ApoE4 significantly enhanced the activation. This ApoE4-specific enhancement of complement activation by A beta may relate to its role in increasing the risk of late-onset AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Apolipoproteins E/pharmacology , Complement Activation/drug effects , Alzheimer Disease/metabolism , Apolipoprotein E2 , Apolipoprotein E3 , Apolipoprotein E4 , Dose-Response Relationship, Drug , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Humans
15.
Proc Natl Acad Sci U S A ; 93(24): 14041-6, 1996 Nov 26.
Article in English | MEDLINE | ID: mdl-8943057

ABSTRACT

The microsomal enzyme acyl-CoA:cholesterol acyltransferase (ACAT; EC 2.3.1.26) catalyzes the esterification of cellular cholesterol with fatty acids to form cholesterol esters. ACAT activity is found in many tissues, including macrophages, the adrenal glands, and the liver. In macrophages, ACAT is thought to participate in foam cell formation and thereby to contribute to atherosclerotic lesion development. Disruption of the gene for ACAT (Acact) in mice resulted in decreased cholesterol esterification in ACAT-deficient fibroblasts and adrenal membranes, and markedly reduced cholesterol ester levels in adrenal glands and peritoneal macrophages; the latter finding will be useful in testing the role of ACAT and macrophage foam cell formation in atherosclerosis. In contrast, the livers of ACAT-deficient mice contained substantial amounts of cholesterol esters and exhibited no reduction in cholesterol esterification activity. These tissue-specific reductions in cholesterol esterification provide evidence that in mammals this process involves more than one form of esterification enzyme.


Subject(s)
Adrenal Glands/metabolism , Cholesterol Esters/metabolism , Cholesterol/metabolism , Sterol O-Acyltransferase/deficiency , Sterol O-Acyltransferase/genetics , Adrenocorticotropic Hormone/pharmacology , Animals , DNA Primers , Dietary Fats , Embryo, Mammalian , Fibroblasts , Heterozygote , Homozygote , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Microsomes/metabolism , Microsomes, Liver/metabolism , Polymerase Chain Reaction , Sterol O-Acyltransferase/biosynthesis
16.
Ann N Y Acad Sci ; 777: 139-45, 1996 Jan 17.
Article in English | MEDLINE | ID: mdl-8624076

ABSTRACT

Apolipoprotein (apo) E is associated with the two characteristic neuropathologic lesions of Alzheimer's disease--extracellular neuritic plaques representing deposits of amyloid beta (A beta) peptide and intracellular neurofibrillary tangles representing filaments of a microtubule-associated protein called tau. Incubation of the apoE4 isoform with the A beta peptide in vitro results in the formation of a dense, stable network of very long monofibrils, while incubation of apoE3 with the A beta peptide results in the formation of a less dense, less stable network. The more complex nature of the plaques formed with the A beta peptide in the presence of apoE4 in vivo may impair the normal clearance process and enhance plaque formation. Alternatively or additionally, apoE may alter the cytoskeletal structure and function and, under certain conditions, may promote the formation of the neurofibrillary tangles. Our studies have demonstrated that apoE3 and apoE4 exert differential effects on neuronal growth (i.e., neurite extension and branching) in vitro. When combined with a source of lipid, apoE3 stimulated neurite extension in peripheral nervous system neurons (dorsal root ganglia), whereas apoE4 inhibited it. Similar results were obtained with central nervous system neurons (murine neuroblastoma Neuro-2a cells). Addition of free apoE3 or apoE4 without beta-VLDL had no effect on neurite outgrowth. There was also differential accumulation of apoE3 and apoE4 by the neuroblastoma cells: apoE3 accumulated within cell bodies and neurites to a greater extent than apoE4. Thus, apoE3 may facilitate cytoskeletal activity, whereas apoE4 may inhibit it, which would be detrimental during synaptic remodeling.


Subject(s)
Alzheimer Disease/physiopathology , Apolipoproteins E/chemistry , Apolipoproteins E/physiology , Amyloid beta-Peptides/physiology , Animals , Apolipoprotein E3 , Apolipoprotein E4 , Humans , Neurites/physiology , Structure-Activity Relationship
17.
J Clin Invest ; 96(6): 2932-46, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8675665

ABSTRACT

The formation of chylomicrons by the intestine is important for the absorption of dietary fats and fat-soluble vitamins (e.g., retinol, alpha-tocopherol). Apo B plays an essential structural role in the formation of chylomicrons in the intestine as well as the VLDL in the liver. We have developed genetically modified mice that express apo B in the liver but not in the intestine. By electron microscopy, the enterocytes of these mice lacked nascent chylomicrons in the endoplasmic reticulum and Golgi apparatus. Because these mice could not form chylomicrons, the intestinal villus enterocytes were massively engorged with fat, which was contained in cytosolic lipid droplets. These mice absorbed D-xylose normally, but there was virtually no absorption of retinol palmitate or cholesterol. The levels of alpha-tocopherol in the plasma were extremely low. Of note, the absence of chylomicron synthesis in the intestine did not appear to have a significant effect on the plasma levels of the apo B-containing lipoproteins produced by the liver. The mice lacking intestinal apo B expression represent the first genetic model of defective absorption of fats and fat-soluble vitamins and provide a useful animal model for studying nutrition and lipoprotein metabolism.


Subject(s)
Apolipoproteins B/biosynthesis , Apolipoproteins B/genetics , Chylomicrons/biosynthesis , Intestinal Mucosa/metabolism , Liver/metabolism , Animals , Chylomicrons/analysis , Crosses, Genetic , Diterpenes , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Female , Genotype , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Heterozygote , Humans , Intestinal Absorption , Intestines/pathology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Models, Genetic , Organ Specificity , Retinyl Esters , Vitamin A/analogs & derivatives , Vitamin A/metabolism , Vitamin E/metabolism
18.
J Biol Chem ; 270(45): 27063-71, 1995 Nov 10.
Article in English | MEDLINE | ID: mdl-7592957

ABSTRACT

Previously, we demonstrated in cultured dorsal root ganglion neurons that, in the presence of beta-migrating very low density lipoproteins (beta-VLDL), apolipoprotein (apo) E4, but not apoE3, suppresses neurite outgrowth. In the current studies, murine neuroblastoma cells (Neuro-2a) were stably transfected with human apoE3 or apoE4 cDNA, and the effect on neurite outgrowth was examined. The stably transfected cells secreted nanogram quantities of apoE (44-89 ng/mg of cell protein in 48 h). In the absence of lipoproteins, neurite outgrowth was similar in the apoE3- and apoE4-secreting cells. The apoE4-secreting cells, when incubated with beta-VLDL, VLDL, cerebrospinal fluid lipoproteins (d < 1.21 g/ml), or with triglyceride/phospholipid (2.7:1 (w/w)) emulsions, showed a reduction in the number of neurites/cell, a decrease in neurite branching, and an inhibition of neurite extension, whereas in the apoE3-secreting cells in the presence of a lipid source, neurite extension was increased. Uptake of beta-VLDL occurred to a similar extent in both the apoE3- and apoE4-secreting cells. With low density lipoproteins or with dimyristoylphosphatidylcholine emulsions, either alone or complexed with cholesterol, no differential effect on neurite outgrowth was observed. A slight differential effect was observed with apoE-containing high density lipoproteins. The differential effect of apoE3 and apoE4 in the presence of beta-VLDL was blocked by incubation of the cells with heparinase and chlorate, with lactoferrin, or with receptor-associated protein, all of which prevent the uptake of lipoproteins by the low density lipoprotein receptor-related protein (LRP). The data suggest that the secreted and/or cell surface-bound apoE interact with the lipoproteins and facilitate their internalization via the heparan sulfate proteoglycan-LRP pathway. The mechanism by which apoE3 and apoE4 exert differential effects on neurite outgrowth remains speculative. However, the data suggest that apoE4, which has been shown to be associated with late onset familial and sporadic Alzheimer's disease, may inhibit neuronal remodeling and contribute to the progression of the disease.


Subject(s)
Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Neurites/physiology , Neurites/ultrastructure , Alzheimer Disease/etiology , Animals , Apolipoprotein E3 , Apolipoprotein E4 , DNA, Complementary/genetics , Gene Expression , Humans , Lipoproteins, VLDL/pharmacology , Liposomes , Mice , Neurites/drug effects , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/ultrastructure , Rabbits , Transfection , Tumor Cells, Cultured
19.
J Clin Invest ; 96(5): 2170-9, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7593602

ABSTRACT

apoE deficiency causes hyperlipidemia and premature atherosclerosis. To determine if macrophage-specific expression of apoE would decrease the extent of atherosclerosis, we expressed human apoE in macrophages of apoE-null mice (apoE-/-) and assessed the effect on lipid accumulation in cells of the arterial wall. Macrophage-specific expression of human apoE in normal mice was obtained by use of the visna virus LTR. These animals were bred with apoE-/- mice to produce animals hemizygous for expression of human apoE in macrophages in the absence of murine apoE (apoE-/-,hTgE+/0). Low levels of human apoE mRNA were present in liver and spleen and high levels in lung and peritoneal macrophages. Human apoE was secreted by peritoneal macrophages and was detected in Kupffer cells of the liver. Human apoE in the plasma of apoE-/-,hTgE+/0 mice (n = 30) was inversely correlated (P < 0.005) with the plasma cholesterol concentration. After 15 wk on a normal chow diet, atherosclerosis was assessed in apoE-/-,hTgE+/0 animals and in apoE-/-,hTgE0/0 littermates matched for plasma cholesterol level (approximately 450 mg/dl) and lipoprotein profile. There was significantly less atherosclerosis in both the aortic sinus and in the proximal aorta (P < 0.0001) in the animals expressing the human apoE transgene. In apo-E-/-,hTgE+/0 animals, which had detectable atherosclerotic lesions, human apoE was detected in the secretory apparatus of macrophage-derived foam cells in the arterial wall. The data demonstrate that expression of apoE by macrophages is antiatherogenic even in the presence of high levels of atherogenic lipoproteins. The data suggest that apoE prevents atherosclerosis by promoting cholesterol efflux from cells of the arterial wall.


Subject(s)
Apolipoproteins E/biosynthesis , Arteriosclerosis/metabolism , Hypercholesterolemia/metabolism , Macrophages/metabolism , Animals , Apolipoproteins E/genetics , Arteriosclerosis/genetics , Arteriosclerosis/pathology , Cells, Cultured , Cholesterol/blood , Female , Foam Cells/metabolism , Gene Expression , Gene Transfer Techniques , Humans , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Male , Mice , Mice, Transgenic
20.
Proc Natl Acad Sci U S A ; 92(21): 9480-4, 1995 Oct 10.
Article in English | MEDLINE | ID: mdl-7568158

ABSTRACT

The epsilon 4 allele of apolipoprotein E (apoE) is a major risk factor for Alzheimer disease, suggesting that apoE may directly influence neurons in the aging brain. Recent data suggest that apoE-containing lipoproteins can influence neurite outgrowth in an isoform-specific fashion. The neuronal mediators of apoE effects have not been clarified. We show here that in a central nervous system-derived neuronal cell line, apoE3 but not apoE4 increases neurite extension. The effect of apoE3 was blocked at low nanomolar concentrations by purified 39-kDa protein that regulates ligand binding to the low density lipoprotein receptor-related protein (LRP). Anti-LRP antibody also completely abolished the neurite-promoting effect of apoE3. Understanding isoform-specific cell biological processes mediated by apoE-LRP interactions in central nervous system neurons may provide insight into Alzheimer disease pathogenesis.


Subject(s)
Apolipoproteins E/metabolism , Hypothalamus/physiology , Neurites/physiology , Receptors, Immunologic/metabolism , Receptors, LDL/metabolism , Apolipoprotein E3 , Carbocyanines , Cell Line , Cross-Linking Reagents , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Hypothalamus/cytology , Low Density Lipoprotein Receptor-Related Protein-1 , Microscopy, Confocal , Microscopy, Fluorescence , Neurites/ultrastructure , Neurons/cytology , Neurons/physiology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...