Skip to main content
Erschienen in: Alzheimer's Research & Therapy 1/2014

Open Access 01.02.2014 | Research

Assessing THK523 selectivity for tau deposits in Alzheimer’s disease and non–Alzheimer’s disease tauopathies

verfasst von: Michelle T Fodero-Tavoletti, Shozo Furumoto, Leanne Taylor, Catriona A McLean, Rachel S Mulligan, Ian Birchall, Ryuichi Harada, Colin L Masters, Kazuhiko Yanai, Yukitsuka Kudo, Christopher C Rowe, Nobuyuki Okamura, Victor L Villemagne

Erschienen in: Alzheimer's Research & Therapy | Ausgabe 1/2014

Abstract

Introduction

The introduction of tau imaging agents such as 18F-THK523 offers new hope for the in vivo assessment of tau deposition in tauopathies such as Alzheimer’s disease (AD), where preliminary 18F-THK523-PET studies have demonstrated significantly higher cortical retention of 18F-THK523 in AD compared to age-matched healthy individuals. In addition to AD, tau imaging with PET may also be of value in assessing non-AD tauopathies, such as corticobasal degeneration (CBD), progressive supranuclear palsy (PSP) and Pick’s disease (PiD).

Methods

To further investigate the ability of THK523 to recognize tau lesions, we undertook immunohistochemical and fluorescence studies in serial brain sections taken from individuals with AD (n = 3), CBD (n = 2), PSP (n = 1), PiD (n = 2) and Parkinson’s disease (PD; n = 2). In addition to the neuropathological analysis, one PSP patient had undergone a 18F-THK523 PET scan 5 months before death.

Results

Although THK523 labelled tau-containing lesions such as neurofibrillary tangles and neuropil threads in the hippocampus and frontal regions of AD brains, it failed to label tau-containing lesions in non-AD tauopathies. Furthermore, though THK523 faintly labelled dense-cored amyloid-β plaques in the AD frontal cortex, it failed to label α-synuclein-containing Lewy bodies in PD brain sections.

Conclusion

The results of this study suggest that 18F-THK523 selectively binds to paired helical filament tau in AD brains but does not bind to tau lesions in non-AD tauopathies, or to α-synuclein in PD brains.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​alzrt240) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

VLV, MTF-T, KY, NO and CLM designed the experiments. SF, RSM, RH, KY, YK and NO designed and manufactured THK523. LT and IB planned and conducted the human brain section immunostaining experiments. CAM planned and conducted the pathological characterisation of human brain samples. VL and CCR planned and coordinated human PET studies. MTF-T and VLV drafted the manuscript. All authors read and approved the final manuscript.
Abkürzungen
AD
Alzheimer’s disease
Amyloid-β
CBD
Corticobasal degeneration
CDR
Clinical Dementia Rating Scale
CDR-SOB
Clinical Dementia Rating Scale–Sum of Boxes
CSF
Cerebrospinal fluid
FTLD
Frontotemporal lobar degeneration
GM
Grey matter
MMSE
Mini Mental State Examination
NFT
Neurofibrillary tangle
PET
Positron emission tomography
PiB
Pittsburgh compound B
PiD
Pick’s disease
PSP
Progressive supranuclear palsy
ROI
Region of interest
SF
Straight filament
SUV
Standardised uptake value
TF
Twisted filament.

Introduction

Alzheimer’s disease (AD) is the most common form of dementia (50% to 70% of dementia cases) [1]. At present, there is no cure for the disease. Age is the greatest risk factor. Despite the existence of distinctive clinical diagnostic criteria, the differential diagnosis of AD and other neurodegenerative disorders is sometimes challenging because of substantial overlap in clinical presentations, especially at the early stages of the disease [2]. Consequently, making the definitive diagnosis of neurodegenerative diseases is still reliant upon postmortem examination of the brain.
AD is pathologically characterised by the presence of (1) extracellular neuritic plaques composed of aggregated β-amyloid (Aβ) and (2) intracellular neurofibrillary tangles (NFTs) composed of the aggregated tau protein [3, 4]. Tau aggregates are a pathological trait of not only AD but also other neurodegenerative conditions, such as corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP), as well as some variants of frontotemporal lobar degeneration (FTLD-tau) [5], such as Pick’s disease (PiD). Whilst the underlying mechanism leading to tau accumulation remains unclear, it is thought to be related to several pathogenic events resulting in hyperphosphorylation, misfolding and aggregation of tau. Tau aggregation in this wide spectrum of tauopathies presents with different morphologies (for example, NFTs in AD, astrocytes in CBD, globose tangles and thorny and tufted astrocytes in PSP and Pick bodies in PiD [69]) and ultrastructural conformations (for example, paired helical filaments in AD, straight filaments in PSP and twisted ribbons and random coils in PiD [6, 10, 11]), which are probably attributable to the combinations of the different tau isoforms and a wide variety of posttranslation modifications [6, 12]. Additionally, the spatial distribution of the tau aggregates in these tauopathies differ from each other, with NFTs in AD being prevalent in the mesial temporal cortex and cortical grey-matter (GM) areas. Tau aggregates are also found in the frontal and striatal brain regions in CBD; in the brainstem, cerebellar white matter and basal ganglia in PSP; and in the frontal and temporal neocortex in PiD [1317]. The diverse distribution of these tau aggregates in the brain can potentially be useful in the differential diagnosis of these tauopathies, assuming the same tau imaging agent binds with similar affinity to the whole spectrum of tau aggregates. Alternatively, the differential diagnosis might require the development of selective tau radiotracers for each specific conformation of tau aggregates.
In recent years, a great deal of interest has been placed on identifying the ideal diagnostic tool for neurodegenerative diseases. Despite the quantitative assessment of Aβ, tau and phospho-tau in cerebrospinal fluid (CSF) [18], lumbar puncture is still considered an invasive procedure for the widespread screening of the ‘at-risk’ population. Additionally, CSF measurements do not provide information on regional brain deposition of Aβ or tau, which may have clear correlates with cognition or regional brain atrophy and might not be able to provide important information regarding the therapeutic outcomes or response to current drugs aimed at modulating the deposition of these misfolded proteins [1923]. Given the sometimes nonspecific nature of clinical symptoms and neuropsychological assessments, modern molecular imaging techniques have proven beneficial in the noninvasive identification of the underlying pathology of these diseases. Considerable effort has been focused on the development of novel Aβ ligands that permit detection of Aβ deposition [24]. The Aβ-specific ligands 18F-AV-45 (florbetapir; (E)-4-(2-(6-(2-(2-(2-([18F]-fluoroethoxy)ethoxy)ethoxy)pyridin-3-yl)vinyl)-N-methyl benzenamine) and Pittsburgh compound B (PiB) [25] are the best characterized and have proven to be suitable positron emission tomography (PET) biomarkers for the in vivo quantitation of cerebral Aβ burden. They have demonstrated a robust difference in retention between AD and healthy individuals [2527]. 18F-AV-45 [27] and flutemetamol 18 (2-[3-fluoranyl-4-(methylamino)phenyl]-1,3-benzothiazol-6-ol) [28] have already been approved for clinical Aβ imaging in the United States. These two agents belong to a second generation of Aβ radiotracers labelled with 18F, which, with a half-life of 110 minutes, allows a wider and more cost-effective application of Aβ imaging.
We recently reported the preclinical characterization of the selective tau radiotracer 18F-THK523 [29], a quinoline derivative pioneered by Okamura and colleagues [30, 31]. Preliminary clinical evaluation of 18F-THK523 has demonstrated that 18F-THK523 retention is significantly higher in the cortical and hippocampal GM of AD patients than in age-matched healthy individuals [32].
To discern whether 18F-THK523 recognises non-AD tau aggregates in addition to NFTs, we evaluated a series of brain sections from AD and non-AD tauopathies to evaluate the binding profile of 18F-THK523.

Methods

Postmortem assessment

Chemicals

All reagents were purchased from Sigma-Aldrich (St Louis, MO, USA) unless otherwise stated.

Tissue collection and characterisation

Tissues were sourced and prepared by the Victorian Brain Bank Network. The AD pathological diagnosis was made according to standard National Institute on Aging/Reagan Institute criteria [5]. Determination of age-matched control cases were subject to the above-described criteria. The pathological diagnoses of PiD, CBD and PSP were all made according to previously described methods [33, 34]. Ten cases were evaluated for this study: AD (n = 3), CBD (n = 1), PiD (n = 2), PD (n = 1) and PSP (n = 3). One of the individuals with PSP had undergone 18F-THK523 PET 5 months before death.

Immunohistochemistry and fluorescence analysis

All brain tissue was fixed in 10% neutral buffered formalin, processed, and embedded in paraffin. For immunohistochemistry, 5-μm serial sections were deparaffinized and treated with 90% formic acid for 5 minutes, and endogenous peroxidase activity was blocked with 5% hydrogen peroxide. Sections were then treated with 0.2% casein in Tris buffer before incubation with primary antibodies to α-synuclein (97/8, 1:2,000 dilution) [35], Aβ (1e8, 1:2,000 dilution; monoclonal antibody recognises Aβ(17–24)) [36] and tau (polyclonal antibody recognises C-terminal tau (amino acids 243 to 441), catalog no. 0024; Dako Denmark, Glostrup, Denmark), for 1 hour at room temperature. Serial 5-μm tissue sections were stained as follows. The first and third sections were immunolabelled with anti-97/8 antibody, anti-1e8 antibody or tau to identify Lewy bodies, Aβ plaques or tau aggregates, respectively. The second serial section was stained with unlabelled THK523 to assess whether THK523 staining colocalised with the immunodetected Lewy bodies and/or Aβ plaques and/or tau aggregates. Detection of antibody binding was achieved using the LSAB kit (labelled streptavidin-biotin, catalog no. K0657; Dako Denmark), then sections were incubated with hydrogen peroxidase diaminobenzidine (H2O2-DAB; Dako Denmark) to visualise the α-synuclein-, Aβ- or tau-positive deposits. Sections were counterstained briefly (15 seconds) with Harris’s haematoxylin. To detect THK523 fluorescence, quenching was first performed whereby sections were first deparaffinized and tissue autofluorescence was minimized by treatment of sections with 0.25% KMnO4 phosphate-buffered saline (PBS) for 20 minutes prior to washing in PBS and incubation with 1% potassium metabisulphite/1% oxalic acid/PBS for 5 minutes. Following autofluorescence quenching, sections were blocked in 2% bovine serum albumin/PBS, pH 7.0, for 10 minutes and stained with 100 μM THK523 for 30 minutes. Sections washed in PBS were then mounted in nonfluorescent mounting medium (catalog no. S3023; Dako Denmark). Epifluorescent images were visualized on a Leica microscope (47-nm cyan fluorescent protein, fluorescence filter set 47 (EM BP 436/20, BS FT 455 and EM BP 480/40); Leica Microsystems, North Ryde, 2113 Australia). Colocalisation of the THK523 and antibody signals were assessed by overlaying images from each of the stained serial tissue sections.

Antemortem assessment

Five months before death, a seventy-nine-year-old patient diagnosed with PSP underwent an Aβ imaging PET scan with 18F-florbetaben and a tau imaging scan with 18F-THK523. Approval of the study was granted by the Austin Health Human Research Ethics Committee, and written informed consent was obtained from all participants and caregivers before the study. The patient was recruited, reviewed and diagnosed on the basis of clinical and neuropsychological assessment by consensus of a neurologist and a neuropsychologist.
As part of the imaging protocol, we performed magnetic resonance imaging (MRI) using a three-dimensional magnetization-prepared rapid acquisition gradient echo sequence and T2-weighted fast spin echo and fluid-attenuated inversion recovery sequences. Both 18F-florbetaben and 18F-THK523 were synthesized at the Centre for PET, Austin Health, as previously described [3739]. PET scans were acquired using a Philips Allegro PET scanner (Philips Healthcare, North Ryde, Australia) at the Austin Health Centre. A transmission scan using a rotating Cs-137 source was taken for attenuation correction immediately prior to obtaining the emission scan. A 60-minute list-mode emission acquisition, followed by a 90- to 120-minute acquisition using 10-minute frames, was performed in three-dimensional mode after injection of 300 MBq of 18F-florbetaben. A 90-minute list-mode emission image acquisition was performed in three-dimensional mode after injection of 200 MBq of 18F-THK523. Images were reconstructed using a three-dimensional row action maximum likelihood algorithm.
PET images were processed using a previously described semiautomatic region of interest (ROI) method [40]. Briefly, coregistration of the patient’s MRI scans with the PET images was performed with Statistical Parametric Mapping 8 (SPM8) software [41]. A narrow cortical ROI template was placed on the coregistered MRI scanner by an operator (VLV) who was blinded to the participant’s clinical status, then it was transferred to the coregistered PET images. The ROI template covered cortical and subcortical GM structures as well as the midbrain and pons. Subcortical white-matter ROIs were placed at the centrum semiovale, and the cerebellar regions were placed over the cerebellar cortex, taking care to avoid white matter. Standardised uptake values (SUVs), defined as the decay-corrected brain radioactivity concentration normalized for injected dose and body weight, were calculated for all regions. In order to avoid arterial blood sampling, a simplified approach was applied using the cerebellar cortex as the reference region. SUVs were used to derive SUV ratios (SUVRs) referenced to the cerebellar cortex soon after the ratio of binding in neocortex to that in the cerebellar cortex reached an apparent steady state. Regional THK523 SUVRs were obtained for all regions sampled. Global tau burden was expressed as the average THK523 SUVR for the following cortical ROIs: frontal (consisting of the dorsolateral prefrontal, ventrolateral prefrontal and orbitofrontal regions), superior parietal, lateral temporal, lateral occipital, and anterior and posterior cingulate. Partial volume correction accounting for both GM atrophy and white-matter spillover was performed using a three-compartment approach with PMOD version 3.1 software (PMOD Technologies, Zurich, Switzerland). To establish whether either 18F-florbetaben or 18F-THK523 retention in the PSP patient was different from age-matched controls, a Z-score was generated for both global and regional retention. The respective Z-scores were generated against ten healthy controls who had 18F-florbetaben studies and ten healthy controls who had 18F-THK523 studies. Conservative Z-scores greater than 1.5, indicating just 1.5 standard deviations (SDs) from the mean of the control participants, were considered abnormal.

Results

Demographic information

The demographics of the patients whose postmortem human brain tissue was utilized for these studies, expressed as mean ± SD, are presented in Table 1. All participants assessed were of similar age with comparable postmortem intervals for tissue collection. The patient with PSP who underwent PET was 79 years old and had 11 years of formal education. A neuropsychological examination revealed the patient had a Mini Mental State Examination score of 26, a Clinical Dementia Rating Scale score of 1 a Clinical Dementia Rating Scale–Sum of Boxes score of 5.5, an episodic memory composite score of -3.22 and a nonmemory composite score of -3.40. The PSP participant died 5 months after the PET scans were taken.
Table 1
Patient demographics a
Diagnosis
No. of patients
Mean age (SD), years
Mean PMI (hours)
AD
3
72.9 ± 6.7
22.8 ± 8.8
PSP
3
73.2 ± 4.6
37.2 ± 18.6
PiD
2
75.4 ± 7.1
47.0 ± 5.2
CBD
1
72.5
11.0
PD
1
70.5
22.5
aAD, Alzheimer’s disease; CBD, Corticobasal degeneration; PD, Parkinson’s disease; PiD, Pick’s disease; PMI, Postmortem interval; PSP, Progressive supranuclear palsy.

Assessment of THK523 binding/fluorescence in non–Alzheimer’s disease tauopathies

The results of our previous postmortem studies [29] have indicated that THK523 labels AD tau lesions, namely, NFTs in the hippocampus of patients with AD (Figure 1). To determine whether THK523 would also bind to non-AD tau lesions, brain sections from non-AD tauopathies were evaluated. For these studies, fixed contiguous serial sections from the striatum (CBD and PSP samples), the frontal cortex (PiD sample) and the pons (PSP sample) were either immunostained with a polyclonal tau antibody for the detection of tau lesions or incubated with the fluorescent compound THK523 to determine whether THK523 bound to non-AD tauopathy aggregates. Immunohistochemical staining of brain tissue regions rich in tau immunoreactivity was detected by light microscopy, and the same tissue region within the adjacent serial section was assessed by fluorescence microscopy to compare and determine whether the immunoreactive tau lesion colocalised with THK523 binding, indicated by a fluorescent signal. Immunohistological assessment of brain sections from CBD and PiD patients revealed the characteristic presence of globose tangles, coiled bodies (as indicated by the arrowheads in the figures; see top left panel of Figure 2) and Pick’s bodies (arrowheads in bottom left panel of Figure 2) in the striatum and frontal cortex. Nonetheless, examination of the same region within the adjacent serial section exhibited no signs of THK523 fluorescence, indicating that THK523 does not bind to these tau lesions. Likewise, immunohistological evaluation of the pons (left, top panel of Figure 3) and striatum (left, bottom panel of Figure 2) in PSP patients revealed that, despite the presence of tau globose tangles, there was no detectable THK523 fluorescence signal in the same region of the adjacent serial brain section, again suggesting that THK523 does not bind to globose tangles. It is noteworthy that one of the three PSP patients evaluated (Figure 3) had had 18F-THK523 and 18F-florbetaben PET scans 5 months prior to death. There was low 18F-florbetaben cortical retention (Figure 4), correlating with postmortem results showing absence of Aβ deposits. There was also low cortical 18F-THK523 retention as well as low 18F-THK523 retention in the basal ganglia, midbrain, pons and cerebellar white matter (Figure 4), which was indistinguishable from age-matched controls and in contrast to the relatively high density of tau lesions observed in the brain, confirming the absence of THK523 binding to globose tangles. Analysis of the PSP patient PET scans showed global and regional Z-scores less than 1.0 for 18F-THK523 and 18F-florbetaben, confirming the visual inspection of the images.

Assessment of THK523 binding/fluorescence in Parkinson’s disease

To further test the selectivity of THK523, we evaluated its ability to bind to Lewy bodies composed of α-synuclein and ubiquitin aggregates sharing a similar β-sheet secondary structure. For these studies, serial sections of the substantia nigra from a PD patient were either immunostained with antibodies raised to α-synuclein, or treated with a fluorescent compound, THK523. Evaluation of these stained serial sections demonstrated that, whilst the presence of Lewy bodies could be clearly identified by immunohistochemistry (Figure 5, left panel), the adjacent serial section was devoid of THK523 fluorescence (Figure 5, right panel), implying that THK523 did not bind to Lewy bodies.

Discussion

In the present study, we further characterized 18F-THK523 as a selective tau imaging agent by testing its ability to recognize the various morphological conformations of tau in a wide spectrum of tauopathies. Whilst in our previous studies we determined that THK523 binds selectively to NFTs in preference to Aβ plaques [29, 32], in this study we also assessed 18F-THK523 binding to other β-sheet structured protein fibrils, namely, α-synuclein-containing Lewy bodies.
Given the morphological and ultrastructural diversity of tau aggregates, it may be unlikely that a single tau imaging agent could be useful for the diagnosis of all tauopathies. In the first instance, tau comprises six isoforms distinguished by their length and number of repeats (R) of microtubule binding domains [6, 42]. AD tau comprises an equal ratio of the 3R and 4R isoforms, which mainly appear as NFTs. The 4R isoform predominates in PSP with tau aggregates comprising tufted-shaped astrocytes, GTs and oligodendroglial coiled bodies [43, 44]. Despite also being a 4R tauopathy, in CBD the tau inclusions appear as astrocytic plaques, neutropil threads and tau pretangles [45]. PiD, a 3R tauopathy, is diagnosed by the presence of ‘Pick bodies’, tau-positive intraneuronal inclusions [46]. Moreover, these tau aggregates are further differentiated by their ultrastructure. NFTs are predominantly composed of paired helical filaments (PHFs), tau inclusions in PSP and CBD are composed predominantly of straight tau filaments (SFs) and twisted tau filaments (TFs) [11], whereas Pick bodies comprise a combination of TFs and random coiled tau filaments [11]. It is noteworthy that, whilst PSP and CBD share SFs, the size of the filaments is significantly different [47]. Despite this diversity, a recent report describing a novel class of tau tracers phenyl/pyridinyl-butadienyl-benzothiazoles/benzothiazoliums (PBB) demonstrated binding to a variety of tau deposits in fluorescence studies of AD, CBD and PSP brain sections [48]. Additionally, that study also demonstrated positive [11C]PBB3 PET scans in both AD and CBD patients [48].
Given the evident differences in THK523 staining, the fluorescence microscopy studies we present herein demonstrate that THK523, even at the very high concentration of 100 μM, does not bind to non-PHF-tau aggregates. The existence of a THK523 binding site on PHFs that is absent in the other conformations is further emphasized by previous computerized cross-sectional and fragmentation studies which indicated that, whilst these types of filaments share a similarly shaped morphological unit, the filament arrangement is different [49].
PHFs appear as two filaments twisted around one another with a cross-over repeat of 80 nm and an apparent width varying between about 10 nm and 22 nm [50]. The resulting aggregate exhibits an amyloid structure characterized by a β-sheet network forming the heart of the protofibril network. This ultrastructural property shared with Aβ and α-synuclein aggregates sometimes results in the nonselective binding of imaging agents [51]. As noted previously [29], in addition to THK523’s binding to NFTs, our fluorescence studies obtained at high tracer concentrations—10,000-fold higher than the concentrations typically achieved during a PET scan—demonstrated inconsistent THK523 staining of Aβ plaques. THK523 stained the dense core of some Aβ plaques in the frontal cortex of AD sections but did not stain dense Aβ plaques in the hippocampus (Figure 1, right panel). It is noteworthy that variable staining of NFTs at high concentrations of PiB has also been reported [52]. In addition to previous reports of in vitro studies [29, 30], several lines of evidence support the notion that THK523 selectively binds to PHF-tau and does not bind to Aβ in vivo: (1) Cortical THK523 retention is significantly higher in AD; (2) THK523 retention follows the known distribution of PHF-tau in the AD brain; (3) PiB and THK523 show different brain regional distribution patterns; (4) hippocampal THK523 retention significantly correlated with cognitive parameters, but hippocampal PiB retention did not; and (5) hippocampal THK523 retention significantly correlated with hippocampal volume, but hippocampal PiB retention did not [32].
The selectivity of THK523 for tau over other β-sheet aggregated proteins was further demonstrated by fluorescence microscopy studies showing the absence of THK523 fluorescence in brain sections exhibiting immunolabelled α-synuclein-containing Lewy bodies (Figure 5, right panel).
The PSP patient showed neither 18F-THK523 nor 18F-florbetaben retention in the brain, suggesting the absence not only of Aβ plaques but also of tau deposits. Neuropathological examination of the brain confirmed the absence of Aβ plaques; however, typical tau lesions were present in different brain regions that were not stained by THK523. Given the ultrastructural diversity of tau aggregates, the information derived from these THK523 studies is highly valuable for the future design of tau imaging ligands.

Conclusion

In the present study, we have demonstrated that THK523 selectively binds to PHF-tau with negligible binding to PSP, CBD and PiD tau aggregates, as well as to Aβ and α-synuclein aggregates. The results of this study also show that novel tracers that bind to non-PHF tau aggregates are needed.

Acknowledgements

We thank Fairlie Hinton and Geoff Pavey from the Victorian Brain Bank Network (VBBN) for sourcing and preparing the human brain tissue (HREC University of Melbourne 9414748). The VBBN is supported by The Florey Institute of Neuroscience and Mental Health, The Alfred and the Victorian Forensic Institute of Medicine, and it is funded by Australia’s National Health & Medical Research Council (NHMRC) and Parkinson’s Victoria. The research was supported in part by Alzheimer’s Drug Discovery Foundation Research Grant 20101208 AFTD and NHMRC project grant 1044361.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

VLV, MTF-T, KY, NO and CLM designed the experiments. SF, RSM, RH, KY, YK and NO designed and manufactured THK523. LT and IB planned and conducted the human brain section immunostaining experiments. CAM planned and conducted the pathological characterisation of human brain samples. VL and CCR planned and coordinated human PET studies. MTF-T and VLV drafted the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Masters CL, Cappai R, Barnham KJ, Villemagne VL: Molecular mechanisms for Alzheimer’s disease: implications for neuroimaging and therapeutics. J Neurochem. 2006, 97: 1700-1725. 10.1111/j.1471-4159.2006.03989.x.CrossRefPubMed Masters CL, Cappai R, Barnham KJ, Villemagne VL: Molecular mechanisms for Alzheimer’s disease: implications for neuroimaging and therapeutics. J Neurochem. 2006, 97: 1700-1725. 10.1111/j.1471-4159.2006.03989.x.CrossRefPubMed
2.
Zurück zum Zitat van der Zee J, Sleegers K, Van Broeckhoven C: Invited article: the Alzheimer disease–frontotemporal lobar degeneration spectrum. Neurology. 2008, 71: 1191-1197. 10.1212/01.wnl.0000327523.52537.86.CrossRefPubMed van der Zee J, Sleegers K, Van Broeckhoven C: Invited article: the Alzheimer disease–frontotemporal lobar degeneration spectrum. Neurology. 2008, 71: 1191-1197. 10.1212/01.wnl.0000327523.52537.86.CrossRefPubMed
3.
Zurück zum Zitat Braak H, Braak E: Morphological criteria for the recognition of Alzheimer’s disease and the distribution pattern of cortical changes related to this disorder. Neurobiol Aging. 1994, 15: 355-356. 10.1016/0197-4580(94)90032-9. discussion 379–380CrossRefPubMed Braak H, Braak E: Morphological criteria for the recognition of Alzheimer’s disease and the distribution pattern of cortical changes related to this disorder. Neurobiol Aging. 1994, 15: 355-356. 10.1016/0197-4580(94)90032-9. discussion 379–380CrossRefPubMed
4.
Zurück zum Zitat Corder EH, Woodbury MA, Volkmann I, Madsen DK, Bogdanovic N, Winblad B: Density profiles of Alzheimer disease regional brain pathology for the Huddinge Brain Bank: pattern recognition emulates and expands upon Braak staging. Exp Gerontol. 2000, 35: 851-864. 10.1016/S0531-5565(00)00147-9.CrossRefPubMed Corder EH, Woodbury MA, Volkmann I, Madsen DK, Bogdanovic N, Winblad B: Density profiles of Alzheimer disease regional brain pathology for the Huddinge Brain Bank: pattern recognition emulates and expands upon Braak staging. Exp Gerontol. 2000, 35: 851-864. 10.1016/S0531-5565(00)00147-9.CrossRefPubMed
5.
Zurück zum Zitat The National Institute on Aging and Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer’s Disease: Consensus recommendations for the postmortem diagnosis of Alzheimer’s disease. Neurobiol Aging. 1997, 18: S1-S2.CrossRef The National Institute on Aging and Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer’s Disease: Consensus recommendations for the postmortem diagnosis of Alzheimer’s disease. Neurobiol Aging. 1997, 18: S1-S2.CrossRef
6.
Zurück zum Zitat Delacourte A: Tauopathies: recent insights into old diseases. Folia Neuropathol. 2005, 43: 244-257.PubMed Delacourte A: Tauopathies: recent insights into old diseases. Folia Neuropathol. 2005, 43: 244-257.PubMed
7.
Zurück zum Zitat King ME, Ghoshal N, Wall JS, Binder LI, Ksiezak-Reding H: Structural analysis of Pick’s disease-derived and in vitro-assembled tau filaments. Am J Pathol. 2001, 158: 1481-1490. 10.1016/S0002-9440(10)64099-0.PubMedCentralCrossRefPubMed King ME, Ghoshal N, Wall JS, Binder LI, Ksiezak-Reding H: Structural analysis of Pick’s disease-derived and in vitro-assembled tau filaments. Am J Pathol. 2001, 158: 1481-1490. 10.1016/S0002-9440(10)64099-0.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Scaravilli T, Tolosa E, Ferrer I: Progressive supranuclear palsy and corticobasal degeneration: lumping versus splitting. Mov Disord. 2005, 20: S21-S28. 10.1002/mds.20536.CrossRefPubMed Scaravilli T, Tolosa E, Ferrer I: Progressive supranuclear palsy and corticobasal degeneration: lumping versus splitting. Mov Disord. 2005, 20: S21-S28. 10.1002/mds.20536.CrossRefPubMed
9.
Zurück zum Zitat Uchihara T, Tsuchiya K: Neuropathology of Pick body disease. Handb Clin Neurol. 2008, 89: 415-430.CrossRefPubMed Uchihara T, Tsuchiya K: Neuropathology of Pick body disease. Handb Clin Neurol. 2008, 89: 415-430.CrossRefPubMed
10.
Zurück zum Zitat Mohorko N, Bresjanac M: [Tau protein and human tauopathies: an overview] [in Slovenian]. Zdrav Vestn. 2008, 77: II-35-II-41. Mohorko N, Bresjanac M: [Tau protein and human tauopathies: an overview] [in Slovenian]. Zdrav Vestn. 2008, 77: II-35-II-41.
11.
Zurück zum Zitat Delacourte A, Buée L: Tau pathology: a marker of neurodegenerative disorders. Curr Opin Neurol. 2000, 13: 371-376. 10.1097/00019052-200008000-00002.CrossRefPubMed Delacourte A, Buée L: Tau pathology: a marker of neurodegenerative disorders. Curr Opin Neurol. 2000, 13: 371-376. 10.1097/00019052-200008000-00002.CrossRefPubMed
12.
Zurück zum Zitat Villemagne VL, Furumoto S, Fodero-Tavoletti M, Harada R, Mulligan RS, Kudo Y, Masters CL, Yanai K, Rowe CC, Okamura N: The challenges of tau imaging. Future Neurol. 2012, 7: 409-421. 10.2217/fnl.12.34.CrossRef Villemagne VL, Furumoto S, Fodero-Tavoletti M, Harada R, Mulligan RS, Kudo Y, Masters CL, Yanai K, Rowe CC, Okamura N: The challenges of tau imaging. Future Neurol. 2012, 7: 409-421. 10.2217/fnl.12.34.CrossRef
13.
Zurück zum Zitat Dickson DW: Neuropathology of Pick’s disease. Neurology. 2001, 56: S16-S20. 10.1212/WNL.56.suppl_4.S16.CrossRefPubMed Dickson DW: Neuropathology of Pick’s disease. Neurology. 2001, 56: S16-S20. 10.1212/WNL.56.suppl_4.S16.CrossRefPubMed
14.
Zurück zum Zitat Josephs KA, Whitwell JL, Dickson DW, Boeve BF, Knopman DS, Petersen RC, Parisi JE, Jack CR: Voxel-based morphometry in autopsy proven PSP and CBD. Neurobiol Aging. 2008, 29: 280-289. 10.1016/j.neurobiolaging.2006.09.019.PubMedCentralCrossRefPubMed Josephs KA, Whitwell JL, Dickson DW, Boeve BF, Knopman DS, Petersen RC, Parisi JE, Jack CR: Voxel-based morphometry in autopsy proven PSP and CBD. Neurobiol Aging. 2008, 29: 280-289. 10.1016/j.neurobiolaging.2006.09.019.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Dickson DW: Neuropathologic differentiation of progressive supranuclear palsy and corticobasal degeneration. J Neurol. 1999, 246: II6-II15.CrossRefPubMed Dickson DW: Neuropathologic differentiation of progressive supranuclear palsy and corticobasal degeneration. J Neurol. 1999, 246: II6-II15.CrossRefPubMed
16.
Zurück zum Zitat Cairns NJ, Bigio EH, Mackenzie IRA, Neumann M, Lee VM, Hatanpaa KJ, White CL, Schneider JA, Grinberg LT, Halliday G, Duyckaerts C, Lowe JS, Holm IE, Tolnay M, Okamoto K, Yokoo H, Murayama S, Woulfe J, Munoz DG, Dickson DW, Ince PG, Trojanowski JQ, Mann DM, Consortium for Frontotemporal Lobar Degeneration: Neuropathologic diagnostic and nosologic criteria for frontotemporal lobar degeneration: consensus of the Consortium for Frontotemporal Lobar Degeneration. Acta Neuropathol. 2007, 114: 5-22. 10.1007/s00401-007-0237-2.PubMedCentralCrossRefPubMed Cairns NJ, Bigio EH, Mackenzie IRA, Neumann M, Lee VM, Hatanpaa KJ, White CL, Schneider JA, Grinberg LT, Halliday G, Duyckaerts C, Lowe JS, Holm IE, Tolnay M, Okamoto K, Yokoo H, Murayama S, Woulfe J, Munoz DG, Dickson DW, Ince PG, Trojanowski JQ, Mann DM, Consortium for Frontotemporal Lobar Degeneration: Neuropathologic diagnostic and nosologic criteria for frontotemporal lobar degeneration: consensus of the Consortium for Frontotemporal Lobar Degeneration. Acta Neuropathol. 2007, 114: 5-22. 10.1007/s00401-007-0237-2.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Boxer AL, Geschwind MD, Belfor N, Gorno-Tempini ML, Schauer GF, Miller BL, Weiner MW, Rosen HJ: Patterns of brain atrophy that differentiate corticobasal degeneration syndrome from progressive supranuclear palsy. Arch Neurol. 2006, 63: 81-86. 10.1001/archneur.63.1.81.CrossRefPubMed Boxer AL, Geschwind MD, Belfor N, Gorno-Tempini ML, Schauer GF, Miller BL, Weiner MW, Rosen HJ: Patterns of brain atrophy that differentiate corticobasal degeneration syndrome from progressive supranuclear palsy. Arch Neurol. 2006, 63: 81-86. 10.1001/archneur.63.1.81.CrossRefPubMed
18.
Zurück zum Zitat Jack CR, Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, Petersen RC, Trojanowski JQ: Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol. 2010, 9: 119-128. 10.1016/S1474-4422(09)70299-6.PubMedCentralCrossRefPubMed Jack CR, Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, Petersen RC, Trojanowski JQ: Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol. 2010, 9: 119-128. 10.1016/S1474-4422(09)70299-6.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Gozes I, Stewart A, Morimoto B, Fox A, Sutherland K, Schmeche D: Addressing Alzheimer’s disease tangles: from NAP to AL-108. Curr Alzheimer Res. 2009, 6: 455-460. 10.2174/156720509789207895.CrossRefPubMed Gozes I, Stewart A, Morimoto B, Fox A, Sutherland K, Schmeche D: Addressing Alzheimer’s disease tangles: from NAP to AL-108. Curr Alzheimer Res. 2009, 6: 455-460. 10.2174/156720509789207895.CrossRefPubMed
20.
Zurück zum Zitat Hampel H, Blennow K, Shaw LM, Hoessler YC, Zetterberg H, Trojanowski JQ: Total and phosphorylated tau protein as biological markers of Alzheimer’s disease. Exp Gerontol. 2010, 45: 30-40. 10.1016/j.exger.2009.10.010.PubMedCentralCrossRefPubMed Hampel H, Blennow K, Shaw LM, Hoessler YC, Zetterberg H, Trojanowski JQ: Total and phosphorylated tau protein as biological markers of Alzheimer’s disease. Exp Gerontol. 2010, 45: 30-40. 10.1016/j.exger.2009.10.010.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Wada T, Miyata T, Sakai H, Kurokawa K: β2-microglobulin and renal bone disease. Perit Dial Int. 1999, 19: S413-S416.PubMed Wada T, Miyata T, Sakai H, Kurokawa K: β2-microglobulin and renal bone disease. Perit Dial Int. 1999, 19: S413-S416.PubMed
22.
Zurück zum Zitat Wider C, Uitti RJ, Wszolek ZK, Fang JY, Josephs KA, Baker MC, Rademakers R, Hutton ML, Dickson DW: Progranulin gene mutation with an unusual clinical and neuropathologic presentation. Mov Disord. 2008, 23: 1168-1173. 10.1002/mds.22065.CrossRefPubMed Wider C, Uitti RJ, Wszolek ZK, Fang JY, Josephs KA, Baker MC, Rademakers R, Hutton ML, Dickson DW: Progranulin gene mutation with an unusual clinical and neuropathologic presentation. Mov Disord. 2008, 23: 1168-1173. 10.1002/mds.22065.CrossRefPubMed
23.
Zurück zum Zitat Wischik C, Staff R: Challenges in the conduct of disease-modifying trials in AD: practical experience from a phase 2 trial of Tau-aggregation inhibitor therapy. J Nutr Health Aging. 2009, 13: 367-369. 10.1007/s12603-009-0046-5.CrossRefPubMed Wischik C, Staff R: Challenges in the conduct of disease-modifying trials in AD: practical experience from a phase 2 trial of Tau-aggregation inhibitor therapy. J Nutr Health Aging. 2009, 13: 367-369. 10.1007/s12603-009-0046-5.CrossRefPubMed
24.
Zurück zum Zitat Cui M: Past and recent progress of molecular imaging probes for β-amyloid plaques in the brain. Curr Med Chem. 2014, 21: 82-112.CrossRefPubMed Cui M: Past and recent progress of molecular imaging probes for β-amyloid plaques in the brain. Curr Med Chem. 2014, 21: 82-112.CrossRefPubMed
25.
Zurück zum Zitat Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, Holt DP, Bergström M, Savitcheva I, Huang GF, Estrada S, Ausén B, Debnath ML, Barletta J, Price JC, Sandell J, Lopresti BJ, Wall A, Koivisto P, Antoni G, Mathis CA, Långström B: Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol. 2004, 55: 306-319. 10.1002/ana.20009.CrossRefPubMed Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, Holt DP, Bergström M, Savitcheva I, Huang GF, Estrada S, Ausén B, Debnath ML, Barletta J, Price JC, Sandell J, Lopresti BJ, Wall A, Koivisto P, Antoni G, Mathis CA, Långström B: Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol. 2004, 55: 306-319. 10.1002/ana.20009.CrossRefPubMed
26.
Zurück zum Zitat Rowe CC, Ng S, Ackermann U, Gong SJ, Pike K, Savage G, Cowie TF, Dickinson KL, Maruff P, Darby D, Smith C, Woodward M, Merory J, Tochon-Danguy H, O’Keefe G, Klunk WE, Mathis CA, Price JC, Masters CL, Villemagne VL: Imaging β-amyloid burden in aging and dementia. Neurology. 2007, 68: 1718-1725. 10.1212/01.wnl.0000261919.22630.ea.CrossRefPubMed Rowe CC, Ng S, Ackermann U, Gong SJ, Pike K, Savage G, Cowie TF, Dickinson KL, Maruff P, Darby D, Smith C, Woodward M, Merory J, Tochon-Danguy H, O’Keefe G, Klunk WE, Mathis CA, Price JC, Masters CL, Villemagne VL: Imaging β-amyloid burden in aging and dementia. Neurology. 2007, 68: 1718-1725. 10.1212/01.wnl.0000261919.22630.ea.CrossRefPubMed
27.
Zurück zum Zitat Clark CM, Schneider JA, Bedell BJ, Beach TG, Bilker WB, Mintun MA, Pontecorvo MJ, Hefti F, Carpenter AP, Flitter ML, Krautkramer MJ, Kung HF, Coleman RE, Doraiswamy PM, Fleisher AS, Sabbagh MN, Sadowsky CH, Reiman EP, Zehntner SP, Skovronsky DM, AV45-A07 Study Group: Use of florbetapir-PET for imaging β-amyloid pathology. JAMA. 2011, 305: 275-283. 10.1001/jama.2010.2008.CrossRefPubMed Clark CM, Schneider JA, Bedell BJ, Beach TG, Bilker WB, Mintun MA, Pontecorvo MJ, Hefti F, Carpenter AP, Flitter ML, Krautkramer MJ, Kung HF, Coleman RE, Doraiswamy PM, Fleisher AS, Sabbagh MN, Sadowsky CH, Reiman EP, Zehntner SP, Skovronsky DM, AV45-A07 Study Group: Use of florbetapir-PET for imaging β-amyloid pathology. JAMA. 2011, 305: 275-283. 10.1001/jama.2010.2008.CrossRefPubMed
28.
Zurück zum Zitat Vandenberghe R, Van Laere K, Ivanoiu A, Salmon E, Bastin C, Triau E, Hasselbalch S, Law I, Andersen A, Korner A, Minthon L, Garraux G, Nelissen N, Bormans G, Buckley C, Owenius R, Thurfjell L, Farrar G, Brooks DJ: 18F-flutemetamol amyloid imaging in Alzheimer disease and mild cognitive impairment: a phase 2 trial. Ann Neurol. 2010, 68: 319-329. 10.1002/ana.22068.CrossRefPubMed Vandenberghe R, Van Laere K, Ivanoiu A, Salmon E, Bastin C, Triau E, Hasselbalch S, Law I, Andersen A, Korner A, Minthon L, Garraux G, Nelissen N, Bormans G, Buckley C, Owenius R, Thurfjell L, Farrar G, Brooks DJ: 18F-flutemetamol amyloid imaging in Alzheimer disease and mild cognitive impairment: a phase 2 trial. Ann Neurol. 2010, 68: 319-329. 10.1002/ana.22068.CrossRefPubMed
29.
Zurück zum Zitat Fodero-Tavoletti MT, Okamura N, Furumoto S, Mulligan RS, Connor AR, McLean CA, Cao D, Rigopoulos A, Cartwright GA, O’Keefe G, Gong S, Adlard PA, Barnham KJ, Rowe CC, Masters CL, Kudo Y, Cappai R, Yanai K, Villemagne VL: 18F-THK523: a novel in vivo tau imaging ligand for Alzheimer’s disease. Brain. 2011, 134: 1089-1100. 10.1093/brain/awr038.CrossRefPubMed Fodero-Tavoletti MT, Okamura N, Furumoto S, Mulligan RS, Connor AR, McLean CA, Cao D, Rigopoulos A, Cartwright GA, O’Keefe G, Gong S, Adlard PA, Barnham KJ, Rowe CC, Masters CL, Kudo Y, Cappai R, Yanai K, Villemagne VL: 18F-THK523: a novel in vivo tau imaging ligand for Alzheimer’s disease. Brain. 2011, 134: 1089-1100. 10.1093/brain/awr038.CrossRefPubMed
30.
Zurück zum Zitat Harada R, Okamura N, Furumoto S, Tago T, Maruyama M, Higuchi M, Yoshikawa T, Arai H, Iwata R, Kudo Y, Yanai K: Comparison of the binding characteristics of [18F]THK-523 and other amyloid imaging tracers to Alzheimer’s disease pathology. Eur J Nucl Med Mol Imaging. 2013, 40: 125-132. 10.1007/s00259-012-2261-2.CrossRefPubMed Harada R, Okamura N, Furumoto S, Tago T, Maruyama M, Higuchi M, Yoshikawa T, Arai H, Iwata R, Kudo Y, Yanai K: Comparison of the binding characteristics of [18F]THK-523 and other amyloid imaging tracers to Alzheimer’s disease pathology. Eur J Nucl Med Mol Imaging. 2013, 40: 125-132. 10.1007/s00259-012-2261-2.CrossRefPubMed
31.
Zurück zum Zitat Okamura N, Suemoto T, Furumoto S, Suzuki M, Shimadzu H, Akatsu H, Yamamoto T, Fujiwara H, Nemoto M, Maruyama M, Arai H, Yanai K, Sawada T, Kudo Y: Quinoline and benzimidazole derivatives: candidate probes for in vivo imaging of tau pathology in Alzheimer’s disease. J Neurosci. 2005, 25: 10857-10862. 10.1523/JNEUROSCI.1738-05.2005.CrossRefPubMed Okamura N, Suemoto T, Furumoto S, Suzuki M, Shimadzu H, Akatsu H, Yamamoto T, Fujiwara H, Nemoto M, Maruyama M, Arai H, Yanai K, Sawada T, Kudo Y: Quinoline and benzimidazole derivatives: candidate probes for in vivo imaging of tau pathology in Alzheimer’s disease. J Neurosci. 2005, 25: 10857-10862. 10.1523/JNEUROSCI.1738-05.2005.CrossRefPubMed
32.
Zurück zum Zitat Villemagne VL, Furumoto S, Fodero-Tavoletti MT, Mulligan RS, Hodges J, Harada R, Yates P, Piguet O, Pejoska S, Doré V, Yanai K, Masters CL, Kudo Y, Rowe CC, Okamura N: In vivo evaluation of a novel tau imaging tracer for Alzheimer's disease. Eur J Nucl Med Mol Imaging. 2014, [Epub ahead of print]. doi:10.1007/s00259-013-2681-7 Villemagne VL, Furumoto S, Fodero-Tavoletti MT, Mulligan RS, Hodges J, Harada R, Yates P, Piguet O, Pejoska S, Doré V, Yanai K, Masters CL, Kudo Y, Rowe CC, Okamura N: In vivo evaluation of a novel tau imaging tracer for Alzheimer's disease. Eur J Nucl Med Mol Imaging. 2014, [Epub ahead of print]. doi:10.1007/s00259-013-2681-7
33.
Zurück zum Zitat Hauw JJ, Daniel SE, Dickson D, Horoupian DS, Jellinger K, Lantos PL, McKee A, Tabaton M, Litvan I: Preliminary NINDS neuropathologic criteria for Steele-Richardson-Olszewski syndrome (progressive supranuclear palsy). Neurology. 1994, 44: 2015-2019. 10.1212/WNL.44.11.2015.CrossRefPubMed Hauw JJ, Daniel SE, Dickson D, Horoupian DS, Jellinger K, Lantos PL, McKee A, Tabaton M, Litvan I: Preliminary NINDS neuropathologic criteria for Steele-Richardson-Olszewski syndrome (progressive supranuclear palsy). Neurology. 1994, 44: 2015-2019. 10.1212/WNL.44.11.2015.CrossRefPubMed
34.
Zurück zum Zitat Lowe J: Part 7: Frontotemporal Lobar Degeneration and Amyotrophic Lateral Sclerosis/Motor Neuron Disease. Chapter 40: Introduction. Neurodegeneration: The Molecular Pathology of Dementia and Movement Disorders. Edited by: Dickson D, Weller RO. 2011, Oxford, UK: Wiley-Blackwell, 389-390. 2 Lowe J: Part 7: Frontotemporal Lobar Degeneration and Amyotrophic Lateral Sclerosis/Motor Neuron Disease. Chapter 40: Introduction. Neurodegeneration: The Molecular Pathology of Dementia and Movement Disorders. Edited by: Dickson D, Weller RO. 2011, Oxford, UK: Wiley-Blackwell, 389-390. 2
35.
Zurück zum Zitat Culvenor JG, McLean CA, Cutt S, Campbell BC, Maher F, Jäkälä P, Hartmann T, Beyreuther K, Masters CL, Li QX: Non-Aβ component of Alzheimer’s disease amyloid (NAC) revisited: NAC and α-synuclein are not associated with Aβ amyloid. Am J Pathol. 1999, 155: 1173-1181. 10.1016/S0002-9440(10)65220-0.PubMedCentralCrossRefPubMed Culvenor JG, McLean CA, Cutt S, Campbell BC, Maher F, Jäkälä P, Hartmann T, Beyreuther K, Masters CL, Li QX: Non-Aβ component of Alzheimer’s disease amyloid (NAC) revisited: NAC and α-synuclein are not associated with Aβ amyloid. Am J Pathol. 1999, 155: 1173-1181. 10.1016/S0002-9440(10)65220-0.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Culvenor JG, Henry A, Hartmann T, Evin G, Galatis D, Friedhuber A, Jayasena UL, Underwood JR, Beyreuther K, Masters CL, Cappai R: Subcellular localization of the Alzheimer’s disease amyloid precursor protein and derived polypeptides expressed in a recombinant yeast system. Amyloid. 1998, 5: 79-89. 10.3109/13506129808995285.CrossRefPubMed Culvenor JG, Henry A, Hartmann T, Evin G, Galatis D, Friedhuber A, Jayasena UL, Underwood JR, Beyreuther K, Masters CL, Cappai R: Subcellular localization of the Alzheimer’s disease amyloid precursor protein and derived polypeptides expressed in a recombinant yeast system. Amyloid. 1998, 5: 79-89. 10.3109/13506129808995285.CrossRefPubMed
37.
Zurück zum Zitat Villemagne V, Fodero-Tavoletti M, Furumoto S, Mulligan RS, Hodges J, Piguet O, Pejoska S, Kudo Y, Masters C, Yanai K, Rowe C, Okamura N: In vivo tau imaging in Alzheimer’s disease and other dementias. Alzheimers Dement. 2012, 8: 699-CrossRef Villemagne V, Fodero-Tavoletti M, Furumoto S, Mulligan RS, Hodges J, Piguet O, Pejoska S, Kudo Y, Masters C, Yanai K, Rowe C, Okamura N: In vivo tau imaging in Alzheimer’s disease and other dementias. Alzheimers Dement. 2012, 8: 699-CrossRef
38.
Zurück zum Zitat Villemagne VL, Fodero-Tavoletti MT, Pike KE, Cappai R, Masters CL, Rowe CC: The ART of loss: Aβ imaging in the evaluation of Alzheimer’s disease and other dementias. Mol Neurobiol. 2008, 38: 1-15. 10.1007/s12035-008-8019-y.CrossRefPubMed Villemagne VL, Fodero-Tavoletti MT, Pike KE, Cappai R, Masters CL, Rowe CC: The ART of loss: Aβ imaging in the evaluation of Alzheimer’s disease and other dementias. Mol Neurobiol. 2008, 38: 1-15. 10.1007/s12035-008-8019-y.CrossRefPubMed
39.
Zurück zum Zitat Rowe CC, Ackerman U, Browne W, Mulligan R, Pike KL, O’Keefe G, Tochon-Danguy H, Chan G, Berlangieri SU, Jones G, Dickinson-Rowe KL, Kung HP, Zhang W, Kung MP, Skovronsky D, Dyrks T, Holl G, Krause S, Friebe M, Lehman L, Lindemann S, Dinkelborg LM, Masters CL, Villemagne VL: Imaging of amyloid β in Alzheimer’s disease with 18F-BAY94-9172, a novel PET tracer: proof of mechanism. Lancet Neurol. 2008, 7: 129-135. 10.1016/S1474-4422(08)70001-2.CrossRefPubMed Rowe CC, Ackerman U, Browne W, Mulligan R, Pike KL, O’Keefe G, Tochon-Danguy H, Chan G, Berlangieri SU, Jones G, Dickinson-Rowe KL, Kung HP, Zhang W, Kung MP, Skovronsky D, Dyrks T, Holl G, Krause S, Friebe M, Lehman L, Lindemann S, Dinkelborg LM, Masters CL, Villemagne VL: Imaging of amyloid β in Alzheimer’s disease with 18F-BAY94-9172, a novel PET tracer: proof of mechanism. Lancet Neurol. 2008, 7: 129-135. 10.1016/S1474-4422(08)70001-2.CrossRefPubMed
40.
Zurück zum Zitat Villemagne VL, Pike KE, Chételat G, Ellis KA, Mulligan RS, Bourgeat P, Ackermann U, Jones G, Szoeke C, Salvado O, Martins R, O’Keefe G, Mathis CA, Klunk WE, Ames D, Masters CL, Rowe CC: Longitudinal assessment of Aβ and cognition in aging and Alzheimer disease. Ann Neurol. 2011, 69: 181-192. 10.1002/ana.22248.PubMedCentralCrossRefPubMed Villemagne VL, Pike KE, Chételat G, Ellis KA, Mulligan RS, Bourgeat P, Ackermann U, Jones G, Szoeke C, Salvado O, Martins R, O’Keefe G, Mathis CA, Klunk WE, Ames D, Masters CL, Rowe CC: Longitudinal assessment of Aβ and cognition in aging and Alzheimer disease. Ann Neurol. 2011, 69: 181-192. 10.1002/ana.22248.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Good CD, Johnsrude IS, Ashburner J, Henson RN, Friston KJ, Frackowiak RS: A voxel-based morphometric study of ageing in 465 normal adult human brains. Neuroimage. 2001, 14: 21-36. 10.1006/nimg.2001.0786.CrossRefPubMed Good CD, Johnsrude IS, Ashburner J, Henson RN, Friston KJ, Frackowiak RS: A voxel-based morphometric study of ageing in 465 normal adult human brains. Neuroimage. 2001, 14: 21-36. 10.1006/nimg.2001.0786.CrossRefPubMed
42.
Zurück zum Zitat Sergeant N, Bretteville A, Hamdane M, Caillet-Boudin ML, Grognet P, Bombois S, Blum D, Delacourte A, Pasquier F, Vanmechelen E, Schraen-Maschke S, Buée L: Biochemistry of Tau in Alzheimer’s disease and related neurological disorders. Expert Rev Proteomics. 2008, 5: 207-224. 10.1586/14789450.5.2.207.CrossRefPubMed Sergeant N, Bretteville A, Hamdane M, Caillet-Boudin ML, Grognet P, Bombois S, Blum D, Delacourte A, Pasquier F, Vanmechelen E, Schraen-Maschke S, Buée L: Biochemistry of Tau in Alzheimer’s disease and related neurological disorders. Expert Rev Proteomics. 2008, 5: 207-224. 10.1586/14789450.5.2.207.CrossRefPubMed
43.
Zurück zum Zitat Williams DR, Holton JL, Strand C, Pittman A, de Silva R, Lees AJ, Revesz T: Pathological tau burden and distribution distinguishes progressive supranuclear palsy-parkinsonism from Richardson’s syndrome. Brain. 2007, 130: 1566-1576. 10.1093/brain/awm104.CrossRefPubMed Williams DR, Holton JL, Strand C, Pittman A, de Silva R, Lees AJ, Revesz T: Pathological tau burden and distribution distinguishes progressive supranuclear palsy-parkinsonism from Richardson’s syndrome. Brain. 2007, 130: 1566-1576. 10.1093/brain/awm104.CrossRefPubMed
44.
Zurück zum Zitat Yamada T, McGeer PL, McGeer EG: Appearance of paired nucleated, Tau-positive glia in patients with progressive supranuclear palsy brain tissue. Neurosci Lett. 1992, 135: 99-102. 10.1016/0304-3940(92)90145-W.CrossRefPubMed Yamada T, McGeer PL, McGeer EG: Appearance of paired nucleated, Tau-positive glia in patients with progressive supranuclear palsy brain tissue. Neurosci Lett. 1992, 135: 99-102. 10.1016/0304-3940(92)90145-W.CrossRefPubMed
45.
Zurück zum Zitat Dickson DW, Bergeron C, Chin SS, Duyckaerts C, Horoupian D, Ikeda K, Jellinger K, Lantos PL, Lippa CF, Mirra SS, Tabaton M, Vonsattel JP, Wakabayashi K, Litvan I, Office of Rare Diseases of the National Institutes of Health: Office of Rare Diseases neuropathologic criteria for corticobasal degeneration. J Neuropathol Exp Neurol. 2002, 61: 935-946.PubMed Dickson DW, Bergeron C, Chin SS, Duyckaerts C, Horoupian D, Ikeda K, Jellinger K, Lantos PL, Lippa CF, Mirra SS, Tabaton M, Vonsattel JP, Wakabayashi K, Litvan I, Office of Rare Diseases of the National Institutes of Health: Office of Rare Diseases neuropathologic criteria for corticobasal degeneration. J Neuropathol Exp Neurol. 2002, 61: 935-946.PubMed
46.
Zurück zum Zitat Buée L, Delacourte A: Comparative biochemistry of tau in progressive supranuclear palsy, corticobasal degeneration, FTDP-17 and Pick’s disease. Brain Pathol. 1999, 9: 681-693. 10.1111/j.1750-3639.1999.tb00550.x.CrossRefPubMed Buée L, Delacourte A: Comparative biochemistry of tau in progressive supranuclear palsy, corticobasal degeneration, FTDP-17 and Pick’s disease. Brain Pathol. 1999, 9: 681-693. 10.1111/j.1750-3639.1999.tb00550.x.CrossRefPubMed
47.
Zurück zum Zitat Arima K: Ultrastructural characteristics of tau filaments in tauopathies: immuno-electron microscopic demonstration of tau filaments in tauopathies. Neuropathology. 2006, 26: 475-483. 10.1111/j.1440-1789.2006.00669.x.CrossRefPubMed Arima K: Ultrastructural characteristics of tau filaments in tauopathies: immuno-electron microscopic demonstration of tau filaments in tauopathies. Neuropathology. 2006, 26: 475-483. 10.1111/j.1440-1789.2006.00669.x.CrossRefPubMed
48.
Zurück zum Zitat Maruyama M, Shimada H, Suhara T, Shinotoh H, Ji B, Maeda J, Zhang MR, Trojanowski JQ, Lee VM, Ono M, Masamoto K, Takano H, Sahara N, Iwata N, Okamura N, Furumoto S, Kudo Y, Chang Q, Saido TC, Takashima A, Lewis J, Jang MK, Aoki I, Ito H, Higuchi M: Imaging of tau pathology in a tauopathy mouse model and in Alzheimer patients compared to normal controls. Neuron. 2013, 79: 1094-1108. 10.1016/j.neuron.2013.07.037.CrossRefPubMed Maruyama M, Shimada H, Suhara T, Shinotoh H, Ji B, Maeda J, Zhang MR, Trojanowski JQ, Lee VM, Ono M, Masamoto K, Takano H, Sahara N, Iwata N, Okamura N, Furumoto S, Kudo Y, Chang Q, Saido TC, Takashima A, Lewis J, Jang MK, Aoki I, Ito H, Higuchi M: Imaging of tau pathology in a tauopathy mouse model and in Alzheimer patients compared to normal controls. Neuron. 2013, 79: 1094-1108. 10.1016/j.neuron.2013.07.037.CrossRefPubMed
49.
Zurück zum Zitat Crowther RA: Straight and paired helical filaments in Alzheimer disease have a common structural unit. Proc Natl Acad Sci U S A. 1991, 88: 2288-2292. 10.1073/pnas.88.6.2288.PubMedCentralCrossRefPubMed Crowther RA: Straight and paired helical filaments in Alzheimer disease have a common structural unit. Proc Natl Acad Sci U S A. 1991, 88: 2288-2292. 10.1073/pnas.88.6.2288.PubMedCentralCrossRefPubMed
50.
Zurück zum Zitat Bulic B, Pickhardt M, Mandelkow EM, Mandelkow E: Tau protein and tau aggregation inhibitors. Neuropharmacology. 2010, 59: 276-289. 10.1016/j.neuropharm.2010.01.016.CrossRefPubMed Bulic B, Pickhardt M, Mandelkow EM, Mandelkow E: Tau protein and tau aggregation inhibitors. Neuropharmacology. 2010, 59: 276-289. 10.1016/j.neuropharm.2010.01.016.CrossRefPubMed
51.
Zurück zum Zitat Agdeppa ED, Kepe V, Liu J, Flores-Torres S, Satyamurthy N, Petric A, Cole GM, Small GW, Huang SC, Barrio JR: Binding characteristics of radiofluorinated 6-dialkylamino-2-naphthylethylidene derivatives as positron emission tomography imaging probes for β-amyloid plaques in Alzheimer’s disease. J Neurosci. 2001, 21: RC189-PubMed Agdeppa ED, Kepe V, Liu J, Flores-Torres S, Satyamurthy N, Petric A, Cole GM, Small GW, Huang SC, Barrio JR: Binding characteristics of radiofluorinated 6-dialkylamino-2-naphthylethylidene derivatives as positron emission tomography imaging probes for β-amyloid plaques in Alzheimer’s disease. J Neurosci. 2001, 21: RC189-PubMed
52.
Zurück zum Zitat Ikonomovic MD, Klunk WE, Abrahamson EE, Mathis CA, Price JC, Tsopelas ND, Lopresti BJ, Ziolko S, Bi W, Paljug WR, Debnath ML, Hope CE, Isanski BA, Hamilton RL, DeKosky ST: Post-mortem correlates of in vivo PiB-PET amyloid imaging in a typical case of Alzheimer’s disease. Brain. 2008, 131: 1630-1645. 10.1093/brain/awn016.PubMedCentralCrossRefPubMed Ikonomovic MD, Klunk WE, Abrahamson EE, Mathis CA, Price JC, Tsopelas ND, Lopresti BJ, Ziolko S, Bi W, Paljug WR, Debnath ML, Hope CE, Isanski BA, Hamilton RL, DeKosky ST: Post-mortem correlates of in vivo PiB-PET amyloid imaging in a typical case of Alzheimer’s disease. Brain. 2008, 131: 1630-1645. 10.1093/brain/awn016.PubMedCentralCrossRefPubMed
Metadaten
Titel
Assessing THK523 selectivity for tau deposits in Alzheimer’s disease and non–Alzheimer’s disease tauopathies
verfasst von
Michelle T Fodero-Tavoletti
Shozo Furumoto
Leanne Taylor
Catriona A McLean
Rachel S Mulligan
Ian Birchall
Ryuichi Harada
Colin L Masters
Kazuhiko Yanai
Yukitsuka Kudo
Christopher C Rowe
Nobuyuki Okamura
Victor L Villemagne
Publikationsdatum
01.02.2014
Verlag
BioMed Central
Erschienen in
Alzheimer's Research & Therapy / Ausgabe 1/2014
Elektronische ISSN: 1758-9193
DOI
https://doi.org/10.1186/alzrt240

Weitere Artikel der Ausgabe 1/2014

Alzheimer's Research & Therapy 1/2014 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Schutz der Synapsen bei Alzheimer

29.05.2024 Morbus Alzheimer Nachrichten

Mit einem Neurotrophin-Rezeptor-Modulator lässt sich möglicherweise eine bestehende Alzheimerdemenz etwas abschwächen: Erste Phase-2-Daten deuten auf einen verbesserten Synapsenschutz.

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Update Neurologie

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