scispace - formally typeset
Search or ask a question

Showing papers by "John Q. Trojanowski published in 2011"


Journal ArticleDOI
Adam C. Naj1, Gyungah Jun2, Gary W. Beecham1, Li-San Wang3  +153 moreInstitutions (38)
TL;DR: The Alzheimer Disease Genetics Consortium performed a genome-wide association study of late-onset Alzheimer disease using a three-stage design consisting of a discovery stage (stage 1), two replication stages (stages 2 and 3), and both joint analysis and meta-analysis approaches were used.
Abstract: The Alzheimer Disease Genetics Consortium (ADGC) performed a genome-wide association study of late-onset Alzheimer disease using a three-stage design consisting of a discovery stage (stage 1) and two replication stages (stages 2 and 3). Both joint analysis and meta-analysis approaches were used. We obtained genome-wide significant results at MS4A4A (rs4938933; stages 1 and 2, meta-analysis P (P(M)) = 1.7 × 10(-9), joint analysis P (P(J)) = 1.7 × 10(-9); stages 1, 2 and 3, P(M) = 8.2 × 10(-12)), CD2AP (rs9349407; stages 1, 2 and 3, P(M) = 8.6 × 10(-9)), EPHA1 (rs11767557; stages 1, 2 and 3, P(M) = 6.0 × 10(-10)) and CD33 (rs3865444; stages 1, 2 and 3, P(M) = 1.6 × 10(-9)). We also replicated previous associations at CR1 (rs6701713; P(M) = 4.6 × 10(-10), P(J) = 5.2 × 10(-11)), CLU (rs1532278; P(M) = 8.3 × 10(-8), P(J) = 1.9 × 10(-8)), BIN1 (rs7561528; P(M) = 4.0 × 10(-14), P(J) = 5.2 × 10(-14)) and PICALM (rs561655; P(M) = 7.0 × 10(-11), P(J) = 1.0 × 10(-10)), but not at EXOC3L2, to late-onset Alzheimer's disease susceptibility.

1,743 citations


Journal ArticleDOI
TL;DR: The Parkinson Progression Marker Initiative (PPMI) is a comprehensive observational, international, multi-center study designed to identify PD progression biomarkers both to improve understanding of disease etiology and course and to provide crucial tools to enhance the likelihood of success of PD modifying therapeutic trials.

1,269 citations


Journal ArticleDOI
06 Oct 2011-Neuron
TL;DR: It is demonstrated that preformed fibrils generated from full-length and truncated recombinant α-syn enter primary neurons, probably by adsorptive-mediated endocytosis, and promote recruitment of soluble endogenousα-syn into insoluble PD-like LBs and LNs.

1,248 citations


Journal ArticleDOI
09 Apr 2011-Brain
TL;DR: Integration of information across disciplines should enhance opportunities for the translation of neuroplasticity and circuit retraining research into effective clinical therapies.
Abstract: Neuroplasticity can be defined as the ability of the nervous system to respond to intrinsic or extrinsic stimuli by reorganizing its structure, function and connections. Major advances in the understanding of neuroplasticity have to date yielded few established interventions. To advance the translation of neuroplasticity research towards clinical applications, the National Institutes of Health Blueprint for Neuroscience Research sponsored a workshop in 2009. Basic and clinical researchers in disciplines from central nervous system injury/stroke, mental/addictive disorders, paediatric/developmental disorders and neurodegeneration/ageing identified cardinal examples of neuroplasticity, underlying mechanisms, therapeutic implications and common denominators. Promising therapies that may enhance training-induced cognitive and motor learning, such as brain stimulation and neuropharmacological interventions, were identified, along with questions of how best to use this body of information to reduce human disability. Improved understanding of adaptive mechanisms at every level, from molecules to synapses, to networks, to behaviour, can be gained from iterative collaborations between basic and clinical researchers. Lessons can be gleaned from studying fields related to plasticity, such as development, critical periods, learning and response to disease. Improved means of assessing neuroplasticity in humans, including biomarkers for predicting and monitoring treatment response, are needed. Neuroplasticity occurs with many variations, in many forms, and in many contexts. However, common themes in plasticity that emerge across diverse central nervous system conditions include experience dependence, time sensitivity and the importance of motivation and attention. Integration of information across disciplines should enhance opportunities for the translation of neuroplasticity and circuit retraining research into effective clinical therapies.

907 citations


Journal ArticleDOI
TL;DR: In this article, Sampathu et al. proposed a new classification system for FTLD-TDP pathology, which is a single harmonized system that replaces the two currently in use.
Abstract: In 2006, two papers were published, each describing pathological heterogeneity in cases of frontotemporal lobar degeneration (FTLD) with ubiquitin-positive, tau-negative inclusions (FTLD-U) [7, 11]. In both studies, large series of cases were evaluated and the investigators felt that they could recognize three distinct histological patterns, based on the morphology and anatomical distribution of ubiquitin immunoreactive neuronal inclusions. The findings of Sampathu et al. were further supported by differential labelling of the pathology, using a panel of novel monoclonal antibodies; whereas, Mackenzie et al. found relatively specific clinicopathological correlations. Most importantly, the pathological features that defined the subtypes in these two studies were almost identical, providing powerful validation of the results. However, because the studies were conducted simultaneously and independently, the numbering of the subtypes, used in the respective papers, did not match (Table 1). Table 1 Proposed new classification system for FTLD-TDP pathology, compared with existing systems Shortly thereafter, further work by one of the two groups led to the identification of the transactive response DNA-binding protein with Mr 43 kD (TDP-43) as the ubiquitinated pathological protein in most cases of FTLD-U as well as the majority of sporadic amyotrophic lateral sclerosis (ALS) and some familial ALS [10]. It was subsequently confirmed that most FTLD-U cases had TDP-43 pathology and that the same pathological patterns could be recognized based on the results of TDP-43 immunohistochemistry (IHC) [1, 2]. By this time, a fourth FTLD-U subtype had been described, specifically associated with the familial syndrome of inclusion body myopathy with Paget’s disease of bone and frontotemporal dementia (IBMPFD) caused by mutations in the valosin-containing protein (VCP) gene [4], and this was also shown to have TDP-43 pathology [9]. As a result, cases of FTLD with TDP-43 pathology are now designated as FTLD-TDP and the term FTLD-U is no longer recommended [8]. The two classification systems for FTLD-U/FTLD-TDP have now gained wide acceptance and have repeatedly been validated by the discovery of additional clinical, genetic and pathological correlations. However, the continued use of two discordant numbering systems proves to be an ongoing source of confusion within the field. Previous attempts, by other groups of authors, to promote one classification over the other have not been successful. To resolve this issue, the principal authors of the original two papers are now proposing a new classification for FTLD-TDP pathology, the sole purpose of which is to provide a single harmonized system that replaces the two currently in use. In developing this new classification, the following principles were adhered to: (1) different pathological subtypes are designated by letters to help distinguish this from the pre-existing number-based systems, (2) the order of subtypes should not exactly match either of the previous systems to avoid any apparent bias, and (3) the order of the subtypes should be based on their relative frequency, with “A” being the most common. The result is summarized in Table 1. Type A is equivalent to type 1 of Mackenzie et al. and type 3 of Sampathu et al., being characterized by numerous short dystrophic neurites (DN) and crescentic or oval neuronal cytoplasmic inclusions (NCI), concentrated primarily in neocortical layer 2. Moderate numbers of lentiform neuronal intranuclear inclusions (NII) are also a common but inconsistent feature of this subtype. Type B matches Mackenzie et al. type 3 and Sampathu et al. type 2, with moderate numbers of NCI, throughout all cortical layers, but very few DN. Type C is the same as Mackenzie et al. type 2 and Sampathu et al. type 1, having a predominance of elongated DN in upper cortical layers, with very few NCI. Finally, Type D refers to the pathology associated with IBMPFD caused by VCP mutations, characterized by numerous short DN and frequent lentiform NII. Based on the results of more recent studies, there are a number of other modifications that we could have considered incorporating into this new system. Additional pathological subtypes could be added; for instance, to describe the TDP-43 pathology that is found in the mesial temporal lobe in a high proportion of cases of Alzheimer’s disease and most other common neurodegenerative conditions [3]. The pathological criteria for each of the subtypes could be expanded to include characteristic findings in subcortical regions [5, 6]. The description of the pathological features could be modified to take into account the greater sensitivity and specificity of TDP-43 IHC, which may demonstrate additional findings, not recognized with the ubiquitin immunostaining techniques upon which the original classifications were based (such as neuronal “pre-inclusions”) [2]. Although these and other recent findings represent important advances in our understanding of FTLD-TDP, most have not yet been broadly replicated or completely defined. Therefore, in order to make the transition to a new classification as simple and widely acceptable as possible and, most importantly, to allow for direct translation with the currently existing systems, we are not proposing any other significant changes, beyond the coding of the subtypes. In summary, we believed that adoption of a single harmonized system for the classification of FTLD-TDP neuropathology would greatly improve communication within the rapidly advancing field of FTLD diagnosis and research. Future attempts to resolve any outstanding issues related to the practical implementation and interpretation of FTLD pathological classification should also benefit. As indicated by their inclusion as co-authors on this paper, this proposal has received the unanimous support of all of the neuropathologists involved in the original two studies [7, 11].

782 citations



Journal ArticleDOI
TL;DR: It is demonstrated that tau acetylation inhibits tau function via impaired tau-microtubule interactions and promotes pathological tau aggregation, and suggests tau K280 acetylations is a potential target for drug discovery and biomarker development for AD and related tauopathies.
Abstract: The microtubule associated protein tau promotes neuronal survival through binding and stabilization of MTs. Phosphorylation regulates tau-microtubule interactions and hyperphosphorylation contributes to the aberrant formation of insoluble tau aggregates in Alzheimer's disease (AD) and related tauopathies. However, other pathogenic post-translational tau modifications have not been well characterized. Here we demonstrate that tau acetylation inhibits tau function via impaired tau-microtubule interactions and promotes pathological tau aggregation. Mass spectrometry analysis identified specific lysine residues, including lysine 280 (K280) within the microtubule-binding motif as the major sites of tau acetylation. Immunohistochemical and biochemical studies of brains from tau transgenic mice and patients with AD and related tauopathies showed that acetylated tau pathology is specifically associated with insoluble, Thioflavin-positive tau aggregates. Thus, tau K280 acetylation in our studies was only detected in diseased tissue, suggesting it may have a role in pathological tau transformation. This study suggests that tau K280 acetylation is a potential target for drug discovery and biomarker development for AD and related tauopathies.

568 citations


Journal ArticleDOI
Günter U. Höglinger1, Nadine M. Melhem2, Dennis W. Dickson3, Patrick M. A. Sleiman4, Li-San Wang4, Lambertus Klei2, Rosa Rademakers3, Rohan de Silva5, Irene Litvan6, David E. Riley7, John C. van Swieten8, Peter Heutink9, Zbigniew K. Wszolek3, Ryan J. Uitti3, Jana Vandrovcova5, Howard I. Hurtig4, Rachel G. Gross4, Walter Maetzler10, Stefano Goldwurm, Eduardo Tolosa11, Barbara Borroni12, Pau Pastor13, Laura B. Cantwell4, Mi Ryung Han4, Allissa Dillman14, Marcel P. van der Brug15, J. Raphael Gibbs5, J. Raphael Gibbs14, Mark R. Cookson14, Dena G. Hernandez14, Dena G. Hernandez5, Andrew B. Singleton14, Matthew J. Farrer16, Chang En Yu17, Lawrence I. Golbe18, Tamas Revesz5, John Hardy5, Andrew J. Lees5, Bernie Devlin2, Hakon Hakonarson4, Ulrich Müller19, Gerard D. Schellenberg4, Roger L. Albin20, Elena Alonso13, Angelo Antonini, Manuela Apfelbacher21, Steven E. Arnold4, Jesús Avila22, Thomas G. Beach, Sherry Beecher4, Daniela Berg23, Thomas D. Bird, Nenad Bogdanovic24, Agnita J.W. Boon8, Yvette Bordelon25, Alexis Brice26, Alexis Brice27, Herbert Budka28, Margherita Canesi, Wang Zheng Chiu8, Roberto Cilia, Carlo Colosimo29, Peter Paul De Deyn30, Justo Garcãa De Yebenes, Laura Donker Kaat8, Ranjan Duara31, Alexandra Durr27, Alexandra Durr26, Sebastiaan Engelborghs30, Giovanni Fabbrini29, Nicole A. Finch3, Robyn Flook32, Matthew P. Frosch33, Carles Gaig11, Douglas Galasko34, Thomas Gasser23, Marla Gearing35, Evan T. Geller4, Bernardino Ghetti36, Neill R. Graff-Radford3, Murray Grossman4, Deborah A. Hall37, Lili-Naz Hazrati38, Matthias Höllerhage1, Joseph Jankovic39, Jorge L. Juncos35, Anna Karydas40, Hans A. Kretzschmar41, Isabelle Leber27, Isabelle Leber26, Virginia M.-Y. Lee4, Andrew P. Lieberman20, Kelly E. Lyons42, Claudio Mariani, Eliezer Masliah34, Luke A. Massey5, Catriona McLean43, Nicoletta Meucci, Bruce L. Miller40, Brit Mollenhauer44, Jens Carsten Möller1, Huw R. Morris45, Christopher Morris46, Sean S. O'Sullivan5, Wolfgang H. Oertel1, Donatella Ottaviani29, Alessandro Padovani12, Rajesh Pahwa42, Gianni Pezzoli, Stuart Pickering-Brown47, Werner Poewe48, Alberto Rábano49, Alex Rajput50, Stephen G. Reich51, Gesine Respondek1, Sigrun Roeber41, Jonathan D. Rohrer5, Owen A. Ross3, Martin N. Rossor5, Giorgio Sacilotto, William W. Seeley40, Klaus Seppi48, Laura Silveira-Moriyama5, Salvatore Spina36, Karin Srulijes23, Peter St George-Hyslop52, Maria Stamelou1, David G. Standaert53, Silvana Tesei, Wallace W. Tourtellotte54, Claudia Trenkwalder44, Claire Troakes55, John Q. Trojanowski4, Juan C. Troncoso56, Vivianna M. Van Deerlin4, Jean Paul G. Vonsattel57, Gregor K. Wenning48, Charles L. White58, Pia Winter19, Chris Zarow59, Anna Zecchinelli 
University of Marburg1, University of Pittsburgh2, Mayo Clinic3, University of Pennsylvania4, University College London5, University of Louisville6, Case Western Reserve University7, Erasmus University Rotterdam8, VU University Amsterdam9, University of Tübingen10, University of Barcelona11, University of Brescia12, University of Navarra13, National Institutes of Health14, Scripps Research Institute15, University of British Columbia16, University of Washington17, Rutgers University18, University of Giessen19, University of Michigan20, University of Würzburg21, Autonomous University of Madrid22, German Center for Neurodegenerative Diseases23, Karolinska Institutet24, University of California, Los Angeles25, French Institute of Health and Medical Research26, Centre national de la recherche scientifique27, Medical University of Vienna28, Sapienza University of Rome29, University of Antwerp30, Mount Sinai Hospital31, Flinders University32, Harvard University33, University of California, San Diego34, Emory University35, Indiana University36, Rush University Medical Center37, University of Toronto38, Baylor College of Medicine39, University of California, San Francisco40, Ludwig Maximilian University of Munich41, University of Kansas42, Mental Health Research Institute43, University of Göttingen44, Cardiff University45, Newcastle University46, University of Manchester47, Innsbruck Medical University48, Carlos III Health Institute49, University of Saskatchewan50, University of Maryland, Baltimore51, University of Cambridge52, University of Alabama at Birmingham53, Veterans Health Administration54, King's College London55, Johns Hopkins University56, Columbia University57, University of Texas Southwestern Medical Center58, University of Southern California59
TL;DR: Two independent variants in MAPT affecting risk for PSP are confirmed, one of which influences MAPT brain expression and the genes implicated encode proteins for vesicle-membrane fusion at the Golgi-endosomal interface and for a myelin structural component.
Abstract: Progressive supranuclear palsy (PSP) is a movement disorder with prominent tau neuropathology. Brain diseases with abnormal tau deposits are called tauopathies, the most common of which is Alzheimer's disease. Environmental causes of tauopathies include repetitive head trauma associated with some sports. To identify common genetic variation contributing to risk for tauopathies, we carried out a genome-wide association study of 1,114 individuals with PSP (cases) and 3,247 controls (stage 1) followed by a second stage in which we genotyped 1,051 cases and 3,560 controls for the stage 1 SNPs that yielded P ≤ 10−3. We found significant previously unidentified signals (P < 5 × 10−8) associated with PSP risk at STX6, EIF2AK3 and MOBP. We confirmed two independent variants in MAPT affecting risk for PSP, one of which influences MAPT brain expression. The genes implicated encode proteins for vesicle-membrane fusion at the Golgi-endosomal interface, for the endoplasmic reticulum unfolded protein response and for a myelin structural component.

504 citations


Journal ArticleDOI
TL;DR: In summary, both SPARE-AD and CSF biomarkers showed high baseline sensitivity, however, many MCI-NC had abnormal baseline Spatial Pattern of Abnormalities for Recognition of Early AD and CSFs, and longer follow-up will elucidate the specificity of baseline measurements.

502 citations


Journal ArticleDOI
Niklas Mattsson1, Ulf Andreasson1, Staffan Persson1, Hiroyuki Arai2, Sat Dev Batish, Sergio Bernardini3, Luisella Bocchio-Chiavetto, Marinus A. Blankenstein4, Maria Berrocal Carrillo5, Sonia Chalbot6, Els Coart7, Davide Chiasserini8, Neal Cutler, Gunilla Dahlfors9, Stefan Duller, Anne M. Fagan10, Orestes Vicente Forlenza11, Giovanni B. Frisoni, Douglas Galasko12, Daniela Galimberti13, Harald Hampel14, Aase Handberg15, Michael T. Heneka16, Adrianna Z. Herskovits17, Sanna-Kaisa Herukka18, David M. Holtzman10, Christian Humpel19, Bradley T. Hyman17, Khalid Iqbal6, Mathias Jucker20, Stephan A. Kaeser20, Elmar Kaiser21, Elisabeth Kapaki22, Daniel Kidd23, Péter Klivényi24, Cindy Soendersoe Knudsen15, Markus P. Kummer16, James Lui25, Albert Lladó, Piotr Lewczuk26, Qiao-Xin Li27, Ralph N. Martins25, Colin L. Masters27, John McAuliffe, Marc Mercken28, Abhay Moghekar29, José Luis Molinuevo, Thomas J. Montine30, William Nowatzke, Richard O'Brien29, Markus Otto31, George P. Paraskevas22, Lucilla Parnetti8, Ronald C. Petersen32, David Prvulovic14, Herman P M de Reus33, Robert A. Rissman12, Elio Scarpini13, Alessandro Stefani, Hilkka Soininen18, Johannes Schröder21, Leslie M. Shaw34, Anders Skinningsrud35, Brith Skrogstad35, Annette Spreer36, Leda Leme Talib11, Charlotte E. Teunissen4, John Q. Trojanowski34, Hayrettin Tumani31, Robert M. Umek, Bianca Van Broeck28, Hugo Vanderstichele7, László Vécsei24, Marcel M. Verbeek33, Manfred Windisch, Jing Zhang30, Henrik Zetterberg1, Kaj Blennow1 
TL;DR: The cerebrospinal fluid biomarkers amyloid β (Aβ)‐42, total‐Tau (T‐tau), and phosphorylated‐t Tau (P‐tAU) demonstrate good diagnostic accuracy for Alzheimer's disease (AD), but there are large variations in biomarker measurements between studies, and between and within laboratories.
Abstract: Background The cerebrospinal fluid (CSF) biomarkers amyloid β (Aβ)-42, total-tau (T-tau), and phosphorylated-tau (P-tau) demonstrate good diagnostic accuracy for Alzheimer’s disease (AD). However, there are large variations in biomarker measurements between studies, and between and within laboratories. The Alzheimer’s Association has initiated a global quality control program to estimate and monitor variability of measurements, quantify batch-to-batch assay variations, and identify sources of variability. In this article, we present the results from the first two rounds of the program.

375 citations


Journal ArticleDOI
TL;DR: It is proposed that aggregation-prone RNA-binding proteins might contribute very broadly to ALS pathogenesis and the genes identified in the yeast functional screen, coupled with prion-like domain prediction analysis, now provide a powerful resource to facilitate ALS disease gene discovery.
Abstract: Amyotrophic lateral sclerosis (ALS) is a devastating and universally fatal neurodegenerative disease. Mutations in two related RNA-binding proteins, TDP-43 and FUS, that harbor prion-like domains, cause some forms of ALS. There are at least 213 human proteins harboring RNA recognition motifs, including FUS and TDP-43, raising the possibility that additional RNA-binding proteins might contribute to ALS pathogenesis. We performed a systematic survey of these proteins to find additional candidates similar to TDP-43 and FUS, followed by bioinformatics to predict prion-like domains in a subset of them. We sequenced one of these genes, TAF15, in patients with ALS and identified missense variants, which were absent in a large number of healthy controls. These disease-associated variants of TAF15 caused formation of cytoplasmic foci when expressed in primary cultures of spinal cord neurons. Very similar to TDP-43 and FUS, TAF15 aggregated in vitro and conferred neurodegeneration in Drosophila, with the ALS-linked variants having a more severe effect than wild type. Immunohistochemistry of postmortem spinal cord tissue revealed mislocalization of TAF15 in motor neurons of patients with ALS. We propose that aggregation-prone RNA-binding proteins might contribute very broadly to ALS pathogenesis and the genes identified in our yeast functional screen, coupled with prion-like domain prediction analysis, now provide a powerful resource to facilitate ALS disease gene discovery.

Journal ArticleDOI
TL;DR: The data suggest that perturbation of endogenous nuclear TDP-43 results in loss of normal T DP-43 function(s) and gene regulatory pathways, culminating in degeneration of selectively vulnerable affected neurons.
Abstract: Fil: Muller Igaz, Lionel Ivan. University of Pennsylvania; Estados Unidos. Consejo Nacional de Investigaciones Cientificas y Tecnicas. Oficina de Coordinacion Administrativa Houssay; Argentina

Journal ArticleDOI
TL;DR: To characterize cognitive and behavioral features, physical findings, and brain atrophy patterns in pathology‐proven corticobasal degeneration (CBD) and corticOBasal syndrome (CBS) with known histopathology.
Abstract: Objective To characterize cognitive and behavioral features, physical findings and brain atrophy patterns in pathology-proven corticobasal degeneration (CBD) and corticobasal syndrome (CBS) with known histopathology.

Journal ArticleDOI
TL;DR: In this paper, a seven-center inter-laboratory standardization study for cerebrospinal fluid (CSF) total tau (t-tau), phospho-Tau (p-tAU(181), and Aβ(1-42) was conducted as part of the Alzheimer's Disease Neuroimaging Initiative (ADNI).
Abstract: The close correlation between abnormally low pre-mortem cerebrospinal fluid (CSF) concentrations of amyloid-β1-42 (Aβ(1-42)) and plaque burden measured by amyloid imaging as well as between pathologically increased levels of CSF tau and the extent of neurodegeneration measured by MRI has led to growing interest in using these biomarkers to predict the presence of AD plaque and tangle pathology. A challenge for the widespread use of these CSF biomarkers is the high variability in the assays used to measure these analytes which has been ascribed to multiple pre-analytical and analytical test performance factors. To address this challenge, we conducted a seven-center inter-laboratory standardization study for CSF total tau (t-tau), phospho-tau (p-tau(181)) and Aβ(1-42) as part of the Alzheimer's Disease Neuroimaging Initiative (ADNI). Aliquots prepared from five CSF pools assembled from multiple elderly controls (n = 3) and AD patients (n = 2) were the primary test samples analyzed in each of three analytical runs by the participating laboratories using a common batch of research use only immunoassay reagents (INNO-BIA AlzBio3, xMAP technology, from Innogenetics) on the Luminex analytical platform. To account for the combined effects on overall precision of CSF samples (fixed effect), different laboratories and analytical runs (random effects), these data were analyzed by mixed-effects modeling with the following results: within center %CV 95% CI values (mean) of 4.0-6.0% (5.3%) for CSF Aβ(1-42); 6.4-6.8% (6.7%) for t-tau and 5.5-18.0% (10.8%) for p-tau(181) and inter-center %CV 95% CI range of 15.9-19.8% (17.9%) for Aβ(1-42), 9.6-15.2% (13.1%) for t-tau and 11.3-18.2% (14.6%) for p-tau(181). Long-term experience by the ADNI biomarker core laboratory replicated this degree of within-center precision. Diagnostic threshold CSF concentrations for Aβ(1-42) and for the ratio t-tau/Aβ(1-42) were determined in an ADNI independent, autopsy-confirmed AD cohort from whom ante-mortem CSF was obtained, and a clinically defined group of cognitively normal controls (NCs) provides statistically significant separation of those who progressed from MCI to AD in the ADNI study. These data suggest that interrogation of ante-mortem CSF in cognitively impaired individuals to determine levels of t-tau, p-tau(181) and Aβ(1-42), together with MRI and amyloid imaging biomarkers, could replace autopsy confirmation of AD plaque and tangle pathology as the "gold standard" for the diagnosis of definite AD in the near future.

Journal ArticleDOI
TL;DR: PiB slightly outperformed FDG in patients with known histopathology, and was more sensitive when interpreted qualitatively or quantitatively when scans were classified quantitatively.
Abstract: Objective: To compare the diagnostic performance of PET with the amyloid ligand Pittsburgh compound B (PiB-PET) to fluorodeoxyglucose (FDG-PET) in discriminating between Alzheimer disease (AD) and frontotemporal lobar degeneration (FTLD). Methods: Patients meeting clinical criteria for AD (n 62) and FTLD (n 45) underwent PiB and FDG-PET. PiB scans were classified as positive or negative by 2 visual raters blinded to clinical diagnosis, and using a quantitative threshold derived from controls (n 25). FDG scans were visually rated as consistent with AD or FTLD, and quantitatively classified based on the region of lowest metabolism relative to controls. Results: PiB visual reads had a higher sensitivity for AD (89.5% average between raters) than FDG visual reads (77.5%) with similar specificity (PiB 83%, FDG 84%). When scans were classified quantitatively, PiB had higher sensitivity (89% vs 73%) while FDG had higher specificity (83% vs 98%). On receiver operating characteristic analysis, areas under the curve for PiB (0.888) and FDG (0.910) were similar. Interrater agreement was higher for PiB ( 0.96) than FDG ( 0.72), as was agreement between visual and quantitative classification (PiB 0.88– 0.92; FDG 0.64–0.68). In patients with known histopathology, overall classification accuracy (2 visual and 1 quantitative classification per patient) was 97% for PiB (n 12 patients) and 87% for FDG (n 10). Conclusions: PiB and FDG showed similar accuracy in discriminating AD and FTLD. PiB was more sensitive when interpreted qualitatively or quantitatively. FDG was more specific, but only when scans were classified quantitatively. PiB slightly outperformed FDG in patients with known histopathology. Neurology ® 2011;77:2034–2042

Journal ArticleDOI
TL;DR: A reduction in the CSF Aβ42 level denotes a pathophysiological process that significantly departs from normality (ie, becomes dynamic) early, whereas theCSF total tau level and the adjusted hippocampal volume are biomarkers of downstream pathophysiology processes.
Abstract: Objective To empirically assess the concept that Alzheimer disease (AD) biomarkers significantly depart from normality in a temporally ordered manner. Design Validation sample. Setting Multisite, referral centers. Participants A total of 401 elderly participants in the Alzheimer's Disease Neuroimaging Initiative who were cognitively normal, who had mild cognitive impairment, or who had AD dementia. We compared the proportions of 3 AD biomarker values (the Aβ42 level in cerebrospinal fluid [CSF], the total tau level in CSF, and the hippocampal volume adjusted for intracranial volume [hereafter referred to as the adjusted hippocampal volume]) that were abnormal as cognitive impairment worsened. Cut points demarcating normal vs abnormal for each biomarker were established by maximizing diagnostic accuracy in independent autopsy samples. Main Outcome Measures Three AD biomarkers (ie, the CSF Aβ42 level, the CSF total tau level, and the adjusted hippocampal volume). Results Within each clinical group of the entire sample (n = 401), the CSF Aβ42 level was abnormal more often than was the CSF total tau level or the adjusted hippocampal volume. Among the 298 participants with both baseline and 12-month data, the proportion of participants with an abnormal Aβ42 level did not change from baseline to 12 months in any group. The proportion of participants with an abnormal total tau level increased from baseline to 12 months in cognitively normal participants (P = .05) but not in participants with mild cognitive impairment or AD dementia. For 209 participants with an abnormal CSF Aβ42 level at baseline, the percentage with an abnormal adjusted hippocampal volume but normal CSF total tau level increased from baseline to 12 months in participants with mild cognitive impairment. No change in the percentage of MCI participants with an abnormal total tau level was seen between baseline and 12 months. Conclusions A reduction in the CSF Aβ42 level denotes a pathophysiological process that significantly departs from normality (ie, becomes dynamic) early, whereas the CSF total tau level and the adjusted hippocampal volume are biomarkers of downstream pathophysiological processes. The CSF total tau level becomes dynamic before the adjusted hippocampal volume, but the hippocampal volume is more dynamic in the clinically symptomatic mild cognitive impairment and AD dementia phases of the disease than is the CSF total tau level.

Journal ArticleDOI
TL;DR: The results implicate HPA dysregulation in AD neuropathogenesis and suggest that prolonged stress may increase Aβ and tau hyperphosphorylation.
Abstract: Because overactivation of the hypothalamic–pituitary–adrenal (HPA) axis occurs in Alzheimer's disease (AD), dysregulation of stress neuromediators may play a mechanistic role in the pathophysiology of AD. However, the effects of stress on tau phosphorylation are poorly understood, and the relationship between corticosterone and corticotropin-releasing factor (CRF) on both β-amyloid (Aβ) and tau pathology remain unclear. Therefore, we first established a model of chronic stress, which exacerbates Aβ accumulation in Tg2576 mice and then extended this stress paradigm to a tau transgenic mouse model with the P301S mutation (PS19) that displays tau hyperphosphorylation, insoluble tau inclusions and neurodegeneration. We show for the first time that both Tg2576 and PS19 mice demonstrate a heightened HPA stress profile in the unstressed state. In Tg2576 mice, 1 month of restraint/isolation (RI) stress increased Aβ levels, suppressed microglial activation, and worsened spatial and fear memory compared with nonstressed mice. In PS19 mice, RI stress promoted tau hyperphosphorylation, insoluble tau aggregation, neurodegeneration, and fear–memory impairments. These effects were not mimicked by chronic corticosterone administration but were prevented by pre-stress administration of a CRF receptor type 1 (CRF1) antagonist. The role for a CRF1-dependent mechanism was further supported by the finding that mice overexpressing CRF had increased hyperphosphorylated tau compared with wild-type littermates. Together, these results implicate HPA dysregulation in AD neuropathogenesis and suggest that prolonged stress may increase Aβ and tau hyperphosphorylation. These studies also implicate CRF in AD pathophysiology and suggest that pharmacological manipulation of this neuropeptide may be a potential therapeutic strategy for AD.

Journal ArticleDOI
TL;DR: Recent findings examining TDP-43 molecular functions are highlighted with particular emphasis on stress-mediated regulation of T DP-43 localization, putative downstream TSPs, target genes and RNAs, as well as TTP-43 interacting proteins, all of which represent viable points of therapeutic intervention for ALS, FTLD-TDP and related proteinopathies.

Journal ArticleDOI
TL;DR: In addition to known candidate genes, APOE, TOMM40, and one hypothetical gene LOC100129500 partially overlapping APOE; one novel gene, EPC2, and several other interesting genes were associated with CSF biomarkers that are related to AD.
Abstract: Objectives: CSF levels of Aβ 1-42 , t-tau, and p-tau 181p are potential early diagnostic markers for probable Alzheimer disease (AD). The influence of genetic variation on these markers has been investigated for candidate genes but not on a genome-wide basis. We report a genome-wide association study (GWAS) of CSF biomarkers (Aβ 1-42 , t-tau, p-tau 181p , p-tau 181p /Aβ 1-42 , and t-tau/Aβ 1-42 ). Methods: A total of 374 non-Hispanic Caucasian participants in the Alzheimer9s Disease Neuroimaging Initiative cohort with quality-controlled CSF and genotype data were included in this analysis. The main effect of single nucleotide polymorphisms (SNPs) under an additive genetic model was assessed on each of 5 CSF biomarkers. The p values of all SNPs for each CSF biomarker were adjusted for multiple comparisons by the Bonferroni method. We focused on SNPs with corrected p p −8 ) and secondarily examined SNPs with uncorrected p values less than 10 −5 to identify potential candidates. Results: Four SNPs in the regions of the APOE , LOC100129500, TOMM40 , and EPC2 genes reached genome-wide significance for associations with one or more CSF biomarkers. SNPs in CCDC134 , ABCG2 , SREBF2 , and NFATC4 , although not reaching genome-wide significance, were identified as potential candidates. Conclusions: In addition to known candidate genes, APOE , TOMM40 , and one hypothetical gene LOC100129500 partially overlapping APOE ; one novel gene, EPC2 , and several other interesting genes were associated with CSF biomarkers that are related to AD. These findings, especially the new EPC2 results, require replication in independent cohorts.

Journal ArticleDOI
TL;DR: A longitudinal study of complementary measures of Aβ pathology (PIB, CSF and plasma Aβ) and other biomarkers in a cohort with an extensive neuropsychological battery is significant because it shows that Aβ measurements have limited value for disease classification and modest value as prognostic factors over the 3-year follow-up as mentioned in this paper.
Abstract: Previous studies of Aβ plasma as a biomarker for Alzheimer’s disease (AD) obtained conflicting results We here included 715 subjects with baseline Aβ1-40 and Aβ1-42 plasma measurement (50% with 4 serial annual measurements): 205 cognitively normal controls (CN), 348 patients mild cognitive impairment (MCI) and 162 with AD We assessed the factors that modified their concentrations and correlated these values with PIB PET, MRI and tau and Aβ1-42 measures in cerebrospinal fluid (CSF) Association between Aβ and diagnosis (baseline and prospective) was assessed A number of health conditions were associated with altered concentrations of plasma Aβ The effect of age differed according to AD stage Plasma Aβ1-42 showed mild correlation with other biomarkers of Aβ pathology and were associated with infarctions in MRI Longitudinal measurements of Aβ1-40 and Aβ1-42 plasma levels showed modest value as a prognostic factor for clinical progression Our longitudinal study of complementary measures of Aβ pathology (PIB, CSF and plasma Aβ) and other biomarkers in a cohort with an extensive neuropsychological battery is significant because it shows that plasma Aβ measurements have limited value for disease classification and modest value as prognostic factors over the 3-year follow-up However, with longer follow-up, within subject plasma Aβ measurements could be used as a simple and minimally invasive screen to identify those at increased risk for AD Our study emphasizes the need for a better understanding of the biology and dynamics of plasma Aβ as well as the need for longer term studies to determine the clinical utility of measuring plasma Aβ

Journal ArticleDOI
TL;DR: The longitudinal patterns support a hypothetical sequence of AD pathology in which amyloid deposition is an early event before hypometabolism or hippocampal atrophy, suggesting that biomarker prediction for cognitive change is stage dependent.
Abstract: Objective To delineate the trajectories of Aβ42 level in cerebrospinal fluid (CSF), fludeoxyglucose F18 (FDG) uptake using positron emission tomography, and hippocampal volume using magnetic resonance imaging and their relative associations with cognitive change at different stages in aging and Alzheimer disease (AD). Design Cohort study. Setting The 59 study sites for the Alzheimer's Disease Neuroimaging Initiative. Participants A total of 819 participants 55 to 90 years of age with normal cognition, mild cognitive impairment, and AD who were followed up during the period from 2005 to 2007. Main Outcome Measures Rates of change in level of Aβ42 in CSF, FDG uptake, hippocampal volume, and the Alzheimer Disease's Assessment Scale–cognitive subscale score during up to 36 months of follow-up by diagnostic group as well as prediction of cognitive change by each biomarker. Results Reductions in the level of Aβ42 in CSF were numerically greater in participants with normal cognition than in participants with mild cognitive impairment or AD; whereas both glucose metabolic decline and hippocampal atrophy were significantly slower in participants with normal cognition than in participants with mild cognitive impairment or AD. Positive APOE4 status accelerated hippocampal atrophic changes in participants with mild cognitive impairment or AD, but did not modify rates of change in level of Aβ42 in CSF or FDG uptake. The Alzheimer Disease's Assessment Scale–cognitive subscale scores were related only to the baseline level of Aβ42 in CSF and the baseline FDG uptake in participants with normal cognition, which were about equally associated with change in FDG uptake and hippocampal volume in participants with mild cognitive impairment and best modeled by change in FDG uptake in participants with AD. Conclusion Trajectories of Aβ42 level in CSF, FDG uptake, and hippocampal volume vary across different cognitive stages. The longitudinal patterns support a hypothetical sequence of AD pathology in which amyloid deposition is an early event before hypometabolism or hippocampal atrophy, suggesting that biomarker prediction for cognitive change is stage dependent.

Journal ArticleDOI
TL;DR: It is shown that the activity of β-site APP cleaving enzyme-1 (BACE1), the rate-limiting enzyme for amyloid-β peptide production, is modulated by S1P in mouse neurons, suggesting that cellular S1p directly modulates BACE1 activity.
Abstract: Sphingosine kinase (SphK) 1 and 2 phosphorylate sphingosine to generate sphingosine-1-phosphate (S1P), a pluripotent lipophilic mediator implicated in a variety of cellular events. Here we show that the activity of β-site APP cleaving enzyme-1 (BACE1), the rate-limiting enzyme for amyloid-β peptide (Aβ) production, is modulated by S1P in mouse neurons. Treatment by SphK inhibitor, RNA interference knockdown of SphK, or overexpression of S1P degrading enzymes decreased BACE1 activity, which reduced Aβ production. S1P specifically bound to full-length BACE1 and increased its proteolytic activity, suggesting that cellular S1P directly modulates BACE1 activity. Notably, the relative activity of SphK2 was upregulated in the brains of patients with Alzheimer's disease. The unique modulatory effect of cellular S1P on BACE1 activity is a novel potential therapeutic target for Alzheimer's disease.

Journal ArticleDOI
TL;DR: The INNOTEST and INNO-BIA CSF platforms perform equally well in identifying individuals with underlying amyloid plaque pathology, however, differences in absolute values point to the need for assay-specific diagnostic cutoff values.
Abstract: Background Cerebrospinal fluid (CSF) biomarkers of Alzheimer disease (AD) are currently being considered for inclusion in revised diagnostic criteria for research and/or clinical purposes to increase the certainty of antemortem diagnosis. Objective To test whether CSF biomarker assays differ in their ability to identify true markers of underlying AD pathology (eg, amyloid plaques and/or neurofibrillary tangles) in living individuals. Design We compared the performances of the 2 most commonly used platforms, INNOTEST enzyme-linked immunosorbent assay and INNO-BIA AlzBio3, for measurement of CSF β-amyloid (Aβ) and tau proteins to identify the presence of amyloid plaques in a research cohort (n=103). Values obtained for CSF Aβ1-42, total tau, and phosphorylated tau 181 (p-tau 181 ) using the 2 assay platforms were compared with brain amyloid load as assessed by positron emission tomography using the amyloid imaging agent Pittsburgh compound B. Setting The Knight Alzheimer's Disease Research Center at Washington University in St Louis, Missouri. Subjects Research volunteers who were cognitively normal or had mild to moderate AD dementia. Results The 2 assay platforms yielded different (approximately 2- to 6-fold) absolute values for the various analytes, but relative values were highly correlated. The CSF Aβ1-42 correlated inversely and tau and p-tau 181 correlated positively with the amount of cortical Pittsburgh compound B binding, albeit to differing degrees. Both assays yielded similar patterns of CSF biomarker correlations with amyloid load. The ratios of total tau to Aβ1-42 and p-tau 181 to Aβ1-42 outperformed any single analyte, including Aβ1-42, in discriminating individuals with vs without cortical amyloid. Conclusions The INNOTEST and INNO-BIA CSF platforms perform equally well in identifying individuals with underlying amyloid plaque pathology. Differences in absolute values, however, point to the need for assay-specific diagnostic cutoff values.

Journal ArticleDOI
15 Jun 2011-PLOS ONE
TL;DR: Assessment of sensorimotor gating could be used to detect the earliest manifestations of tauopathies exemplified by prodromal AD, in which abnormal tau protein may play critical roles in the onset of neuronal dysfunctions.
Abstract: Tauopathies are neurodegenerative disorders characterized by the accumulation of abnormal tau protein leading to cognitive and/or motor dysfunction. To understand the relationship between tau pathology and behavioral impairments, we comprehensively assessed behavioral abnormalities in a mouse tauopathy model expressing the human P301S mutant tau protein in the early stage of disease to detect its initial neurological manifestations. Behavioral abnormalities, shown by open field test, elevated plus-maze test, hot plate test, Y-maze test, Barnes maze test, Morris water maze test, and/or contextual fear conditioning test, recapitulated the neurological deficits of human tauopathies with dementia. Furthermore, we discovered that prepulse inhibition (PPI), a marker of sensorimotor gating, was enhanced in these animals concomitantly with initial neuropathological changes in associated brain regions. This finding provides evidence that our tauopathy mouse model displays neurofunctional abnormalities in prodromal stages of disease, since enhancement of PPI is characteristic of amnestic mild cognitive impairment, a transitional stage between normal aging and dementia such as Alzheimer's disease (AD), in contrast with attenuated PPI in AD patients. Therefore, assessment of sensorimotor gating could be used to detect the earliest manifestations of tauopathies exemplified by prodromal AD, in which abnormal tau protein may play critical roles in the onset of neuronal dysfunctions.

Journal ArticleDOI
TL;DR: The HCI, alone or in combination with certain other biomarker measurements, has the potential to help characterize AD and predict subsequent rates of clinical decline and could be applied to a range of imaging modalities and voxel-based image-analysis algorithms.

Journal ArticleDOI
TL;DR: These data are the first to demonstrate the concordance of ¹⁸F-flutemetamol PET imaging with histopathology, supporting its sensitivity to detect amyloid and potential use in the study and detection of Alzheimer disease.
Abstract: Objective To determine the correspondence of in vivo quantitative estimates of brain uptake of fluorine 18–labeled flutemetamol with immunohistochemical estimates of amyloid levels in patients who underwent previous biopsy. Design Cross-sectional study of 18 F-flutemetamol positron emission tomography (PET) findings in patients with prior cortical biopsy specimen stained for the presence or absence of amyloid plaques. Setting University hospital. Patients Seven patients who previously had a prior right frontal cortical biopsy at the site of ventriculoperitoneal placement for presumed normal pressure hydrocephalus were recruited. Inclusion criteria included an adequate biopsy specimen for detection and quantification of β-amyloid pathology and age older than 50 years. Intervention All patients underwent an 18 F-flutemetamol PET scan. Main Outcome Measures Quantitative measures of 18 F-flutemetamol uptake (standardized uptake value ratio, a ratio of mean target cortex activity divided by that in a cerebellar reference region) were made at a location contralateral to the biopsy site and compared with estimates of amyloid load based on immunohistochemical and histological staining. Results There was complete agreement between visual reads of 18 F-flutemetamol PET scans (3 blinded readers with majority rule) and histology. A regression model, including time from biopsy as a covariate, demonstrated a significant relationship (P = .01) between 18 F-flutemetamol uptake and percentage of area of amyloid measured by a monoclonal antibody raised against amyloid (NAB228). Similar results were found with the amyloid-specific monoclonal antibody 4G8 and Thioflavin S. Conclusion To our knowledge, these data are the first to demonstrate the concordance of 18 F-flutemetamol PET imaging with histopathology, supporting its sensitivity to detect amyloid and potential use in the study and detection of Alzheimer disease.

Journal ArticleDOI
TL;DR: Several MT-stabilizing compounds from the taxane and epothilone natural product families were examined to assess their membrane permeability and to determine whether they act as substrates or inhibitors of P-glycoprotein, and whether brain-penetrant compounds could stabilize mouse CNS MTs.

Journal ArticleDOI
TL;DR: A large number of plasma‐based biomarkers were sought for CI in PD and the timing of onset nor the severity of cognitive symptoms can be accurately predicted.
Abstract: Objective Most people with Parkinson disease (PD) eventually develop cognitive impairment (CI). However, neither the timing of onset nor the severity of cognitive symptoms can be accurately predicted. We sought plasma-based biomarkers for CI in PD. Methods A discovery cohort of 70 PD patients was recruited. Cognitive status was evaluated with the Mattis Dementia Rating Scale-2 (DRS) at baseline and on annual follow-up visits, and baseline plasma levels of 102 proteins were determined with a bead-based immunoassay. Using linear regression, we identified biomarkers of CI in PD, that is, proteins whose levels correlated with cognitive performance at baseline and/or cognitive decline at follow-up. We then replicated the association between cognitive performance and levels of the top biomarker, using a different technical platform, with a separate cohort of 113 PD patients. Results Eleven proteins exhibited plasma levels correlating with baseline cognitive performance in the discovery cohort. The best candidate was epidermal growth factor (EGF, p < 0.001); many of the other 10 analytes covaried with EGF across samples. Low levels of EGF not only correlated with poor cognitive test scores at baseline, but also predicted an 8-fold greater risk of cognitive decline to dementia-range DRS scores at follow-up for those with intact baseline cognition. A weaker, but still significant, relationship between plasma EGF levels and cognitive performance was found in an independent replication cohort of 113 PD patients. Interpretation Our data suggest that plasma EGF may be a biomarker for progression to CI in PD. Ann Neurol 2010

Journal ArticleDOI
01 May 2011-Brain
TL;DR: Investigating how a measure of educational and occupational attainment, a component of cognitive reserve, modifies the relationship between biomarkers of pathology and cognition in Alzheimer's disease found that in cognitively normal subjects, none of the biomarkers correlated with the measures of cognition, whereas American National Adult Reading Test scores were significantly correlated with Boston naming and mini-mental state examination results.
Abstract: The objective of this study was to investigate how a measure of educational and occupational attainment, a component of cognitive reserve, modifies the relationship between biomarkers of pathology and cognition in Alzheimer's disease. The biomarkers evaluated quantified neurodegeneration via atrophy on magnetic resonance images, neuronal injury via cerebral spinal fluid t-tau, brain amyloid-β load via cerebral spinal fluid amyloid-β1–42 and vascular disease via white matter hyperintensities on T2/proton density magnetic resonance images. We included 109 cognitively normal subjects, 192 amnestic patients with mild cognitive impairment and 98 patients with Alzheimer's disease, from the Alzheimer's Disease Neuroimaging Initiative study, who had undergone baseline lumbar puncture and magnetic resonance imaging. We combined patients with mild cognitive impairment and Alzheimer's disease in a group labelled ‘cognitively impaired’ subjects. Structural Abnormality Index scores, which reflect the degree of Alzheimer's disease-like anatomic features on magnetic resonance images, were computed for each subject. We assessed Alzheimer's Disease Assessment Scale (cognitive behaviour section) and mini-mental state examination scores as measures of general cognition and Auditory–Verbal Learning Test delayed recall, Boston naming and Trails B scores as measures of specific domains in both groups of subjects. The number of errors on the American National Adult Reading Test was used as a measure of environmental enrichment provided by educational and occupational attainment, a component of cognitive reserve. We found that in cognitively normal subjects, none of the biomarkers correlated with the measures of cognition, whereas American National Adult Reading Test scores were significantly correlated with Boston naming and mini-mental state examination results. In cognitively impaired subjects, the American National Adult Reading Test and all biomarkers of neuronal pathology and amyloid load were independently correlated with all cognitive measures. Exceptions to this general conclusion were absence of correlation between cerebral spinal fluid amyloid-β1–42 and Boston naming and Trails B. In contrast, white matter hyperintensities were only correlated with Boston naming and Trails B results in the cognitively impaired. When all subjects were included in a flexible ordinal regression model that allowed for non-linear effects and interactions, we found that the American National Adult Reading Test had an independent additive association such that better performance was associated with better cognitive performance across the biomarker distribution. Our main conclusions included: (i) that in cognitively normal subjects, the variability in cognitive performance is explained partly by the American National Adult Reading Test and not by biomarkers of Alzheimer's disease pathology; (ii) in cognitively impaired subjects, the American National Adult Reading Test, biomarkers of neuronal pathology (structural magnetic resonance imaging and cerebral spinal fluid t-tau) and amyloid load (cerebral spinal fluid amyloid-β1–42) all independently explain variability in general cognitive performance; and (iii) that the association between cognition and the American National Adult Reading Test was found to be additive rather than to interact with biomarkers of Alzheimer's disease pathology.

Journal ArticleDOI
TL;DR: The data corroborate the utility of TSPO-PET imaging as a biomarker for tau-triggered toxicity, and as a complement to amyloid scans for diagnostic assessment of tauopathies with and without Aβ pathologies.
Abstract: Core pathologies of Alzheimer's disease (AD) are aggregated amyloid-β peptides (Aβ) and tau, and the latter is also characteristic of diverse neurodegenerative tauopathies. These amyloid lesions provoke microglial activation, and recent neuroimaging technologies have enabled visualization of this response in living brains using radioligands for the peripheral benzodiazepine receptor also known as the 18 kDa translocator protein (TSPO). Here, we elucidated contributions of Aβ and tau deposits to in vivo TSPO signals in pursuit of mechanistic and diagnostic significance of TSPO imaging in AD and other tauopathies. A new antibody to human TSPO revealed induction of TSPO-positive microgliosis by tau fibrils in tauopathy brains. Emergence of TSPO signals before occurrence of brain atrophy and thioflavin-S-positive tau amyloidosis was also demonstrated in living mice transgenic for mutant tau by positron emission tomography (PET) with two classes of TSPO radioligands, [(11)C]AC-5216 and [(18)F]fluoroethoxy-DAA1106. Meanwhile, only modest TSPO elevation was observed in aged mice modeling Aβ plaque deposition, despite the notably enhanced in vivo binding of amyloid radiotracer, [(11)C]Pittsburgh Compound-B, to plaques. In these animals, [(11)C]AC-5216 yielded better TSPO contrasts than [(18)F]fluoroethoxy-DAA1106, supporting the possibility of capturing early neurotoxicity with high-performance TSPO probes. Furthermore, an additional line of mice modeling intraneuronal Aβ accumulation displayed elevated TSPO signals following noticeable neuronal loss, unlike TSPO upregulation heralding massive neuronal death in tauopathy model mice. Our data corroborate the utility of TSPO-PET imaging as a biomarker for tau-triggered toxicity, and as a complement to amyloid scans for diagnostic assessment of tauopathies with and without Aβ pathologies.