Skip to main content
Erschienen in: Acta Neuropathologica Communications 1/2016

Open Access 01.12.2016 | Research

Increased tauopathy drives microglia-mediated clearance of beta-amyloid

verfasst von: Wesley Chen, Edsel A. Abud, Stephen T. Yeung, Anita Lakatos, Trevor Nassi, Jane Wang, David Blum, Luc Buée, Wayne W. Poon, Mathew Blurton-Jones

Erschienen in: Acta Neuropathologica Communications | Ausgabe 1/2016

Abstract

Alzheimer disease is characterized by the accumulation of β-amyloid (Aβ) plaques and tau-laden neurofibrillary tangles. Emerging studies suggest that in neurodegenerative diseases, aggregation of one protein species can promote other proteinopathies and that inflammation plays an important role in this process. To study the interplay between Aβ deposition, tau pathology, and microgliosis, we established a new AD transgenic mouse model by crossing 5xfAD mice with Thy-Tau22 transgenic mice. The resulting ‘T5x’ mice exhibit a greater than three-fold increase in misfolded and hyperphosphorylated tau and further substantiates the hypothesis that Aβ accelerates tau pathology. Surprisingly, T5x mice exhibit a 40-50 % reduction in Aβ plaque load and insoluble Aβ species when compared with aged-matched 5xfAD littermates. T5x mice exhibit significant changes in cytokine production, an almost doubling of microglial number, and a dramatic shift in microglia activation state. Furthermore, T5x microglia exhibit increased phagocytic capacity that enhances the clearance of insoluble Aβ and decreasing plaque load. Therefore, our results suggest that strategies to increase the phagocytic ability of microglia can be employed to reduce Aβ and that tau-induced changes in microglial activation state can promote the clearance of Aβ.
Begleitmaterial
Additional files 2: Figure S2. T5x mice exhibit increased levels of Tau phosphorylation and misfolding. (A) The ratio of phosphorylated human tau (65 kDa) to unphosphorylated human tau (60 kDa) was quantified by western blot. Analysis revealed significantly higher 65:60 kDa HT7 ratio in T5x mice relative to Tau22 littermates, suggesting that tau hyperphosphorylation is exacerbated by the presence of Aβ in T5x mice. (B) Equivalent hippocampal sections were examined from 7-month old T5x and Thy-Tau22 mice and immunohistochemically labeled with the conformational-specific tau antibody; MC-1 (see Figs. 2 and 3). Quantification revealed a significant increase in the numbers of MC-1-positive neurons in hippocampus CA1 (p < 0.0001) and an even greater increase in MC-1-immunoreactive cells within the perirhinal/entorhinal cortex. Data are represented as mean ± SEM of optical density (O.D.), n ≥ 8 mice/group. * Indicates p < 0.05 for both ANOVA and Fisher’s protected least-significant difference (PLSD) post hoc tests. (PDF 427 kb)
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s40478-016-0336-1) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interest.

Authors’ contributions

WC, WWP, EMA, and M B-J. designed research; WC, AL, and MB-J performed research; WC, AL, and MB-J. analyzed data; LB and DB provided the Thy-Tau22 mice; WC, WWP, EMA, and MB-J wrote the paper. All authors read and approved the final manuscript.

Introduction

Alzheimer’s Disease (AD) is the leading cause of age-related dementia, affecting over 5 million people in the United States alone [5]. AD pathology is characterized by two primary lesions: extracellular amyloid plaques and intraneuronal tau-laden neurofibrillary tangles. The mechanisms that drive AD remain unclear, but the ‘amyloid cascade hypothesis’ first proposed by Hardy and Higgins posits that beta-amyloid (Aβ) accumulation is the initiating factor in AD pathogenesis [8, 31, 33]. Increased deposition of Aβ, in turn, is thought to promote the hyperphosphorylation of tau leading to neurofibrillary tangle (NFT) formation [2]. Together, Aβ and tau pathologies disrupt critical biological functions such as axonal transport and synaptic function and promote neuroinflammation, ultimately leading to widespread synaptic and neuronal loss [65, 83].
The role of neuroinflammation in the development and progression of AD has been studied for several decades [26, 27, 34]. However, the recent identification of AD risk polymorphisms in several microglial-enriched genes such as TREM2, MS4A, and CD33, has intensified this area of research [9, 15, 25, 28, 32, 42, 51, 86]. Microglia serve as one of the brain’s primary mechanisms of Aβ clearance, but also play critical roles in neuronal homeostasis and synaptic plasticity [63, 69, 76]. In response to Aβ, microglia increase their expression of pro-inflammatory cytokines, which has been shown to promote tau hyperphosphorylation and NFT pathology [10, 49] and contribute to synaptic and neuronal dysfunction [3, 30, 69].
To better understand the role of microglia in the interactions between Aβ and tau pathologies we crossed two transgenic AD models, 5xfAD and Thy-Tau22 mice, to create a novel bigenic line termed ‘T5x’ mice. 5xfAD mice express three APP mutations (Swedish, Florida, London) and two PS1 mutations (M146L, L286V) that are co-inherited and driven under control of the neuronal-specific Thy1.2 promoter [61]. 5xfAD exhibit intraneuronal Aβ accumulation beginning at 1.5 months, amyloid plaque deposition and gliosis starting at 2 months, synaptic loss by 6 months, and neuronal death beginning at 9 months of age [22]. Amyloid deposition in these mice is most prominent in the hippocampus, subiculum, deep cortical layers, and the basal lateral amygdala. Increased neuroinflammation and microglial activation have also been shown to play an essential role in mediating disease progression in this model [43]. Thus, 5xfAD mice provide an aggressive amyloidogenic model that exhibit robust AD-associated plaque pathology and microgliosis. In comparison, Thy-Tau22 mice express human four repeat tau with two mutations (G272V, P301S) driven under the Thy1.2 promoter and progressively develop hippocampal hyperphosphorylated tau, neurofibrillary tangles, and gliosis [72]. Studies examining the effects of exercise, caffeine and A2A receptor modulation in Thy-Tau22 mice suggest important roles for microglia and neuroinflammatory responses in the accumulation of tau pathology [7, 44].
Interactions between Aβ and tau in mouse models were first reported in two seminal studies in 2001 that clearly demonstrated that Aβ accumulation could accelerate the development of tau pathology [24, 47]. Subsequently, the 3xTg-AD model was generated and extended our understanding of the influence of Aβ on tau and was used to explore the role of microglial inflammation in this process [39, 62]. Since then several other bigenic models have been created and studies have continued to investigate the effects of amyloid on tau and suggested that the amyloid cascade follows a unidirectional pathway [18, 35, 71]. However, many of these models exhibit far less pathology than occurs in human AD cases and thus more complex interactions that might occur over decades in the human brain or at later stages of advanced disease may not be faithfully recapitulated in many of these models [85]. Furthermore, the potential role of inflammation in these interactions between Aβ and tau remains greatly understudied.
In the present study, we combined a model of Aβ accumulation (5xfAD mice) with a progressive model of neurofibrillary tangle pathology (Thy-Tau22 mice). The resulting ‘T5x’ mice were generated to provide insight into the later stages of disease progression akin to that observed in clinically-diagnosed AD patients and the interactions and consequences of advanced amyloid pathology on tau and visa-versa. By comparing T5x bigenic mice to their single transgenic littermates we have uncovered potential new roles for tau in the modulation of Aβ and neuroinflammatory response. Most notably, we identify and examine how amyloid and tau synergize to alter microglial activity and promote Aβ clearance. Thus, these data provide additional insight into the relationship between AD pathology and neuroinflammatory response and suggest that tau can exhibit reciprocal interactions with amyloid.

Material and Methods

Generation of T5x mice

Thy-Tau22 mice express human 4 repeat tau with two frontotemporal dementia-associated point mutations (G272V and P301S) under control of the neuronal driven promoter Thy1.2 and are maintained on a C57Bl6/J background [72]. The 5xfAD mice used in this study are also maintained on a congenic C57Bl6/J background and were purchased from the Mutant Mouse Resource and Research Center (MMRRC, stock# 034848-JAX). The 5xfAD model co-expresses human amyloid precursor protein (APP695) carrying the Swedish, Florida, and London mutations and a human presenilin-1 (PS1) transgene carrying the M146L and L286V mutations under the Thy-1 promoter. Both APP and PS1 transgenes are co-integrated and thus co-inherited. Heterozygous Thy-Tau22 and 5xfAD mice were crossed to create Thy-Tau22-5xfAD (T5x) mice, as well as Thy-Tau22, 5xfAD, and WT littermates that were genotyped via PCR amplification of human tau, PS1, and APP transgenes. The number of mice from each sex-balanced genotype was: WT (n = 8), 5x (n = 11), Tau (n = 14), and T5x (n = 9). All mice were maintained on a purebred C57Bl/6 background and group housed (2–4 mice/cage) on a 12 h/12 h light/dark cycle with access to food and water ad libitum. All animal procedures were performed in strict accordance to the National Institutes of Health and University of California Institutional Animal Care and Use Committee.

Tissue preparation and neuropathological analysis

Following behavioral testing, all mice were sedated with euthasol and sacrificed by cardiac perfusion with 0.1 M phosphate buffered saline (PBS). Brains were removed and one hemisphere was snap frozen on dry ice while the other hemisphere was postfixed in 4 % paraformaldehyde for 48 h then stored in PBS + 0.05 % sodium azide. Fixed half-brains were placed in 30 % sucrose for at least 48 h before being cut in the coronal plane (40 μm sections) using a freezing sliding microtome.

Immunohistological staining

Brain sections were rinsed in PBS three times and blocked in PBS + 0.05 % Triton-X with 5 % donkey or goat serum for one hour. Primary antibodies used included: 6E10 (Covance; 1:500) and 82E1 (ABL America; 1:500) against Aβ, total Tau (human + mouse; Dako; 1:1000), human tau (HT7, ThermoFisher; 1:1000), phosho-tau epitopes AT8 (Pierce; 1:500) and PHF-1, and conformational tau epitope MC-1 (generously provided by Peter Davies; 1:1000). Analysis of gliosis, phagocytosis, and dendritic architecture utilized CD68 (Abcam; 1:200), IBA1 (Wako; 1:1000), GFAP (Abcam; 1:1000), Beta3Tubullin (Covance; 1:1000). Sections were incubated in primary antibodies at 4° Celsius overnight. Sections were then washed three times with PBS and incubated with appropriate Alexa fluor-conjugated secondary antibodies at room temperature for one hour. Sections were then rinsed three additional times, mounted on slides and coverslipped with Fluoromount-G with DAPI.

Biochemical analysis

Hippocampus and cortex was microdissected from frozen brains and processed to collect both soluble and insoluble extracts. Briefly, microdissected tissue was homogenized in TPER (ThermoFisher) and centrifuged at 12,000 RPM for 15 min. Supernatant was collected as the soluble fraction and the pellet was treated with 70 % formic acid and spun down at 25,000 rpm. The resulting supernatant was collected as the insoluble fraction. Insoluble protein samples were neutralized for Western blotting and further precipitated with trichloroacetic acid (TCA) when probing for insoluble tau. Protein samples were denatured at 95 °C for 15 min before being loaded onto 4-20 % TGX precast polyacrylamide gels (Bio-rad). Antibodies used for western blotting include: HT7 (1:1000), PS199 (Abcam; 1:1000), PS202 (Abcam; 1:1000), AT100 (ThermoFisher; 1:1000), AT270 (ThermoFisher; 1:1000), PHF1 (1:1000), 6E10 (1:1000), GFAP (1:1000). Mesoscale Discovery immunoassay kits (Mesoscale Diagnostics) were used for cytokine (K15048G) and Aβ38, 40, and 42 (K15199E) quantification of cortical samples following the manufacturers protocols. The proinflammatory MSD was able to detect levels of that were within the standard curve, whereas brain levels of IFN-γ, IL-12p70, CXCL1 and IL-4 were below the threshold of detection.

Confocal microscopy and quantification

Equivalent brain sections were picked and immunofluorescent sections were imaged using Olympus FX1200 confocal microscope. Amyloid plaque burden identified by 82E1 were visualized through a Z-stack image taken through the entire depth of the section at 1 μm intervals. Confocal files were then rendered in 3D and analyzed by a blinded observer using the volume function of IMARIS software (Bitplane). Microglia and astrocyte quantification by confocal microscopy was also analyzed using IMARIS. Microglia number was quantified using IMARIS spot function and process morphology was measured using filament length and branching functions. Astrocyte IMARIS quantification was performed using the volumetric function. Microglia phagocytosis of Aβ was quantified using a combined immunofluorescent staining of IBA1, 82E1, and CD68. High magnification Z-stack images were taken of randomly selected plaques while being blinded to IBA1 microglial staining. Quantification of internalized Aβ was done according to previously described protocols [40, 54]. To account for varying total microglia numbers across images, the internalized Aβ index was normalized to the number of microglia per image. Quantification of neuronal degeneration was performed in equivalent hippocampal brain sections using immunohistochemical labeling of Beta-III Tubulin (B3T). Four randomly selected square sub-areas were selected in pyramidal, radiatum, or molecular layers of hippocampus CA1 and quantified by optical density using ImageJ.

Nanostring analysis

RNA was isolated from microdissected hippocampi of WT, Thy-Tau22, 5xfAD, and T5x mice using RNeasy Plus Universal Mini Kit (Qiagen). RNA samples were run a custom Nanostring panel (Nanostring Technologies) examining mouse AD-linked genes. To evaluate mouse amyloidogenesis, we compared levels of murine RNA for genes APP, BACE1, BACE2, ADAM10, PSEN1, and PSEN2.

Statistical analysis

Statistical analysis was performed using StatView software (SAS Institute Inc.). Statistical comparisons between multiple groups were performed using ANOVA followed by Fischer’s PLSD post-hoc tests. Error bars represent the standard error of the mean. Groups were considered statistically significant when *p < 0.05 for both ANOVA and posthoc analysis.

Results

Aβ pathology induces robust tau hyperphosphorylation and neurofibrillary tangle formation in T5x mice

Brains of WT, Thy-Tau22, 5xfAD, and T5x mice (7 months) were examined by immunocytochemistry to assess accumulation of tau and beta-amyloid. Robust accumulation of tau (green) and Aβ (red) plaques was detected throughout the brain of T5x mice with extensive extracellular amyloid plaque pathology and intraneuronal tau accumulation observed within the hippocampus, neocortex and amygdala (Fig. 1). Next, biochemical approaches were used to quantify changes in tau pathology induced by Aβ accumulation. Soluble levels of total human tau (HT7 antibody) were quantified in the hippocampus and cortex of 7-month old littermates of each genotype (Fig. 2, Additional file 1: Figure S1). As expected, human tau was detectable only in mice that expressed the human tau transgene; Thy-Tau22 and T5x. Interestingly, levels of the 60 kDa human tau species were not significantly different between T5x and Thy-Tau22 littermates in either the hippocampus (p = 0.33) or cortex (p = 0.13). In contrast, the HT7 antibody also recognized a higher molecular weight tau species (65 kDa) that exhibited marked increases in T5x versus Thy-Tau22 mice in both the hippocampus (p = 0.0007; Fig. 2a) and cortex (p = 0.01; Additional file 1: Figure S1A). The ratio of the phosphorylated 65 kDa tau band to the unphosphorylated 60 kDa tau band was also significantly higher in T5x mice relative to Tau22 littermates in hippocampus (p = 0.004; Additional file 2: Figure S2A) and cortex (p = 0.001; Additional file 2: Figure S2A). Because the hyperphosphorylation of tau results in an electrophoretic shift in migration, these data provide evidence that Aβ accumulation leads to increased soluble hyperphosphorylated tau in T5x mice.
To further characterize how Aβ alters tau hyperphosphorylation, soluble hippocampal and cortical lysates were probed for multiple pathological tau epitopes (i.e. AT100, AT270, PHF-1). For many of the phospho-epitopes, two bands were often detected, likely representing tau species with varying degrees of hyperphosphorylation at multiple sites. Quantification of these phospho-epitopes within both the hippocampus and cortex, demonstrate that many exhibit a 2–3 fold elevation in T5x mice compared to Thy-Tau22 littermates (Fig. 2, Additional file 1: Figure S1, p < 0.001). As tau becomes hyperphosphorylated, it aggregates and becomes increasing insoluble. Therefore, we examined whether insoluble tau was also increased within T5x mice. While total levels of insoluble human tau (HT7) were unchanged between T5x and Thy-Tau22 mice, T5x mice exhibited a dramatic increase in several hyperphosphorylated insoluble tau species, as AT100, AT270, and PHF1 levels were elevated 5–8 fold in T5x mice compared to Thy-Tau22 littermates (Fig. 2, Additional file 1: Figure S1, p < 0.001).
Increased tau hyperphosphorylation within T5x brain was further confirmed immunohistochemically. The brains of T5x mice exhibited increased numbers and density of AT8, AT100, and PHF1 immunoreactive neurons within both the hippocampus and cortex when compared to Thy-Tau22 (Figs. 2c-d, Additional file 1: Figure S1C-D). In AD, it is hypothesized that tau misfolds prior to hyperphosphorylation and this change can be detected with the conformation-specific antibody MC-1 [38]. Indeed, MC-1 immunoreactivity was more prevalent in the brains of T5x versus Thy-Tau22 mice, exhibiting a more than three-fold increase in the hippocampus and cortex (Figs. 2e-f, Additional file 1: Figure S1E-F and 2B). Thus, consistent with previous reports, it appears that the presence of beta-amyloid pathology accelerates the accumulation, misfolding, and hyperphosphorylation of tau in T5x mice.

T5x mice exhibit differential levels of cytokines in brain

Cytokines have been implicated in the modulation of microglia number and function in the presence of beta-amyloid and tau pathologies. To determine the combined effects of tau and amyloid pathology on neuroinflammation, we quantified the protein levels of IL-10, IL-1β, IL-2, IL-5, IL-6, and TNF-α within the cortex (Fig. 3). In T5x mice, a surprising three-fold decrease in IL-1β relative to 5xfAD mice was observed (p < 0.0001; Fig. 3). Interestingly, IL-5, IL-6, and IL-10 levels were also decreased in T5x mice compared to 5xfAD littermates (p < 0.05; Fig. 3). In contrast, T5x mice showed an approximately 7-fold increase in both IL-2 and TNF-α compared to wild-type and transgenic littermates (p < 0.0001). Because IL-2 and TNF-α levels highly correlate with PHF-1 (Additional file 3: Figure S3, IL-2: PHF-1 R2 = 0.481, TNF-α: PHF-1 R2 = 0.557), this suggests that increasing tau pathology likely plays a role in the induction of these cytokines. Whereas IL-2 and TNF-α promotes microglial activation and proliferation, IL-10 has been shown to inhibit microglial Aβ phagocytosis [19, 29, 57, 89]. Taken together, the cytokine profile of T5x mice suggested that microglial function might be differentially affected by varying amounts of tau and amyloid and led us to investigate whether microglia number or activation state were also altered in T5x mice.

Tau and Aβ synergistically modulate astrocyte and microglia number and morphology

To determine whether neuroinflammatory changes in T5x mice were associated with astrogliosis, astrocyte morphology was examined in all genotypes (Additional file 4: Figure S4A). Astrocyte total volume, as assessed by GFAP immunoreactivity, was increased 2–3 fold in T5x mice compared to 5xfAD and Thy-Tau22 littermates within CA1 and the cortex (Additional file 4: Figure S4B, ANOVA and PLSD p < 0.05). Western blot analysis of cortical fractions further corroborated these results revealing an approximately two-fold increase in GFAP protein levels in T5x lysates (Additional file 4: Figure S4C, ANOVA and PLSD p < 0.05). This increase in astrogliosis is suggestive of a synergistic immune response to the combination of tau and beta-amyloid pathology in T5x mice.
Next, we sought to determine the effect of amyloid and tau on microgliosis as microglia are considered the predominant immune cells of the brain, are tasked with the bulk phagocytosis of CNS material including beta-amyloid, and are known to play a prominent role in AD pathogenesis [1, 48, 50, 64]. To investigate how microglia are affected by AD pathology in T5x mice, we quantified microglial number and characterized their morphology within the CA1, the dentate gyrus (DG), and the parietal association cortex (PAC) from all four genotypes (Fig. 4a, b, c, respectively). Our analysis revealed a more than two-fold increase in the number of microglia (IBA1+) within T5x mice compared to the other genotypes in CA1 (p < 0.0001) and this increase was similarly observed within the DG and PAC (Fig. 4a, d). Activated pro-inflammatory microglia are typified by short processes and decreased branching [60]. In T5x mice, microglia process length per microglia was significantly decreased compared to all other genotypes, in all three analyzed regions: CA1, DG, and PAC (Fig. 4e, ANOVA and PLSD p < 0.05). T5x mice also displayed significant decreases in microglial branching per microglia compared to all genotypes, in each of the same three brain regions (Fig. 4 f, ANOVA and PLSD, p < 0.05). 5xfAD microglia also exhibited significantly shorter processes and decreased branching compared to WT and Thy-Tau22 in PAC and DG, supporting previous studies demonstrating that amyloid accumulation can drive microglia activation [12, 55]. However, T5x microglia exhibited shorter processes and less process branching in CA1 relative to their age-matched 5xfAD counterparts-suggesting that tau plays a synergistic role with amyloid in the modulation of microglia activation state and that the dramatic increase in tau pathology in T5x mice further exacerbates microgliosis.
Additional evidence of tau-mediated microglial activation was observed by examination of the morphology of microglial populations within CA1 of the hippocampus-an area that exhibits abundant tauopathy [16, 72]. T5x brain sections labeled for Beta-III tubulin (B3T) and IBA1 revealed a distinct microglia population uniquely surrounding the dendrites of pyramidal neurons (Fig. 4h-k). These microglia are characterized by long rod-like morphology with shortened processes and were observed in T5x mice but not 5xfAD, Thy-Tau22, or WT littermates (Fig. 4a). Rod-shaped microglia of similar morphology have recently been reported to accumulate in areas of acute neuronal injury and axonal degeneration, further supporting the notion that accumulating tau pathology further alters microglial activation in T5x mice [82, 87].

Amyloid plaque burden is significantly reduced in T5x mice versus 5xfAD littermates

Although the effects of amyloid on tau hyperphosphorylation have been extensively studied, far fewer reports have examined whether tau pathology could potentially influence amyloid burden. While we hypothesized that no differences in Aβ plaque pathology would be detected between T5x and 5xfAD littermates, changes in microgliosis suggested that microglial interactions with Aβ could potentially be altered. We therefore examined Aβ plaque load using IMARIS 3-D volumetric quantification. Surprisingly, this analysis revealed a ~50 % reduction in Aβ plaque volume within multiple brain regions of T5x versus 5xfAD mice (Fig. 5a-b, p < 0.05). Amyloid plaque deposition was significantly lower in the CA1 of the hippocampus, the retrosplenial cortex (RC), and parietal association cortex (PAC) of T5x versus 5xfAD littermates (ANOVA and PSLD p < 0.05). Within the dentate gyrus of the hippocampus, T5x mice exhibited a non-significant trend towards decreased plaque volume (p = 0.07, Fig. 5b).
To examine whether the decrease in amyloid burden resulted from a down-regulation of the APP transgene, APP soluble protein levels were quantified by Western blot analysis in both the cortex (Fig. 5c-e) and hippocampus (data not shown). Our results revealed no changes in APP holoprotein (p = 0.81) and also no change in C99, the C-terminal fragment produced by beta-secretase cleavage of APP (p = 0.86) between T5x and 5xfAD littermates. To determine whether alterations in Aβ might be influenced by changes in endogenous amyloidogenic enzymes we also examined the mRNA expression of BACE1, BACE2, ADAM10, PSEN1, and PSEN2 and found no significant differences across genotypes (Additional file 5: Figure S5). These data therefore support the notion that changes in Aβ load may be mediated by alterations in Aβ clearance mechanisms, rather than altered Aβ production.
To further validate the observed effects on Aβ plaque load we used a Mesoscale Discovery (MSD) V-Plex Aβ Peptide ELISA to quantify both soluble and insoluble levels of Aβ38, Aβ40, and Aβ42. MSD analysis of soluble cortical fractions revealed no significant differences between T5x and 5xfAD littermates for Aβ38 Aβ40, or Aβ42, consistent with the notion that production of soluble Aβ is unchanged (Fig. 5 f). In contrast, levels of insoluble Aβ that contribute to fibrillar Aβ plaques exhibited a significant ~2-fold decrease in T5x Aβ 38 (p = 0.03), Aβ40 (p = 0.01) and Aβ42 (p = 0.02) versus 5xfAD mice (Fig. 6g). Thus, in T5x mice, there is a surprising two-fold decrease in insoluble Aβ species and total plaque volume when compared to 5xfAD littermates.

T5x microglia exhibit increased Aβ phagocytosis

While greater microglial numbers alone could potentially promote Aβ clearance, the observed changes in T5x microglial morphology and decreased brain IL-10 levels might also indicate increased microglial phagocytic capacity that could further contribute to Aβ reduction. Altered brain cytokine profiles indicate changes in microglia activation state, which can influence amyloid phagocytosis [23, 68]. For example, decreased IL-10 levels is associated with increased microglial Aβ phagocytosis [40, 54]. To test the hypothesis that reduced Aβ in T5x mice is the result of increased phagocytic capacity of T5x microglia, microglial Aβ internalization was assessed using volumetric IMARIS analysis in matching brain sections of T5x and 5xfAD mice. Microglia were identified by IBA1 immunoreactivity and Aβ phagocytosis was determined by co-localization of Aβ (82E1) and CD68 which is only present in microglial phagolysosomes (Fig. 6a). While the total volume of phagolysosome labeling per microglia was not significantly different between 5xfAD and T5x littermates (Fig. 6b, p = 0.18), quantification of the proportion of Aβ internalized within CD68+ phagolysosomes was significantly different between T5x and 5xfAD groups. In fact, this analysis revealed a nearly 30 % increase in the ability of T5x microglia to internalize Aβ versus 5xfAD littermates (Fig. 6c, p = 0.01). Taken together, these data sug\gest that increases in tau pathology and neuroinflammation can affect microglial Aβ phagocytosis, leading to a profound increase in amyloid clearance and reduction of plaque load in T5x mice.

Discussion

Although the notion that amyloid increases tau pathology is well established, very few studies have in contrast examined whether a reciprocal relationship between tau and the development of amyloid pathology might exist. Beginning in 2001, two seminal studies demonstrated that Aβ could exacerbate the development of tau pathology in transgenic AD tau models [24, 47]. Subsequently, a similar causal relationship between Aβ and tau pathology was established in several other AD models [13, 17, 39, 62]. Even in the absence of the expression of a human tau transgene, many AD transgenic mice exhibit some degree of Aβ-induced tau hyperphosphorylation, a prerequisite for the development of neurofibrillary tangles. Likewise, in humans, triplication of APP, either in some family pedigrees or in trisomy 21, leads to elevated levels of Aβ and the development of tau pathology [53, 79]. Independent of Aβ, alterations in Presenilin-1 have also been shown to increase tau hyperphosphorylation in transgenic mice expressing mutant tau [14, 78]. Thus, it is quite likely that the presence of a mutant PS1 transgene in 5xfAD influences tau phosphorylation both directly and indirectly by enhancing Aβ42 generation.
As expected, the accumulation of Aβ in T5x mice leads to a dramatic increase in tau hyperphosphorylation and increased accumulation of neurofibrillary tangle pathology within the hippocampus and neocortex. Yet surprisingly, T5x mice also exhibited a 50 % reduction in amyloid plaque burden and insoluble Aβ species versus 5xfAD littermates. Although the expression of both the APP and tau transgenes are driven by the Thy1 promoter, the expression of APP in T5x mice was unchanged and therefore, decreased Aβ levels were not the result of altered transgene expression. Furthermore, levels of soluble Aβ and APP processing enzymes were unaffected; suggesting that the observed decreases in plaques and insoluble Aβ was not due to effects on APP processing.
However, the combination of amyloid and tau pathology did produce a dramatic effect on neuroinflammatory processes, included increased microgliosis and astrogliosis in T5x versus 5xfAD or Thy-Tau22 littermates. Therefore, we examined whether changes in specific cytokines could explain the increase in neuroinflammation. Our data show that the combined presence of amyloid and tau pathology exacerbates the levels of both TNF-α and IL-2 (Fig. 4). In contrast, the observed decreases in IL-1β, IL-5, and IL-6 in T5x mice versus 5xfAD littermates may reflect the predominant influence of amyloid on the release and maturation of certain pro-inflammatory cytokines. Although IL-1β has been shown to increase tau phosphorylation, amyloid has also been reported to promote astrocytic and microglial release of IL-1β [20, 36, 40, 74]. Thus, while one may expect elevated IL-1β in T5x mice in association with increased tau hyperphosphorylation [49], the influence of reduced amyloid burden on IL1β induction may take precedence over effects caused by changes in tau. Therefore, it may be the case that IL-1β, IL-5 and IL-6 are less elevated in T5x than 5xfAD mice as a consequence, rather than a cause of reduced amyloid burden. However, future studies that examine the progression of pathology and inflammation in T5x across multiple ages will likely help to resolve these questions.
TNF-α, a pro-inflammatory cytokine that is released by activated microglia was one of the most upregulated cytokines observed in T5x mice [41, 55]. Although TNF-α does not directly increase microglial proliferation, TNF-α promotes astrocyte proliferation and GM-CSF release, which in turn, can stimulate microglial proliferation [6, 45, 73]. Taken together, elevated astrocyte numbers and a ~5-fold increase in TNF-α in T5x mice relative to 5xfAD and Thy-Tau22 littermates suggests that the combined stress of amyloid and tau pathology work synergistically to promote both astrocytic and microglial proliferation. Furthermore, the significant decrease in IL-10 expression in T5x mice suggested that microglial phagocytosis of Aβ may also be altered as recent studies have shown that deletion of IL-10 can enhance microglial Aβ phagocytosis [29]. Our finding that T5x mice exhibit a similar decrease in IL-10 relative to Thy-Tau22 and 5xFAD littermates suggest that decreased IL-10 is a result of the combined stress of tau and amyloid pathology. Subsequent analysis confirmed that T5x microglia indeed have increased Aβ phagocytosis, providing further support that tau pathology contributes to microglial regulation of amyloid. It should be noted that while our results support strong interactions between tau pathology and microglial activation, the exact temporal relationship between the two is still to be determined. Additional longitudinal studies will be needed to determine whether accumulation of tau pathology first promotes microglial activation or whether changes in microglia precede and drive increases in tau hyperphosphorylation.
While cytokines are often determined by the extent of AD pathology, they can also control the progression of the disease through the modulation of microglial response. The influence of microglia in AD has received growing attention with the recent discovery of genetic risk polymorphisms in several microglial-enriched genes [15]. Some of these risk factor genes such as CD33 have themselves been implicated in microglial Aβ phagocytosis [25, 81]. Thus, the role of microglia in limiting Aβ pathology continues to garner new attention. Microglia have long been implicated in the modulation of tau hyperphosphorylation and misfolding through cytokine-mediated neuroinflammatory activation of tau kinases [49, 84]. More recently, studies have shown that the misfolding of tau can promote microglial activation [88], and tau oligomers and fibrils have been found to interact directly with microglia [56]. In addition, microglia have also recently been implicated in the propagation of tau within the brain [4]. The fact that microglia exhibit the capacity to directly phagocytose the same tau oligomers that can promote their activation, highlights the possibility that tau pathology is not only exacerbated by microglial activation, but likely in turn modulates microglial responses within the brain [52]. We reasoned that changes in cytokine levels observed in T5x mice would affect microgliosis and our current findings suggest that tau and amyloid pathology in T5x mice synergistically recruit a robust microglial response. Significantly increased activated amoeboid microglia in T5x mice versus Thy-Tau22 littermates suggests that amyloid pathology is the strongest determinant of microglial activation. Amyloid holds an advantage over predominately intracellular tau aggregates in its potential influence over microglial activation as evident of amyloid soluble oligomers released from abundant extracellular plaques [55, 70, 77]. However, our findings of increased microglial activation, phagocytosis, and numbers in T5x mice, suggest that tau pathology can also contribute to or further modulate microglia activation state.
The remaining question is exactly how development of tau pathology influences the neuroinflammatory response. One possibility is that amyloid driven tau hyperphosphorylation and subsequent release of intracellular factors and tau oligomers from degenerating neurons leads to altered microglial activation states, potentially increasing Aβ phagocytosis and thereby providing a somewhat beneficial response. Previous studies have reported the activation of microglia by the release of apoptotic factors and intracellular molecules such as ADP or ATP [21, 37, 58]. When we examined T5x mice for dendritic degeneration, we observed a reduction in hippocampal Beta-III tubulin staining in T5x mice compared to transgenic and WT littermates (Additional file 6: Figure S6). Notably, hippocampal regions displaying the significant reduction in Beta-III tubulin were accompanied by the previously described rod microglia (Fig. 4 g-j) [80]. Although not extensively characterized, rod microglia have been found in human AD cases and exhibit abnormally frequent interactions with synaptic clefts [46, 59, 82]. T5x rod-like microglia are similar in morphology to recently described rod-microglia which appear highly activated and respond to diffuse neuronal injury [87]. The association between reduced β3-Tubullin density and rod microglia presence therefore suggests that degenerating dendrites and subsequent release of intracellular factors such as ADP may contribute to the altered microglia activation state in T5x mice. However, further studies are clearly needed to elucidate the exact mechanisms by which tau synergizes with Aβ to modulate microglial activation.
It is important to acknowledge that our findings both corroborate and differ from other recently examined amyloid-tau crosses. We report a significant reduction in amyloid plaque burden and insoluble Aβ in T5x mice relative to 5xfAD littermates that appears to be mediated via increased microglial-mediated Aβ clearance. Using a similar model, one group has shown that 5xfAD crossed with Tg30 tau mice likewise exhibit decreased Aβ pathology, although the potential role of microglia in this finding was not examined [35]. In contrast, two other studies crossed 5xfAD mice with other tau models and reported no change in Aβ plaques by optical densitometry, although only 3 or 4 mice per group were compared versus group sizes of 9–10 mice for the current Aβ ELISA analysis [75]. Yet, one of these studies nevertheless showed a non-significant 45 % reduction in Aβ plaque load at 3 months of age. It is therefore quite possible that the use of 3D plaque volume quantification, ELISA, or a larger sample size would have revealed a similar significant reduction in Aβ to that observed in the current study. In another study, Tg2576 APP mice were crossed with mutant tau VLW mice. Surprisingly, this study reported a significant increase in amyloid deposition in bigenic mice [67]. Likewise, a recent study that examined a cross between rTgTauEC and APP/PS1 mice also reported increased amyloid burden [66].
These varying reports suggest that perhaps the magnitude of tau pathology and/or microgliosis can influence the effects on Aβ pathology. In support of this notion, we find that T5x mice exhibit a dramatic increase in microglial number, shift in morphology, and change in cytokines that is not apparent in Thy-Tau22 littermates with lower levels of tau pathology (Figs. 4 and 5). Interestingly, in other studies that crossed different tau models with 5xfAD mice [35, 75], there were either no significant changes or a similar decrease in amyloid pathology. In contrast, one study reported an increase in amyloid burden when Tg2576 mice were crossed with VLW tau mice [67]. 5xfAD mice, which carry three APP and two PS1 mutations, generate considerably more amyloid pathology than Tg2576 mice and the presence of a PS1 transgene may further promote tau hyperphosphorylation both directly [14, 78] and indirectly by enhancing Aβ42 generation. Thus, differences in levels of tau, the type and magnitude of amyloid pathology, and their corresponding effects on microglial activation state might explain these discrepancies. For example, it is quite possible that the more aggregation prone Aβ42 leads to a greater increase in tau pathology and a differential activation of microglia that combine to promote microglial phagocytosis. Another possible explanation for these contrasting findings is the potential impact of background strain on pathology. In the current study we crossed congenic C57/Bl6 5xfAD mice with congenic C57/Bl6 Thy-Tau22 mice, thus background strain was identical between all four genotypes examined. In some of the contrasting studies, the cross-bred mice exhibited differential mixed background strains that include Bl6, CH3, and FVB [66] and thus variability in background genetics even between littermates could influence the development of pathology. Although our current data clearly suggests that tau pathology can reduce the accumulation of Aβ, these prior reports that in many cases corroborate our findings but in others contrast our findings clearly suggest that this remains a highly complex question that will need additional examination. For example, it is quite possible that changes in APP transport or alterations in autophagy also play a role in this process and thus studies that further examine these additional potential mechanisms are also needed.

Conclusion

In conclusion, we have established an AD model that provides further insight into the potential reciprocal relationships between amyloid, tau and neuroinflammation. The combination of these pathologies significantly increases tau hyperphosphorylation and microgliosis, yet decreases amyloid burden. Our data also suggest that tau-induced increases in microglial number, and phagocytic activity may explain the reductions in amyloid burden. The changes observed in amyloid accumulation and microgliosis in the presence of tau pathology suggests that therapies targeting tauopathy could have increased benefit towards treating additional underlying proponents of AD. Therefore, our study adds to the growing understanding of the role of microglia in AD pathogenesis and suggests that differential activation of these cells in response to Aβ and tau pathology can have both detrimental as well as beneficial effects on disease progression.

Acknowledgments

This work was supported in part by: NIH AG048099 and Alzheimer’s Association BFG-14-317000 (M.B-J.), NIH AG16573 (M.B.J. and W.W.P.), and CIRM TG NS082174- (W.C.). L.B. and D.B. are supported by ANR (ADORATAU, CYTOKALZ, SPREADTAU), Association France Alzheimer, LECMA/Alzheimer Forschung Initiative, Fondation Plan Alzheimer and from the programs Investissements d’avenir LabEx (excellence laboratory) DISTALZ (Development of Innovative Strategies for a Transdisciplinary approach to ALZheimer’s disease), Inserm, CNRS, Université Lille 2, UPMC, Lille Métropole Communauté Urbaine, Région Nord/Pas-de-Calais, FEDER, DN2M and FUI MEDIALZ.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interest.

Authors’ contributions

WC, WWP, EMA, and M B-J. designed research; WC, AL, and MB-J performed research; WC, AL, and MB-J. analyzed data; LB and DB provided the Thy-Tau22 mice; WC, WWP, EMA, and MB-J wrote the paper. All authors read and approved the final manuscript.
Anhänge

Additional files

Additional files 2: Figure S2. T5x mice exhibit increased levels of Tau phosphorylation and misfolding. (A) The ratio of phosphorylated human tau (65 kDa) to unphosphorylated human tau (60 kDa) was quantified by western blot. Analysis revealed significantly higher 65:60 kDa HT7 ratio in T5x mice relative to Tau22 littermates, suggesting that tau hyperphosphorylation is exacerbated by the presence of Aβ in T5x mice. (B) Equivalent hippocampal sections were examined from 7-month old T5x and Thy-Tau22 mice and immunohistochemically labeled with the conformational-specific tau antibody; MC-1 (see Figs. 2 and 3). Quantification revealed a significant increase in the numbers of MC-1-positive neurons in hippocampus CA1 (p < 0.0001) and an even greater increase in MC-1-immunoreactive cells within the perirhinal/entorhinal cortex. Data are represented as mean ± SEM of optical density (O.D.), n ≥ 8 mice/group. * Indicates p < 0.05 for both ANOVA and Fisher’s protected least-significant difference (PLSD) post hoc tests. (PDF 427 kb)
Literatur
2.
Zurück zum Zitat Amadoro G et al. Endogenous Aβ causes cell death via early tau hyperphosphorylation. Neurobiol Aging. 2011;32(6):969–90.CrossRefPubMed Amadoro G et al. Endogenous Aβ causes cell death via early tau hyperphosphorylation. Neurobiol Aging. 2011;32(6):969–90.CrossRefPubMed
3.
Zurück zum Zitat Apelt J, Schliebs R. β-Amyloid-induced glial expression of both pro-and anti-inflammatory cytokines in cerebral cortex of aged transgenic Tg2576 mice with Alzheimer plaque pathology. Brain Res. 2001;894(1):21–30.CrossRefPubMed Apelt J, Schliebs R. β-Amyloid-induced glial expression of both pro-and anti-inflammatory cytokines in cerebral cortex of aged transgenic Tg2576 mice with Alzheimer plaque pathology. Brain Res. 2001;894(1):21–30.CrossRefPubMed
4.
Zurück zum Zitat Asai H. et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nature neuroscience. 2015;18(11):1584–93. Asai H. et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nature neuroscience. 2015;18(11):1584–93.
5.
Zurück zum Zitat Association A.s. 2014 Alzheimer’s disease facts and figures. Alzheimers Dement. 2014;10(2):e47–92.CrossRef Association A.s. 2014 Alzheimer’s disease facts and figures. Alzheimers Dement. 2014;10(2):e47–92.CrossRef
6.
Zurück zum Zitat Barna BP et al. Human astrocytes proliferate in response to tumor necrosis factor alpha. J Neuroimmunol. 1990;30(2–3):239–43.CrossRefPubMed Barna BP et al. Human astrocytes proliferate in response to tumor necrosis factor alpha. J Neuroimmunol. 1990;30(2–3):239–43.CrossRefPubMed
7.
Zurück zum Zitat Belarbi K et al. Beneficial effects of exercise in a transgenic mouse model of Alzheimer’s disease-like Tau pathology. Neurobiol Dis. 2011;43(2):486–94.CrossRefPubMed Belarbi K et al. Beneficial effects of exercise in a transgenic mouse model of Alzheimer’s disease-like Tau pathology. Neurobiol Dis. 2011;43(2):486–94.CrossRefPubMed
8.
Zurück zum Zitat Benilova I, Karran E, De Strooper B. The toxic A [beta] oligomer and Alzheimer’s disease: an emperor in need of clothes. Nat Neurosci. 2012;15(3):349–57.CrossRefPubMed Benilova I, Karran E, De Strooper B. The toxic A [beta] oligomer and Alzheimer’s disease: an emperor in need of clothes. Nat Neurosci. 2012;15(3):349–57.CrossRefPubMed
9.
Zurück zum Zitat Benitez BA et al. Missense variant in TREML2 protects against Alzheimer’s disease. Neurobiol Aging. 2014;35(6):1510.e19–26.CrossRef Benitez BA et al. Missense variant in TREML2 protects against Alzheimer’s disease. Neurobiol Aging. 2014;35(6):1510.e19–26.CrossRef
11.
Zurück zum Zitat Bisht K. et al. Dark microglia: A new phenotype predominantly associated with pathological states. Glia. 2016;64(5):826–39. Bisht K. et al. Dark microglia: A new phenotype predominantly associated with pathological states. Glia. 2016;64(5):826–39.
12.
Zurück zum Zitat Block ML, Zecca L, Hong J-S. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8(1):57–69.CrossRefPubMed Block ML, Zecca L, Hong J-S. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8(1):57–69.CrossRefPubMed
13.
Zurück zum Zitat Blurton-Jones M, Laferla FM. Pathways by which Abeta facilitates tau pathology. Curr Alzheimer Res. 2006;3(5):437–48.CrossRefPubMed Blurton-Jones M, Laferla FM. Pathways by which Abeta facilitates tau pathology. Curr Alzheimer Res. 2006;3(5):437–48.CrossRefPubMed
14.
Zurück zum Zitat Boutajangout A et al. Increased tau phosphorylation but absence of formation of neurofibrillary tangles in mice double transgenic for human tau and Alzheimer mutant (M146L) presenilin-1. Neurosci Lett. 2002;318(1):29–33.CrossRefPubMed Boutajangout A et al. Increased tau phosphorylation but absence of formation of neurofibrillary tangles in mice double transgenic for human tau and Alzheimer mutant (M146L) presenilin-1. Neurosci Lett. 2002;318(1):29–33.CrossRefPubMed
15.
Zurück zum Zitat Bradshaw EM et al. CD33 Alzheimer’s disease locus: altered monocyte function and amyloid biology. Nat Neurosci. 2013;16(7):848–50.PubMedPubMedCentral Bradshaw EM et al. CD33 Alzheimer’s disease locus: altered monocyte function and amyloid biology. Nat Neurosci. 2013;16(7):848–50.PubMedPubMedCentral
16.
Zurück zum Zitat Bramblett G, Trojanowski J, Lee V. Regions with abundant neurofibrillary pathology in human brain exhibit a selective reduction in levels of binding-competent tau and accumulation of abnormal tau-isoforms (A68 proteins). Laboratory investigation; a journal of technical methods and pathology. 1992;66(2):212–22.PubMed Bramblett G, Trojanowski J, Lee V. Regions with abundant neurofibrillary pathology in human brain exhibit a selective reduction in levels of binding-competent tau and accumulation of abnormal tau-isoforms (A68 proteins). Laboratory investigation; a journal of technical methods and pathology. 1992;66(2):212–22.PubMed
18.
Zurück zum Zitat Chabrier MA et al. Synergistic effects of amyloid-beta and wild-type human tau on dendritic spine loss in a floxed double transgenic model of Alzheimer’s disease. Neurobiol Dis. 2014;64:107–17.CrossRefPubMedPubMedCentral Chabrier MA et al. Synergistic effects of amyloid-beta and wild-type human tau on dendritic spine loss in a floxed double transgenic model of Alzheimer’s disease. Neurobiol Dis. 2014;64:107–17.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Chakrabarty P et al. IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron. 2015;85(3):519–33.CrossRefPubMedPubMedCentral Chakrabarty P et al. IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron. 2015;85(3):519–33.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Combs CK et al. β-Amyloid stimulation of microglia and monocytes results in TNFα-dependent expression of inducible nitric oxide synthase and neuronal apoptosis. J Neurosci. 2001;21(4):1179–88.PubMed Combs CK et al. β-Amyloid stimulation of microglia and monocytes results in TNFα-dependent expression of inducible nitric oxide synthase and neuronal apoptosis. J Neurosci. 2001;21(4):1179–88.PubMed
21.
Zurück zum Zitat Davalos D et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8(6):752–8.CrossRefPubMed Davalos D et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8(6):752–8.CrossRefPubMed
22.
Zurück zum Zitat Eimer WA, Vassar R. Neuron loss in the 5XFAD mouse model of Alzheimer’s disease correlates with intraneuronal Aβ42 accumulation and Caspase-3 activation. Mol Neurodegener. 2013;8(1):1–12.CrossRef Eimer WA, Vassar R. Neuron loss in the 5XFAD mouse model of Alzheimer’s disease correlates with intraneuronal Aβ42 accumulation and Caspase-3 activation. Mol Neurodegener. 2013;8(1):1–12.CrossRef
23.
Zurück zum Zitat Fiala M et al. Phagocytosis of amyloid-β and inflammation: two faces of innate immunity in Alzheimer’s disease. J Alzheimers Dis. 2007;11(4):457–63.PubMed Fiala M et al. Phagocytosis of amyloid-β and inflammation: two faces of innate immunity in Alzheimer’s disease. J Alzheimers Dis. 2007;11(4):457–63.PubMed
24.
Zurück zum Zitat Gotz J et al. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science. 2001;293(5534):1491–5.CrossRefPubMed Gotz J et al. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science. 2001;293(5534):1491–5.CrossRefPubMed
26.
Zurück zum Zitat Griffin W. IL-1 and the cytokine cycle in Alzheimer’s disease. in Journal of Neurochemistry. 2000. Lippincott Williams & Wilkins 530 Walnut ST, Philadelphia. PA 19106–3621 USA. Griffin W. IL-1 and the cytokine cycle in Alzheimer’s disease. in Journal of Neurochemistry. 2000. Lippincott Williams & Wilkins 530 Walnut ST, Philadelphia. PA 19106–3621 USA.
27.
Zurück zum Zitat Griffin W et al. Glial‐neuronal interactions in Alzheimer’s disease: The potential role of a ‘cytokine cycle’in disease progression. Brain Pathol. 1998;8(1):65–72.CrossRefPubMed Griffin W et al. Glial‐neuronal interactions in Alzheimer’s disease: The potential role of a ‘cytokine cycle’in disease progression. Brain Pathol. 1998;8(1):65–72.CrossRefPubMed
28.
Zurück zum Zitat Guerreiro R et al. TREM2 variants in Alzheimer’s disease. N Engl J Med. 2013;368(2):117–27.CrossRefPubMed Guerreiro R et al. TREM2 variants in Alzheimer’s disease. N Engl J Med. 2013;368(2):117–27.CrossRefPubMed
29.
30.
Zurück zum Zitat Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–6.CrossRefPubMed Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–6.CrossRefPubMed
31.
Zurück zum Zitat Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184.CrossRefPubMed Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184.CrossRefPubMed
32.
Zurück zum Zitat Harold D et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat Genet. 2009;41(10):1088–93.CrossRefPubMedPubMedCentral Harold D et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat Genet. 2009;41(10):1088–93.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Hauss-Wegrzyniak B et al. Chronic neuroinflammation in rats reproduces components of the neurobiology of Alzheimer’s disease. Brain Res. 1998;780(2):294–303.CrossRefPubMed Hauss-Wegrzyniak B et al. Chronic neuroinflammation in rats reproduces components of the neurobiology of Alzheimer’s disease. Brain Res. 1998;780(2):294–303.CrossRefPubMed
34.
Zurück zum Zitat Heneka MT et al. Neuroinflammatory processes in Alzheimer’s disease. J Neural Transm. 2010;117(8):919–47.CrossRefPubMed Heneka MT et al. Neuroinflammatory processes in Alzheimer’s disease. J Neural Transm. 2010;117(8):919–47.CrossRefPubMed
35.
Zurück zum Zitat Héraud C et al. Increased misfolding and truncation of tau in APP/PS1/tau transgenic mice compared to mutant tau mice. Neurobiol Dis. 2014;62:100–12.CrossRefPubMed Héraud C et al. Increased misfolding and truncation of tau in APP/PS1/tau transgenic mice compared to mutant tau mice. Neurobiol Dis. 2014;62:100–12.CrossRefPubMed
36.
Zurück zum Zitat Hu J et al. Amyloid-β peptide activates cultured astrocytes: morphological alterations, cytokine induction and nitric oxide release. Brain Res. 1998;785(2):195–206.CrossRefPubMed Hu J et al. Amyloid-β peptide activates cultured astrocytes: morphological alterations, cytokine induction and nitric oxide release. Brain Res. 1998;785(2):195–206.CrossRefPubMed
37.
38.
Zurück zum Zitat Jicha GA et al. Alz‐50 and MC‐1, a new monoclonal antibody raised to paired helical filaments, recognize conformational epitopes on recombinant tau. J Neurosci Res. 1997;48(2):128–32.CrossRefPubMed Jicha GA et al. Alz‐50 and MC‐1, a new monoclonal antibody raised to paired helical filaments, recognize conformational epitopes on recombinant tau. J Neurosci Res. 1997;48(2):128–32.CrossRefPubMed
39.
Zurück zum Zitat Kitazawa M et al. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer’s disease. J Neurosci. 2005;25(39):8843–53.CrossRefPubMed Kitazawa M et al. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer’s disease. J Neurosci. 2005;25(39):8843–53.CrossRefPubMed
40.
Zurück zum Zitat Koenigsknecht-Talboo J, Landreth GE. Microglial phagocytosis induced by fibrillar β-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J Neurosci. 2005;25(36):8240–9.CrossRefPubMed Koenigsknecht-Talboo J, Landreth GE. Microglial phagocytosis induced by fibrillar β-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J Neurosci. 2005;25(36):8240–9.CrossRefPubMed
41.
Zurück zum Zitat Kuno R et al. Autocrine activation of microglia by tumor necrosis factor-α. J Neuroimmunol. 2005;162(1):89–96.CrossRefPubMed Kuno R et al. Autocrine activation of microglia by tumor necrosis factor-α. J Neuroimmunol. 2005;162(1):89–96.CrossRefPubMed
42.
Zurück zum Zitat Lambert J-C et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet. 2013;45(12):1452–8.CrossRefPubMedPubMedCentral Lambert J-C et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet. 2013;45(12):1452–8.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Landel V et al. Temporal gene profiling of the 5XFAD transgenic mouse model highlights the importance of microglial activation in Alzheimer’s disease. Mol Neurodegener. 2014;9(1):1–18.CrossRef Landel V et al. Temporal gene profiling of the 5XFAD transgenic mouse model highlights the importance of microglial activation in Alzheimer’s disease. Mol Neurodegener. 2014;9(1):1–18.CrossRef
44.
Zurück zum Zitat Laurent C et al. Beneficial effects of caffeine in a transgenic model of Alzheimer’s disease-like tau pathology. Neurobiol Aging. 2014;35(9):2079–90.CrossRefPubMed Laurent C et al. Beneficial effects of caffeine in a transgenic model of Alzheimer’s disease-like tau pathology. Neurobiol Aging. 2014;35(9):2079–90.CrossRefPubMed
45.
Zurück zum Zitat Lee SC et al. GM‐CSF promotes proliferation of human fetal and adult microglia in primary cultures. Glia. 1994;12(4):309–18.CrossRefPubMed Lee SC et al. GM‐CSF promotes proliferation of human fetal and adult microglia in primary cultures. Glia. 1994;12(4):309–18.CrossRefPubMed
46.
Zurück zum Zitat Lewandowska E et al. Ultrastructural evaluation of activated forms of microglia in human brain in selected neurological diseases (SSPE, Wilson’s disease and Alzheimer’s disease). Folia Neuropathol. 2003;42(2):81–91. Lewandowska E et al. Ultrastructural evaluation of activated forms of microglia in human brain in selected neurological diseases (SSPE, Wilson’s disease and Alzheimer’s disease). Folia Neuropathol. 2003;42(2):81–91.
47.
Zurück zum Zitat Lewis J et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science. 2001;293(5534):1487–91.CrossRefPubMed Lewis J et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science. 2001;293(5534):1487–91.CrossRefPubMed
48.
Zurück zum Zitat Li C et al. Astrocytes: implications for neuroinflammatory pathogenesis of Alzheimer’s disease. Current Alzheimer Research. 2011;8(1):67–80.CrossRefPubMed Li C et al. Astrocytes: implications for neuroinflammatory pathogenesis of Alzheimer’s disease. Current Alzheimer Research. 2011;8(1):67–80.CrossRefPubMed
49.
Zurück zum Zitat Li Y et al. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003;23(5):1605–11.PubMedPubMedCentral Li Y et al. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003;23(5):1605–11.PubMedPubMedCentral
50.
Zurück zum Zitat Liu B, Hong J-S. Role of microglia in inflammation-mediated neurodegenerative diseases: mechanisms and strategies for therapeutic intervention. J Pharmacol Exp Ther. 2003;304(1):1–7.CrossRefPubMed Liu B, Hong J-S. Role of microglia in inflammation-mediated neurodegenerative diseases: mechanisms and strategies for therapeutic intervention. J Pharmacol Exp Ther. 2003;304(1):1–7.CrossRefPubMed
51.
Zurück zum Zitat Lucin KM et al. Microglial beclin 1 regulates retromer trafficking and phagocytosis and is impaired in Alzheimer’s disease. Neuron. 2013;79(5):873–86.CrossRefPubMedPubMedCentral Lucin KM et al. Microglial beclin 1 regulates retromer trafficking and phagocytosis and is impaired in Alzheimer’s disease. Neuron. 2013;79(5):873–86.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Mann DM. The pathological association between Down syndrome and Alzheimer disease. Mech Ageing Dev. 1988;43(2):99–136.CrossRefPubMed Mann DM. The pathological association between Down syndrome and Alzheimer disease. Mech Ageing Dev. 1988;43(2):99–136.CrossRefPubMed
54.
Zurück zum Zitat Marsh SE et al. The adaptive immune system restrains Alzheimer’s disease pathogenesis by modulating microglial function. Proc Natl Acad Sci. 2016;113(9):E1316–25.CrossRefPubMedPubMedCentral Marsh SE et al. The adaptive immune system restrains Alzheimer’s disease pathogenesis by modulating microglial function. Proc Natl Acad Sci. 2016;113(9):E1316–25.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Meda L et al. Activation of microglial cells by β-amyloid protein and interferon-γ. Nature. 1995;374(6523):647–50.CrossRefPubMed Meda L et al. Activation of microglial cells by β-amyloid protein and interferon-γ. Nature. 1995;374(6523):647–50.CrossRefPubMed
56.
Zurück zum Zitat Morales I et al. Tau oligomers and fibrils induce activation of microglial cells. J Alzheimers Dis. 2013;37(4):849–56.PubMed Morales I et al. Tau oligomers and fibrils induce activation of microglial cells. J Alzheimers Dis. 2013;37(4):849–56.PubMed
57.
Zurück zum Zitat Nakajima K, Kohsaka S. Microglia: activation and their significance in the central nervous system. J Biochem. 2001;130(2):169–75.CrossRefPubMed Nakajima K, Kohsaka S. Microglia: activation and their significance in the central nervous system. J Biochem. 2001;130(2):169–75.CrossRefPubMed
58.
Zurück zum Zitat Nakamura Y. Regulating factors for microglial activation. Biol Pharm Bull. 2002;25(8):945–53.CrossRefPubMed Nakamura Y. Regulating factors for microglial activation. Biol Pharm Bull. 2002;25(8):945–53.CrossRefPubMed
59.
Zurück zum Zitat Nelson PT, Soma LA, Lavi E. Microglia in diseases of the central nervous system. Ann Med. 2002;34(7):491–500.CrossRefPubMed Nelson PT, Soma LA, Lavi E. Microglia in diseases of the central nervous system. Ann Med. 2002;34(7):491–500.CrossRefPubMed
60.
Zurück zum Zitat Neumann H et al. Tumor necrosis factor inhibits neurite outgrowth and branching of hippocampal neurons by a rho-dependent mechanism. J Neurosci. 2002;22(3):854–62.PubMed Neumann H et al. Tumor necrosis factor inhibits neurite outgrowth and branching of hippocampal neurons by a rho-dependent mechanism. J Neurosci. 2002;22(3):854–62.PubMed
61.
Zurück zum Zitat Oakley H et al. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J Neurosci. 2006;26(40):10129–40.CrossRefPubMed Oakley H et al. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J Neurosci. 2006;26(40):10129–40.CrossRefPubMed
62.
Zurück zum Zitat Oddo S et al. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron. 2003;39(3):409–21.CrossRefPubMed Oddo S et al. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron. 2003;39(3):409–21.CrossRefPubMed
63.
64.
Zurück zum Zitat Perry VH, Nicoll JA, Holmes C. Microglia in neurodegenerative disease. Nat Rev Neurol. 2010;6(4):193–201.CrossRefPubMed Perry VH, Nicoll JA, Holmes C. Microglia in neurodegenerative disease. Nat Rev Neurol. 2010;6(4):193–201.CrossRefPubMed
65.
Zurück zum Zitat Pigino G et al. Disruption of fast axonal transport is a pathogenic mechanism for intraneuronal amyloid beta. Proc Natl Acad Sci. 2009;106(14):5907–12.CrossRefPubMedPubMedCentral Pigino G et al. Disruption of fast axonal transport is a pathogenic mechanism for intraneuronal amyloid beta. Proc Natl Acad Sci. 2009;106(14):5907–12.CrossRefPubMedPubMedCentral
66.
67.
Zurück zum Zitat Ribé EM et al. Accelerated amyloid deposition, neurofibrillary degeneration and neuronal loss in double mutant APP/tau transgenic mice. Neurobiol Dis. 2005;20(3):814–22.CrossRefPubMed Ribé EM et al. Accelerated amyloid deposition, neurofibrillary degeneration and neuronal loss in double mutant APP/tau transgenic mice. Neurobiol Dis. 2005;20(3):814–22.CrossRefPubMed
68.
Zurück zum Zitat Rogers J, Lue L-F. Microglial chemotaxis, activation, and phagocytosis of amyloid β-peptide as linked phenomena in Alzheimer’s disease. Neurochem Int. 2001;39(5):333–40.CrossRefPubMed Rogers J, Lue L-F. Microglial chemotaxis, activation, and phagocytosis of amyloid β-peptide as linked phenomena in Alzheimer’s disease. Neurochem Int. 2001;39(5):333–40.CrossRefPubMed
69.
Zurück zum Zitat Rogers J et al. Microglia and inflammatory mechanisms in the clearance of amyloid β peptide. Glia. 2002;40(2):260–9.CrossRefPubMed Rogers J et al. Microglia and inflammatory mechanisms in the clearance of amyloid β peptide. Glia. 2002;40(2):260–9.CrossRefPubMed
70.
Zurück zum Zitat Sasaki A et al. Microglial activation in early stages of amyloid β protein deposition. Acta Neuropathol. 1997;94(4):316–22.CrossRefPubMed Sasaki A et al. Microglial activation in early stages of amyloid β protein deposition. Acta Neuropathol. 1997;94(4):316–22.CrossRefPubMed
71.
Zurück zum Zitat Saul A et al. Accelerated tau pathology with synaptic and neuronal loss in a novel triple transgenic mouse model of Alzheimer’s disease. Neurobiol Aging. 2013;34(11):2564–73.CrossRefPubMed Saul A et al. Accelerated tau pathology with synaptic and neuronal loss in a novel triple transgenic mouse model of Alzheimer’s disease. Neurobiol Aging. 2013;34(11):2564–73.CrossRefPubMed
72.
Zurück zum Zitat Schindowski K et al. Alzheimer’s disease-like tau neuropathology leads to memory deficits and loss of functional synapses in a novel mutated tau transgenic mouse without any motor deficits. Am J Pathol. 2006;169(2):599–616.CrossRefPubMedPubMedCentral Schindowski K et al. Alzheimer’s disease-like tau neuropathology leads to memory deficits and loss of functional synapses in a novel mutated tau transgenic mouse without any motor deficits. Am J Pathol. 2006;169(2):599–616.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Selmaj K et al. Proliferation of astrocytes in vitro in response to cytokines. A primary role for tumor necrosis factor. J Immunol. 1990;144(1):129–35.PubMed Selmaj K et al. Proliferation of astrocytes in vitro in response to cytokines. A primary role for tumor necrosis factor. J Immunol. 1990;144(1):129–35.PubMed
74.
Zurück zum Zitat Shaftel SS et al. Sustained hippocampal IL-1β overexpression mediates chronic neuroinflammation and ameliorates Alzheimer plaque pathology. J Clin Invest. 2007;117(6):1595–604.CrossRefPubMedPubMedCentral Shaftel SS et al. Sustained hippocampal IL-1β overexpression mediates chronic neuroinflammation and ameliorates Alzheimer plaque pathology. J Clin Invest. 2007;117(6):1595–604.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Stancu IC et al. Tauopathy contributes to synaptic and cognitive deficits in a murine model for Alzheimer’s disease. Faseb j. 2014;28(6):2620–31.CrossRefPubMed Stancu IC et al. Tauopathy contributes to synaptic and cognitive deficits in a murine model for Alzheimer’s disease. Faseb j. 2014;28(6):2620–31.CrossRefPubMed
76.
Zurück zum Zitat Stephan AH, Barres BA, Stevens B. The complement system: an unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci. 2012;35:369–89.CrossRefPubMed Stephan AH, Barres BA, Stevens B. The complement system: an unexpected role in synaptic pruning during development and disease. Annu Rev Neurosci. 2012;35:369–89.CrossRefPubMed
77.
Zurück zum Zitat Tan J et al. Microglial activation resulting from CD40-CD40L interaction after β-amyloid stimulation. Science. 1999;286(5448):2352–5.CrossRefPubMed Tan J et al. Microglial activation resulting from CD40-CD40L interaction after β-amyloid stimulation. Science. 1999;286(5448):2352–5.CrossRefPubMed
78.
Zurück zum Zitat Tanemura K et al. Formation of tau inclusions in knock-in mice with familial Alzheimer disease (FAD) mutation of presenilin 1 (PS1). J Biol Chem. 2006;281(8):5037–41.CrossRefPubMed Tanemura K et al. Formation of tau inclusions in knock-in mice with familial Alzheimer disease (FAD) mutation of presenilin 1 (PS1). J Biol Chem. 2006;281(8):5037–41.CrossRefPubMed
79.
Zurück zum Zitat Tanzi RE et al. Amyloid beta protein gene: cDNA, mRNA distribution, and genetic linkage near the Alzheimer locus. Science. 1987;235(4791):880–4.CrossRefPubMed Tanzi RE et al. Amyloid beta protein gene: cDNA, mRNA distribution, and genetic linkage near the Alzheimer locus. Science. 1987;235(4791):880–4.CrossRefPubMed
81.
82.
Zurück zum Zitat Wierzba-Bobrowicz T et al. Morphological analysis of active microglia--rod and ramified microglia in human brains affected by some neurological diseases (SSPE, Alzheimer’s disease and Wilson’s disease). Folia Neuropathol. 2001;40(3):125–31. Wierzba-Bobrowicz T et al. Morphological analysis of active microglia--rod and ramified microglia in human brains affected by some neurological diseases (SSPE, Alzheimer’s disease and Wilson’s disease). Folia Neuropathol. 2001;40(3):125–31.
83.
Zurück zum Zitat Wirths O, Multhaup G, Bayer TA. A modified β‐amyloid hypothesis: intraneuronal accumulation of the β‐amyloid peptide–the first step of a fatal cascade. J Neurochem. 2004;91(3):513–20.CrossRefPubMed Wirths O, Multhaup G, Bayer TA. A modified β‐amyloid hypothesis: intraneuronal accumulation of the β‐amyloid peptide–the first step of a fatal cascade. J Neurochem. 2004;91(3):513–20.CrossRefPubMed
84.
Zurück zum Zitat Yoshiyama Y et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53(3):337–51.CrossRefPubMed Yoshiyama Y et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53(3):337–51.CrossRefPubMed
85.
Zurück zum Zitat Zahs KR, Ashe KH. Too much good news’–are Alzheimer mouse models trying to tell us how to prevent, not cure, Alzheimer’s disease? Trends Neurosci. 2010;33(8):381–9.CrossRefPubMed Zahs KR, Ashe KH. Too much good news’–are Alzheimer mouse models trying to tell us how to prevent, not cure, Alzheimer’s disease? Trends Neurosci. 2010;33(8):381–9.CrossRefPubMed
86.
Zurück zum Zitat Zhang B et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell. 2013;153(3):707–20.CrossRefPubMedPubMedCentral Zhang B et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell. 2013;153(3):707–20.CrossRefPubMedPubMedCentral
87.
Zurück zum Zitat Ziebell JM et al. Rod microglia: elongation, alignment, and coupling to form trains across the somatosensory cortex after experimental diffuse brain injury. J Neuroinflammation. 2012;9(1):247.CrossRefPubMedPubMedCentral Ziebell JM et al. Rod microglia: elongation, alignment, and coupling to form trains across the somatosensory cortex after experimental diffuse brain injury. J Neuroinflammation. 2012;9(1):247.CrossRefPubMedPubMedCentral
88.
Zurück zum Zitat Zilka N et al. Human misfolded truncated tau protein promotes activation of microglia and leukocyte infiltration in the transgenic rat model of tauopathy. J Neuroimmunol. 2009;209(1):16–25.CrossRefPubMed Zilka N et al. Human misfolded truncated tau protein promotes activation of microglia and leukocyte infiltration in the transgenic rat model of tauopathy. J Neuroimmunol. 2009;209(1):16–25.CrossRefPubMed
89.
Zurück zum Zitat Zujovic V et al. Fractalkine modulates TNF‐α secretion and neurotoxicity induced by microglial activation. Glia. 2000;29(4):305–15.CrossRefPubMed Zujovic V et al. Fractalkine modulates TNF‐α secretion and neurotoxicity induced by microglial activation. Glia. 2000;29(4):305–15.CrossRefPubMed
Metadaten
Titel
Increased tauopathy drives microglia-mediated clearance of beta-amyloid
verfasst von
Wesley Chen
Edsel A. Abud
Stephen T. Yeung
Anita Lakatos
Trevor Nassi
Jane Wang
David Blum
Luc Buée
Wayne W. Poon
Mathew Blurton-Jones
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Acta Neuropathologica Communications / Ausgabe 1/2016
Elektronische ISSN: 2051-5960
DOI
https://doi.org/10.1186/s40478-016-0336-1

Weitere Artikel der Ausgabe 1/2016

Acta Neuropathologica Communications 1/2016 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.