Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1788(2): 442-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19121285

ABSTRACT

Supramolecular structures, particularly micelles and liposomes equipped with uptake-mediating address compounds, have attracted much attention as pharmaceutical formulations. Their development requires an understanding of the mechanism by which the carrier systems interact with and translocate into the target cells. We developed an apolipoprotein E-derived peptide, called A2, that efficiently translocates across cell membranes. Upon coupling of two palmitoyl chains (P2), the highly cationic sequence acquires detergent-like properties such as a strong tendency to self-associate and the ability to integrate into lipid bilayers. Confocal laser scanning microscopy and fluorescence activated cell sorting were used to compare the internalization of the fluorescence-labeled monomeric A2 with the uptake of the colloidal P2A2 micelles and P2A2-tagged liposomes into endothelial cells of blood vessels. Specific inhibitors of endocytosis were used to identify the underlying mechanisms. b.End3 and BAEC cells as example of endothelial cells of small capillaries and large vascular vessels, respectively, were examined. The uptake of monomeric A2 was characterized by poor cellular selectivity. A2 was efficiently internalized into both cell lines via at least two different mechanisms. Besides an endocytotic uptake route, a second passive pathway exists, that leads to a rapid distribution of A2 within the cytoplasm. Also liposomes tagged with P2A2 were non-selectively internalized into both b.End3 and BAEC cells. Their nonselective uptake was mediated by clathrin- and caveolin-independent endocytosis. In contrast, micellar P2A2 entered b.End3 cells via clathrin-mediated endocytosis, while no uptake of P2A2 into BAEC cells was observed. In conclusion, the specific clathrin-mediated uptake mode of P2A2 micelles might provide the basis for a blood brain barrier-specific targeting.


Subject(s)
Apolipoproteins E/chemistry , Apolipoproteins E/metabolism , Endothelial Cells/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Apolipoproteins E/toxicity , Cell Line , Cell Survival/drug effects , Colloids/chemistry , Colloids/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Liposomes/chemistry , Mice , Micelles , Molecular Sequence Data , Peptide Fragments/toxicity , Protein Binding , Transferrin/metabolism
2.
Curr Opin Drug Discov Devel ; 9(5): 627-41, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17002223

ABSTRACT

Apolipoprotein (apo) E is a multifunctional protein that has central roles in lipid metabolism and neurobiology. It has three common isoforms (apoE2, apoE3 and apoE4) that have different effects on lipid and neuronal homneostasis. Unlike apoE3, tie most common isoform, apoE4 is a major risk factor for Alzheimer's disease (AD). Although the mechanisms underlying the actions of apoE4 in AD pathogenesis are still poorly understood, emerging data strongly suggest that apoE4, with its multiple cellular origins and multiple structural and biophysical properties, contributes to this disease by interacting with different factors through various pathways. Thus, multiple molecular and cellular mechanisms should be considered in developing anti-AD drugs that target apoE4.


Subject(s)
Alzheimer Disease/drug therapy , Apolipoproteins E/toxicity , Neurotoxins , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Apolipoprotein E4 , Apolipoproteins E/biosynthesis , Apolipoproteins E/genetics , Humans , Polymorphism, Genetic
3.
J Mol Biol ; 361(5): 932-44, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16890957

ABSTRACT

The three isoforms of apolipoprotein (apo) E are strongly associated with different risks for Alzheimer's disease: apoE4>apoE3>apoE2. Here, we show at physiological salt concentrations and pH that native tetramers of apoE form soluble aggregates in vitro that bind the amyloid dyes thioflavin T and Congo red. However, unlike classic amyloid fibrils, the aggregates adopt an irregular protofilament-like morphology and are seemingly highly alpha-helical. The aggregates formed at substantially different rates (apoE4>apoE3>apoE2) and were significantly more toxic to cultured neuronal cells than the tetramer. Since the three isoforms have large differences in conformational stability that can influence aggregation and amyloid pathways, we tested the effects of mutations that increased or decreased stability. Decreasing the conformational stability of the amino-terminal domain of apoE increased aggregation rates and vice versa. Our findings provide a new perspective for an isoform-specific pathogenic role for apoE aggregation in which differences in the conformational stability of the amino-terminal domain mediate neurodegeneration.


Subject(s)
Apolipoproteins E/chemistry , Apolipoproteins E/toxicity , Neurons/drug effects , Animals , Apolipoprotein E2 , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/metabolism , Apolipoproteins E/ultrastructure , Benzothiazoles , Cells, Cultured , Congo Red , Mice , Models, Biological , Neurons/pathology , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Isoforms/toxicity , Protein Isoforms/ultrastructure , Protein Structure, Quaternary , Protein Structure, Tertiary , Structure-Activity Relationship , Thermodynamics , Thiazoles
4.
Proc Natl Acad Sci U S A ; 102(51): 18694-9, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16344479

ABSTRACT

Apolipoprotein (apo) E4, a 299-aa protein and a major risk factor for Alzheimer's disease, can be cleaved to generate C-terminal-truncated fragments that cause neurotoxicity in vitro and neurodegeneration and behavioral deficits in transgenic mice. To investigate this neurotoxicity, we expressed apoE4 with C- or N-terminal truncations or mutations in transfected Neuro-2a cells. ApoE4 (1-272) was neurotoxic, but full-length apoE4(1-299) and apoE4(1-240) were not, suggesting that the lipid-binding region (amino acids 241-272) mediates the neurotoxicity and that amino acids 273-299 are protective. A quadruple mutation in the lipid-binding region (I250A, F257A, W264R, and V269A) abolished the neurotoxicity of apoE4(1-272), and single mutations in the region of amino acids 273-299 (L279Q, K282A, or Q284A) made full-length apoE4 neurotoxic. Immunofluorescence staining showed that apoE4(1-272) formed filamentous inclusions containing phosphorylated tau in some cells and interacted with mitochondria in others, leading to mitochondrial dysfunction as determined by MitoTracker staining and flow cytometry. ApoE4(241-272) did not cause mitochondrial dysfunction or neurotoxicity, suggesting that the lipid-binding region alone is insufficient for neurotoxicity. Truncation of N-terminal sequences (amino acids 1-170) containing the receptor-binding region (amino acids 135-150) and triple mutations within that region (R142A, K146A, and R147A) abolished the mitochondrial interaction and neurotoxicity of apoE4(1-272). Further analysis showed that the receptor-binding region is required for escape from the secretory pathway and that the lipid-binding region mediates mitochondrial interaction. Thus, the lipid- and receptor-binding regions in apoE4 fragments act together to cause mitochondrial dysfunction and neurotoxicity, which may be important in Alzheimer's disease pathogenesis.


Subject(s)
Apolipoproteins E/metabolism , Apolipoproteins E/toxicity , Lipids , Mitochondria/drug effects , Neurons/drug effects , Neurons/pathology , Receptors, Lipoprotein/metabolism , Alzheimer Disease/metabolism , Animals , Apolipoprotein E4 , Apolipoproteins E/genetics , Cell Line, Tumor , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Mice , Mitochondria/metabolism , Mutation/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/toxicity
5.
Biochemistry ; 44(6): 2021-9, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15697227

ABSTRACT

A promising strategy to solve the problems of insufficient membrane penetration of drugs and low target specificity is the localization of targeting and uptake-facilitating ligands on the surface of drug-carrier systems. This study investigated the role of a peptide derived from the LDL receptor (LDLr)-binding domain of apolipoprotein E (apoE) in initiating endocytosis in brain capillary endothelial cells. The highly cationic tandem dimer of apoE residues (141-150) was coupled covalently onto poly(ethylene glycol)-derivatized liposomes. Membrane binding and cellular uptake was monitored qualitatively by confocal-laser-scanning microscopy as well as quantitatively using a fluorescence assay. The peptide mediated an efficient, energy-dependent translocation of liposomes across the membrane of brain capillary endothelial cells. Liposomes without surface-located peptides displayed neither membrane accumulation nor cellular uptake. Low peptide affinity to LDLr and internalization of the complex into fibroblasts with up- and down-regulated receptor expression levels, as well as complex translocation into cells incubated with an antibody against the LDLr, pointed to a dominating role of an LDLr-independent transport route. Enzymatic digestion of heparan sulfate proteoglycan (HSPG) with heparinase I and addition of heparin and poly-l-lysin as competitors of HSPG and HSPG ligands, respectively, resulted in a significant loss in liposome internalization. The results suggested that HSPG played a major role in the apoE-peptide-mediated uptake of liposomes into endothelial cells of brain microvessels.


Subject(s)
Apolipoproteins E/chemistry , Brain/blood supply , Brain/metabolism , Endothelium, Vascular/chemistry , Endothelium, Vascular/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Apolipoproteins E/toxicity , Binding, Competitive , Biological Transport , Capillaries/chemistry , Capillaries/cytology , Capillaries/metabolism , Cell Survival/drug effects , Cells, Cultured , Drug Delivery Systems/methods , Endothelium, Vascular/cytology , Heparan Sulfate Proteoglycans/chemistry , Humans , Iodine Radioisotopes/metabolism , Lipoproteins, LDL/metabolism , Liposomes , Microscopy, Confocal , Molecular Sequence Data , Peptide Fragments/toxicity , Protein Binding , Rats , Receptors, LDL/metabolism , Repetitive Sequences, Amino Acid , Stereoisomerism
6.
Biochemistry ; 41(25): 8203-11, 2002 Jun 25.
Article in English | MEDLINE | ID: mdl-12069613

ABSTRACT

Apolipoprotein E (apoE) has been genetically linked to late-onset Alzheimer's disease (AD). The role of this lipid-transport protein in AD remains to be established. One hypothesis is that apoE, particularly the apoE4 isoform, may have neurotoxic effects as demonstrated using apoE-related synthetic peptides and the N-terminal fragment of apoE. ApoE is a heparan-sulfate binding protein, and apoE peptide neurotoxicity can be blocked by heparin and prevented by degrading heparan sulfate or inhibiting its biosynthesis. The possibility that heparin inhibition of toxicity is mediated by a specific oligosaccharide sequence was investigated using a bioassay to determine the inhibition of apoE peptide toxicity by glycosaminoglycans and purified glycosaminoglycan oligosaccharides. Studies on modified heparins showed that the presence of N-sulfo groups and either 2- or 6-O sulfo groups were required for inhibition of toxicity. Heparin oligosaccharides with eight or more saccharide residues with seven O-sulfo groups and four N-sulfo groups exhibited potent inhibition. Larger oligosaccharides, and heparin and heparan sulfate polymers, afforded comparable, or somewhat better, protective effects but also caused clumping and detachment of cells when administrated alone.


Subject(s)
Apolipoproteins E/antagonists & inhibitors , Apolipoproteins E/toxicity , Glycosaminoglycans/pharmacology , Neurons/drug effects , Oligosaccharides/pharmacology , Animals , Apolipoproteins E/metabolism , Binding, Competitive/drug effects , Cell Death/drug effects , Cells, Cultured , Chick Embryo , Dermatan Sulfate/metabolism , Dermatan Sulfate/pharmacology , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/metabolism , Glycosaminoglycans/metabolism , Heparan Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/pharmacology , Heparin/metabolism , Heparin/pharmacology , Heparin, Low-Molecular-Weight/metabolism , Heparin, Low-Molecular-Weight/pharmacology , Neurons/metabolism , Oligosaccharides/metabolism , Partial Thromboplastin Time , Protein Binding/drug effects , Swine
7.
J Neurosci Res ; 67(3): 379-87, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11813243

ABSTRACT

The association of the E4 allele of apolipoprotein E (apoE4) as a genetic risk factor for Alzheimer's disease (AD) has been well established. Although recent studies in neuronal cell lines and transgenic mice have shown that apoE4 promotes neurodegeneration, the mechanisms through which apoE4 impairs neuronal viability are not completely understood. In this context, the main objective of the present study was to determine whether the neurotoxic effects of apoE4 are mediated by an alteration in calcium homeostasis. For this purpose, effects of recombinant apoE3 and apoE4 on cell viability and intracellular calcium levels were analyzed in a murine hippocampal cell line (HT22) and in primary rat cortical neurons, in the presence or absence of calcium inhibitors. Under basal conditions, apoE4-treated cells displayed increased levels of cytosolic calcium associated with cell death in a dose-dependent manner. Furthermore, apoE4 treatment potentiated the rise in cytosolic calcium and cell death following the administration of a calcium ionophore. The effects of apoE4 on cell viability and calcium homeostasis were inhibited by calcium chelators or by blocking calcium channels, but not by inhibitors of intracellular calcium reserves. Taken together, these results indicate that the neurotoxic effects of apoE4 are dependent on extracellular calcium influx via calcium channels.


Subject(s)
Apolipoproteins E/toxicity , Calcium/metabolism , Homeostasis/drug effects , Neurons/drug effects , Neurons/metabolism , Animals , Apolipoprotein E4 , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Cell Membrane/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chelating Agents/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Ionophores/pharmacology , LDL-Receptor Related Protein-Associated Protein/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice , Neurons/cytology , Rats
8.
Neuroreport ; 12(2): 201-6, 2001 Feb 12.
Article in English | MEDLINE | ID: mdl-11209921

ABSTRACT

The toxic effects of beta-amyloid (A beta) (1-42), apolipoprotein E (apoE) isoforms, and apoE/A beta complexes were studied in human SH-SY5Y neuroblastoma cells and fibroblasts using MTT reduction. In SH-SY5Y cells, A beta(1-42) gave time-dependent toxicity over 2-48 h, which was reduced by co-incubation with rabbit beta-very low density lipoproteins (beta-VLDL). Human recombinant apoE3 and E4 isoforms were also toxic by themselves and also potentiated A beta effects when used alone, but not when associated with beta-VLDL. None of the treatments were toxic to human fibroblasts. These results suggest that beta-VLDL has a protective role on A beta-induced neurotoxicity and that the status of apoE or the conformation of lipoprotein containing apoE particles may be important for determining the contribution of apoE to neurodegeneration.


Subject(s)
Amyloid beta-Peptides/toxicity , Apolipoproteins E/toxicity , Lipoproteins, VLDL/pharmacology , Neuroblastoma , Neurons/drug effects , Peptide Fragments/toxicity , Coloring Agents , Fibroblasts/cytology , Humans , Neurons/cytology , Neuroprotective Agents/pharmacology , Skin/cytology , Tetrazolium Salts , Thiazoles , Tumor Cells, Cultured
9.
Neurobiol Dis ; 8(1): 78-89, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11162242

ABSTRACT

The association of the E4 isoform of apolipoprotein E (apoE) as a genetic risk factor for late onset Alzheimer's disease (AD) has been well established. Central nervous system (CNS) neurons are specifically affected so that defining the mechanisms by which two of the major human apoE isoforms act within CNS neurons is important to our understanding of their effect on neuronal maintenance and function. We have developed a cell culture model using human brain tissue to characterize exogenous apoE transport. We have tracked the association of apoE3 and E4 with CD63, the GTP-binding protein rab5a and the acidic hydrolase cathepsin D, which localize lysosomes, early endosomes, and late endosomes/lysosomes, respectively. Double immunostaining and confocal laser scanning microscopy revealed by z-series that after 30 min most intraneuronal apoE colocalized with rab5a, whereas no astrocyte apoE/rab5a colocalization was detected. Conversely, apoE3 and CD63 did not colocalize in neurons, even after 1 h, but was colocalized in astrocytes. Also, there was approximately 9% apoE3 colocalization with cathepsin D in neurons, whereas up to 87% of apoE4 vesicles were colocalized. In astrocytes, the proportion of apoE3 colocalized with cathepsin D was greater than that in neurons, but still significantly different from that found with apoE4. These immunohistological data demonstrate that, in neurons, apoE can be endocytosed via a rab5a-regulated vesicle-mediated pathway and that beyond this stage there may be isoform specific differences in apoE trafficking present in both neurons and astrocytes.


Subject(s)
Apolipoproteins E/metabolism , Astrocytes/metabolism , Brain Chemistry , Neurons/metabolism , Androstadienes/pharmacology , Anti-Inflammatory Agents/pharmacology , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/antagonists & inhibitors , Apolipoproteins E/toxicity , Astrocytes/drug effects , Blotting, Western , Brain/cytology , Brain/drug effects , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Endocytosis , Humans , Immunohistochemistry , Lysosomes/metabolism , Lysosomes/ultrastructure , Microscopy, Confocal , Neurons/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Wortmannin
10.
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
11.
J Neurochem ; 72(3): 1069-80, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10037478

ABSTRACT

The apolipoprotein E (apoE)-derived peptide (141-155)2 has a neurotoxic effect, implying that apoE itself could be a source of toxicity in Alzheimer's disease brain. We characterized the toxicity of this peptide on superior cervical ganglion (SCG) neurons and compared the death with the apoptotic death that occurs after nerve growth factor (NGF) deprivation in these cells. A dose of 10 microM apoE (141-155)2 resulted in the death of approximately 50% of the neurons within 24 h. Nuclear condensation and DNA fragmentation preceded the death. However, most inhibitors of NGF deprivation-induced death, including the caspase inhibitor Boc-aspartyl(O-methyl)fluoromethyl ketone and genetic deletion of bax-/-, had no effect on the toxicity. Inclusion of depolarizing levels of potassium did block the toxicity. Receptor-associated peptide (RAP), an antagonist for apoE receptors, did not protect cells in either SCG or hippocampal cultures. In addition, RAP had no effect on internalization of the apoE peptide. These data support the observation that apoE (141-155)2 is neurotoxic but suggest that the neurotoxicity is distinct from classical apoptosis or necrosis. Furthermore, these results indicate that the toxic effect may occur independently of members of the low-density lipoprotein receptor gene family.


Subject(s)
Apolipoproteins E/physiology , Apolipoproteins E/toxicity , Peptide Fragments/toxicity , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cells, Cultured , Hippocampus/cytology , Hippocampus/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Molecular Sequence Data , Phosphorylation , Potassium/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Sprague-Dawley , Receptors, LDL/physiology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/drug effects
12.
J Neurosci Res ; 54(1): 58-67, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9778150

ABSTRACT

The presence of the apolipoprotein E (apoE) allele epsilon4 is a major risk factor for the development of Alzheimer's disease (AD); however, the molecular mechanism underlying the acceleration of AD development in individuals with epsilon4 remains to be determined. To investigate the isoform-specific effects of apoE on neurons, primary neuron cultures were prepared from fetal rat cerebral cortices. Inhibition by compactin, a 3-hydroxyl-3-methylglutaryl coenzyme A reductase inhibitor of de novo cholesterol synthesis, induced premature neuronal cell death in a dose-dependent manner. In the presence of compactin at a sublethal dose to the cells, rabbit beta-migrating very low density lipoprotein (beta-VLDL) with human apoE4 (the product of epsilon4) induced premature neuronal cell death, while that with apoE3 (the product of epsilon3) did not. Neurons cultured in the presence of apoE4, beta-VLDL, and compactin were shrunken and spherical, containing condensed chromatin and fragmented DNA, features characteristic of apoptosis. The addition of intermediate metabolites of the cholesterol biosynthetic pathway, including mevalonate and squalene, rescued neuronal cells incubated with apoE4 and beta-VLDL, in the presence of compactin. These results strongly suggest that a reduction in the level of endogenously synthesized cholesterol is a prerequisite for apoE4-induced neuronal cell death.


Subject(s)
Apolipoproteins E/toxicity , Apoptosis/drug effects , Cholesterol/metabolism , Neurons/drug effects , Animals , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cholesterol/biosynthesis , Cholesterol, VLDL/pharmacology , DNA Fragmentation , In Situ Nick-End Labeling , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Mevalonic Acid/pharmacology , Neurons/cytology , Rats , Rats, Sprague-Dawley , Squalene/pharmacology
13.
J Neurosci ; 17(15): 5678-86, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9221767

ABSTRACT

Potent neurotoxicity is associated with both apolipoprotein E (apoE)-related synthetic peptides and the 22 kDa N-terminal thrombin-cleavage fragment of apoE. Furthermore, the E4 isoform of the 22 kDa fragment is significantly more toxic than the same fragment derived from the E3 isoform, suggesting the possibility of a direct role of apoE-associated neurotoxicity in the pathophysiology of Alzheimer's disease. In the present study, the potential role of cell surface receptors in mediating neurotoxicity was assessed by using a variety of agents that should block the heparin-binding and receptor-binding activity of apoE. Effective inhibitors of neurotoxicity of both the apoE peptides and the apoE fragment include heparin, heparan sulfate, sodium chlorate and heparinase, the low-density lipoprotein (LDL) receptor-related protein receptor-associated protein, and a polyclonal anti-LDL receptor-related protein antibody. These results suggest that the neurotoxicity of the 22 kDa thrombin cleavage fragment of apoE and related peptides is receptor-mediated, and that the most likely candidate receptor is a heparan sulfate proteoglycan-LDL receptor-related protein complex.


Subject(s)
Apolipoproteins E/toxicity , Cell Death/drug effects , Heparitin Sulfate/toxicity , Receptors, Lipoprotein/drug effects , Animals , Cells, Cultured/drug effects , Chick Embryo , Dose-Response Relationship, Drug , Low Density Lipoprotein Receptor-Related Protein-1 , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...