Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 18(1): 214, 2021 Sep 18.
Article in English | MEDLINE | ID: mdl-34537055

ABSTRACT

BACKGROUND: APOE4 is the strongest genetic risk factor for Alzheimer's disease (AD), and obesity is a strong environmental risk factor for AD. These factors result in multiple central nervous system (CNS) disturbances and significantly increase chances of AD. Since over 20% of the US population carry the APOE4 allele and over 40% are obese, it is important to understand how these risk factors interact to affect neurons and glia in the CNS. METHODS: We fed male and female APOE3 and APOE4 knock-in mice a high-fat diet (HFD-45% kcal fat) or a "control" diet (CD-10% kcal fat) for 12 weeks beginning at 6 months of age. At the end of the 12 weeks, brains were collected and analyzed for gliosis, neuroinflammatory genes, and neuronal integrity. RESULTS: APOE3 mice on HFD, but not APOE4 mice, experienced increases in gliosis as measured by GFAP and Iba1 immunostaining. APOE4 mice on HFD showed a stronger increase in the expression of Adora2a than APOE3 mice. Finally, APOE3 mice on HFD, but not APOE4 mice, also showed increased neuronal expression of immediate early genes cFos and Arc. CONCLUSIONS: These findings demonstrate that APOE genotype and obesity interact in their effects on important processes particularly related to inflammation and neuronal plasticity in the CNS. During the early stages of obesity, the APOE3 genotype modulates a response to HFD while the APOE4 genotype does not. This supports a model where early dysregulation of inflammation in APOE4 brains could predispose to CNS damages from various insults and later result in the increased CNS damage normally associated with the APOE4 genotype.


Subject(s)
Apolipoprotein E3/biosynthesis , Apolipoprotein E4/biosynthesis , Brain/metabolism , Diet, High-Fat/adverse effects , Genes, Immediate-Early/physiology , Gliosis/metabolism , Animals , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Female , Gene Expression , Gene Knock-In Techniques , Gliosis/etiology , Gliosis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
2.
Sci Rep ; 11(1): 3438, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33564035

ABSTRACT

ApoEε4 is a major genetic risk factor for Alzheimer's disease (AD), a disease hallmarked by extracellular amyloid-beta (Aß) plaques and intracellular neurofibrillary tangles (NFTs). The presence of the ApoEε4 allele is associated with increased Aß deposition and a role for ApoEε4 in the potentiation of tau pathology has recently emerged. This study focused on comparing the effects of adeno-associated virus (AAV)-mediated overexpression of the three predominant human ApoE isoforms within astrocytes. The isoform-specific effects of human ApoE were evaluated within in vitro models of tau pathology within neuron/astrocyte co-cultures, as well as in a transgenic tau mouse model. Tau aggregation, accumulation, and phosphorylation were measured to determine if the three isoforms of human ApoE had differential effects on tau. Astrocytic overexpression of the human ApoEε4 allele increased phosphorylation and misfolding of overexpressed neuronal tau in multiple models, including the aggregation and accumulation of added tau oligomers, in an isoform-specific manner. The ability of ApoEε4 to increase tau aggregation could be inhibited by an ApoEε4-specific antibody. This study indicates that astrocytic expression of ApoEε4 can potentiate tau aggregation and phosphorylation within neurons and supports a gain of toxic function hypothesis for the effect of hApoEε4 on tau.


Subject(s)
Alleles , Alzheimer Disease/metabolism , Apolipoprotein E4/biosynthesis , Astrocytes/metabolism , Gene Expression Regulation , Protein Aggregates , tau Proteins , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Animals , Apolipoprotein E4/genetics , Astrocytes/pathology , Disease Models, Animal , Rats , Rats, Sprague-Dawley , tau Proteins/genetics , tau Proteins/metabolism
3.
J Neurosci ; 39(37): 7408-7427, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31331998

ABSTRACT

In blood, apolipoprotein E (ApoE) is a component of circulating lipoproteins and mediates the clearance of these lipoproteins from blood by binding to ApoE receptors. Humans express three genetic ApoE variants, ApoE2, ApoE3, and ApoE4, which exhibit distinct ApoE receptor-binding properties and differentially affect Alzheimer's disease (AD), such that ApoE2 protects against, and ApoE4 predisposes to AD. In brain, ApoE-containing lipoproteins are secreted by activated astrocytes and microglia, but their functions and role in AD pathogenesis are largely unknown. Ample evidence suggests that ApoE4 induces microglial dysregulation and impedes Aß clearance in AD, but the direct neuronal effects of ApoE variants are poorly studied. Extending previous studies, we here demonstrate that the three ApoE variants differentially activate multiple neuronal signaling pathways and regulate synaptogenesis. Specifically, using human neurons (male embryonic stem cell-derived) cultured in the absence of glia to exclude indirect glial mechanisms, we show that ApoE broadly stimulates signal transduction cascades. Among others, such stimulation enhances APP synthesis and synapse formation with an ApoE4>ApoE3>ApoE2 potency rank order, paralleling the relative risk for AD conferred by these ApoE variants. Unlike the previously described induction of APP transcription, however, ApoE-induced synaptogenesis involves CREB activation rather than cFos activation. We thus propose that in brain, ApoE acts as a glia-secreted signal that activates neuronal signaling pathways. The parallel potency rank order of ApoE4>ApoE3>ApoE2 in AD risk and neuronal signaling suggests that ApoE4 may in an apparent paradox promote AD pathogenesis by causing a chronic increase in signaling, possibly via enhancing APP expression.SIGNIFICANCE STATEMENT Humans express three genetic variants of apolipoprotein E (ApoE), ApoE2, ApoE3, and ApoE4. ApoE4 constitutes the most important genetic risk factor for Alzheimer's disease (AD), whereas ApoE2 protects against AD. Significant evidence suggests that ApoE4 impairs microglial function and impedes astrocytic Aß clearance in brain, but the direct neuronal effects of ApoE are poorly understood, and the differences between ApoE variants in these effects are unclear. Here, we report that ApoE acts on neurons as a glia-secreted signaling molecule that, among others, enhances synapse formation. In activating neuronal signaling, the three ApoE variants exhibit a differential potency of ApoE4>ApoE3>ApoE2, which mirrors their relative effects on AD risk, suggesting that differential signaling by ApoE variants may contribute to AD pathogenesis.


Subject(s)
Alzheimer Disease/genetics , Apolipoprotein E2/genetics , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Embryonic Stem Cells/physiology , Genetic Predisposition to Disease/genetics , Alzheimer Disease/metabolism , Animals , Animals, Newborn , Apolipoprotein E2/biosynthesis , Apolipoprotein E3/biosynthesis , Apolipoprotein E4/biosynthesis , Cells, Cultured , Double-Blind Method , Female , Genetic Variation/physiology , HEK293 Cells , Humans , Male , Mice , Neurons/physiology , Random Allocation , Signal Transduction/physiology
4.
Ann Neurol ; 85(5): 726-739, 2019 05.
Article in English | MEDLINE | ID: mdl-30840313

ABSTRACT

OBJECTIVE: The apolipoprotein E (APOE) E4 isoform is the strongest genetic risk factor for sporadic Alzheimer disease (AD). Although APOE is predominantly expressed by astrocytes in the central nervous system, neuronal expression of APOE is of increasing interest in age-related cognitive impairment, neurological injury, and neurodegeneration. Here, we show that endogenous expression of E4 in stem-cell-derived neurons predisposes them to injury and promotes the release of phosphorylated tau. METHODS: Induced pluripotent stem cells from 2 unrelated AD patients carrying the E4 allele were corrected to the E3/E3 genotype with the CRISPR/Cas9 system and differentiated into pure cultures of forebrain excitatory neurons without contamination from other cells types. RESULTS: Compared to unedited E4 neurons, E3 neurons were less susceptible to ionomycin-induced cytotoxicity. Biochemically, E4 cells exhibited increased tau phosphorylation and ERK1/2 phosphoactivation. Moreover, E4 neurons released increased amounts of phosphorylated tau extracellularly in an isoform-dependent manner by a heparin sulfate proteoglycan-dependent mechanism. INTERPRETATION: Our results demonstrate that endogenous expression of E4 by stem-cell-derived forebrain excitatory neurons predisposes neurons to calcium dysregulation and ultimately cell death. This change is associated with increased cellular tau phosphorylation and markedly enhanced release of phosphorylated tau. Importantly, these effects are independent of glial APOE. These findings suggest that E4 accelerates spreading of tau pathology and neuron death in part by neuron-specific, glia-independent mechanisms. Ann Neurol 2019;85:726-739.


Subject(s)
Alzheimer Disease/metabolism , Apolipoprotein E4/biosynthesis , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Cell Death/physiology , Female , Humans , Induced Pluripotent Stem Cells/pathology , Male , Neurons/pathology , Phosphorylation/physiology
5.
Brain ; 142(1): 163-175, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30496349

ABSTRACT

In addition to being the greatest genetic risk factor for Alzheimer's disease, expression of the ɛ4 allele of apolipoprotein E can lead to cognitive decline during ageing that is independent of Alzheimer's amyloid-ß and tau pathology. In human post-mortem tissue and mouse models humanized for apolipoprotein E, we examined the impact of apolipoprotein E4 expression on brain exosomes, vesicles that are produced within and secreted from late-endocytic multivesicular bodies. Compared to humans or mice homozygous for the risk-neutral ɛ3 allele we show that the ɛ4 allele, whether homozygous or heterozygous with an ɛ3 allele, drives lower exosome levels in the brain extracellular space. In mice, we show that the apolipoprotein E4-driven change in brain exosome levels is age-dependent: while not present at age 6 months, it is detectable at 12 months of age. Expression levels of the exosome pathway regulators tumor susceptibility gene 101 (TSG101) and Ras-related protein Rab35 (RAB35) were found to be reduced in the brain at the protein and mRNA levels, arguing that apolipoprotein E4 genotype leads to a downregulation of exosome biosynthesis and release. Compromised exosome production is likely to have adverse effects, including diminishing a cell's ability to eliminate materials from the endosomal-lysosomal system. This reduction in brain exosome levels in 12-month-old apolipoprotein E4 mice occurs earlier than our previously reported brain endosomal pathway changes, arguing that an apolipoprotein E4-driven failure in exosome production plays a primary role in endosomal and lysosomal deficits that occur in apolipoprotein E4 mouse and human brains. Disruption of these interdependent endosomal-exosomal-lysosomal systems in apolipoprotein E4-expressing individuals may contribute to amyloidogenic amyloid-ß precursor protein processing, compromise trophic signalling and synaptic function, and interfere with a neuron's ability to degrade material, all of which are events that lead to neuronal vulnerability and higher risk of Alzheimer's disease development. Together, these data suggest that exosome pathway dysfunction is a previously unappreciated component of the brain pathologies that occur as a result of apolipoprotein E4 expression.


Subject(s)
Apolipoprotein E4/biosynthesis , Brain/metabolism , Exosomes/metabolism , Aged , Aged, 80 and over , Aging/metabolism , Alleles , Animals , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , DNA-Binding Proteins/biosynthesis , Down-Regulation , Endosomal Sorting Complexes Required for Transport/biosynthesis , Exosomes/ultrastructure , Extracellular Space/metabolism , Female , Genotype , Humans , Lipid Metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Transcription Factors/biosynthesis , rab GTP-Binding Proteins/biosynthesis
6.
J Neurosci ; 34(42): 14069-78, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25319703

ABSTRACT

Apolipoprotein (apo) E4 is expressed in many types of brain cells, is associated with age-dependent decline of learning and memory in humans, and is the major genetic risk factor for AD. To determine whether the detrimental effects of apoE4 depend on its cellular sources, we generated human apoE knock-in mouse models in which the human APOE gene is conditionally deleted in astrocytes, neurons, or GABAergic interneurons. Here we report that deletion of apoE4 in astrocytes does not protect aged mice from apoE4-induced GABAergic interneuron loss and learning and memory deficits. In contrast, deletion of apoE4 in neurons does protect aged mice from both deficits. Furthermore, deletion of apoE4 in GABAergic interneurons is sufficient to gain similar protection. This study demonstrates a detrimental effect of endogenously produced apoE4 on GABAergic interneurons that leads to learning and memory deficits in mice and provides a novel target for drug development for AD related to apoE4.


Subject(s)
Apolipoprotein E4/biosynthesis , GABAergic Neurons/metabolism , Interneurons/metabolism , Learning/physiology , Memory Disorders/metabolism , Animals , Female , GABAergic Neurons/pathology , Humans , Interneurons/pathology , Memory Disorders/pathology , Mice , Mice, Transgenic
7.
J Alzheimers Dis ; 33(4): 1009-19, 2013.
Article in English | MEDLINE | ID: mdl-23064258

ABSTRACT

The aim of our study was to elucidate whether specific patterns of gray matter loss were associated with apolipoprotein E ε4 (APOE ε4) and microtubule-associated protein tau (MAPT)-H1) genetic variants in subjects with mild cognitive impairment (MCI) at a baseline visit. Gray matter voxel-based morphometry analysis of T1 magnetic resonance imaging scans were performed in 65 amnestic-MCI subjects. MCI APOE ε4 carriers compared with non-carriers showed increased brain atrophy in right hippocampus and rostral amygdala, superior and middle temporal gyrus, and right parietal operculum, including inferior frontal gyrus, inferior parietal, and supramarginal gyrus. MAPT-H1/H1 MCI carriers showed an increased bilateral atrophy in superior frontal gyri (including frontal eye fields and left prefrontal cortex) and precentral gyrus but also unilateral left atrophy in the inferior temporal gyrus and calcarine gyrus. In addition, MCI subjects carrying both APOE ε4 and MAPT-H1/H1 variants showed gray matter loss in the supplementary motor area and right pre- and postcentral gyri. The effect of APOE ε4 on gray matter loss in right hippocampus suggests that, at least in some AD sub-types, the neuronal vulnerability could be increased in the right hemisphere. The pattern of frontal gray matter loss observed among MCI MAPT H1/H1 carriers has also been found in other tauopathies, suggesting that MCI may share etiological factors with other tauopathies. Frontal and parietal cortex vulnerability was found when adding MAPT H1/H1 and APOE ε4 effects, suggesting a synergistic effect of these variants. These results could be due to changes in APOE ε4 and MAPT expression.


Subject(s)
Apolipoprotein E4/genetics , Cerebral Cortex/pathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , tau Proteins/genetics , Aged , Apolipoprotein E4/biosynthesis , Atrophy/genetics , Female , Follow-Up Studies , Gene Expression Regulation , Genetic Variation/genetics , Humans , Magnetic Resonance Imaging/methods , Male , Polymorphism, Single Nucleotide/genetics , tau Proteins/biosynthesis
8.
Electrophoresis ; 33(24): 3745-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23161654

ABSTRACT

Apolipoprotein E (ApoE) is a major lipid carrier protein. In humans, ApoE is expressed in three polymorphic isoforms, which are encoded by three different alleles APOE2, APOE3, and APOE4. In the brains of Alzheimer's disease (AD) patients, each one of these three allelic isoforms is found in several "isoelectric" protein isoforms (qPI), i.e. protein isoforms resulting from PTMs altering the net charge (q) of the polypeptide. AD is a complex disease in which multiple causes and several risk factors affect the onset and disease outcome. A major risk factor for AD is ApoE4; therefore, it is important to characterize the different ApoE qPIs. We have implemented a detergent-based method for isolation and quantitation of protein isoforms, and we found differences in the solubility of protein isoforms depending on the type of solvent used. In this manuscript, we describe these methods and applied them to young human-ApoE targeted replacement mice. Our results indicate that there are no significant differences in the hippocampus proteome of these mice as a function of the APOE genotype.


Subject(s)
Apolipoprotein E3/biosynthesis , Apolipoprotein E4/biosynthesis , Proteome/analysis , Analysis of Variance , Animals , Apolipoprotein E3/analysis , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/analysis , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Creatine Kinase/analysis , Creatine Kinase/chemistry , Creatine Kinase/metabolism , Electrophoresis, Gel, Two-Dimensional , Genotype , Hippocampus/chemistry , Hippocampus/metabolism , Humans , Mice , Mice, Transgenic , Phosphopyruvate Hydratase/analysis , Phosphopyruvate Hydratase/chemistry , Phosphopyruvate Hydratase/metabolism , Protein Isoforms , Proteome/chemistry , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Solubility
9.
Arterioscler Thromb Vasc Biol ; 32(5): 1116-23, 2012 May.
Article in English | MEDLINE | ID: mdl-22441102

ABSTRACT

OBJECTIVE: Apolipoprotein (apo) E4 is an established risk factor for atherosclerosis, but the structural components underlying this association remain unclear. ApoE4 is characterized by 2 biophysical properties: domain interaction and molten globule state. Substituting Arg-61 for Thr-61 in mouse apoE introduces domain interaction without molten globule state, allowing us to delineate potential proatherogenic effects of domain interaction in vivo. METHODS AND RESULTS: We studied atherosclerosis susceptibility of hypomorphic Apoe mice expressing either Thr-61 or Arg-61 apoE (ApoeT(h/h) or ApoeR(h/h)mice). On a chow diet, both mouse models were normolipidemic with similar levels of plasma apoE and lipoproteins. However, on a high-cholesterol diet, ApoeR(h/h) mice displayed increased levels of total plasma cholesterol and very-low-density lipoprotein as well as larger atherosclerotic plaques in the aortic root, arch, and descending aorta compared with ApoeT(h/h) mice. In addition, evidence of cellular dysfunction was identified in peritoneal ApoeR(h/h) macrophages which released lower amounts of apoE in culture medium and displayed increased expression of major histocompatibility complex class II molecules. CONCLUSIONS: These data indicate that domain interaction mediates proatherogenic effects of apoE4 in part by modulating lipoprotein metabolism and macrophage biology. Pharmaceutical targeting of domain interaction could lead to new treatments for atherosclerosis in apoE4 individuals.


Subject(s)
Apolipoprotein E4/genetics , Atherosclerosis/genetics , DNA/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Animals , Apolipoprotein E4/biosynthesis , Atherosclerosis/etiology , Atherosclerosis/metabolism , Diet, Atherogenic/adverse effects , Disease Models, Animal , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Neurobiol Aging ; 33(3): 622.e17-28, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21482441

ABSTRACT

We examined the associations between extracerebral markers of cholesterol homeostasis and cognitive decline over 6 years of follow-up, and studied the modifying effect of apolipoprotein E (ApoE) e4. Data were collected in the Longitudinal Aging Study Amsterdam (n = 967, with longitudinal data on cognition, ages ≥ 65 years) and analyzed using linear mixed models. General cognition (Mini-Mental State Examination; MMSE), memory (Auditory Verbal Learning Test), and information processing speed (Coding task) were measured. The results show that ApoE e4 was a significant effect modifier. Significant associations were found only in ApoE e4 noncarriers (n = 718). We found a nonlinear negative association between the ratio of lanosterol to cholesterol (≤ 189.96 ng/mg), a marker for cholesterol synthesis, and general cognition. Lower cholesterol absorption, i.e., lower ratios of campesterol and sitosterol to cholesterol, as well as a higher rate of cholesterol synthesis relative to absorption were associated with lower information processing speed. In ApoE e4 carriers, the negative association between the ratio of campesterol to cholesterol and memory reached borderline significance. Future research should focus on the interaction between (disturbed) cholesterol homeostasis and ApoE e4 status with respect to dementia.


Subject(s)
Apolipoprotein E4/physiology , Brain Chemistry , Cholesterol/physiology , Cognition Disorders/metabolism , Homeostasis/genetics , Aged , Aged, 80 and over , Apolipoprotein E4/biosynthesis , Apolipoprotein E4/genetics , Brain Chemistry/genetics , Cholesterol/analogs & derivatives , Cholesterol/genetics , Cognition Disorders/genetics , Down-Regulation/genetics , Female , Follow-Up Studies , Genetic Carrier Screening , Humans , Longitudinal Studies , Male , Middle Aged , Phytosterols/genetics , Phytosterols/physiology
11.
Ann Med ; 43(4): 302-11, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21604997

ABSTRACT

INTRODUCTION. We have studied the functions of truncated apoE4 forms in vitro and in vivo in order to identify the domains of apoE4 required for the biogenesis of apoE-containing high-density lipoprotein (HDL). RESULTS. We have found that apoE4-185, -202, -229, or -259 could promote ATP-binding cassette transporter A1 (ABCA1)-dependent cholesterol efflux in vitro, although less efficiently than Full-length apoE4, and had diminished capacity to activate lecithin cholesterol acyltransferase (LCAT). Formation of HDL in vivo was assessed by various methods following gene transfer in apolipoprotein A-I(-/-) × apoE(-/-) mice. Fast protein liquid chromatography of plasma showed that the truncated apoE forms, except apoE4-185, generated an apoE-containing HDL peak. Two-dimensional gel electrophoresis of plasma and electron microscopy showed that truncated apoE forms generated distinct HDL subpopulations and formed discoidal HDL particles which could be converted to spherical by co-administration of truncated apoE4-202 and LCAT. CONCLUSION. Overall, the in-vivo and in-vitro data are consistent and indicate that apoE4-185 is the shortest truncated form that supports formation of discoidal apoE4-containing HDL particles.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Apolipoprotein A-I/physiology , Apolipoprotein E4/chemistry , Apolipoprotein E4/physiology , Apolipoproteins E/biosynthesis , Lipoproteins, HDL/biosynthesis , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Adenoviridae/genetics , Animals , Apolipoprotein A-I/metabolism , Apolipoprotein E4/biosynthesis , Apolipoprotein E4/metabolism , Apolipoproteins E/metabolism , Female , Humans , Lipoproteins/biosynthesis , Lipoproteins/metabolism , Lipoproteins, HDL/metabolism , Male , Metabolic Networks and Pathways , Mice , Mice, Knockout/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , RNA, Messenger , Transduction, Genetic
12.
Neurobiol Aging ; 32(7): 1236-48, 2011 Jul.
Article in English | MEDLINE | ID: mdl-19713000

ABSTRACT

The apolipoprotein E (ApoE) ε4 allele has consistently been established as an Alzheimer's disease (AD) risk factor, but its pathological contribution to AD is obscure. Certain butyrylcholinesterase (BuChE) polymorphisms together with the ApoE ε4 allele synergistically increase the risk of AD. In addition, AD risk factors, i.e. advanced age, female gender and ApoE ε4 are associated with different levels of CSF BuChE in AD patients, and BuChE protein attenuates Aß fibrillization in vitro. Here we investigated the roles of ApoE and BuChE gene products as modulators of pathological features of AD in vivo. We found that AD risk factors were associated with different levels of ApoE protein in the CSF of AD patients (n=115). Women and ApoE ε4 carriers had the highest levels of ApoE protein (up by 50-120%, p<0.01-0.0001), which were increased with age (r=0.30, p<0.0006). The CSF surrogate markers of pathological features of AD, i.e. high tau and P-tau, low Aß(42) and high tau/Aß(42) ratio, were associated with high levels of ApoE protein. Intriguingly, high ApoE protein levels were not only associated with low amounts of BuChE, but they also altered the aging and activity of this enzyme in concentration- and isoform-dependent manners, particularly in the presence of Aß peptides. Both ApoE and BuChE levels were also differentially related to levels of the proinflammatory cytokine IL-1ß. In conclusion, ApoE ε4 might impart its pathological role through high protein expression and interaction with BuChE, which in turn might modulate central cholinergic activity and Aß load in the brain.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Apolipoprotein E4/genetics , Butyrylcholinesterase/cerebrospinal fluid , Butyrylcholinesterase/genetics , Aged , Alleles , Alzheimer Disease/enzymology , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Apolipoprotein E4/biosynthesis , Apolipoprotein E4/cerebrospinal fluid , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Brain/enzymology , Brain/metabolism , Brain/pathology , Butyrylcholinesterase/biosynthesis , Female , Gene Expression Regulation, Enzymologic , Humans , Male , Protein Interaction Mapping , Risk Factors
13.
Neurobiol Aging ; 31(8): 1340-54, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20570401

ABSTRACT

Previously it was reported that Alzheimer's disease (AD) patients have reduced beta amyloid (Abeta(1-42)) and elevated total tau (t-tau) and phosphorylated tau (p-tau(181p)) in the cerebrospinal fluid (CSF), suggesting that these same measures could be used to detect early AD pathology in healthy elderly individuals and those with mild cognitive impairment (MCI). In this study, we tested the hypothesis that there would be an association among rates of regional brain atrophy, the CSF biomarkers Abeta(1-42), t-tau, and p-tau(181p) and apolipoprotein E (ApoE) epsilon4 status, and that the pattern of this association would be diagnosis-specific. Our findings primarily showed that lower CSF Abeta(1-42) and higher tau concentrations were associated with increased rates of regional brain tissue loss and the patterns varied across the clinical groups. Taken together, these findings demonstrate that CSF biomarker concentrations are associated with the characteristic patterns of structural brain changes in healthy elderly and mild cognitive impairment subjects that resemble to a large extent the pathology seen in AD. Therefore, the finding of faster progression of brain atrophy in the presence of lower Abeta(1-42) levels and higher tau levels supports the hypothesis that CSF Abeta(1-42) and tau are measures of early AD pathology. Moreover, the relationship among CSF biomarkers, ApoE epsilon4 status, and brain atrophy rates are regionally varying, supporting the view that the genetic predisposition of the brain to beta amyloid and tau mediated pathology is regional and disease stage specific.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Apolipoprotein E4/genetics , Brain/pathology , Peptide Fragments/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Apolipoprotein E4/biosynthesis , Atrophy , Biomarkers/cerebrospinal fluid , Cognition Disorders/cerebrospinal fluid , Cognition Disorders/genetics , Cognition Disorders/pathology , Female , Gene Expression Profiling/methods , Humans , Longitudinal Studies , Magnetic Resonance Imaging/methods , Male , Prospective Studies
14.
J Proteome Res ; 7(8): 3535-42, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18557639

ABSTRACT

We report a cell-free approach for expressing and inserting integral membrane proteins into water-soluble particles composed of discoidal apolipoprotein-lipid bilayers. Proteins are inserted into the particles, circumventing the need of extracting and reconstituting the product into membrane vesicles. Moreover, the planar nature of the membrane support makes the protein freely accessible from both sides of the lipid bilayer. Complexes are successfully purified by means of the apoplipoprotein component or by the carrier protein. The method significantly enhances the solubility of a variety of membrane proteins with different functional roles and topologies. Analytical assays for a subset of model membrane proteins indicate that proteins are correctly folded and active. The approach provides a platform amenable to high-throughput structural and functional characterization of a variety of traditionally intractable drug targets.


Subject(s)
Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Antiporters/biosynthesis , Antiporters/chemistry , Antiporters/genetics , Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Apolipoprotein E4/biosynthesis , Apolipoprotein E4/chemistry , Apolipoprotein E4/genetics , Bacteriorhodopsins/biosynthesis , Bacteriorhodopsins/chemistry , Bacteriorhodopsins/genetics , Chromatography, Gel , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Microscopy, Atomic Force , Solubility
15.
Neurodegener Dis ; 5(3-4): 186-9, 2008.
Article in English | MEDLINE | ID: mdl-18322386

ABSTRACT

BACKGROUND: The apolipoprotein E (APOE) epsilon4 allele is a risk factor for Alzheimer's disease. Earlier studies have shown differences in brain structure according to the APOE epsilon4 status. OBJECTIVE: To assess possible differences in brain structure according to the APOE epsilon4 status in mild cognitive impairment (MCI) subjects in relation to conversion to dementia. METHODS: In a follow-up study of 56 MCI subjects, 13 MCI subjects progressed to dementia (PMCI) during a mean follow-up time of 31 months. Brain structure differences in both stable MCI (SMCI) and PMCI epsilon4 carriers and noncarriers in the baseline MRI scan were assessed with voxel-based morphometry. RESULTS: The SMCI epsilon4 carriers had atrophy in the amygdala and hippocampus compared to the SMCI noncarriers. The PMCI epsilon4 carriers revealed atrophy of the left inferior frontal gyrus and parietal cortex compared to the PMCI noncarriers. CONCLUSION: The rate of brain atrophy in certain brain areas may be increased in epsilon4-positive MCI subjects progressing to dementia.


Subject(s)
Alleles , Apolipoprotein E4/genetics , Cerebral Cortex/pathology , Cognition Disorders/genetics , Cognition Disorders/pathology , Dementia/genetics , Aged , Aged, 80 and over , Apolipoprotein E4/biosynthesis , Atrophy , Brain Mapping/methods , Cerebral Cortex/physiology , Cognition Disorders/psychology , Cohort Studies , Dementia/pathology , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Middle Aged , Risk Factors
16.
Proc Natl Acad Sci U S A ; 105(4): 1343-6, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18212130

ABSTRACT

Convergent evidence has revealed an association between insulin resistance and Alzheimer's disease (AD), and the peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonist, rosiglitazone, an insulin sensitizer and mitochondrial activator, improves cognition in patients with early or mild-to-moderate AD. Apolipoprotein (apo) E4, a major genetic risk factor for AD, exerts neuropathological effects through multiple pathways, including impairment of dendritic spine structure and mitochondrial function. Here we show that rosiglitazone significantly increased dendritic spine density in a dose-dependent manner in cultured primary cortical rat neurons. This effect was abolished by the PPAR-gamma-specific antagonist, GW9662, suggesting that rosiglitazone exerts this effect by activating the PPAR-gamma pathway. Furthermore, the C-terminal-truncated fragment of apoE4 significantly decreased dendritic spine density. Rosiglitazone rescued this detrimental effect. Thus, rosiglitazone might improve cognition in AD patients by increasing dendritic spine density.


Subject(s)
Apolipoprotein E4/physiology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Dendrites/drug effects , Dendritic Spines/drug effects , Neurons/metabolism , Thiazolidinediones/pharmacology , Animals , Animals, Newborn , Apolipoprotein E4/biosynthesis , Cell Count , Cells, Cultured , Cerebral Cortex/pathology , Neurons/pathology , Rats , Rosiglitazone
17.
Biochim Biophys Acta ; 1768(8): 1943-51, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17321494

ABSTRACT

One of the fundamental questions regarding the pathogenesis of Alzheimer's disease (AD) is how the monomeric, nontoxic amyloid beta-protein (Abeta) is converted to its toxic assemblies in the brain. A unique Abeta species was identified previously in an AD brain, which is characterized by its binding to the GM1 ganglioside (GM1). On the basis of the molecular characteristics of this GM1-bound Abeta (GAbeta), it was hypothesized that Abeta adopts an altered conformation through its binding to GM1, and GAbeta acts as a seed for Abeta fibrillogenesis in an AD brain. To date, various in vitro and in vivo studies of GAbeta have been performed, and their results support the hypothesis. Using a novel monoclonal antibody specific to GAbeta, it was confirmed that GAbeta is endogenously generated in the brain. Regarding the role of gangliosides in the facilitation of Abeta assembly, it has recently been reported that region-specific deposition of hereditary variant-type Abetas is determined by local gangliosides in the brain. Furthermore, it is likely that risk factors for AD, including aging and the expression of apolipoprotein E4, alter GM1 distribution on the neuronal surface, leading to GAbeta generation.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoprotein E4/biosynthesis , Cell Membrane/metabolism , G(M1) Ganglioside/metabolism , Neurons/metabolism , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Apolipoprotein E4/genetics , Brain/metabolism , Brain/pathology , Cell Membrane/genetics , Cell Membrane/pathology , G(M1) Ganglioside/genetics , Humans , Neurons/pathology , Protein Binding/genetics , Risk Factors
18.
Article in English | MEDLINE | ID: mdl-16961266

ABSTRACT

The carboxyl-terminal amino acids 272-299-truncated apoE4 (delta272-299) is the main fragments of apoE4 hydrolysate in neurons. The effects of truncated-ApoE4 (delta272-299) overexpression on tau phosphorylation in cultured N2a cells were investigated. The truncated-apoE4 (delta272-299) cDNA was subcloned into pEGFP-c3 to form recombinant pEGFP-T-apoE4. pEGFP-c3, pEGFP-T-apoE4 and pEGFP-apoE4 were transfected into N2a cells respectively by lipofectamine 2000 method. After 24--48 h, tau phosphorylation was detected by Western blot assay and glycogen synthase kinase-3 (GSK-3) activity by using GSK-3 activity assay. The results showed that the overexpression of both full length-apoE4 and truncated apoE4 fragments in N2a cells induced a dramatic increase in phosphorylation of tau at Ser202 sites and the activation of GSK-3 as compared with untransfected cells, most significantly in the cells transfected with pEGFP-T-apoE4 (P < 0.05). It was concluded that in vitro overexpression of truncated-ApoE4 (delta272-299) can result in tau hyperphosphorylation in N2a cells by activating GSK-3, suggesting truncated-ApoE4 (delta272-299) might contribute the pathogenesis of Alzheimer disease.


Subject(s)
Apolipoprotein E4/biosynthesis , tau Proteins/metabolism , Animals , Apolipoprotein E4/genetics , Blotting, Western , Cell Line, Tumor , Glycogen Synthase Kinase 3/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phosphorylation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...