Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2011

Open Access 01.12.2011 | Research

Apolipoprotein E expression is elevated by interleukin 1 and other interleukin 1-induced factors

verfasst von: Ling Liu, Orwa Aboud, Richard A Jones, Robert E Mrak, W Sue T Griffin, Steven W Barger

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2011

Abstract

Background

We have previously outlined functional interactions, including feedback cycles, between several of the gene products implicated in the pathogenesis of Alzheimer's disease. A number of Alzheimer-related stressors induce neuronal expression of apolipoprotein E (ApoE), β-amyloid precursor protein (βAPP), and fragments of the latter such as amyloid β-peptide (Aβ) and secreted APP (sAPP). These stressors include interleukin-1 (IL-1)-mediated neuroinflammation and glutamate-mediated excitotoxicity. Such circumstances are especially powerful when they transpire in the context of an APOE ε4 allele.

Methods

Semi-quantitative immunofluorescence imaging was used to analyze rat brains implanted with IL-1β slow-release pellets, sham pellets, or no pellets. Primary neuronal or NT2 cell cultures were treated with IL-1β, glutamate, Aβ, or sAPP; relative levels of ApoE mRNA and protein were measured by RT-PCR, qRT-PCR, and western immunoblot analysis. Cultures were also treated with inhibitors of multi-lineage kinases--in particular MAPK-p38 (SB203580), ERK (U0126), or JNK (SP600125)--prior to exposure of cultures to IL-1β, Aβ, sAPP, or glutamate.

Results

Immunofluorescence of tissue sections from pellet-implanted rats showed that IL-1β induces expression of βAPP, IL-1α, and ApoE; the latter was confirmed by western blot analysis. These protein changes were mirrored by increases in their mRNAs, as well as in those encoding IL-1β, IL-1β-converting enzyme (ICE), and tumor necrosis factor (TNF). IL-1β also increased ApoE expression in neuronal cultures. It stimulated release of sAPP and glutamate in these cultures too, and both of these agents--as well as Aβ--stimulated ApoE expression themselves, suggesting that they may contribute to the effect of IL-1β on ApoE levels. Inhibitors of MAPK-p38, ERK, and JNK inhibited ApoE induction by all these agents except glutamate, which was sensitive only to inhibitors of ERK and JNK.

Conclusion

Conditions of glial activation and hyperexcitation can elevate proinflammatory cytokines, ApoE, glutamate, βAPP, and its secreted fragments. Because each of these factors promotes glial activation and neuronal hyperexcitation, these relationships have the potential to sustain self-propagating neurodegenerative cycles that could culminate in a progressive neurodegenerative disorder such as Alzheimer's disease.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1742-2094-8-175) contains supplementary material, which is available to authorized users.

Competing interests

S.W.B. receives royalty payments from Sigma-Aldrich Corp., which manufactures some of the reagents utilized in this study. The authors have no other conflicts to declare.

Authors' contributions

LL performed the cell culture experiments and contributed to writing the first draft of the manuscript. OA performed the immunofluorescence and assisted with western blots and writing. RAJ performed the experiments with rat brain tissue. REM contributed to interpretation of the results and to writing. WSTG contributed to the design of the study, interpretation of the results, and writing. SWB made essential contributions to the design of the study and interpretation of the results and completed the final draft of the manuscript. All authors read and approved the final manuscript.

Introduction

The pluripotent glial cytokine interleukin-1 (IL-1) and the CNS-abundant, lipid-cholesterol-carrying protein apolipoprotein E (ApoE) are key participants in the pathogenesis of Alzheimer's disease (AD). ApoE contributes both to learning and to recovery from neural injury [1], perhaps by enhancing synaptogenesis by influencing Reelin signaling [2, 3]. In humans, single-nucleotide polymorphisms in the coding region of the ApoE gene (APOE) yield three alleles (ε2, ε3, ε4) that translate into three distinct protein sequences, ApoE2, ApoE3, and ApoE4. Inheritance of the particular isoform of ApoE encoded by the ε4 variant of the APOE gene confers significant risk for precocious development of AD [4, 5]: those with two copies of the ε4 allele of APOE have a 50-90% chance of developing AD by the age of 85, and even one copy confers a three-fold increase in risk over individuals with no ε4 alleles [6]. Though ApoE is primarily expressed in astrocytes in the healthy brain, stressors can induce its expression in neurons [7, 8].
Although not as strongly associated with AD risk as possession of ApoE4 sequences, specific polymorphisms in the genes encoding IL-1α and IL-1β are also associated with increased AD risk. Specifically, variations in the promoter region of IL1A and in the coding region of IL1B influence AD risk when homozygous in one gene or heterozygous in both [913]. Glial activation marked by excess production of both IL-1α and β is a constant feature in several conditions associated with increased risk for precocious development of AD: i) traumatic brain injury (TBI) [14], ii) systemic viral disease, e.g., AIDS [15]; iii) the neuronal hyperexcitability of epilepsy [1619]; iv) chromosome 21 anomalies such as Down's syndrome [20]; and v) advancing age [2123]. Each of these stressors is associated with precocious development of AD [18, 24, 25], especially in those who have inherited one or more ε4 alleles of APOE [1, 2629].
Excess production and secretion of IL-1β elevates neuronal expression of the precursors of each of the changes characteristic of AD. These neurodegeneration-related precursors include β-amyloid precursor protein (βAPP), which may lead in vivo to deposition of Aβ [30] and further induction of IL-1β [31]; ApoE, which is present in plaques [32] and necessary for the accumulation of Aβ deposits [33]; and hyperphosphorylated tau [5], the principal component of neurofibrillary tangles. IL-1 also induces α-synuclein [34], the Lewy body precursor.
Despite the potential for contributing to the production of Aβ, elevations of βAPP may participate in compensatory responses. βAPP is elevated in response to stressors beyond IL-1β, including excitotoxins and age itself, yet AD pathology is correlated with a deficiency in βAPP expression [35]. ApoE appears to mediate the compensatory induction of βAPP; blocking ApoE synthesis or its receptors inhibits the effect of glutamate on βAPP [35]. βAPP knockout mice show learning and memory deficits [36] and die prematurely [37]; secreted βAPP (sAPP) is generally neuroprotective [38]. Taken together, these findings suggest that possession of an ε4 allele or ApoE insufficiency compromises neurological parameters and exacerbates injury-induced deficits at least in part by limiting inductions of βAPP. ApoE, especially ApoE3, may also serve to keep inflammatory reactions in check [3941]. A possible mechanism is suggested by the ability of ApoE to suppress the proinflammatory activity of sAPP [31].
In AD, activated microglia overexpressing IL-1 are present in diffuse Aβ deposits prior to the appearance of ApoE [32]. With normal aging, the brain shows increased microglial activation and expression of IL-1 [21], as well as neuronal expression of both ApoE and βAPP [35]. The ability of IL-1β to induce βAPP expression [30, 42] raises the question of whether this is a direct mechanism or an indirect phenomenon resulting from ApoE induction, similar to the effect of glutamate [35]. In view of the relations between the AD-related stressors and the importance of ApoE in risk for development of AD, together with the neuropathological changes observed in AD patients, we tested the hypothesis that ApoE would be elevated in CNS neurons secondary to several AD-related stressors associated with excessive expression of IL-1. Specifically, rat primary cortical neurons and a neuropotent human cell line (NTera2) were assessed for ApoE expression after treatment with IL-1β, sAPP, glutamate, or Aβ. To delineate the roles of multi-lineage kinase (MLK) pathways in the induction of neuronal ApoE expression, we utilized inhibitors of p38-MAPK, ERK, and JNK pathways. To determine if such changes in ApoE expression might be observed in vivo, and the potential relationship of such changes to other proteins that are induced by IL-1, we measured the expression of ApoE, βAPP, and other neuroinflammatory proteins in rat brains exposed to excess IL-1β.

Materials and methods

Pellet Implantation

Pellets (1.5 mm in diameter, designed for controlled slow release of compounds over a 21-day period; Innovative Research of America, Sarasota, FL) impregnated with IL-1β (100 ng recombinant mouse IL-1β; Sigma Chemical Company, St. Louis, MO) and 'control' pellets (with bovine serum albumin) were implanted 2.8 mm caudal to bregma, 4.5 mm right of the midline, and 2.5 mm below the pial surface. Twenty-one male Sprague-Dawley rats, weighing 264 ± 6 g, were randomly assigned to three groups. Eight rats received implants of 21-day timed-release IL-1β-containing pellets, seven rats received sham (vehicle) pellets, and six rats served as unoperated controls. Twenty-one days after implantation, cortices from left hemispheres were collected for protein and mRNA isolation. For histological study, brain tissues were fixed in 10% formalin, embedded in paraffin, sectioned at 7 μm, and prepared for immunohistochemical analysis. All animal studies were conducted in accordance with a protocol reviewed and approved by the Institutional Animal Care and Use Committee of the Central Arkansas Veterans Healthcare System.

Reagents

Rat recombinant mature IL-1β (IL-1β holoprotein cleavage product) was purchased from Sigma (St. Louis MO), secreted APP (sAPP) was purified from a recombinant expression system as described previously [42], and L-glutamate was from Sigma (St. Louis MO). Aβ1-42, from US Peptide Inc. (Rancho Cucamonga CA), was dissolved in DMSO and then incubated at 4°C overnight prior to use. Rabbit anti-mouse IL-1β antibody was from Chemicon (Temecula CA); goat anti-human apolipoprotein E was from Calbiochem (Sunnyvale CA). Inhibitors of the p38-MAPK (SB203580), ERK (U0126), and JNK (SP600125) pathways were from Calbiochem. Medium, serum, and B27 supplement for cell cultures were from Invitrogen/Life Technologies (Grand Island NY). The antibodies used were rabbit anti-human IL-1α (Peprotech 4:1000), goat anti-human APP (ADI 1:50), goat anti-Human APO E (Invitrogen 1:50), diluted in antibody diluent (Dako, Carpiteria CA).

Immunofluorescence

For immunofluorescent analysis of brain tissues, paraffin blocks were sectioned at 7 μm and placed on pre-cleaned microscope slides (Fischer). Then, sections were deparaffinized in xylene, rehydrated in graduated ethanol solutions to deionized water. For IL-1α immunoreactions, sections were placed in boiling sodium citrate buffer (0.01 M, pH 6.0) for 20 minutes. Sections for βAPP and ApoE were placed in trypsin solution for 10 minutes at 37°C, all sections were blocked using protein block (Dako). For each antibody, sections were incubated overnight at room temperature. The secondary antibodies, Alexa Fluor donkey anti-goat and donkey anti-rabbit were diluted in antibody diluent (Dako) and sections were incubated for 60 minutes. The sections were then washed in three changes 5 minutes each of distilled H2O and then coverslipped with prolong Gold with DAPI (Invitrogen).

Image Analysis

Similar to our previous study [35], a quantitative approach was used to examine mean intensities of immunoreactions. Three representative images per slide (40× magnification) from IL-1-pellet, sham, and unoperated rat brains were obtained at identical exposure settings, using a Nikon Eclipse E600 microscope equipped with a Coolsnap monochrome camera. Each of the three images in each tissue section spanned a total area of 37241.5 μm2. These images were from hippocampal CA1 and two cortical regions, one at the midline and another at the superior aspects of the temporal cortex and were acquired and analyzed using NIS-Elements BR3 software (Nikon.com). All cells of a type were captured, and images were thresholded. Data obtained from cells in each of the three regions were averaged, thus providing a single value for each image, and this value was used for statistical analysis. Data were analyzed by ANOVA to assess difference among groups. A statistical value of p ≤ 0.05 was defined as being significant.

Cell Cultures

Primary neuronal cultures were derived from cerebral cortex of fetal Spraque-Dawley rats (embryonic day 18), as previously described [43]. Experiments using primary neuronal cell cultures were performed after 10-14 days in culture. Highly purified cultures of rat microglia and astrocytes were generated from the cortical tissue of neonatal (0-3 days) Sprague-Dawley rats, as described previously [43, 44]. The NTera2 human cell line (Stratagene, La Jolla, CA) was maintained in Dulbecco's modified Eagle medium (DMEM; Invitrogen/Life Technologies, Grand Island, NY) supplemented to 10% with fetal bovine serum (FBS). For specific experiments, SB203580 (5 μM), U0126 (5 μM), or SP600125 (5 μM) was applied to cultures one hour before application of a stimulus. Glutamate released in the culture medium was assayed with a kit that utilizes a glutamate dehydrogenase-coupled color reaction (Roche Diagnostics, Indianapolis IN).

Reverse Transcription (RT) Reaction and Polymerase Chain Reaction (PCR) Amplification

Total RNA was extracted from cultured cells using TriReagent™ RNA (Molecular Research Center Inc., Cincinnati OH) according to the manufacturer's instructions. Gel-based RT-PCR was performed as described previously [45]. Briefly, RT reactions were performed simultaneously using reagents from a single master mix, and PCR was performed using reagents from Clontech (Palo Alto CA). Aliquots of the product were resolved on agarose gels, ethidium bromide staining was captured by digital camera, and pixel intensities were quantified with Scion Image 4.0.3.2. Conditions were established to ensure that maximal cycle number fell within the linear phase of amplification. Real-time (quantitative) RT-PCR was performed as described previously [35]. RT utilized random hexamers for priming, and PCR was performed with the Power SYBR-Green PCR Master Mix in an ABI 7900 HT Fast Real-time PCR System (Applied Biosystems, Foster City, CA). Signals were interpolated within standard curve reactions performed for each primer set, and the result for ApoE was expressed as a fraction of the 18S signal for each sample. All primer sequences, annealing temperatures, and number of cycles are provided in Table 1.
Table 1
RT-PCR primers
Gene
Sequences
Annealing
temp. (°C)
Cycle no.
Gel-based PCR, rat
ApoE
F: TGT TGG TCC CAT TGC TGA CAG GAT
60
25
 
R: TGG TGT TTA CCT CGT TGC GGT ACT
  
βAPP
F: CCC TGA CGC AGT CGA CAA GT
61
25
 
R: TGT CAT AAC CTG GGA CCG GAT
  
GAPDH
F: CAA CGG ATT TGG CCG TAT TG
61
25
 
R: TGG GGG TAG GAA CAC GGA A
  
ICE
F: ACA AAG AAG GTG GCG CAT TTC
61
28
 
R: CCT TGT TTC TCT CCA CGG C
  
IL-1α
F: GAG TCA ACT CAT TGG CGC TTG
60
27
 
R: GGG CTG ATT GAA ACT TAG CCG
  
IL-1β
F: TGA CTC GTG GGA TGA TGA CG
61
27
 
R: CTG GAG ACT GCC CAT TCT CG
  
TNF
F: GCA CAG AAA GCA TGA TCC GAG
64
28
 
R: CCT GGT ATG AAG TGG CAA ATC G
  
Gel-based PCR, human
ApoE
F: TTC TGT GGG CTG CGT TGC TG
60
25
 
R: TAC ACT GCC AGG CGC TTC TG
  
GAPDH
F: AGG TCG GAG TCA ACG GAT TTG
61
25
 
R: TGG CAG GTT TTT CTA GAC GGC
  
Real-time PCR (rat)
ApoE
F: CTG GTT CGA GCC GCT AGT G
60
N.A.
 
R: CCT GTA TCT TCT CCA TTA GGT TTG C
  
18S
F: TTC GAA CGT CTG CCC TAT CAA
60
N.A.
 
R: ATG GTA GGC ACG GCG ACT A
  

Western Immunoblot Assay

Cellular fractions were prepared by application of a lysis buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1% Nonidet P40, 0.5% sodium deoxycholate and 0.1% SDS) to the cultures after a wash with cold PBS. Tissue samples were prepared by homogenization in RIPA buffer (Cell Bioscience) as described previously [35, 42]. Lysates were quantified using a Micro BCA assay reagent kit (Pierce, Rockford IL) as described previously [43]. Aliquots (100 μg each) were resolved by SDS-PAGE, subjected to electrophoresis at 70 V for 20 minutes and 90 V for 1.5 h, and transferred to nitrocellulose membranes. After transfer, each blot was stained with Ponceau S to ensure even loading of protein across lanes. Blots were then blocked in I-Block Buffer (Applied Biosystem Inc., Bedford, MA) for 45 minutes, then incubated overnight at 4°C with goat anti-human ApoE (1:2000) primary antibody, incubated for 1 h at room temperature with alkaline phosphatase-conjugated secondary antibody, and developed using the Western-Light™ Chemiluminescent Detection System (Applied Biosystem Inc) and exposure to x-ray film. Digital images were captured and analyzed using NIH Image software, version 1.60.

Statistical Analysis

Comparisons between two conditions were made via unpaired t-test, and experiments with a greater number of variables were subjected to ANOVA with Fisher's post hoc test. Differences were considered significant at p-values ≤0.05.

Results

Chronic IL-1β increases the expression of ApoE, βAPP, and neuroinflammatory factors in rat brain

Rats were implanted with either slow-release (21-day) IL-1β-impregnated pellets or vehicle-impregnated sham pellets. Cerebral cortices from these rats, as well as unoperated control rats, were processed for protein or mRNA tissue level analyses or were fixed and processed for immunofluorescent image analyses. Rat brains implanted with IL-1β-containing pellets had markedly elevated steady-state levels of ApoE mRNA (Figure 1A,B) and of ApoE protein (Figure 1C,D) compared to those in rats implanted with sham pellets or to unoperated controls (p < 0.01).
Neuroinflammatory conditions and models thereof often exhibit chain reactions of multiple effectors working sequentially, in parallel, or in feedback loops fomenting a persistent and progressive situation. In this vein, the ability of IL-1β to elevate the levels of IL-α prompted an examination of gene-expression indices of neuroinflammation in this chronic IL-1β delivery paradigm. The increase in IL-1α immunofluorescence noted above was found to be reflected at the mRNA level (Figure 2). Chronic IL-1β also elevated mRNA levels of endogenous IL-1β, as well as its cleavage enzyme ICE (Figure 2). Along with these changes in IL-1-related molecules, the mRNA for the proinflammatory cytokine TNF was elevated. These proinflammatory changes were accompanied by induction of βAPP mRNA (Figure 2), consistent with the immunofluorescence results and prior studies of IL-1/βAPP interactions.
The induction of ApoE in the cortex by IL-1β pellets was also detectable by immunofluorescence (Figure 3A), which demonstrated neuronal localization. IL-1β pellets also elevated expression of IL-1α in the CA1 of hippocampus (Figure 3B). This IL-1α induction was localized principally to cells with astrocytic morphology. Pyramidal neurons of the CA1 overexpressed βAPP in response to the chronic delivery of IL-1β (Figure 3C).

Tissue culture studies reveal potential for indirect impacts of IL-1β on ApoE

To examine the impact of IL-1β on ApoE expression in greater temporal and mechanistic detail, we utilized two types of neuronal cell culture: primary cultures of rat cortical neurons and the human NTera2 (NT2) cell line. We previously demonstrated that glutamate elevates βAPP expression via a mechanism that requires the biological activity of ApoE [35]. Moreover, IL-1β has been shown to influence the processing of βAPP [46]. Therefore, we tested whether ApoE expression was responsive to these agents and another derivative of βAPP: Aβ1-42. In both culture types, expression of ApoE mRNA was elevated approximately two-fold by exposure to IL-1β, Aβ1-42, or glutamate for 20 h; the induction by sAPP exceeded six-fold (Figure 4A). All of these agents were found to elevate ApoE protein levels as well (Figure 4B).
The ability of glutamate and βAPP fragments to impact ApoE was given additional relevance by demonstration of impacts of IL-1β on these agents. Levels of glutamate released into neuronal culture medium was elevated by IL-1β (Figure 5A). Likewise, IL-1β elevated the levels of sAPP in the culture medium of primary neurons in a dose-dependent fashion (Figure 5B). Glutamate induction of ApoE in primary neurons was confirmed by immunofluorescence, which also documented a larger induction by Aβ1-42 (Figure 5C,D). Intriguingly, coapplication of glutamate in combination with Aβ1-42 reduced the induction to one on par with that of glutamate alone (Figure 5D).

Regulation of ApoE expression by IL-1β, Aβ, sAPP, and glutamate is via multi-lineage kinase pathways

Each of the IL-1β-induced entities, sAPP and glutamate, as well as Aβ, were shown to elevate ApoE expression in both primary neurons and NT2 cells (Figure 5). To begin investigating the mechanisms involved in the induction of such ApoE expression, we focused on multi-lineage kinases (MLKs) previously shown to regulate cytokine-induced AD-related proteins [47]. Primary neurons and NT2 cells were incubated with inhibitors of three principle MLK pathways, viz., the MEK/ERK (U0126), MAPK-p38 (SB203580), and JNK (SP600125) pathways. Constitutive expression of ApoE in both primary neurons and NT2 cells was unaffected by treatment with these inhibitors (data not shown). However, each of these MLK inhibitors suppressed induction of ApoE by IL-1β (Figure 6A), Aβ1-42 (Figure 6B), and sAPP (Figure 6C) in both types of culture. Induction of ApoE by glutamate in both NT2 and primary neurons was not inhibited by SB203580, a MAPK-p38 inhibitor. Thus, regulation of ApoE expression by MLK pathways appears to be somewhat selective and dependent on the effector of its induction; in the case of glutamate, ERK and JNK activity is involved but not MAPK-p38 (Figure 6D).

Discussion

The neuroinflammagenic potential of IL-1β is shown here through its induction of synthesis of itself and other proinflammatory cytokines including TNF, IL-1α, IL-1β, as well as the latter's maturation enzyme ICE. The additional impact of IL-1β on neuronal ApoE production shown here suggests that in neurological conditions where the expression of proinflammatory cytokines is elevated, the expression of IL-1-driven AD-related proteins such as ApoE would be elevated as well. Multiple MLKs--ERK, p38-MAPK, and JNK--were shown to be involved in elevated expression of ApoE in neurons exposed to IL-1β, Aβ, or sAPP. The increased expression of ApoE induced by glutamate was mediated by ERK and JNK, but not by MAPK-p38. Together, these findings have several implications for AD pathogenesis, particularly with respect to conditions in which neuroinflammation is prominent, especially those influenced by APOE genotype.
The actions of IL-1 and the other agents tested here--sAPP, Aβ, and glutamate--create the possibility for complex loops of influence analogous to the vicious circle of neuroinflammatory events we have termed the Cytokine Cycle [48]. Glutamate can elevate neuronal expression of βAPP and its conversion to sAPP [42]. βAPP is elevated in dystrophic neurites in and around plaques [49]; its breakdown into both sAPP and Aβ can result in induction of IL-1β in microglia [31, 50]. In addition to inducing IL-1β expression and release, sAPP and Aβ also stimulate microglia to release biologically relevant levels of glutamate and its cooperative excitatory amino acid D-serine [44, 51, 52]. Thus, future studies should address the potential for each of these agents to act indirectly through the elaboration of a key agent or agents that can directly stimulate ApoE expression.
Key to interpretation of our findings--and, indeed, to the role of APOE genotype in AD--is determining whether elevation of ApoE levels would be beneficial or harmful. Possession of the ε4 allele is associated with enhanced deposition of Aβ [53], consistent with in vitro studies wherein ApoE was shown to enhance Aβ fibrillogenesis [54]. In this regard, ApoE4 has been shown to be more effective than ApoE3, fostering speculation that replacement of the ε3 allele by ε4 merely enhances an activity already present in ApoE3. This has been described as a toxic gain of function, implying that overabundance of any ApoE--even ApoE2 or ApoE3--would also create a gain in this function and thus be detrimental. Moreover, transient increases in cellular ApoE occur in response to injuries that promote AD, e.g., traumatic brain injury [55] and stroke [56]. ApoE4 is generally reported to be present at higher steady-state levels than ApoE3 in CSF or brain parenchyma [5761], though some studies have reported lower levels of total ApoE in ε4-positive individuals [62, 63].
In contrast to these connections to pathology, ApoE provides neuroprotection in many paradigms, and ApoE deficiency has proved detrimental in several respects [64]. Therefore, inductions of ApoE by the stimuli we tested may represent a compensatory response, meaning that the distinction between ApoE3 and ApoE4 represents loss of a beneficial function. ApoE has anti-inflammatory effects, and even its interaction with Aβ can attenuate glial activation by the latter [65]. However, ApoE3 is more effective than ApoE4 as an anti-inflammatory agent [31, 65, 66], so this putative compensatory response may be inadequate in ε4-positive individuals and thus allow more extensive propagation of the Cytokine Cycle. Such an allele-specific compensatory response may also extend to direct neuroprotective activity. We previously reported that ApoE3 induces βAPP expression but ApoE4 does not [35], confirming the findings of Ezra et al. [67]. In this regard, elevations of ApoE by the process of neuroinflammation, or other stressors, would reflect a requisite role for the lipoprotein in enhancing the beneficial roles of βAPP and/or other acute-phase response proteins. Thus, it would be the inability of ApoE4 to participate in this chain of salutary events that makes it detrimental. We have previously shown that the increase in ApoE brain levels that occurs with aging continues to occur in AD, with a large fraction being deposited in plaques [35]. This increase in ApoE levels is distinguishable from changes in βAPP, which rises with age but declines markedly in AD [35]. This disease-associated severance of the coordinate regulation of ApoE and βAPP further strengthens the correlation of brain health with the coregulation of these two proteins; to wit, with ApoE expression itself, provided that the ApoE is not ApoE4.
Multi-lineage kinase pathways may be invoked in the regulation of ApoE expression, and can themselves be invoked by ApoE [68, 69], suggesting a feedback loop between MLK pathways and ApoE expression in neurons. Our findings of involvement of multiple MLKs--ERK, p38-MAPK, and JNK--in expression of ApoE in neurons exposed to IL-1β, Aβ, or sAPP, together with previous reports of ERK pathway invocation of ApoE expression and vice versa, are consistent with the existence of a complex feedback system that may be important in acute-phase responses to neuronal injury as well as potential exacerbation of neurodegenerative events. Our finding that glutamate regulates ApoE expression via ERK and JNK, but not by p38-MAPK pathways may be indicative of a correlation between glutamatergic induction of ApoE and neuronal survival. Excitotoxic effects of glutamate are largely dependent upon activation of extrasynaptic NMDA receptors, p38-MAPK, and the inhibition of ERK signaling; synaptic receptors, on the other hand, appear to activate ERK and promote survival [7072].
In conclusion, the induction of neuronal ApoE by either neuroinflammatory or excitotoxic agents or neurotoxins, acting through MLK pathways suggests that alterations in these signaling pathways, together with other neuropathological entities in AD brain, may have consequences for ApoE expression. Differences in this expression may be critical, considering the role of APOE genotype in AD risk. The response of ApoE to IL-1β we show here in rodent brain suggests that elevation of IL-1 leads to the increases in ApoE that we and others have observed in the AD brain. This may have added significance with regard to the self-propagating nature of IL-1-driven cascades, especially when such cascades are instigated in the context of an ε4 allele of APOE. While induction of ApoE2 or ApoE3 may be anti-inflammatory or neuroprotective, and thereby act as a self-limiting influence on IL-1-driven cascades, ApoE4 may fail to participate and leave the brain vulnerable to prolonged activation of a maladaptive cycle.

Acknowledgements

The authors thank John McGinness, Quinton Palmer, Dr. Jin G. Sheng, Sue Woodward, and Weiwen Ye for technical support. This work was supported in part by NIH grants AG12411, AG19606, HD37989, AG17498, and AG033215; by a grant from the Alzheimer's Association; and by endowments from The Alexa and William T. Dillard, the Windgate Foundation, and the Donald W. Reynolds Foundation.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

S.W.B. receives royalty payments from Sigma-Aldrich Corp., which manufactures some of the reagents utilized in this study. The authors have no other conflicts to declare.

Authors' contributions

LL performed the cell culture experiments and contributed to writing the first draft of the manuscript. OA performed the immunofluorescence and assisted with western blots and writing. RAJ performed the experiments with rat brain tissue. REM contributed to interpretation of the results and to writing. WSTG contributed to the design of the study, interpretation of the results, and writing. SWB made essential contributions to the design of the study and interpretation of the results and completed the final draft of the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Blackman JA, Worley G, Strittmatter WJ: Apolipoprotein E and brain injury: implications for children. Dev Med Child Neurol. 2005, 47: 64-70.CrossRefPubMed Blackman JA, Worley G, Strittmatter WJ: Apolipoprotein E and brain injury: implications for children. Dev Med Child Neurol. 2005, 47: 64-70.CrossRefPubMed
2.
Zurück zum Zitat D'Arcangelo G: Apoer2: a reelin receptor to remember. Neuron. 2005, 47: 471-473. 10.1016/j.neuron.2005.08.001.CrossRefPubMed D'Arcangelo G: Apoer2: a reelin receptor to remember. Neuron. 2005, 47: 471-473. 10.1016/j.neuron.2005.08.001.CrossRefPubMed
3.
Zurück zum Zitat D'Arcangelo G, Homayouni R, Keshvara L, Rice DS, Sheldon M, Curran T: Reelin is a ligand for lipoprotein receptors. Neuron. 1999, 24: 471-479. 10.1016/S0896-6273(00)80860-0.CrossRefPubMed D'Arcangelo G, Homayouni R, Keshvara L, Rice DS, Sheldon M, Curran T: Reelin is a ligand for lipoprotein receptors. Neuron. 1999, 24: 471-479. 10.1016/S0896-6273(00)80860-0.CrossRefPubMed
4.
Zurück zum Zitat Strittmatter WJ, Bova Hill C: Molecular biology of apolipoprotein E. Curr Opin Lipidol. 2002, 13: 119-123. 10.1097/00041433-200204000-00002.CrossRefPubMed Strittmatter WJ, Bova Hill C: Molecular biology of apolipoprotein E. Curr Opin Lipidol. 2002, 13: 119-123. 10.1097/00041433-200204000-00002.CrossRefPubMed
5.
Zurück zum Zitat Strittmatter WJ, Saunders AM, Goedert M, Weisgraber KH, Dong LM, Jakes R, Huang DY, Pericak-Vance M, Schmechel D, Roses AD: Isoform-specific interactions of apolipoprotein E with microtubule-associated protein tau: implications for Alzheimer disease. Proc Natl Acad Sci USA. 1994, 91: 11183-11186. 10.1073/pnas.91.23.11183.PubMedCentralCrossRefPubMed Strittmatter WJ, Saunders AM, Goedert M, Weisgraber KH, Dong LM, Jakes R, Huang DY, Pericak-Vance M, Schmechel D, Roses AD: Isoform-specific interactions of apolipoprotein E with microtubule-associated protein tau: implications for Alzheimer disease. Proc Natl Acad Sci USA. 1994, 91: 11183-11186. 10.1073/pnas.91.23.11183.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Bu G: Apolipoprotein E and its receptors in Alzheimer's disease: pathways, pathogenesis and therapy. Nat Rev Neurosci. 2009 Bu G: Apolipoprotein E and its receptors in Alzheimer's disease: pathways, pathogenesis and therapy. Nat Rev Neurosci. 2009
7.
Zurück zum Zitat Boschert U, Merlo-Pich E, Higgins G, Roses AD, Catsicas S: Apolipoprotein E expression by neurons surviving excitotoxic stress. Neurobiol Dis. 1999, 6: 508-514. 10.1006/nbdi.1999.0251.CrossRefPubMed Boschert U, Merlo-Pich E, Higgins G, Roses AD, Catsicas S: Apolipoprotein E expression by neurons surviving excitotoxic stress. Neurobiol Dis. 1999, 6: 508-514. 10.1006/nbdi.1999.0251.CrossRefPubMed
8.
Zurück zum Zitat Xu Q, Bernardo A, Walker D, Kanegawa T, Mahley RW, Huang Y: Profile and regulation of apolipoprotein E (ApoE) expression in the CNS in mice with targeting of green fluorescent protein gene to the ApoE locus. J Neurosci. 2006, 26: 4985-4994. 10.1523/JNEUROSCI.5476-05.2006.CrossRefPubMed Xu Q, Bernardo A, Walker D, Kanegawa T, Mahley RW, Huang Y: Profile and regulation of apolipoprotein E (ApoE) expression in the CNS in mice with targeting of green fluorescent protein gene to the ApoE locus. J Neurosci. 2006, 26: 4985-4994. 10.1523/JNEUROSCI.5476-05.2006.CrossRefPubMed
9.
Zurück zum Zitat Du Y, Dodel RC, Eastwood BJ, Bales KR, Gao F, Lohmuller F, Muller U, Kurz A, Zimmer R, Evans RM, et al: Association of an interleukin 1 alpha polymorphism with Alzheimer's disease. Neurology. 2000, 55: 480-483.CrossRefPubMed Du Y, Dodel RC, Eastwood BJ, Bales KR, Gao F, Lohmuller F, Muller U, Kurz A, Zimmer R, Evans RM, et al: Association of an interleukin 1 alpha polymorphism with Alzheimer's disease. Neurology. 2000, 55: 480-483.CrossRefPubMed
10.
Zurück zum Zitat Grimaldi LM, Casadei VM, Ferri C, Veglia F, Licastro F, Annoni G, Biunno I, De Bellis G, Sorbi S, Mariani C, et al: Association of early-onset Alzheimer's disease with an interleukin-1alpha gene polymorphism. Ann Neurol. 2000, 47: 361-365. 10.1002/1531-8249(200003)47:3<361::AID-ANA12>3.0.CO;2-N.CrossRefPubMed Grimaldi LM, Casadei VM, Ferri C, Veglia F, Licastro F, Annoni G, Biunno I, De Bellis G, Sorbi S, Mariani C, et al: Association of early-onset Alzheimer's disease with an interleukin-1alpha gene polymorphism. Ann Neurol. 2000, 47: 361-365. 10.1002/1531-8249(200003)47:3<361::AID-ANA12>3.0.CO;2-N.CrossRefPubMed
11.
Zurück zum Zitat Nicoll JA, Mrak RE, Graham DI, Stewart J, Wilcock G, MacGowan S, Esiri MM, Murray LS, Dewar D, Love S, et al: Association of interleukin-1 gene polymorphisms with Alzheimer's disease. Ann Neurol. 2000, 47: 365-368. 10.1002/1531-8249(200003)47:3<365::AID-ANA13>3.0.CO;2-G.PubMedCentralCrossRefPubMed Nicoll JA, Mrak RE, Graham DI, Stewart J, Wilcock G, MacGowan S, Esiri MM, Murray LS, Dewar D, Love S, et al: Association of interleukin-1 gene polymorphisms with Alzheimer's disease. Ann Neurol. 2000, 47: 365-368. 10.1002/1531-8249(200003)47:3<365::AID-ANA13>3.0.CO;2-G.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Rebeck GW: Confirmation of the genetic association of interleukin-1A with early onset sporadic Alzheimer's disease. Neurosci Lett. 2000, 293: 75-77. 10.1016/S0304-3940(00)01487-7.CrossRefPubMed Rebeck GW: Confirmation of the genetic association of interleukin-1A with early onset sporadic Alzheimer's disease. Neurosci Lett. 2000, 293: 75-77. 10.1016/S0304-3940(00)01487-7.CrossRefPubMed
13.
Zurück zum Zitat Bertram L, Tanzi RE: Thirty years of Alzheimer's disease genetics: the implications of systematic meta-analyses. Nat Rev Neurosci. 2008, 9: 768-778. 10.1038/nrn2494.CrossRefPubMed Bertram L, Tanzi RE: Thirty years of Alzheimer's disease genetics: the implications of systematic meta-analyses. Nat Rev Neurosci. 2008, 9: 768-778. 10.1038/nrn2494.CrossRefPubMed
14.
Zurück zum Zitat Griffin WS, Sheng JG, Gentleman SM, Graham DI, Mrak RE, Roberts GW: Microglial interleukin-1 alpha expression in human head injury: correlations with neuronal and neuritic beta-amyloid precursor protein expression. Neurosci Lett. 1994, 176: 133-136. 10.1016/0304-3940(94)90066-3.PubMedCentralCrossRefPubMed Griffin WS, Sheng JG, Gentleman SM, Graham DI, Mrak RE, Roberts GW: Microglial interleukin-1 alpha expression in human head injury: correlations with neuronal and neuritic beta-amyloid precursor protein expression. Neurosci Lett. 1994, 176: 133-136. 10.1016/0304-3940(94)90066-3.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Stanley LC, Mrak RE, Woody RC, Perrot LJ, Zhang S, Marshak DR, Nelson SJ, Griffin WS: Glial cytokines as neuropathogenic factors in HIV infection: pathogenic similarities to Alzheimer's disease. J Neuropathol Exp Neurol. 1994, 53: 231-238. 10.1097/00005072-199405000-00003.CrossRefPubMed Stanley LC, Mrak RE, Woody RC, Perrot LJ, Zhang S, Marshak DR, Nelson SJ, Griffin WS: Glial cytokines as neuropathogenic factors in HIV infection: pathogenic similarities to Alzheimer's disease. J Neuropathol Exp Neurol. 1994, 53: 231-238. 10.1097/00005072-199405000-00003.CrossRefPubMed
16.
Zurück zum Zitat Sheng JG, Boop FA, Mrak RE, Griffin WS: Increased neuronal beta-amyloid precursor protein expression in human temporal lobe epilepsy: association with interleukin-1 alpha immunoreactivity. J Neurochem. 1994, 63: 1872-1879.PubMedCentralCrossRefPubMed Sheng JG, Boop FA, Mrak RE, Griffin WS: Increased neuronal beta-amyloid precursor protein expression in human temporal lobe epilepsy: association with interleukin-1 alpha immunoreactivity. J Neurochem. 1994, 63: 1872-1879.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Griffin WS, Yeralan O, Sheng JG, Boop FA, Mrak RE, Rovnaghi CR, Burnett BA, Feoktistova A, Van Eldik LJ: Overexpression of the neurotrophic cytokine S100 beta in human temporal lobe epilepsy. J Neurochem. 1995, 65: 228-233.PubMedCentralCrossRefPubMed Griffin WS, Yeralan O, Sheng JG, Boop FA, Mrak RE, Rovnaghi CR, Burnett BA, Feoktistova A, Van Eldik LJ: Overexpression of the neurotrophic cytokine S100 beta in human temporal lobe epilepsy. J Neurochem. 1995, 65: 228-233.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Mackenzie IR, Miller LA: Senile plaques in temporal lobe epilepsy. Acta Neuropathol (Berl). 1994, 87: 504-510. 10.1007/BF00294177.CrossRef Mackenzie IR, Miller LA: Senile plaques in temporal lobe epilepsy. Acta Neuropathol (Berl). 1994, 87: 504-510. 10.1007/BF00294177.CrossRef
19.
Zurück zum Zitat Vezzani A, Balosso S, Ravizza T: The role of cytokines in the pathophysiology of epilepsy. Brain Behav Immun. 2008, 22: 797-803. 10.1016/j.bbi.2008.03.009.CrossRefPubMed Vezzani A, Balosso S, Ravizza T: The role of cytokines in the pathophysiology of epilepsy. Brain Behav Immun. 2008, 22: 797-803. 10.1016/j.bbi.2008.03.009.CrossRefPubMed
20.
Zurück zum Zitat Griffin WS, Stanley LC, Ling C, White L, MacLeod V, Perrot LJ, White CL, Araoz C: Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc Natl Acad Sci USA. 1989, 86: 7611-7615. 10.1073/pnas.86.19.7611.PubMedCentralCrossRefPubMed Griffin WS, Stanley LC, Ling C, White L, MacLeod V, Perrot LJ, White CL, Araoz C: Brain interleukin 1 and S-100 immunoreactivity are elevated in Down syndrome and Alzheimer disease. Proc Natl Acad Sci USA. 1989, 86: 7611-7615. 10.1073/pnas.86.19.7611.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Sheng JG, Mrak RE, Griffin WS: Enlarged and phagocytic, but not primed, interleukin-1 alpha-immunoreactive microglia increase with age in normal human brain. Acta Neuropathol (Berl). 1998, 95: 229-234. 10.1007/s004010050792.CrossRef Sheng JG, Mrak RE, Griffin WS: Enlarged and phagocytic, but not primed, interleukin-1 alpha-immunoreactive microglia increase with age in normal human brain. Acta Neuropathol (Berl). 1998, 95: 229-234. 10.1007/s004010050792.CrossRef
22.
Zurück zum Zitat Blasko I, Stampfer-Kountchev M, Robatscher P, Veerhuis R, Eikelenboom P, Grubeck-Loebenstein B: How chronic inflammation can affect the brain and support the development of Alzheimer's disease in old age: the role of microglia and astrocytes. Aging Cell. 2004, 3: 169-176. 10.1111/j.1474-9728.2004.00101.x.CrossRefPubMed Blasko I, Stampfer-Kountchev M, Robatscher P, Veerhuis R, Eikelenboom P, Grubeck-Loebenstein B: How chronic inflammation can affect the brain and support the development of Alzheimer's disease in old age: the role of microglia and astrocytes. Aging Cell. 2004, 3: 169-176. 10.1111/j.1474-9728.2004.00101.x.CrossRefPubMed
23.
Zurück zum Zitat Gee JR, Ding Q, Keller JN: Age-related alterations of Apolipoprotein E and interleukin-1beta in the aging brain. Biogerontology. 2006, 7: 69-79. 10.1007/s10522-005-6039-9.CrossRefPubMed Gee JR, Ding Q, Keller JN: Age-related alterations of Apolipoprotein E and interleukin-1beta in the aging brain. Biogerontology. 2006, 7: 69-79. 10.1007/s10522-005-6039-9.CrossRefPubMed
24.
25.
Zurück zum Zitat Graham DI, Gentleman SM, Nicoll JA, Royston MC, McKenzie JE, Roberts GW, Griffin WS: Altered beta-APP metabolism after head injury and its relationship to the aetiology of Alzheimer's disease. Acta Neurochir Suppl. 1996, 66: 96-102.PubMed Graham DI, Gentleman SM, Nicoll JA, Royston MC, McKenzie JE, Roberts GW, Griffin WS: Altered beta-APP metabolism after head injury and its relationship to the aetiology of Alzheimer's disease. Acta Neurochir Suppl. 1996, 66: 96-102.PubMed
26.
Zurück zum Zitat Gouras GK, Relkin NR, Sweeney D, Munoz DG, Mackenzie IR, Gandy S: Increased apolipoprotein E epsilon 4 in epilepsy with senile plaques. Ann Neurol. 1997, 41: 402-404. 10.1002/ana.410410317.CrossRefPubMed Gouras GK, Relkin NR, Sweeney D, Munoz DG, Mackenzie IR, Gandy S: Increased apolipoprotein E epsilon 4 in epilepsy with senile plaques. Ann Neurol. 1997, 41: 402-404. 10.1002/ana.410410317.CrossRefPubMed
27.
Zurück zum Zitat Teasdale GM, Murray GD, Nicoll JA: The association between APOE epsilon4, age and outcome after head injury: a prospective cohort study. Brain. 2005, 128: 2556-2561. 10.1093/brain/awh595.CrossRefPubMed Teasdale GM, Murray GD, Nicoll JA: The association between APOE epsilon4, age and outcome after head injury: a prospective cohort study. Brain. 2005, 128: 2556-2561. 10.1093/brain/awh595.CrossRefPubMed
28.
Zurück zum Zitat Corder EH, Robertson K, Lannfelt L, Bogdanovic N, Eggertsen G, Wilkins J, Hall C: HIV-infected subjects with the E4 allele for APOE have excess dementia and peripheral neuropathy. Nat Med. 1998, 4: 1182-1184. 10.1038/2677.CrossRefPubMed Corder EH, Robertson K, Lannfelt L, Bogdanovic N, Eggertsen G, Wilkins J, Hall C: HIV-infected subjects with the E4 allele for APOE have excess dementia and peripheral neuropathy. Nat Med. 1998, 4: 1182-1184. 10.1038/2677.CrossRefPubMed
29.
Zurück zum Zitat Prasher VP, Farrer MJ, Kessling AM, Fisher EM, West RJ, Barber PC, Butler AC: Molecular mapping of Alzheimer-type dementia in Down's syndrome. Ann Neurol. 1998, 43: 380-383. 10.1002/ana.410430316.CrossRefPubMed Prasher VP, Farrer MJ, Kessling AM, Fisher EM, West RJ, Barber PC, Butler AC: Molecular mapping of Alzheimer-type dementia in Down's syndrome. Ann Neurol. 1998, 43: 380-383. 10.1002/ana.410430316.CrossRefPubMed
30.
Zurück zum Zitat Sheng JG, Ito K, Skinner RD, Mrak RE, Rovnaghi CR, Van Eldik LJ, Griffin WS: In vivo and in vitro evidence supporting a role for the inflammatory cytokine interleukin-1 as a driving force in Alzheimer pathogenesis. Neurobiol Aging. 1996, 17: 761-766. 10.1016/0197-4580(96)00104-2.PubMedCentralCrossRefPubMed Sheng JG, Ito K, Skinner RD, Mrak RE, Rovnaghi CR, Van Eldik LJ, Griffin WS: In vivo and in vitro evidence supporting a role for the inflammatory cytokine interleukin-1 as a driving force in Alzheimer pathogenesis. Neurobiol Aging. 1996, 17: 761-766. 10.1016/0197-4580(96)00104-2.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat Barger SW, Harmon AD: Microglial activation by Alzheimer amyloid precursor protein and modulation by apolipoprotein E. Nature. 1997, 388: 878-881. 10.1038/42257.CrossRefPubMed Barger SW, Harmon AD: Microglial activation by Alzheimer amyloid precursor protein and modulation by apolipoprotein E. Nature. 1997, 388: 878-881. 10.1038/42257.CrossRefPubMed
32.
Zurück zum Zitat Sheng JG, Mrak RE, Griffin WS: Apolipoprotein E distribution among different plaque types in Alzheimer's disease: implications for its role in plaque progression. Neuropathol Appl Neurobiol. 1996, 22: 334-341. 10.1111/j.1365-2990.1996.tb01112.x.CrossRefPubMed Sheng JG, Mrak RE, Griffin WS: Apolipoprotein E distribution among different plaque types in Alzheimer's disease: implications for its role in plaque progression. Neuropathol Appl Neurobiol. 1996, 22: 334-341. 10.1111/j.1365-2990.1996.tb01112.x.CrossRefPubMed
33.
Zurück zum Zitat Bales KR, Verina T, Cummins DJ, Du Y, Dodel RC, Saura J, Fishman CE, DeLong CA, Piccardo P, Petegnief V, et al: Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimer's disease. Proc Natl Acad Sci USA. 1999, 96: 15233-15238. 10.1073/pnas.96.26.15233.PubMedCentralCrossRefPubMed Bales KR, Verina T, Cummins DJ, Du Y, Dodel RC, Saura J, Fishman CE, DeLong CA, Piccardo P, Petegnief V, et al: Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimer's disease. Proc Natl Acad Sci USA. 1999, 96: 15233-15238. 10.1073/pnas.96.26.15233.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Griffin WS, Liu L, Li Y, Mrak RE, Barger SW: Interleukin-1 mediates Alzheimer and Lewy body pathologies. J Neuroinflammation. 2006, 3: 5-10.1186/1742-2094-3-5.PubMedCentralCrossRefPubMed Griffin WS, Liu L, Li Y, Mrak RE, Barger SW: Interleukin-1 mediates Alzheimer and Lewy body pathologies. J Neuroinflammation. 2006, 3: 5-10.1186/1742-2094-3-5.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Barger SW, DeWall KM, Liu L, Mrak RE, Griffin WS: Relationships between expression of apolipoprotein E and beta-amyloid precursor protein are altered in proximity to Alzheimer beta-amyloid plaques: potential explanations from cell culture studies. J Neuropathol Exp Neurol. 2008, 67: 773-783. 10.1097/NEN.0b013e318180ec47.PubMedCentralCrossRefPubMed Barger SW, DeWall KM, Liu L, Mrak RE, Griffin WS: Relationships between expression of apolipoprotein E and beta-amyloid precursor protein are altered in proximity to Alzheimer beta-amyloid plaques: potential explanations from cell culture studies. J Neuropathol Exp Neurol. 2008, 67: 773-783. 10.1097/NEN.0b013e318180ec47.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Senechal Y, Kelly PH, Dev KK: Amyloid precursor protein knockout mice show age-dependent deficits in passive avoidance learning. Behav Brain Res. 2008, 186: 126-132. 10.1016/j.bbr.2007.08.003.CrossRefPubMed Senechal Y, Kelly PH, Dev KK: Amyloid precursor protein knockout mice show age-dependent deficits in passive avoidance learning. Behav Brain Res. 2008, 186: 126-132. 10.1016/j.bbr.2007.08.003.CrossRefPubMed
37.
Zurück zum Zitat Dawson GR, Seabrook GR, Zheng H, Smith DW, Graham S, O'Dowd G, Bowery BJ, Boyce S, Trumbauer ME, Chen HY, et al: Age-related cognitive deficits, impaired long-term potentiation and reduction in synaptic marker density in mice lacking the beta-amyloid precursor protein. Neuroscience. 1999, 90: 1-13. 10.1016/S0306-4522(98)00410-2.CrossRefPubMed Dawson GR, Seabrook GR, Zheng H, Smith DW, Graham S, O'Dowd G, Bowery BJ, Boyce S, Trumbauer ME, Chen HY, et al: Age-related cognitive deficits, impaired long-term potentiation and reduction in synaptic marker density in mice lacking the beta-amyloid precursor protein. Neuroscience. 1999, 90: 1-13. 10.1016/S0306-4522(98)00410-2.CrossRefPubMed
38.
Zurück zum Zitat Mattson MP: Cellular actions of beta-amyloid precursor protein and its soluble and fibrillogenic derivatives. Physiol Rev. 1997, 77: 1081-1132.PubMed Mattson MP: Cellular actions of beta-amyloid precursor protein and its soluble and fibrillogenic derivatives. Physiol Rev. 1997, 77: 1081-1132.PubMed
39.
Zurück zum Zitat Laskowitz DT, Matthew WD, Bennett ER, Schmechel D, Herbstreith MH, Goel S, McMillian MK: Endogenous apolipoprotein E suppresses LPS-stimulated microglial nitric oxide production. Neuroreport. 1998, 9: 615-618. 10.1097/00001756-199803090-00010.CrossRefPubMed Laskowitz DT, Matthew WD, Bennett ER, Schmechel D, Herbstreith MH, Goel S, McMillian MK: Endogenous apolipoprotein E suppresses LPS-stimulated microglial nitric oxide production. Neuroreport. 1998, 9: 615-618. 10.1097/00001756-199803090-00010.CrossRefPubMed
40.
Zurück zum Zitat Lynch JR, Morgan D, Mance J, Matthew WD, Laskowitz DT: Apolipoprotein E modulates glial activation and the endogenous central nervous system inflammatory response. J Neuroimmunol. 2001, 114: 107-113. 10.1016/S0165-5728(00)00459-8.CrossRefPubMed Lynch JR, Morgan D, Mance J, Matthew WD, Laskowitz DT: Apolipoprotein E modulates glial activation and the endogenous central nervous system inflammatory response. J Neuroimmunol. 2001, 114: 107-113. 10.1016/S0165-5728(00)00459-8.CrossRefPubMed
41.
Zurück zum Zitat Koistinaho M, Kettunen MI, Holtzman DM, Kauppinen RA, Higgins LS, Koistinaho J: Expression of human apolipoprotein E downregulates amyloid precursor protein-induced ischemic susceptibility. Stroke. 2002, 33: 1905-1910. 10.1161/01.STR.0000020124.61998.BC.CrossRefPubMed Koistinaho M, Kettunen MI, Holtzman DM, Kauppinen RA, Higgins LS, Koistinaho J: Expression of human apolipoprotein E downregulates amyloid precursor protein-induced ischemic susceptibility. Stroke. 2002, 33: 1905-1910. 10.1161/01.STR.0000020124.61998.BC.CrossRefPubMed
42.
Zurück zum Zitat Li Y, Liu L, Kang J, Sheng JG, Barger SW, Mrak RE, Griffin WS: Neuronal-glial interactions mediated by interleukin-1 enhance neuronal acetylcholinesterase activity and mRNA expression. J Neurosci. 2000, 20: 149-155.PubMed Li Y, Liu L, Kang J, Sheng JG, Barger SW, Mrak RE, Griffin WS: Neuronal-glial interactions mediated by interleukin-1 enhance neuronal acetylcholinesterase activity and mRNA expression. J Neurosci. 2000, 20: 149-155.PubMed
43.
Zurück zum Zitat Li Y, Wang J, Sheng JG, Liu L, Barger SW, Jones RA, Van Eldik LJ, Mrak RE, Griffin WS: S100 beta increases levels of beta-amyloid precursor protein and its encoding mRNA in rat neuronal cultures. J Neurochem. 1998, 71: 1421-1428.CrossRefPubMed Li Y, Wang J, Sheng JG, Liu L, Barger SW, Jones RA, Van Eldik LJ, Mrak RE, Griffin WS: S100 beta increases levels of beta-amyloid precursor protein and its encoding mRNA in rat neuronal cultures. J Neurochem. 1998, 71: 1421-1428.CrossRefPubMed
44.
Zurück zum Zitat Wu SZ, Bodles AM, Porter MM, Griffin WS, Basile AS, Barger SW: Induction of serine racemase expression and D-serine release from microglia by amyloid beta-peptide. J Neuroinflammation. 2004, 1: 2-10.1186/1742-2094-1-2.PubMedCentralCrossRefPubMed Wu SZ, Bodles AM, Porter MM, Griffin WS, Basile AS, Barger SW: Induction of serine racemase expression and D-serine release from microglia by amyloid beta-peptide. J Neuroinflammation. 2004, 1: 2-10.1186/1742-2094-1-2.PubMedCentralCrossRefPubMed
45.
Zurück zum Zitat Li Y, Liu L, Barger SW, Mrak RE, Griffin WS: Vitamin E suppression of microglial activation is neuroprotective. J Neurosci Res. 2001, 66: 163-170. 10.1002/jnr.1208.PubMedCentralCrossRefPubMed Li Y, Liu L, Barger SW, Mrak RE, Griffin WS: Vitamin E suppression of microglial activation is neuroprotective. J Neurosci Res. 2001, 66: 163-170. 10.1002/jnr.1208.PubMedCentralCrossRefPubMed
46.
Zurück zum Zitat Buxbaum JD, Oishi M, Chen HI, Pinkas-Kramarski R, Jaffe EA, Gandy SE, Greengard P: Cholinergic agonists and interleukin 1 regulate processing and secretion of the Alzheimer beta/A4 amyloid protein precursor. Proc Natl Acad Sci USA. 1992, 89: 10075-10078. 10.1073/pnas.89.21.10075.PubMedCentralCrossRefPubMed Buxbaum JD, Oishi M, Chen HI, Pinkas-Kramarski R, Jaffe EA, Gandy SE, Greengard P: Cholinergic agonists and interleukin 1 regulate processing and secretion of the Alzheimer beta/A4 amyloid protein precursor. Proc Natl Acad Sci USA. 1992, 89: 10075-10078. 10.1073/pnas.89.21.10075.PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Sheng JG, Jones RA, Zhou XQ, McGinness JM, Van Eldik LJ, Mrak RE, Griffin WS: Interleukin-1 promotion of MAPK-p38 overexpression in experimental animals and in Alzheimer's disease: potential significance for tau protein phosphorylation. Neurochem Int. 2001, 39: 341-348. 10.1016/S0197-0186(01)00041-9.PubMedCentralCrossRefPubMed Sheng JG, Jones RA, Zhou XQ, McGinness JM, Van Eldik LJ, Mrak RE, Griffin WS: Interleukin-1 promotion of MAPK-p38 overexpression in experimental animals and in Alzheimer's disease: potential significance for tau protein phosphorylation. Neurochem Int. 2001, 39: 341-348. 10.1016/S0197-0186(01)00041-9.PubMedCentralCrossRefPubMed
48.
Zurück zum Zitat Griffin WS, Sheng JG, Royston MC, Gentleman SM, McKenzie JE, Graham DI, Roberts GW, Mrak RE: Glial-neuronal interactions in Alzheimer's disease: the potential role of a 'cytokine cycle' in disease progression. Brain Pathol. 1998, 8: 65-72.CrossRefPubMed Griffin WS, Sheng JG, Royston MC, Gentleman SM, McKenzie JE, Graham DI, Roberts GW, Mrak RE: Glial-neuronal interactions in Alzheimer's disease: the potential role of a 'cytokine cycle' in disease progression. Brain Pathol. 1998, 8: 65-72.CrossRefPubMed
49.
Zurück zum Zitat Mrak RE, Sheng JG, Griffin WS: Correlation of astrocytic S100 beta expression with dystrophic neurites in amyloid plaques of Alzheimer's disease. J Neuropathol Exp Neurol. 1996, 55: 273-279. 10.1097/00005072-199603000-00002.PubMedCentralCrossRefPubMed Mrak RE, Sheng JG, Griffin WS: Correlation of astrocytic S100 beta expression with dystrophic neurites in amyloid plaques of Alzheimer's disease. J Neuropathol Exp Neurol. 1996, 55: 273-279. 10.1097/00005072-199603000-00002.PubMedCentralCrossRefPubMed
50.
Zurück zum Zitat Meda L, Baron P, Prat E, Scarpini E, Scarlato G, Cassatella MA, Rossi F: Proinflammatory profile of cytokine production by human monocytes and murine microglia stimulated with beta-amyloid[25-35]. J Neuroimmunol. 1999, 93: 45-52. 10.1016/S0165-5728(98)00188-X.CrossRefPubMed Meda L, Baron P, Prat E, Scarpini E, Scarlato G, Cassatella MA, Rossi F: Proinflammatory profile of cytokine production by human monocytes and murine microglia stimulated with beta-amyloid[25-35]. J Neuroimmunol. 1999, 93: 45-52. 10.1016/S0165-5728(98)00188-X.CrossRefPubMed
51.
Zurück zum Zitat Barger SW, Mattson MP: Isoform-specific modulation by apolipoprotein E of the activities of secreted beta-amyloid precursor protein. J Neurochem. 1997, 69: 60-67.CrossRefPubMed Barger SW, Mattson MP: Isoform-specific modulation by apolipoprotein E of the activities of secreted beta-amyloid precursor protein. J Neurochem. 1997, 69: 60-67.CrossRefPubMed
52.
Zurück zum Zitat Wu S, Basile AS, Barger SW: Induction of serine racemase expression and D-serine release from microglia by secreted amyloid precursor protein (sAPP). Curr Alzheimer Res. 2007, 4: 243-251.CrossRefPubMed Wu S, Basile AS, Barger SW: Induction of serine racemase expression and D-serine release from microglia by secreted amyloid precursor protein (sAPP). Curr Alzheimer Res. 2007, 4: 243-251.CrossRefPubMed
53.
Zurück zum Zitat Schmechel DE, Saunders AM, Strittmatter WJ, Crain BJ, Hulette CM, Joo SH, Pericak-Vance MA, Goldgaber D, Roses AD: Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc Natl Acad Sci USA. 1993, 90: 9649-9653. 10.1073/pnas.90.20.9649.PubMedCentralCrossRefPubMed Schmechel DE, Saunders AM, Strittmatter WJ, Crain BJ, Hulette CM, Joo SH, Pericak-Vance MA, Goldgaber D, Roses AD: Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc Natl Acad Sci USA. 1993, 90: 9649-9653. 10.1073/pnas.90.20.9649.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Wisniewski T, Castano EM, Golabek A, Vogel T, Frangione B: Acceleration of Alzheimer's fibril formation by apolipoprotein E in vitro. Am J Pathol. 1994, 145: 1030-1035.PubMedCentralPubMed Wisniewski T, Castano EM, Golabek A, Vogel T, Frangione B: Acceleration of Alzheimer's fibril formation by apolipoprotein E in vitro. Am J Pathol. 1994, 145: 1030-1035.PubMedCentralPubMed
55.
Zurück zum Zitat Horsburgh K, Fitzpatrick M, Nilsen M, Nicoll JA: Marked alterations in the cellular localisation and levels of apolipoprotein E following acute subdural haematoma in rat. Brain Res. 1997, 763: 103-110. 10.1016/S0006-8993(97)00411-3.CrossRefPubMed Horsburgh K, Fitzpatrick M, Nilsen M, Nicoll JA: Marked alterations in the cellular localisation and levels of apolipoprotein E following acute subdural haematoma in rat. Brain Res. 1997, 763: 103-110. 10.1016/S0006-8993(97)00411-3.CrossRefPubMed
56.
Zurück zum Zitat Kay A, Petzold A, Kerr M, Keir G, Thompson E, Nicoll J: Temporal alterations in cerebrospinal fluid amyloid beta-protein and apolipoprotein E after subarachnoid hemorrhage. Stroke. 2003, 34: e240-243. 10.1161/01.STR.0000100157.88508.2F.CrossRefPubMed Kay A, Petzold A, Kerr M, Keir G, Thompson E, Nicoll J: Temporal alterations in cerebrospinal fluid amyloid beta-protein and apolipoprotein E after subarachnoid hemorrhage. Stroke. 2003, 34: e240-243. 10.1161/01.STR.0000100157.88508.2F.CrossRefPubMed
57.
Zurück zum Zitat Lambert JC, Perez-Tur J, Dupire MJ, Galasko D, Mann D, Amouyel P, Hardy J, Delacourte A, Chartier-Harlin MC: Distortion of allelic expression of apolipoprotein E in Alzheimer's disease. Hum Mol Genet. 1997, 6: 2151-2154. 10.1093/hmg/6.12.2151.CrossRefPubMed Lambert JC, Perez-Tur J, Dupire MJ, Galasko D, Mann D, Amouyel P, Hardy J, Delacourte A, Chartier-Harlin MC: Distortion of allelic expression of apolipoprotein E in Alzheimer's disease. Hum Mol Genet. 1997, 6: 2151-2154. 10.1093/hmg/6.12.2151.CrossRefPubMed
58.
Zurück zum Zitat Hesse C, Larsson H, Fredman P, Minthon L, Andreasen N, Davidsson P, Blennow K: Measurement of apolipoprotein E (apoE) in cerebrospinal fluid. Neurochem Res. 2000, 25: 511-517. 10.1023/A:1007516210548.CrossRefPubMed Hesse C, Larsson H, Fredman P, Minthon L, Andreasen N, Davidsson P, Blennow K: Measurement of apolipoprotein E (apoE) in cerebrospinal fluid. Neurochem Res. 2000, 25: 511-517. 10.1023/A:1007516210548.CrossRefPubMed
59.
Zurück zum Zitat Fukumoto H, Ingelsson M, Garevik N, Wahlund LO, Nukina N, Yaguchi Y, Shibata M, Hyman BT, Rebeck GW, Irizarry MC: APOE epsilon 3/epsilon 4 heterozygotes have an elevated proportion of apolipoprotein E4 in cerebrospinal fluid relative to plasma, independent of Alzheimer's disease diagnosis. Exp Neurol. 2003, 183: 249-253. 10.1016/S0014-4886(03)00088-8.CrossRefPubMed Fukumoto H, Ingelsson M, Garevik N, Wahlund LO, Nukina N, Yaguchi Y, Shibata M, Hyman BT, Rebeck GW, Irizarry MC: APOE epsilon 3/epsilon 4 heterozygotes have an elevated proportion of apolipoprotein E4 in cerebrospinal fluid relative to plasma, independent of Alzheimer's disease diagnosis. Exp Neurol. 2003, 183: 249-253. 10.1016/S0014-4886(03)00088-8.CrossRefPubMed
60.
Zurück zum Zitat Bray NJ, Jehu L, Moskvina V, Buxbaum JD, Dracheva S, Haroutunian V, Williams J, Buckland PR, Owen MJ, O'Donovan MC: Allelic expression of APOE in human brain: effects of epsilon status and promoter haplotypes. Hum Mol Genet. 2004, 13: 2885-2892. 10.1093/hmg/ddh299.CrossRefPubMed Bray NJ, Jehu L, Moskvina V, Buxbaum JD, Dracheva S, Haroutunian V, Williams J, Buckland PR, Owen MJ, O'Donovan MC: Allelic expression of APOE in human brain: effects of epsilon status and promoter haplotypes. Hum Mol Genet. 2004, 13: 2885-2892. 10.1093/hmg/ddh299.CrossRefPubMed
61.
Zurück zum Zitat Darreh-Shori T, Modiri N, Blennow K, Baza S, Kamil C, Ahmed H, Andreasen N, Nordberg A: The apolipoprotein E varepsilon4 allele plays pathological roles in AD through high protein expression and interaction with butyrylcholinesterase. Neurobiol Aging. 2009 Darreh-Shori T, Modiri N, Blennow K, Baza S, Kamil C, Ahmed H, Andreasen N, Nordberg A: The apolipoprotein E varepsilon4 allele plays pathological roles in AD through high protein expression and interaction with butyrylcholinesterase. Neurobiol Aging. 2009
62.
Zurück zum Zitat Beffert U, Cohn JS, Petit-Turcotte C, Tremblay M, Aumont N, Ramassamy C, Davignon J, Poirier J: Apolipoprotein E and beta-amyloid levels in the hippocampus and frontal cortex of Alzheimer's disease subjects are disease-related and apolipoprotein E genotype dependent. Brain Res. 1999, 843: 87-94. 10.1016/S0006-8993(99)01894-6.CrossRefPubMed Beffert U, Cohn JS, Petit-Turcotte C, Tremblay M, Aumont N, Ramassamy C, Davignon J, Poirier J: Apolipoprotein E and beta-amyloid levels in the hippocampus and frontal cortex of Alzheimer's disease subjects are disease-related and apolipoprotein E genotype dependent. Brain Res. 1999, 843: 87-94. 10.1016/S0006-8993(99)01894-6.CrossRefPubMed
63.
Zurück zum Zitat Bertrand P, Poirier J, Oda T, Finch CE, Pasinetti GM: Association of apolipoprotein E genotype with brain levels of apolipoprotein E and apolipoprotein J (clusterin) in Alzheimer disease. Brain Res Mol Brain Res. 1995, 33: 174-178.CrossRefPubMed Bertrand P, Poirier J, Oda T, Finch CE, Pasinetti GM: Association of apolipoprotein E genotype with brain levels of apolipoprotein E and apolipoprotein J (clusterin) in Alzheimer disease. Brain Res Mol Brain Res. 1995, 33: 174-178.CrossRefPubMed
64.
65.
Zurück zum Zitat Guo L, LaDu MJ, Van Eldik LJ: A dual role for apolipoprotein e in neuroinflammation: anti- and pro-inflammatory activity. J Mol Neurosci. 2004, 23: 205-212. 10.1385/JMN:23:3:205.CrossRefPubMed Guo L, LaDu MJ, Van Eldik LJ: A dual role for apolipoprotein e in neuroinflammation: anti- and pro-inflammatory activity. J Mol Neurosci. 2004, 23: 205-212. 10.1385/JMN:23:3:205.CrossRefPubMed
66.
Zurück zum Zitat Colton CA, Brown CM, Cook D, Needham LK, Xu Q, Czapiga M, Saunders AM, Schmechel DE, Rasheed K, Vitek MP: APOE and the regulation of microglial nitric oxide production: a link between genetic risk and oxidative stress. Neurobiol Aging. 2002, 23: 777-785. 10.1016/S0197-4580(02)00016-7.CrossRefPubMed Colton CA, Brown CM, Cook D, Needham LK, Xu Q, Czapiga M, Saunders AM, Schmechel DE, Rasheed K, Vitek MP: APOE and the regulation of microglial nitric oxide production: a link between genetic risk and oxidative stress. Neurobiol Aging. 2002, 23: 777-785. 10.1016/S0197-4580(02)00016-7.CrossRefPubMed
67.
Zurück zum Zitat Ezra Y, Oron L, Moskovich L, Roses AD, Beni SM, Shohami E, Michaelson DM: Apolipoprotein E4 decreases whereas apolipoprotein E3 increases the level of secreted amyloid precursor protein after closed head injury. Neuroscience. 2003, 121: 315-325. 10.1016/S0306-4522(03)00436-6.CrossRefPubMed Ezra Y, Oron L, Moskovich L, Roses AD, Beni SM, Shohami E, Michaelson DM: Apolipoprotein E4 decreases whereas apolipoprotein E3 increases the level of secreted amyloid precursor protein after closed head injury. Neuroscience. 2003, 121: 315-325. 10.1016/S0306-4522(03)00436-6.CrossRefPubMed
68.
Zurück zum Zitat Hoe HS, Harris DC, Rebeck GW: Multiple pathways of apolipoprotein E signaling in primary neurons. J Neurochem. 2005, 93: 145-155. 10.1111/j.1471-4159.2004.03007.x.CrossRefPubMed Hoe HS, Harris DC, Rebeck GW: Multiple pathways of apolipoprotein E signaling in primary neurons. J Neurochem. 2005, 93: 145-155. 10.1111/j.1471-4159.2004.03007.x.CrossRefPubMed
69.
Zurück zum Zitat Ohkubo N, Mitsuda N, Tamatani M, Yamaguchi A, Lee YD, Ogihara T, Vitek MP, Tohyama M: Apolipoprotein E4 stimulates cAMP response element-binding protein transcriptional activity through the extracellular signal-regulated kinase pathway. J Biol Chem. 2001, 276: 3046-3053. 10.1074/jbc.M005070200.CrossRefPubMed Ohkubo N, Mitsuda N, Tamatani M, Yamaguchi A, Lee YD, Ogihara T, Vitek MP, Tohyama M: Apolipoprotein E4 stimulates cAMP response element-binding protein transcriptional activity through the extracellular signal-regulated kinase pathway. J Biol Chem. 2001, 276: 3046-3053. 10.1074/jbc.M005070200.CrossRefPubMed
70.
Zurück zum Zitat Hardingham GE, Fukunaga Y, Bading H: Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nat Neurosci. 2002, 5: 405-414.PubMed Hardingham GE, Fukunaga Y, Bading H: Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nat Neurosci. 2002, 5: 405-414.PubMed
71.
Zurück zum Zitat Soriano FX, Martel MA, Papadia S, Vaslin A, Baxter P, Rickman C, Forder J, Tymianski M, Duncan R, Aarts M, et al: Specific targeting of pro-death NMDA receptor signals with differing reliance on the NR2B PDZ ligand. J Neurosci. 2008, 28: 10696-10710. 10.1523/JNEUROSCI.1207-08.2008.PubMedCentralCrossRefPubMed Soriano FX, Martel MA, Papadia S, Vaslin A, Baxter P, Rickman C, Forder J, Tymianski M, Duncan R, Aarts M, et al: Specific targeting of pro-death NMDA receptor signals with differing reliance on the NR2B PDZ ligand. J Neurosci. 2008, 28: 10696-10710. 10.1523/JNEUROSCI.1207-08.2008.PubMedCentralCrossRefPubMed
72.
Zurück zum Zitat Xu J, Kurup P, Zhang Y, Goebel-Goody SM, Wu PH, Hawasli AH, Baum ML, Bibb JA, Lombroso PJ: Extrasynaptic NMDA receptors couple preferentially to excitotoxicity via calpain-mediated cleavage of STEP. J Neurosci. 2009, 29: 9330-9343. 10.1523/JNEUROSCI.2212-09.2009.PubMedCentralCrossRefPubMed Xu J, Kurup P, Zhang Y, Goebel-Goody SM, Wu PH, Hawasli AH, Baum ML, Bibb JA, Lombroso PJ: Extrasynaptic NMDA receptors couple preferentially to excitotoxicity via calpain-mediated cleavage of STEP. J Neurosci. 2009, 29: 9330-9343. 10.1523/JNEUROSCI.2212-09.2009.PubMedCentralCrossRefPubMed
Metadaten
Titel
Apolipoprotein E expression is elevated by interleukin 1 and other interleukin 1-induced factors
verfasst von
Ling Liu
Orwa Aboud
Richard A Jones
Robert E Mrak
W Sue T Griffin
Steven W Barger
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2011
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-8-175

Weitere Artikel der Ausgabe 1/2011

Journal of Neuroinflammation 1/2011 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.