Skip to main content
Erschienen in: Acta Neuropathologica Communications 1/2014

Open Access 01.12.2014 | Research article

Frequent and symmetric deposition of misfolded tau oligomers within presynaptic and postsynaptic terminals in Alzheimer’s disease

verfasst von: Hwan-Ching Tai, Bo Y Wang, Alberto Serrano-Pozo, Matthew P Frosch, Tara L Spires-Jones, Bradley T Hyman

Erschienen in: Acta Neuropathologica Communications | Ausgabe 1/2014

Abstract

The accumulation of neurofibrillary tangles in Alzheimer’s disease (AD) propagates with characteristic spatiotemporal patterns which follow brain network connections, implying trans-synaptic transmission of tauopathy. Since misfolded tau has been shown to transmit across synapses in AD animal models, we hypothesized that synapses in AD patients may contain misfolded tau. By immunofluorescence imaging of bipartite synapses from AD subjects, we detected tau protein in 38.4% of presynaptic and 50.9% of postsynaptic terminals. The pre/post distribution for hyperphosphorylated tau was 26.9%/30.7%, and for misfolded tau 18.3%/19.3%. In the temporal cortex, microscopic aggregates of tau, containing ultra-stable oligomers, were estimated to accumulate within trillions of synapses, outnumbering macroscopic tau aggregates such as tangles by 10000 fold. Non-demented elderly also showed considerable synaptic tau hyperphosphorylation and some misfolding, implicating the synapse as one of the first subcellular compartments affected by tauopathy. Misfolding of tau protein appeared to occur in situ inside synaptic terminals, without mislocalizing or mistrafficking. Misfolded tau at synapses may represent early signs of neuronal degeneration, mediators of synaptotoxicity, and anatomical substrates for transmitting tauopathy, but its actual role in these processes remain to be elucidated.

Introduction

The pathological hallmarks of Alzheimer’s disease (AD) are senile plaques and neurofibrillary tangles (NFT) [1],[2]. The accumulation of NFT, composed of misfolded, hyperphosphorylated tau proteins [3], follows a hierarchical spatiotemporal pattern which is consistent with anatomical connections in the brain [4],[5]. It therefore appears that NFT deposition spreads from one brain region to the next along major axonal projections, but the underlying mechanism remains unclear. Given the ability of misfolded tau to induce the misfolding of normal tau molecules via a seeding mechanism [6]-[10], it has been hypothesized that trans-synaptic transmission of misfolded tau molecules may underlie the spread of tauopathy [11]-[13], perhaps analogous to the spread of prion proteins within the brain [14]. Some have proposed that many neurodegenerative disorders may share the general feature of "prion-like" propagation of misfolded proteins [15],[16].
Studies in animal models have demonstrated that tauopathy can spread in the living brain, using either transgenic mice that express mutant human tau proteins specifically in the entorhinal cortex [11],[17], or by injecting tau aggregates into specific brain regions [18],[19]. However, these processes are relatively inefficient, taking weeks to months to observe, and occur only in situations of high levels of exogenous or transgenic tau. Tau has historically been identified as a microtubule-associated protein localized to the axon of mature neurons [20],[21], and a prerequisite for trans-synaptic propagation would be the localization of tau specifically at the synapse. We therefore hypothesized that, for the propagation model to be credible in human diseases, tau would need to be found at the synapse (at least in the disease state); if present at the synapse, the identification of tau species differentially present in pre- or post-synaptic elements, and in AD compared to controls, will test the further hypotheses that misfolded tau accumulates presynaptically before "release" into postsynaptic space, and that tau is mislocalized to the synapse in AD compared to normal neurons.
To test these hypotheses, we isolated and visualized intact, bipartite human neuronal synapses from cortical tissues of control and AD subjects. Using immunofluorescence to detect different forms of tau at bipartite synapses, we found normal tau protein to be symmetrically distributed across presynaptic and postsynaptic terminals in the normal human brain. Misfolded tau in AD-affected brains was also symmetrically distributed on both sides of the synapse, forming sodium dodecyl sulfate (SDS)-resistant oligomers. These data suggest that synaptic tau becomes hyperphosphorylated and misfolded in situ without significant spatial redistribution. Microscopic aggregates of misfolded tau deposited within synapses may represent early signs of neuronal degeneration, agents of synaptic toxicity, and anatomical substrates responsible for the transmission of tauopathy.

Materials and methods

Reagents

Protease inhibitor (cOmplete tablet) was purchased from Roche (Indianapolis, IN). Phosphatase inhibitor cocktails 2 and 3 were purchased from Sigma (St. Louis, MO) and used in 1:1 combination. High-quality Triton X-100 (glass ampule packaging) was purchased from Pierce (Rockford, IL). Mouse monoclonal antibodies DA9 (total tau), PHF1 (pS396/pS404 tau), and Alz-50 (misfolded tau) were gifts of Peter Davies (Albert Einstein College of Medicine). In accordance with original studies of Alz-50 antibody [22], we found Alz-50 to be weakly reactive against denatured tau in Western blotting after SDS-PAGE. So Alz-50 is a misfolded-conformation-specific tau antibody only under non-denaturing conditions, suitable for immunostaining of fixed cells/tissues.
Rabbit anti-tau (A20024) was purchased from Dako (Glostrup, Denmark); Rabbit anti-PSD95 (#2507) from Cell Signaling (Danvers, MA); Mouse anti-actin (A4700), rabbit anti-actin (A5060), and mouse anti-MAP2 (M4403) from Sigma; Mouse anti-synaptophysin (AB8049) from Abcam (Cambridge, MA); Rabbit anti-histone H3 (05-928) from Millipore (Billerica, MA); Mouse anti-GFAP (MS-1376) from Thermo (Waltham, MA).

Human subjects

Brains from human subjects with a diagnosis of Alzheimer disease or no cognitive deficits were obtained through the Massachusetts Alzheimer’s Disease Research Center and Massachusetts General Hospital Neuropathology Department. All donor tissue was obtained in accord with local and national IRB regulations. Characteristics of control and AD subjects are listed in Table 1.
Table 1
Characteristics of control and AD subjects examined in this study
Case #
Age (y)
Gender
Diagnosis
Disease duration (y)
ApoE genotype
PMI (h)
Braak stage
C1
76
F
Control
NA
3/3
24
1
C2
80
F
Control
NA
2/4
54
1
C3
76
M
Control
NA
3/4
48
1
C4
89
F
Control
NA
2/3
13
2
C5
91
F
Control
NA
3/3
19
2
C6
71
M
Control
NA
NA
5
0
C7
87
M
Control
NA
NA
36
1
A1
85
F
AD
4
3/4
10
5
A2
73
F
AD
19
3/3
14
5
A3
84
F
AD
16
3/4
12
5
A4
92
M
AD
22
4/4
12
5
A5
83
F
AD
13
3/4
12
5
A6
82
M
AD
6
3/4
7
6
A7
91
F
AD
14
3/4
9
5
A8
95
M
AD
NA
3/3
11
6
NA = not applicable or not available; PMI = postmortem interval.

Subcellular fractionation and protein extraction

Frozen human cortical tissue was dissected to separate grey matter from white matter, and 200-300 mg of thoroughly thawed grey matter was gently ground in a Potter-Elvehjem homogenizer with 1.5 mL ice-cold buffer A (25 mM HEPES pH 7.5, 120 mM NaCl, 5 mM KCl, 1 mM MgCl2, 2 mM CaCl2), supplemented with 2 mM DTT, protease inhibitors and phosphatase inhibitors. The homogenate was passed through two layers of 80 μm nylon filters (Millipore) to remove tissue debris, and a 200 μL aliquot was saved. The saved aliquot was mixed with 200 μL of water and 70 μL of 10% SDS, passed through a 27 gauge needle several times to shear DNA, and boiled for 5 min to prepare the total extract, followed by centrifugation at 15,000×g for 10 min to remove insoluble matter.
To prepare filtered synaptoneurosomes, the rest of the homogenate was passed through a 5 μm Supor membrane filter (PALL, Port Washington, NY) to remove large organelles and nuclei, and centrifuged at 1,000×g for 5 min to sediment synaptic terminals. Each pellet was resuspended in buffer A, split into two aliquots, and centrifuged again to yield two synaptoneurosome pellets. Supernatant from the first centrifugation step was clarified by centrifugation at 100,000×g for 1 h to obtain the cytosol fraction. Cytosolic extract was prepared by adding 1.5% SDS and boiling for 5 min. To prepare synaptoneurosome extracts, each synaptoneurosome pellet was mixed with 250 μL buffer B (50 mM Tris pH 7.5, 1.5% SDS, 2 mM DTT) and boiled for 5 min, followed by centrifugation at 15,000×g for 10 min to remove insoluble matter. Synaptoneurosome pellets may be snap frozen in liquid nitrogen and stored at -80°C.

Immunostaining of synaptoneurosomes

Four-well Lab-Tek II CC2 (polyamine pre-coated) chamber slides (Nunc, Rochester, NY) were used for fixing and imaging synaptoneurosomes. Each synaptoneurosome pellet was resuspended in 5 mL of ice-cold buffer C (10 mM HEPES pH 7.9, 0.3 M sucrose) by gentle pipetting. Using a syringe with 27 gauge needle, 200 μL of synaptoneurosome suspension (containing about 25 μg of total protein) was transferred to each chamber well, followed by the addition of 200 μL 2% paraformaldehyde in ice-cold PBS-MC (phosphate buffered saline with 1 mM MgCl2 and 1 mM CaCl2). After 10 min of incubation with 1% paraformaldehyde at 4°C, synaptoneurosomes became fixed and crosslinked to the glass surface. Fixed synaptoneurosomes were washed with PBS-MC (room temperature from this point on) for three times (5 min each), and permeabilized for 10 min with 0.05% Triton X-100 in PBS-MC with 3% bovine serum albumin (BSA, Sigma), followed by three more washes. Slides were blocked with 4% normal goat serum (Invitrogen) and 3% BSA in PBS-MC for 30 min, and then incubated with primary antibodies diluted in PBS-MC with 3% BSA for 90 min, followed by three washes. Secondary antibodies diluted in PBS-MC with 3% BSA were added for 50 min, followed by three washes. The slide was mounted with #1.5 glass coverslip and Prolong Gold Antifade reagent (Invitrogen, Carlsbad, CA).
Primary antibodies for immunostaining included guinea pig anti-VGluT1 (Millipore AB5905, 1:150), chicken anti-MAP2 (Abcam AB5392, 1:100), DA9 (mouse IgG, 1:150), PHF1 (mouse IgG, 1:80), and Alz-50 (mouse IgM, 1:30). Fluorescent secondary donkey antibodies were purchased from Jackson Immunoresearch (West Grove, PA) and used at 1:100 dilutions (anti-guinea pig DyLight 649, anti-chicken Cy3, anti-mouse IgG Alexa 488, and anti-mouse IgM Alexa 488).

Image acquisition and analysis

Immunofluorescence and brightfield images of synaptoneurosomes were acquired on an AxioImager Z1 epifluorescence microscope (Carl Zeiss, Oberkochen, Germany) equipped with a 63x oil immersion objective (N.A. = 1.40). Images were deconvolved with the Iterative Deconvolution plugin (by Bob Dougherty, OptiNav Inc.) in ImageJ software (version 1.44). This 2D deconvolution program required a point spread function (PSF) generated by Diffraction Limit PSF plugin (Bob Dougherty, OptiNav Inc.). For the brightfield image we used a 400 nm PSF; for green fluorescence channel a 509 nm PSF; for red channel a 550 nm PSF; for far red channel a 650 nm PSF. The optimal iteration number was empirically determined to be 12 for brightfield images and 16 for fluorescence images, with the LP filter diameter set at 1.5 pixels. After deconvolution, brightfield and fluorescence images were overlaid in ImageJ (using hyperstacks) and protein colocalization was determined by manual inspection. The cutoff threshold for fluorescence signals (after deconvolution) was generally set as two standard deviations above the mean. For stereological counting, we created a 12×8 grid image which was overlaid with microscope images, and devised a rule to randomly select different grid areas to look for synaptic terminals until the target number was reached. Two-way ANOVA was computed using Graphpad Prism software and t-tests were computed using Excel software.

Classification of synapse morphology

During stereological counting, we defined a synaptic terminal as a brightfield object in the size range of 300-1000 nm with immunofluorescence signals for either presynaptic (VGlut1) or postsynaptic (MAP2) marker. Brightfield objects negative for both synaptic markers were regarded as non-synaptic organelles/vesicles, and these were not quantified, even if they were positively stained by tau antibodies. To qualify as a bipartite synapse, the brightfield object should show a snowman-like structure or a non-spherical shape being elongated or protruded, and pre/post makers needed to be non-overlapping (judged by the centers of mass of the puncta) and overlaid with the brightfield object in a reasonable manner to be considered as adjacent presynaptic and postsynaptic terminals. When synapses were clustered together with other objects under the brightfield, we did not attempt to classify them as bipartite or hemi synapses. There were also tau-positive puncta which did not overlap with brightfield objects and they were excluded from our analysis.

Transmission electron microscopy

Synaptoneurosome pellets were fixed in 2% glutaraldehyde and 2% paraformaldehyde in PBS overnight at 4°C, rinsed, post-fixed in 1% osmium tetroxide, and embedded in LR White resin (Electron Microscopy Sciences, Hatfield, PA) according to manufacturer's protocols. Embedded blocks were cut into 70-nm thin sections on an Ultracut Microtome (Leica, Nussloch, Germany). Images were acquired on a JEOL1011 transmission electron microscope equipped with an ATM digital camera (JEOL USA, Peabody, MA).

Gel electrophoresis and immunoblotting

SDS-denatured protein extracts were subjected to BCA assay (Pierce) to determine protein concentrations. Extracts were boiled again for 3 min after adding 5x sample buffer (250 mM Tris pH 7.5, 5% SDS, 400 mM DTT, 50% glycerol, 0.2% Orange G). Samples were resolved by SDS-PAGE using Bis-Tris 4-12% precast gels (Invitrogen), and transferred to low-fluorescence PVDF or nitrocellulose membranes (Millipore) for immunoblotting, detected using an Odyssey laser scanner (Li-Cor, Lincoln, NE). Blocking buffer and fluorescent secondary antibodies were purchased from Li-Cor and used according to manufacturer's protocols.

Results

Isolation and visualization of synaptic terminals from postmortem tissues

We recently developed a method for isolating human synaptic terminals from frozen (unfixed) postmortem brain tissues, based on homogenization, 80 and 5 μm filtration, and low-speed centrifugation (Figure 1A) [23]. The resulting subcellular fraction, called filtered synaptoneurosomes [24], was enriched in synaptic marker proteins (synaptophysin and PSD95) by immunoblotting (Figure 1B), and enriched in synaptic structures under transmission electron microscopy (Figure 1C, Additional file 1: Figure S1 and S2). Using a gentle formaldehyde fixation protocol to crosslink synaptoneurosomes onto polyamine-coated glass slides, we could visualize individual synaptic or organelle structures by brightfield microscopy (Figure 1D). Three channel immunofluorescence was utilized to detect the presence of tau (or its modified forms), the presynaptic marker (vesicular glutamate transporter 1, VGluT1), and the postsynaptic marker (microtubule-associated protein 2, MAP2).
A significant proportion of brightfield objects were negative for synaptic markers (like object 2 in Figure 1D), and by electron microscopy we observed non-synaptic organelles such as mitochondria and myelin in the synaptoneurosome preparation (Additional file 1: Figure S1). Synaptophysin/PSD95 are commonly used pre/post-synaptic markers, but we have previously observed that their respective puncta often showed significant spatial overlap [23], making it difficult to colocalize with a third protein. So instead we chose the VGlut1/MAP2 marker pair, which showed better puncta separation (see Figure 1D for instance). Presynaptic marker VGlut1 is expected to label the majority of glutamatergic presynaptic terminals in the cortex [25], and we have previously shown its usefulness for immunostaining human presynaptic terminals in array tomography experiments [26]. Although MAP2 is often used as a subcellular marker for dendrites, its distribution in dendritic spines has been clearly demonstrated by ultrastructural studies [27],[28]. In the absence of interfering signals from dendrites, we found it to be a useful marker for immunostaining postsynaptic terminals [23].
The enrichment of MAP2 in both mouse and human synaptoneurosomes was further demonstrated by immunoblotting with a different MAP2 antibody (mouse monoclonal, Additional file 1: Figure S3), apart from the chicken anti-MAP2 used for immunofluorescence. Low molecular weight MAP2 (<100 kDa) appeared to be more highly enriched in synaptoneurosomes than common synaptic markers synatophysin and PSD95 (Figure 1B), confirming the synaptic localization of MAP2. We have also tried phalloidin reagents to stain postsynaptic terminals but they worked poorly (data not shown) compared to anti-MAP2.

Biochemical and immunocytochemical detection of synaptic tau misfolding

Since misfolded tau proteins are generally believed to be seeds for transmitting proteinopathy [12],[13], we tried to detect their presence in synaptoneurosomes by two independent methods. The first method was SDS-polyacrylamide gel electrophoresis (PAGE) analysis, followed by immunoblotting (Figure 2A). In control cases, normal tau protein migrated as a monomer around 60 kDa, being more enriched in the cytosol (which included the axonal cytoplasm) than the synaptoneurosome, but with minimal phosphorylation. AD cytosolic extracts similarly contained mostly monomeric, non-phosphorylated tau. However, in AD synaptoneurosomes, tau became hyperphosphorylated and reactive against PHF1 antibody (pS396/pS404) [29], and misfolded into SDS-resistant oligomers that migrated as high molecular weight smears in the gel.
The second method utilized a conformation-specific antibody, Alz-50 [22], which recognized NFTs and dystrophic neurites in AD-affected brains [30],[31]. Immunostaining against Alz-50 was strongly elevated in synaptoneurosomes isolated from AD subjects compared to non-demented controls (Figure 2B). By stereological counting, we detected about 15% increase in Alz-50-positive terminals in AD subjects over controls (Figure 2C). Altogether, our data showed that tau underwent misfolding, hyperphosphorylation, and oligomerization at AD-affected synapses.
Stereological counting in Figure 2C was conducted over all synaptic terminals observable within chosen areas, but it could be potentially confounded by the uneven morphological distribution of synapses. So we further classified observed synapses into three common morphology types, as shown in Figure 3. The first morphology class was the intact, bipartite synapse, represented by snowman-like or elongated/protruded structures under brightfield, with immunostaining for VGluT1 and MAP2 on opposite sides. The second class was the isolated hemi-synapse, represented by a single object under brightfield, with immunostaining against either VGluT1 or MAP2. These represented detached presynaptic or postsynaptic (mushroom dendritic spine) terminals. The third class had multiple synaptic or non-synaptic structures clustered together (see Materials and methods section for detailed classification criteria).
To better understand the spatial distribution of tau in synaptic structures, we analyzed bipartite and hemi synapses separately (Figures 4B and 5B), in addition to summing over mixed morphologies (Figure 2C). In particular, bipartite synapses allowed us to unambiguously assign tau, or its phosphorylated or misfolded forms, into three distribution patterns: presynaptic-only, postsynaptic-only, or both sides.

Tau localizes to synapses to a similar extent in normal aging and AD

In non-demented elderly, normal tau proteins were detected by DA9 antibody at 76.0 ± 2.9% (mean ± S.E.M.) of bipartite synapses, with a distribution ratio of 33.7%/37.5%/4.8% (pre-only/post-only/both) (Figure 4). It was surprising to find a slightly higher (not significant) fraction of postsynaptic terminals containing tau compared to presynaptic terminals, despite tau being concentrated in the axonal cytoplasm of mature neurons [20],[21].
We also examined isolated hemi-synapses from control subjects, finding tau inside 67.2 ± 2.3% of presynaptic terminals and 71.9 ± 2.1% of post synaptic terminals (Figure 5). By multiplying these two numbers, we would expect 44.0% of bipartite synapses to be tau-positive on both sides, if they arose from random associations of hemi-synapses. But the actual percentage of double-positive bipartite synapses was much lower, only 4.8 ± 1.8%, and the difference was highly significant (p < 10-5, two-tailed t-test). Altogether, our results clearly established that tau was abundantly distributed inside dendritic spines, in addition to being axonal and presynaptic.
DA9 antibody could recognize both phosphorylated and non-phosphorylated tau forms [32], and in AD samples 85.6 ± 5.1% of bipartite synapses were tau (DA9)-positive, with a distribution ratio of 34.6%:47.1%:3.8% (pre-only/post-only/both) (Figure 4B). It revealed that tau proteins did not mistraffic or mislocalize to synapses in AD, since they were abundantly present in synapses before AD.

Abnormal tau forms increase in AD-affected synapses

In bipartite synapses of AD subjects (Figure 4B), immunostaining with Alz-50 antibody detected misfolded tau at both presynaptic and postsynaptic terminals, and the distribution ratio was 15.4%:16.4%:2.9% (presynaptic-only/postsynaptic-only/both), showing bilateral symmetry across the synaptic cleft. As such, tau misfolding occurred at 34.6 ± 5.7% (mean ± S.E.M.) of bipartite synapses from the temporal lobe, a region marked by high NFT burdens in AD [33]. The fraction containing hyperphosphorylated tau was even higher at 53.8 ± 4.7%, with a distribution ratio of 23.1%:26.9%:3.8% (pre-only/post-only/both). Tau hyperphosphorylation and misfolding were significantly higher in AD compared to non-demented controls (by two-way ANOVA, Table 2).
Table 2
Summary of two-way ANOVA analyses of tau detection frequencies at bipartite synapses and hemi-synapses
Tau detection type
Total tau
Phosphorylated tau
Misfolded tau
Synapse type
Bipartite
Hemi
Bipartite
Hemi
Bipartite
Hemi
P value
Con vs. AD
n.s.
n.s.
0.0002
< 0.0001
0.0014
0.0004
(Con < AD)
(Con < AD)
(Con < AD)
(Con < AD)
pre vs. post
0.0236
n.s.
n.s.
n.s.
n.s.
0.0475
(pre < post)
(pre < post)
n.s. = not significant.
By observing more frequent tau hyperphosphorylation than misfolding in bipartite synapses (p < 0.05, two-tailed t-test, Figure 4B), our data supported the observation that tau phosphorylation preceded fibril formation in the brain and in cellular models [1],[34],[35]. The probability of tau becoming hyperphosphorylated or misfolded was roughly equivalent at presynaptic and postsynaptic sites. This was also confirmed by counting hyperphosphorylated tau (PHF1) and misfolded tau (Alz-50) in isolated hemi-synapses from AD cases. The presynaptic:postsynaptic ratios were 44.6%:46.6% (phospho) and 22.6%:32.8% (misfolded), respectively (Figure 5B).

Comparing control and AD synaptoneurosomes

The percentage of synapses (bipartite and hemi) positive for total tau was similar between control and AD subjects, but abnormal forms of tau was elevated in the latter (Figures 4B and 5B). However, appreciable amounts of abnormal tau were detectable in control synaptoneurosomes, with hyperphosphorylation found in 20.9%:26.8% (pre/post) and misfolding in 10.5%:13.7% (pre/post) of hemi-synapses (Figure 5B). It appeared that tau abnormality occurred at neocortical synapses in aging brains before apparent AD pathology (Braak stages 1 or 2). AD would lead to even more synaptic tau molecules being hyperphosphorylated and misfolded (Table 2), but the initial trigger might have occurred in the preclinical stage of AD.
All forms of tau examined in this study, normal or abnormal, generally showed similar frequencies on both sides of bipartite synapses. This was true in both control and AD subjects. Nor did we notice obvious intensity differences between presynaptic and postsynaptic signals after inspecting thousands of synaptic terminals, although we did not formally quantify fluorescence signals. The pre/post symmetry of tau distribution appeared to be an invariant feature of normal and AD-affected brains.

Discussion

Misfolded tau oligomers deposited at synapses have the potential to spread tauopathy

In this study, we detected misfolded tau in a significant fraction of presynaptic and postsynaptic terminals in AD subjects, which had two significant functional implications: (1) it could impair brain network function; (2) it could represent the anatomical substrate of tau synaptic transmission.
The "transmission theory" of AD neurofibrillary pathology [12],[13] predicts that there should be misfolded, seeding-capable tau molecules localized inside synaptic terminals. The true identity of tau species that seed intracellular/trans-cellular tau aggregation remains unknown in the human brain, but studies in cellular and animal models have suggested that they may be oligomers or protofibrils [36]-[39]. Recently, Lasagna-Reeves et al. [18] have immunopurified tau oligomers from AD brains that could propagate tauopathy in living mice, and such oligomers shared several similarities with synaptic tau oligomers found in this study: (1) they migrated as smears between 75-300 kDa in SDS-PAGE; (2) they were microscopic, amorphous aggregates instead of fibrillar.
Lasagna-Reeves et al. observed tau oligomers isolated from AD brains forming amorphous globular aggregates 4-8 nm in diameter [18], while Maeda et al. also reported granular tau oligomers from AD subjects in the range of 5-50 nm [40]. Although we could not resolve the size or number of microscopic aggregates within each synaptic terminal by optical microscopy, under electron microscopy we did not observe any fibril within synaptic structures (Additional file 1: Figure S2). It was likely that synaptic tau inclusions were also amorphous, granular oligomers smaller than 50 nm, which appeared to be the typical state of pre-fibrillar tau aggregates [41]. SDS-PAGE analysis showed that the core components of these aggregates were ultra-stable oligomers. We also observed that synaptic tau oligomers shared certain conformational features (recognized by Alz-50 antibody) with larger fibrillar inclusions like NFTs and neuropil threads [22],[30],[31]. Hence, it is conceivable that the former may be able to seed the formation of the latter.

Tau is present at synapses in sufficient quantities to plausibly impact neural system function

The number of synapses and neurons in the human temporal cortex has been estimated to be around 40 trillion and 6 billion, respectively [42]. In temporal cortices of end-stage AD subjects, we estimate that several trillion synaptic terminals contain microscopic aggregates of misfolded tau. On the other hand, the number of NFTs was estimated to be a few hundred million in this brain region [33]. Given the extraordinary number of synapses carrying misfolded tau, which may easily reach many billions in a tauopathy-affected area, the spread of misfolded tau into interconnected brain regions may be quite appreciable even if the probability of transmission is exceedingly low, such as one millionth per day at each synapse.
Given the importance of synaptic plasticity in neural transmission and memory encoding, the frequent deposition of misfolded tau at synapses may contribute significantly to cognitive decline in AD. This was supported by our previous observation that tau oligomer levels, but not Aβ oligomer levels, correlated with the disruption of the ubiquitin-proteasome system at synapses [23]. Moreover, in a special group of non-demented elderly with AD-like neuropathology marked by high plaque and NFT burdens, we observed that they did not develop synaptic tau oligomers like AD subjects. In these cases, synaptic tau oligomers appeared to be better correlated with dementia than synaptic Aβ oligomers, NFTs, or plaques [43].

Synaptic compartments may exhibit early signs of tauopathy

Although tau is highly enriched in axons as a microtubule binding protein, its presence in other cellular compartments such as somas, dendrites, and nuclei has been clearly documented in healthy rodent and monkey brains [44]-[46]. Tau also appears to interact with actin [47],[48] and various membranous structures within the neuron [49],[50], which may potentially explain its partitioning into postsynaptic terminals instead of just presynaptic terminals. With the progression of AD, misfolded tau accumulates in the somatodendritic compartment [30],[51], but we found that the percentage of pre- and post-synaptic terminals with total tau did not increase significantly with AD—only phosphorylation and misfolding did. We found all forms of tau to be symmetrically distributed across the synapse, with or without AD. These data argue against the spatial redistribution of synaptic tau molecules during AD pathogenesis, but instead, tau hyperphosphorylation and misfolding probably occurred in situ within each synaptic terminal.
Inside individual terminals, hyperphosphorylation probably preceded misfolding, because more synapses showed hyperphosphorylation than misfolding in AD brains. In non-demented elderly, tau hyperphosphorylation was observation in ~20% of hemi-synapses (Figure 5B), which may be an early sign of tau abnormality. We do not know if this process began at a much younger age, or if it was the result of abnormal tau being synaptically transmitted from tangle-bearing regions. Most elderly over 65 would be characterized as Braak Stage 1 or 2, with tangles already present in the transentorhinal region [4],[30].
Hyperphosphorylation is known to precede macroscopic tau aggregation during AD pathogenesis [1],[30], but the subcellular origin of tau abnormality remains unclear. Our data suggest that one of the early signs of tau abnormality may be hyperphosphorylation inside synaptic terminals, even in non-demented elderly. Hyperphosphorylated tau loses its ability to bind microtubules [34],[52],[53], thereby gaining the tendency to misfold.

Possible directionality in synaptic tau transmission

Recent studies using tauopathy mouse models have shown that normal and misfolded tau both transmit across brain regions via synaptic connectivity instead of spatial proximity [11],[17],[19],[54]. Moreover, both anterograde and retrograde spreading of tauopathy have been observed along axonal tracts, suggesting that tauopathy also transmits bidirectionally at synapses [19]. Our data appear to be consistent with the possibility of bidirectional transmission, since misfolded tau is equally likely to be presynaptic or postsynaptic within a bipartite synapse, but infrequently on both sides.
Recent studies have identified tau in secreted vesicles of neuronal and non-neuronal cells [49],[55], and in the vesicle fraction of cerebrospinal fluids of early AD patients [56]. Others have shown that membrane-free tau aggregates added to culture media could also enter neurons to induce further aggregation [36],[37],[39],[57]. Hence, there are two possible routes for misfolded tau to cross into another neuron via synaptic contacts, which are illustrated in Figure 6.
The first pathway (Figure 6A) involves vesicular transport between presynaptic and postsynaptic terminals [58]. The symmetric distribution of misfolded tau we observed suggests that either direction of transmission seems plausible. The second model (Figure 6B) is prompted by the astronomical number of synaptic tau aggregates observed. As a tauopathy-stricken neuron dies, what happens to the hundreds of microscopic tau inclusions inside its synapses, especially in distal neurites? Micro-aggregates containing SDS-resistant tau oligomers are unlikely to be completely removed by the proteolytic systems of a failing neuron. Some aggregates may remain in its original place after the neurite disintegrates, much like a ghost tangle that stands in the place of a disintegrated neuron. It is conceivable that oligomeric tau debris may be taken up by neighboring neurites/synapses, causing tauopathy to spread in both retrograde and anterograde directions.
Had we observed misfolded tau depositing at both sides of the bipartite synapse with higher probability than chance, it might have provided indirect evidence for the vesicular transmission model in Figure 6A. This was one of the original goals of our experimental design. However, in our synaptoneurosome preparation, only a few percent of bipartite synapses contained tau protein on both sides, much lower than the expected value (Figure 4B), and the underlying reason remains unclear. Since well-characterized bipartite synapses were also rare in our preparation, the sample number was too small for estimating transmission probability. Further studies will be required to provide supporting evidence for the models proposed above.

Conclusion

In this study we developed a new procedure to image intact synapses isolated from postmortem brain tissues and conducted the first quantitative assessment of synaptic tauopathy in AD. We demonstrated that tau misfolding occurs symmetrically across the synapse with high frequencies of oligomer deposition, revealing for the first time the extraordinary number of synaptic terminals containing microscopic tau inclusions. This methodology would be equally applicable to other neurodegenerative tauopathies such as progressive supranuclear palsy, certain forms of frontotemporal dementia, or corticobasal degeneration [59]. The underlying reason that AD, but not other tauopathies [51],[60], shows a hierarchical pattern of NFT deposition originating in transentorhinal regions still remains unanswered. Comparing the synaptic distribution of tau and analyzing its biochemical state in different tauopathies may provide insights into their respective pathogenesis mechanisms. The discovery of misfolded tau oligomers inside synaptic terminals highlights novel targets for pathogenesis studies and therapeutic interventions. For instance, tau immunotherapy may be able to sequester extracellular misfolded tau after it escapes from pre- or post-synaptic compartments but before it enters another neuron. It remains to be seen if synaptic deposition of misfolded proteins is a common mechanism of neurodegenerative proteinopathies, and imaging of isolated human synapses will be very useful for such investigations.

Authors’ contributions

HCT carried out staining and biochemical experiments and drafted the manuscript. BYW conducted image and statistical analysis. AS and MPF contributed to case selection and tissue preparation. TLS carried out electron microscopy. BTH conceived the study, designed the experiments, and revised the manuscript. All authors discussed and approved the manuscript.

Additional file

Acknowledgment

We thank Marian DiFiglia and the Philly Dake Electron Microscopy Center for access to the TEM. We thank Karlotta Fitch for assistance with brain tissues. We thank Peter Davies for anti-tau antibodies PHF1, CP13, and DA9. This work is partially supported by grant NSC101-2113-M-002-019-MY2 from Ministry of Science and Technology, Taiwan.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. 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 interests.
Literatur
1.
Zurück zum Zitat Braak E, Griffing K, Arai K, Bohl J, Bratzke H, Braak H: Neuropathology of Alzheimer’s disease: what is new since A. Alzheimer? Eur Arch Psychiatry Clin Neurosci 1999, 249(Suppl 3):14–22. 10.1007/PL00014168CrossRefPubMed Braak E, Griffing K, Arai K, Bohl J, Bratzke H, Braak H: Neuropathology of Alzheimer’s disease: what is new since A. Alzheimer? Eur Arch Psychiatry Clin Neurosci 1999, 249(Suppl 3):14–22. 10.1007/PL00014168CrossRefPubMed
2.
Zurück zum Zitat Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT (2011) Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med 1:a006189 Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT (2011) Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med 1:a006189
3.
Zurück zum Zitat Wood JG, Mirra SS, Pollock NJ, Binder LI: Neurofibrillary tangles of Alzheimer disease share antigenic determinants with the axonal microtubule-associated protein tau. Proc Natl Acad Sci U S A 1986, 83: 4040–4043. 10.1073/pnas.83.11.4040CrossRefPubMedPubMedCentral Wood JG, Mirra SS, Pollock NJ, Binder LI: Neurofibrillary tangles of Alzheimer disease share antigenic determinants with the axonal microtubule-associated protein tau. Proc Natl Acad Sci U S A 1986, 83: 4040–4043. 10.1073/pnas.83.11.4040CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Braak H, Braak E: Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 1991, 82: 239–259. 10.1007/BF00308809CrossRefPubMed Braak H, Braak E: Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 1991, 82: 239–259. 10.1007/BF00308809CrossRefPubMed
5.
Zurück zum Zitat Arnold SE, Hyman BT, Flory J, Damasio AR, Van Hoesen GW: The topographical and neuroanatomical distribution of neurofibrillary tangles and neuritic plaques in the cerebral cortex of patients with Alzheimer’s disease. Cereb Cortex 1991, 1: 103–116. 10.1093/cercor/1.1.103CrossRefPubMed Arnold SE, Hyman BT, Flory J, Damasio AR, Van Hoesen GW: The topographical and neuroanatomical distribution of neurofibrillary tangles and neuritic plaques in the cerebral cortex of patients with Alzheimer’s disease. Cereb Cortex 1991, 1: 103–116. 10.1093/cercor/1.1.103CrossRefPubMed
6.
Zurück zum Zitat Friedhoff P, von Bergen M, Mandelkow EM, Davies P, Mandelkow E: A nucleated assembly mechanism of Alzheimer paired helical filaments. Proc Natl Acad Sci U S A 1998, 95: 15712–15717. 10.1073/pnas.95.26.15712CrossRefPubMedPubMedCentral Friedhoff P, von Bergen M, Mandelkow EM, Davies P, Mandelkow E: A nucleated assembly mechanism of Alzheimer paired helical filaments. Proc Natl Acad Sci U S A 1998, 95: 15712–15717. 10.1073/pnas.95.26.15712CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Montejo de Garcini E, Serrano L, Avila J: Self assembly of microtubule associated protein tau into filaments resembling those found in Alzheimer disease. Biochem Biophys Res Commun 1986, 141: 790–796. 10.1016/S0006-291X(86)80242-XCrossRefPubMed Montejo de Garcini E, Serrano L, Avila J: Self assembly of microtubule associated protein tau into filaments resembling those found in Alzheimer disease. Biochem Biophys Res Commun 1986, 141: 790–796. 10.1016/S0006-291X(86)80242-XCrossRefPubMed
8.
Zurück zum Zitat Morozova OA, March ZM, Robinson AS, Colby DW: Conformational features of tau fibrils from Alzheimer’s disease brain are faithfully propagated by unmodified recombinant protein. Biochemistry 2013, 52: 6960–6967. 10.1021/bi400866wCrossRefPubMedPubMedCentral Morozova OA, March ZM, Robinson AS, Colby DW: Conformational features of tau fibrils from Alzheimer’s disease brain are faithfully propagated by unmodified recombinant protein. Biochemistry 2013, 52: 6960–6967. 10.1021/bi400866wCrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Patterson KR, Remmers C, Fu Y, Brooker S, Kanaan NM, Vana L, Ward S, Reyes JF, Philibert K, Glucksman MJ, Binder LI: Characterization of prefibrillar Tau oligomers in vitro and in Alzheimer disease. J Biol Chem 2011, 286: 23063–23076. 10.1074/jbc.M111.237974CrossRefPubMedPubMedCentral Patterson KR, Remmers C, Fu Y, Brooker S, Kanaan NM, Vana L, Ward S, Reyes JF, Philibert K, Glucksman MJ, Binder LI: Characterization of prefibrillar Tau oligomers in vitro and in Alzheimer disease. J Biol Chem 2011, 286: 23063–23076. 10.1074/jbc.M111.237974CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Rankin CA, Sun Q, Gamblin TC (2007) Tau phosphorylation by GSK-3beta promotes tangle-like filament morphology. Mol Neurodegener 2:12 Rankin CA, Sun Q, Gamblin TC (2007) Tau phosphorylation by GSK-3beta promotes tangle-like filament morphology. Mol Neurodegener 2:12
11.
Zurück zum Zitat de Calignon A, Polydoro M, Suarez-Calvet M, William C, Adamowicz DH, Kopeikina KJ, Pitstick R, Sahara N, Ashe KH, Carlson GA, Spires-Jones TL, Hyman BT: Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron 2012, 73: 685–697. 10.1016/j.neuron.2011.11.033CrossRefPubMedPubMedCentral de Calignon A, Polydoro M, Suarez-Calvet M, William C, Adamowicz DH, Kopeikina KJ, Pitstick R, Sahara N, Ashe KH, Carlson GA, Spires-Jones TL, Hyman BT: Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron 2012, 73: 685–697. 10.1016/j.neuron.2011.11.033CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Mohamed NV, Herrou T, Plouffe V, Piperno N, Leclerc N: Spreading of tau pathology in Alzheimer’s disease by cell-to-cell transmission. Eur J Neurosci 2013, 37: 1939–1948. 10.1111/ejn.12229CrossRefPubMed Mohamed NV, Herrou T, Plouffe V, Piperno N, Leclerc N: Spreading of tau pathology in Alzheimer’s disease by cell-to-cell transmission. Eur J Neurosci 2013, 37: 1939–1948. 10.1111/ejn.12229CrossRefPubMed
13.
Zurück zum Zitat Walker LC, Diamond MI, Duff KE, Hyman BT: Mechanisms of protein seeding in neurodegenerative diseases. JAMA Neurol 2013, 70: 304–310. 10.1001/jamaneurol.2013.1453CrossRefPubMed Walker LC, Diamond MI, Duff KE, Hyman BT: Mechanisms of protein seeding in neurodegenerative diseases. JAMA Neurol 2013, 70: 304–310. 10.1001/jamaneurol.2013.1453CrossRefPubMed
14.
Zurück zum Zitat Colby DW, Prusiner SB (2011) Prions. Cold Spring Harb Perspect Biol 3:a006833 Colby DW, Prusiner SB (2011) Prions. Cold Spring Harb Perspect Biol 3:a006833
15.
Zurück zum Zitat Frost B, Diamond MI: Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci 2010, 11: 155–159.PubMed Frost B, Diamond MI: Prion-like mechanisms in neurodegenerative diseases. Nat Rev Neurosci 2010, 11: 155–159.PubMed
16.
Zurück zum Zitat Jucker M, Walker LC: Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 2013, 501: 45–51. 10.1038/nature12481CrossRefPubMedPubMedCentral Jucker M, Walker LC: Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 2013, 501: 45–51. 10.1038/nature12481CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Liu L, Drouet V, Wu JW, Witter MP, Small SA, Clelland C, Duff K (2012) Trans-synaptic spread of tau pathology in vivo. PLoS One 7:e31302 Liu L, Drouet V, Wu JW, Witter MP, Small SA, Clelland C, Duff K (2012) Trans-synaptic spread of tau pathology in vivo. PLoS One 7:e31302
18.
Zurück zum Zitat Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Guerrero-Munoz MJ, Kiritoshi T, Neugebauer V, Jackson GR, Kayed R (2012) Alzheimer brain-derived tau oligomers propagate pathology from endogenous tau. Sci Rep 2:700 Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Guerrero-Munoz MJ, Kiritoshi T, Neugebauer V, Jackson GR, Kayed R (2012) Alzheimer brain-derived tau oligomers propagate pathology from endogenous tau. Sci Rep 2:700
19.
Zurück zum Zitat Ahmed Z, Cooper J, Murray TK, Garn K, McNaughton E, Clarke H, Parhizkar S, Ward MA, Cavallini A, Jackson S, Bose S, Clavaguera F, Tolnay M, Lavenir I, Goedert M, Hutton ML, O'Neill MJ: A novel in vivo model of tau propagation with rapid and progressive neurofibrillary tangle pathology: the pattern of spread is determined by connectivity, not proximity. Acta Neuropathol 2014, 127: 667–683. 10.1007/s00401-014-1254-6CrossRefPubMedPubMedCentral Ahmed Z, Cooper J, Murray TK, Garn K, McNaughton E, Clarke H, Parhizkar S, Ward MA, Cavallini A, Jackson S, Bose S, Clavaguera F, Tolnay M, Lavenir I, Goedert M, Hutton ML, O'Neill MJ: A novel in vivo model of tau propagation with rapid and progressive neurofibrillary tangle pathology: the pattern of spread is determined by connectivity, not proximity. Acta Neuropathol 2014, 127: 667–683. 10.1007/s00401-014-1254-6CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Binder LI, Frankfurter A, Rebhun LI: The distribution of tau in the mammalian central nervous system. J Cell Biol 1985, 101: 1371–1378. 10.1083/jcb.101.4.1371CrossRefPubMed Binder LI, Frankfurter A, Rebhun LI: The distribution of tau in the mammalian central nervous system. J Cell Biol 1985, 101: 1371–1378. 10.1083/jcb.101.4.1371CrossRefPubMed
21.
Zurück zum Zitat Kosik KS, Finch EA: MAP2 and tau segregate into dendritic and axonal domains after the elaboration of morphologically distinct neurites: an immunocytochemical study of cultured rat cerebrum. J Neurosci 1987, 7: 3142–3153.PubMed Kosik KS, Finch EA: MAP2 and tau segregate into dendritic and axonal domains after the elaboration of morphologically distinct neurites: an immunocytochemical study of cultured rat cerebrum. J Neurosci 1987, 7: 3142–3153.PubMed
22.
Zurück zum Zitat Wolozin B, Davies P: Alzheimer-related neuronal protein A68: specificity and distribution. Ann Neurol 1987, 22: 521–526. 10.1002/ana.410220412CrossRefPubMed Wolozin B, Davies P: Alzheimer-related neuronal protein A68: specificity and distribution. Ann Neurol 1987, 22: 521–526. 10.1002/ana.410220412CrossRefPubMed
23.
Zurück zum Zitat Tai HC, Serrano-Pozo A, Hashimoto T, Frosch MP, Spires-Jones TL, Hyman BT: The synaptic accumulation of hyperphosphorylated tau oligomers in Alzheimer disease is associated with dysfunction of the ubiquitin-proteasome system. Am J Pathol 2012, 181: 1426–1435. 10.1016/j.ajpath.2012.06.033CrossRefPubMedPubMedCentral Tai HC, Serrano-Pozo A, Hashimoto T, Frosch MP, Spires-Jones TL, Hyman BT: The synaptic accumulation of hyperphosphorylated tau oligomers in Alzheimer disease is associated with dysfunction of the ubiquitin-proteasome system. Am J Pathol 2012, 181: 1426–1435. 10.1016/j.ajpath.2012.06.033CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Hollingsworth EB, McNeal ET, Burton JL, Williams RJ, Daly JW, Creveling CR: Biochemical characterization of a filtered synaptoneurosome preparation from guinea pig cerebral cortex: cyclic adenosine 3':5'-monophosphate-generating systems, receptors, and enzymes. J Neurosci 1985, 5: 2240–2253.PubMed Hollingsworth EB, McNeal ET, Burton JL, Williams RJ, Daly JW, Creveling CR: Biochemical characterization of a filtered synaptoneurosome preparation from guinea pig cerebral cortex: cyclic adenosine 3':5'-monophosphate-generating systems, receptors, and enzymes. J Neurosci 1985, 5: 2240–2253.PubMed
25.
Zurück zum Zitat Micheva KD, Busse B, Weiler NC, O’Rourke N, Smith SJ: Single-synapse analysis of a diverse synapse population: proteomic imaging methods and markers. Neuron 2010, 68: 639–653. 10.1016/j.neuron.2010.09.024CrossRefPubMedPubMedCentral Micheva KD, Busse B, Weiler NC, O’Rourke N, Smith SJ: Single-synapse analysis of a diverse synapse population: proteomic imaging methods and markers. Neuron 2010, 68: 639–653. 10.1016/j.neuron.2010.09.024CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Kay KR, Smith C, Wright AK, Serrano-Pozo A, Pooler AM, Koffie R, Bastin ME, Bak TH, Abrahams S, Kopeikina KJ, McGuone D, Frosch MP, Gillingwater TH, Hyman BT, Spires-Jones TL: Studying synapses in human brain with array tomography and electron microscopy. Nat Protoc 2013, 8: 1366–1380. 10.1038/nprot.2013.078CrossRefPubMedPubMedCentral Kay KR, Smith C, Wright AK, Serrano-Pozo A, Pooler AM, Koffie R, Bastin ME, Bak TH, Abrahams S, Kopeikina KJ, McGuone D, Frosch MP, Gillingwater TH, Hyman BT, Spires-Jones TL: Studying synapses in human brain with array tomography and electron microscopy. Nat Protoc 2013, 8: 1366–1380. 10.1038/nprot.2013.078CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Caceres A, Binder LI, Payne MR, Bender P, Rebhun L, Steward O: Differential subcellular localization of tubulin and the microtubule-associated protein MAP2 in brain tissue as revealed by immunocytochemistry with monoclonal hybridoma antibodies. J Neurosci 1984, 4: 394–410.PubMed Caceres A, Binder LI, Payne MR, Bender P, Rebhun L, Steward O: Differential subcellular localization of tubulin and the microtubule-associated protein MAP2 in brain tissue as revealed by immunocytochemistry with monoclonal hybridoma antibodies. J Neurosci 1984, 4: 394–410.PubMed
28.
Zurück zum Zitat Morales M, Fifkova E: Distribution of MAP2 in dendritic spines and its colocalization with actin. An immunogold electron-microscope study. Cell Tissue Res 1989, 256: 447–456. 10.1007/BF00225592CrossRefPubMed Morales M, Fifkova E: Distribution of MAP2 in dendritic spines and its colocalization with actin. An immunogold electron-microscope study. Cell Tissue Res 1989, 256: 447–456. 10.1007/BF00225592CrossRefPubMed
29.
Zurück zum Zitat Greenberg SG, Davies P, Schein JD, Binder LI: Hydrofluoric acid-treated tau PHF proteins display the same biochemical properties as normal tau. J Biol Chem 1992, 267: 564–569.PubMed Greenberg SG, Davies P, Schein JD, Binder LI: Hydrofluoric acid-treated tau PHF proteins display the same biochemical properties as normal tau. J Biol Chem 1992, 267: 564–569.PubMed
30.
Zurück zum Zitat Braak E, Braak H, Mandelkow EM: A sequence of cytoskeleton changes related to the formation of neurofibrillary tangles and neuropil threads. Acta Neuropathol 1994, 87: 554–567. 10.1007/BF00293315CrossRefPubMed Braak E, Braak H, Mandelkow EM: A sequence of cytoskeleton changes related to the formation of neurofibrillary tangles and neuropil threads. Acta Neuropathol 1994, 87: 554–567. 10.1007/BF00293315CrossRefPubMed
31.
Zurück zum Zitat Hyman BT, Van Hoesen GW, Wolozin BL, Davies P, Kromer LJ, Damasio AR: Alz-50 antibody recognizes Alzheimer-related neuronal changes. Ann Neurol 1988, 23: 371–379. 10.1002/ana.410230410CrossRefPubMed Hyman BT, Van Hoesen GW, Wolozin BL, Davies P, Kromer LJ, Damasio AR: Alz-50 antibody recognizes Alzheimer-related neuronal changes. Ann Neurol 1988, 23: 371–379. 10.1002/ana.410230410CrossRefPubMed
32.
Zurück zum Zitat Rojo LE, Alzate-Morales J, Saavedra IN, Davies P, Maccioni RB: Selective interaction of lansoprazole and astemizole with tau polymers: potential new clinical use in diagnosis of Alzheimer’s disease. J Alzheimers Dis 2010, 19: 573–589.PubMedPubMedCentral Rojo LE, Alzate-Morales J, Saavedra IN, Davies P, Maccioni RB: Selective interaction of lansoprazole and astemizole with tau polymers: potential new clinical use in diagnosis of Alzheimer’s disease. J Alzheimers Dis 2010, 19: 573–589.PubMedPubMedCentral
33.
Zurück zum Zitat Gomez-Isla T, Hollister R, West H, Mui S, Growdon JH, Petersen RC, Parisi JE, Hyman BT: Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer’s disease. Ann Neurol 1997, 41: 17–24. 10.1002/ana.410410106CrossRefPubMed Gomez-Isla T, Hollister R, West H, Mui S, Growdon JH, Petersen RC, Parisi JE, Hyman BT: Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer’s disease. Ann Neurol 1997, 41: 17–24. 10.1002/ana.410410106CrossRefPubMed
34.
Zurück zum Zitat Mandelkow EM, Mandelkow E (2012) Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb Perspect Med 2:a006247 Mandelkow EM, Mandelkow E (2012) Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb Perspect Med 2:a006247
35.
Zurück zum Zitat Bancher C, Brunner C, Lassmann H, Budka H, Jellinger K, Wiche G, Seitelberger F, Grundke-Iqbal I, Iqbal K, Wisniewski HM: Accumulation of abnormally phosphorylated tau precedes the formation of neurofibrillary tangles in Alzheimer’s disease. Brain Res 1989, 477: 90–99. 10.1016/0006-8993(89)91396-6CrossRefPubMed Bancher C, Brunner C, Lassmann H, Budka H, Jellinger K, Wiche G, Seitelberger F, Grundke-Iqbal I, Iqbal K, Wisniewski HM: Accumulation of abnormally phosphorylated tau precedes the formation of neurofibrillary tangles in Alzheimer’s disease. Brain Res 1989, 477: 90–99. 10.1016/0006-8993(89)91396-6CrossRefPubMed
36.
Zurück zum Zitat Frost B, Jacks RL, Diamond MI: Propagation of tau misfolding from the outside to the inside of a cell. J Biol Chem 2009, 284: 12845–12852. 10.1074/jbc.M808759200CrossRefPubMedPubMedCentral Frost B, Jacks RL, Diamond MI: Propagation of tau misfolding from the outside to the inside of a cell. J Biol Chem 2009, 284: 12845–12852. 10.1074/jbc.M808759200CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Guo JL, Lee VM: Seeding of normal Tau by pathological Tau conformers drives pathogenesis of Alzheimer-like tangles. J Biol Chem 2011, 286: 15317–15331. 10.1074/jbc.M110.209296CrossRefPubMedPubMedCentral Guo JL, Lee VM: Seeding of normal Tau by pathological Tau conformers drives pathogenesis of Alzheimer-like tangles. J Biol Chem 2011, 286: 15317–15331. 10.1074/jbc.M110.209296CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Nonaka T, Watanabe ST, Iwatsubo T, Hasegawa M: Seeded aggregation and toxicity of α-synuclein and tau: cellular models of neurodegenerative diseases. J Biol Chem 2010, 285: 34885–34898. 10.1074/jbc.M110.148460CrossRefPubMedPubMedCentral Nonaka T, Watanabe ST, Iwatsubo T, Hasegawa M: Seeded aggregation and toxicity of α-synuclein and tau: cellular models of neurodegenerative diseases. J Biol Chem 2010, 285: 34885–34898. 10.1074/jbc.M110.148460CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Wu JW, Herman M, Liu L, Simoes S, Acker CM, Figueroa H, Steinberg JI, Margittai M, Kayed R, Zurzolo C, Di Paolo G, Duff KE: Small misfolded Tau species are internalized via bulk endocytosis and anterogradely and retrogradely transported in neurons. J Biol Chem 2013, 288: 1856–1870. 10.1074/jbc.M112.394528CrossRefPubMed Wu JW, Herman M, Liu L, Simoes S, Acker CM, Figueroa H, Steinberg JI, Margittai M, Kayed R, Zurzolo C, Di Paolo G, Duff KE: Small misfolded Tau species are internalized via bulk endocytosis and anterogradely and retrogradely transported in neurons. J Biol Chem 2013, 288: 1856–1870. 10.1074/jbc.M112.394528CrossRefPubMed
40.
Zurück zum Zitat Maeda S, Sahara N, Saito Y, Murayama S, Ikai A, Takashima A: Increased levels of granular tau oligomers: an early sign of brain aging and Alzheimer’s disease. Neurosci Res 2006, 54: 197–201. 10.1016/j.neures.2005.11.009CrossRefPubMed Maeda S, Sahara N, Saito Y, Murayama S, Ikai A, Takashima A: Increased levels of granular tau oligomers: an early sign of brain aging and Alzheimer’s disease. Neurosci Res 2006, 54: 197–201. 10.1016/j.neures.2005.11.009CrossRefPubMed
41.
Zurück zum Zitat Maeda S, Sahara N, Saito Y, Murayama M, Yoshiike Y, Kim H, Miyasaka T, Murayama S, Ikai A, Takashima A: Granular tau oligomers as intermediates of tau filaments. Biochemistry 2007, 46: 3856–3861. 10.1021/bi061359oCrossRefPubMed Maeda S, Sahara N, Saito Y, Murayama M, Yoshiike Y, Kim H, Miyasaka T, Murayama S, Ikai A, Takashima A: Granular tau oligomers as intermediates of tau filaments. Biochemistry 2007, 46: 3856–3861. 10.1021/bi061359oCrossRefPubMed
42.
Zurück zum Zitat Tang Y, Nyengaard JR, De Groot DM, Gundersen HJ: Total regional and global number of synapses in the human brain neocortex. Synapse 2001, 41: 258–273. 10.1002/syn.1083CrossRefPubMed Tang Y, Nyengaard JR, De Groot DM, Gundersen HJ: Total regional and global number of synapses in the human brain neocortex. Synapse 2001, 41: 258–273. 10.1002/syn.1083CrossRefPubMed
43.
Zurück zum Zitat Perez-Nievas BG, Stein TD, Tai HC, Dols-Icardo O, Scotton TC, Barroeta-Espar I, Fernandez-Carballo L, de Munain EL, Perez J, Marquie M, Serrano-Pozo A, Frosch MP, Lowe V, Parisi JE, Petersen RC, Ikonomovic MD, Lopez OL, Klunk W, Hyman BT, Gomez-Isla T: Dissecting phenotypic traits linked to human resilience to Alzheimer’s pathology. Brain 2013, 136: 2510–2526. 10.1093/brain/awt171CrossRefPubMedPubMedCentral Perez-Nievas BG, Stein TD, Tai HC, Dols-Icardo O, Scotton TC, Barroeta-Espar I, Fernandez-Carballo L, de Munain EL, Perez J, Marquie M, Serrano-Pozo A, Frosch MP, Lowe V, Parisi JE, Petersen RC, Ikonomovic MD, Lopez OL, Klunk W, Hyman BT, Gomez-Isla T: Dissecting phenotypic traits linked to human resilience to Alzheimer’s pathology. Brain 2013, 136: 2510–2526. 10.1093/brain/awt171CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Papasozomenos SC, Binder LI: Phosphorylation determines two distinct species of Tau in the central nervous system. Cell Motil Cytoskeleton 1987, 8: 210–226. 10.1002/cm.970080303CrossRefPubMed Papasozomenos SC, Binder LI: Phosphorylation determines two distinct species of Tau in the central nervous system. Cell Motil Cytoskeleton 1987, 8: 210–226. 10.1002/cm.970080303CrossRefPubMed
45.
Zurück zum Zitat Tashiro K, Hasegawa M, Ihara Y, Iwatsubo T: Somatodendritic localization of phosphorylated tau in neonatal and adult rat cerebral cortex. Neuroreport 1997, 8: 2797–2801. 10.1097/00001756-199708180-00029CrossRefPubMed Tashiro K, Hasegawa M, Ihara Y, Iwatsubo T: Somatodendritic localization of phosphorylated tau in neonatal and adult rat cerebral cortex. Neuroreport 1997, 8: 2797–2801. 10.1097/00001756-199708180-00029CrossRefPubMed
46.
Zurück zum Zitat Gartner U, Janke C, Holzer M, Vanmechelen E, Arendt T: Postmortem changes in the phosphorylation state of tau-protein in the rat brain. Neurobiol Aging 1998, 19: 535–543. 10.1016/S0197-4580(98)00094-3CrossRefPubMed Gartner U, Janke C, Holzer M, Vanmechelen E, Arendt T: Postmortem changes in the phosphorylation state of tau-protein in the rat brain. Neurobiol Aging 1998, 19: 535–543. 10.1016/S0197-4580(98)00094-3CrossRefPubMed
47.
Zurück zum Zitat He HJ, Wang XS, Pan R, Wang DL, Liu MN, He RQ (2009) The proline-rich domain of tau plays a role in interactions with actin. BMC Cell Biol 10:81 He HJ, Wang XS, Pan R, Wang DL, Liu MN, He RQ (2009) The proline-rich domain of tau plays a role in interactions with actin. BMC Cell Biol 10:81
48.
Zurück zum Zitat Fulga TA, Elson-Schwab I, Khurana V, Steinhilb ML, Spires TL, Hyman BT, Feany MB: Abnormal bundling and accumulation of F-actin mediates tau-induced neuronal degeneration in vivo. Nat Cell Biol 2007, 9: 139–148. 10.1038/ncb1528CrossRefPubMed Fulga TA, Elson-Schwab I, Khurana V, Steinhilb ML, Spires TL, Hyman BT, Feany MB: Abnormal bundling and accumulation of F-actin mediates tau-induced neuronal degeneration in vivo. Nat Cell Biol 2007, 9: 139–148. 10.1038/ncb1528CrossRefPubMed
49.
Zurück zum Zitat Lee S, Kim W, Li Z, Hall GF (2012) Accumulation of vesicle-associated human tau in distal dendrites drives degeneration and tau secretion in an in situ cellular tauopathy model. Int J Alzheimers Dis 2012:172837 Lee S, Kim W, Li Z, Hall GF (2012) Accumulation of vesicle-associated human tau in distal dendrites drives degeneration and tau secretion in an in situ cellular tauopathy model. Int J Alzheimers Dis 2012:172837
50.
Zurück zum Zitat Pooler AM, Hanger DP: Functional implications of the association of tau with the plasma membrane. Biochem Soc Trans 2010, 38: 1012–1015. 10.1042/BST0381012CrossRefPubMed Pooler AM, Hanger DP: Functional implications of the association of tau with the plasma membrane. Biochem Soc Trans 2010, 38: 1012–1015. 10.1042/BST0381012CrossRefPubMed
51.
Zurück zum Zitat Tolnay M, Probst A: REVIEW: tau protein pathology in Alzheimer’s disease and related disorders. Neuropathol Appl Neurobiol 1999, 25: 171–187. 10.1046/j.1365-2990.1999.00182.xCrossRefPubMed Tolnay M, Probst A: REVIEW: tau protein pathology in Alzheimer’s disease and related disorders. Neuropathol Appl Neurobiol 1999, 25: 171–187. 10.1046/j.1365-2990.1999.00182.xCrossRefPubMed
52.
Zurück zum Zitat Stoothoff WH, Johnson GV: Tau phosphorylation: physiological and pathological consequences. Biochim Biophys Acta 2005, 1739: 280–297. 10.1016/j.bbadis.2004.06.017CrossRefPubMed Stoothoff WH, Johnson GV: Tau phosphorylation: physiological and pathological consequences. Biochim Biophys Acta 2005, 1739: 280–297. 10.1016/j.bbadis.2004.06.017CrossRefPubMed
53.
Zurück zum Zitat Iqbal K, Zaidi T, Bancher C, Grundke-Iqbal I: Alzheimer paired helical filaments. Restoration of the biological activity by dephosphorylation. FEBS Lett 1994, 349: 104–108. 10.1016/0014-5793(94)00650-4CrossRefPubMed Iqbal K, Zaidi T, Bancher C, Grundke-Iqbal I: Alzheimer paired helical filaments. Restoration of the biological activity by dephosphorylation. FEBS Lett 1994, 349: 104–108. 10.1016/0014-5793(94)00650-4CrossRefPubMed
54.
Zurück zum Zitat Dujardin S, Lecolle K, Caillierez R, Begard S, Zommer N, Lachaud C, Carrier S, Dufour N, Auregan G, Winderickx J, Hantraye P, Deglon N, Colin M, Buee L (2014) Neuron-to-neuron wild-type Tau protein transfer through a trans-synaptic mechanism: relevance to sporadic tauopathies. Acta Neuropathol Commun 2:14 Dujardin S, Lecolle K, Caillierez R, Begard S, Zommer N, Lachaud C, Carrier S, Dufour N, Auregan G, Winderickx J, Hantraye P, Deglon N, Colin M, Buee L (2014) Neuron-to-neuron wild-type Tau protein transfer through a trans-synaptic mechanism: relevance to sporadic tauopathies. Acta Neuropathol Commun 2:14
55.
Zurück zum Zitat Simon D, Garcia-Garcia E, Gomez-Ramos A, Falcon-Perez JM, Diaz-Hernandez M, Hernandez F, Avila J: Tau overexpression results in its secretion via membrane vesicles. Neurodegener Dis 2012, 10: 73–75. 10.1159/000334915CrossRefPubMed Simon D, Garcia-Garcia E, Gomez-Ramos A, Falcon-Perez JM, Diaz-Hernandez M, Hernandez F, Avila J: Tau overexpression results in its secretion via membrane vesicles. Neurodegener Dis 2012, 10: 73–75. 10.1159/000334915CrossRefPubMed
56.
Zurück zum Zitat Saman S, Kim W, Raya M, Visnick Y, Miro S, Saman S, Jackson B, McKee AC, Alvarez VE, Lee NC, Hall GF: Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J Biol Chem 2012, 287: 3842–3849. 10.1074/jbc.M111.277061CrossRefPubMed Saman S, Kim W, Raya M, Visnick Y, Miro S, Saman S, Jackson B, McKee AC, Alvarez VE, Lee NC, Hall GF: Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease. J Biol Chem 2012, 287: 3842–3849. 10.1074/jbc.M111.277061CrossRefPubMed
57.
Zurück zum Zitat Kfoury N, Holmes BB, Jiang H, Holtzman DM, Diamond MI: Trans-cellular propagation of Tau aggregation by fibrillar species. J Biol Chem 2012, 287: 19440–19451. 10.1074/jbc.M112.346072CrossRefPubMedPubMedCentral Kfoury N, Holmes BB, Jiang H, Holtzman DM, Diamond MI: Trans-cellular propagation of Tau aggregation by fibrillar species. J Biol Chem 2012, 287: 19440–19451. 10.1074/jbc.M112.346072CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Gendreau KL, Hall GF (2013) Tangles, Toxicity, and Tau Secretion in AD - New Approaches to a Vexing Problem. Front Neurol 4:160 Gendreau KL, Hall GF (2013) Tangles, Toxicity, and Tau Secretion in AD - New Approaches to a Vexing Problem. Front Neurol 4:160
59.
Zurück zum Zitat Spires-Jones TL, Stoothoff WH, de Calignon A, Jones PB, Hyman BT: Tau pathophysiology in neurodegeneration: a tangled issue. Trends Neurosci 2009, 32: 150–159. 10.1016/j.tins.2008.11.007CrossRefPubMed Spires-Jones TL, Stoothoff WH, de Calignon A, Jones PB, Hyman BT: Tau pathophysiology in neurodegeneration: a tangled issue. Trends Neurosci 2009, 32: 150–159. 10.1016/j.tins.2008.11.007CrossRefPubMed
60.
Zurück zum Zitat Avila J, Lucas JJ, Perez M, Hernandez F: Role of tau protein in both physiological and pathological conditions. Physiol Rev 2004, 84: 361–384. 10.1152/physrev.00024.2003CrossRefPubMed Avila J, Lucas JJ, Perez M, Hernandez F: Role of tau protein in both physiological and pathological conditions. Physiol Rev 2004, 84: 361–384. 10.1152/physrev.00024.2003CrossRefPubMed
Metadaten
Titel
Frequent and symmetric deposition of misfolded tau oligomers within presynaptic and postsynaptic terminals in Alzheimer’s disease
verfasst von
Hwan-Ching Tai
Bo Y Wang
Alberto Serrano-Pozo
Matthew P Frosch
Tara L Spires-Jones
Bradley T Hyman
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Acta Neuropathologica Communications / Ausgabe 1/2014
Elektronische ISSN: 2051-5960
DOI
https://doi.org/10.1186/s40478-014-0146-2

Weitere Artikel der Ausgabe 1/2014

Acta Neuropathologica Communications 1/2014 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.