Skip to main content
Erschienen in: Acta Neuropathologica 5/2017

Open Access 12.01.2017 | Original Paper

Tau interactome mapping based identification of Otub1 as Tau deubiquitinase involved in accumulation of pathological Tau forms in vitro and in vivo

verfasst von: Peng Wang, Gerard Joberty, Arjan Buist, Alexandre Vanoosthuyse, Ilie-Cosmin Stancu, Bruno Vasconcelos, Nathalie Pierrot, Maria Faelth-Savitski, Pascal Kienlen-Campard, Jean-Noël Octave, Marcus Bantscheff, Gerard Drewes, Diederik Moechars, Ilse Dewachter

Erschienen in: Acta Neuropathologica | Ausgabe 5/2017

Abstract

Dysregulated proteostasis is a key feature of a variety of neurodegenerative disorders. In Alzheimer’s disease (AD), progression of symptoms closely correlates with spatiotemporal progression of Tau aggregation, with “early” oligomeric Tau forms rather than mature neurofibrillary tangles (NFTs) considered to be pathogenetic culprits. The ubiquitin–proteasome system (UPS) controls degradation of soluble normal and abnormally folded cytosolic proteins. The UPS is affected in AD and is identified by genomewide association study (GWAS) as a risk pathway for AD. The UPS is determined by balanced regulation of ubiquitination and deubiquitination. In this work, we performed isobaric tags for relative and absolute quantitation (iTRAQ)-based Tau interactome mapping to gain unbiased insight into Tau pathophysiology and to identify novel Tau-directed therapeutic targets. Focusing on Tau deubiquitination, we here identify Otub1 as a Tau-deubiquitinating enzyme. Otub1 directly affected Lys48-linked Tau deubiquitination, impairing Tau degradation, dependent on its catalytically active cysteine, but independent of its noncanonical pathway modulated by its N-terminal domain in primary neurons. Otub1 strongly increased AT8-positive Tau and oligomeric Tau forms and increased Tau-seeded Tau aggregation in primary neurons. Finally, we demonstrated that expression of Otub1 but not its catalytically inactive form induced pathological Tau forms after 2 months in Tau transgenic mice in vivo, including AT8-positive Tau and oligomeric Tau forms. Taken together, we here identified Otub1 as a Tau deubiquitinase in vitro and in vivo, involved in formation of pathological Tau forms, including small soluble oligomeric forms. Otub1 and particularly Otub1 inhibitors, currently under development for cancer therapies, may therefore yield interesting novel therapeutic avenues for Tauopathies and AD.

Introduction

Alzheimer’s disease (AD) is the most prevalent form of dementia, being characterized by progressive accumulation of amyloid plaques and neurofibrillary tangles, composed of aggregated Aβ and hyperphosphorylated Tau, respectively [9, 69]. Over the last decade, Tau has emerged as an important therapeutic target. This is underscored by (i) the close correlation of Tau pathology with the progression of symptoms and (ii) the existence of a family of Tauopathies which are neurodegenerative disorders characterized by Tau aggregation spatiotemporally correlating with brain dysfunction and associated symptoms, as well as, most importantly, (iii) the identification of mutations in Tau causally linked to Tauopathies, demonstrating a causal executive role of Tau in Tauopathies. Although Tau aggregation is closely linked to symptom progression, smaller oligomeric Tau forms rather than mature full-blown neurofibrillary tangles (NFTs) are generally considered to be pathogenetic culprits in the disease process [6, 11, 12, 15, 20, 33, 37, 39, 40, 59, 60, 67, 70]. Accumulation of misfolded and aggregated proteins is a key feature of a variety of neurodegenerative disorders, raising interest in understanding clearance and degradation mechanisms of these pathogenetic culprits [7, 22, 28, 35, 42, 43]. The UPS selectively degrades normal and abnormally folded soluble proteins, which are tagged by ubiquitin for elimination [22], while the autophagic–lysosomal system (ALS) mainly degrades large protein aggregates or inclusions and organelles. With smaller soluble Tau forms considered to be toxic culprits, the UPS represents an important target for therapeutic interventions and is the main focus of this work.
The UPS is reported to be downregulated in various neurodegenerative disorders, with increased proteasome activity shown to be beneficial in related disease models [15, 28, 42, 52]. The presence of ubiquitinated Tau in pathological lesions of AD provided the first lead implicating the UPS in AD and Tauopathies [49, 50]. In AD patients’ brain, paired helical filaments (PHFs) were found to be associated with impaired proteasome activity in a brain-region specific way [15, 28, 34, 35]. Furthermore, a ubiquitin mutant called UBB+1, with a 19-AA extension, has been identified in neurons from AD patients and suppresses UPS function [38, 45, 71]. UBB+1-induced proteasome dysfunction resulted in Tau pathology and related neuronal dysfunction in transgenic mice expressing UBB+1 [29]. Accumulating evidence indicates that the UPS is implicated in clearance of different Tau forms and its dysfunction is closely related to Tau aggregation and accumulation [1, 1316, 25, 46, 57, 68]. Conversely, Tau accumulation was recently shown to impair proteasomal degradation, suggesting a vicious circle [52]. Degradation of target proteins through the UPS is regulated by ubiquitination, with differently linked (Lys6, Lys11, Lys48, and Lys63) polyubiquitinated Tau identified in AD brains and models [13, 4951, 56]. Different Tau forms can be ubiquitinated by E3 ligases CHIP [57, 63], TRAF6 [1], and MARCH7 [19], with particularly CHIP/Hsp70-dependent polyubiquitination characterized in detail in vitro and in vivo. Ubiquitination is highly dynamic and reversible by the action of deubiquitinating enzymes known as deubiquitinases (DUBs), hence being determined by balanced regulation between ubiquitination and deubiquitination. While Tau ubiquitination has been studied intensively, its deubiquitination remains less well explored.
Deubiquitinases are attracting increasing interest as therapeutic targets. Otub1 is an ovarian-tumor domain cysteine protease deubiquitinating enzyme [2] with strong preference for Lys48-linked polyubiquitin chains over Lys11-, Lys29-, or Lys63-linked polyubiquitin chains in the canonical pathway, dependent on its highly conserved active-site cysteine (C91) [17, 48, 74]. Recently, a noncanonical pathway of Otub1, involved in DNA double-strand break response by modulating Lys63 polyubiquitin chain formation, was highlighted [31, 53, 77, 78]. Otub1 has been implicated in a broad range of cellular pathways such as IL-1β-induced inflammation and stabilization of proteins including c-IAP1, p53, TRAF3/6, SMAD2/3, and RhoA, indicating that it is an important regulator in different physiological processes [23, 26, 32, 44, 66], while its role in the nervous system remains unknown.
In this study, we performed iTRAQ-based Tau interactome mapping to identify Tau-interacting proteins with Tau-modifying potential as therapeutic targets. This highlighted Otub1 as a potential entry point to better understand the link between the UPS and Tauopathies. We demonstrate here for the first time that Tau is deubiquitinated by Otub1. We demonstrate that Otub1 regulates levels of Lys48-linked ubiquitin-conjugated Tau and increases pathological forms of Tau in vitro and in vivo.

Materials and methods

Animals

TauP301S (PS19) mice [81] were bred and used in our laboratory as reported previously [64, 65, 72]. The mice bred in our laboratory develop Tau pathology and a neurodegenerative phenotype starting at around ~10–11 months. In this study, stereotactic injections were performed at P0 and age-matched littermates were used for analysis 2 months postinjection. At the age of 2 months, no Tau pathology or AT8-positive staining is detected in TauP301S mice in our colony. All experiments were performed in compliance with protocols approved by the UCLouvain Ethical Committee for Animal Welfare.

Reagents and antibodies

MG132 and cycloheximide were purchased from Sigma. Phosphatase inhibitor (PhosSTOP™) and protease inhibitor cocktails (cOmplete™, Mini, EDTA-free) were obtained from Roche. Primary antibodies used in this study included antibodies directed against human Tau (Dako), Otub1, Ubiquitin-Lys48-specific, Ubiquitin-Lys63-specific (Abcam), Phospho-Tau (Ser202/Thr205) AT8, Phospho-Tau (Thr231) AT180, Phospho-Tau (Thr212/Ser214) AT100 (Thermo Fisher Scientific), and Oligomeric-Tau T22 (Merck), applied in combination with appropriate horseradish peroxidase (HRP) or Alexa Fluor® (488/568/647) coupled secondary antibodies.

Cell culture and transfection

Human kidney-derived QBI-293 were originally obtained from QBiogene (Carlsbad, CA, USA) and HEK293 cells from American Type Culture Collection (ATCC), and were cultured according to the manufacturer’s protocol. All cells were maintained at 37 °C in humidified atmosphere with 5% CO2. Cells were transfected with plasmids expressing Otub1, USP9x, USP5 or empty vector using FuGENE® 6 (Roche) or short interfering RNAs (siRNAs) using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s protocol. For analysis of the effects on Tau or on Tau-seeded Tau aggregation, transfection was performed 24 h before starting the assay.

In vivo gene delivery

Adeno-associated viral (AAV) vectors expressing wild-type Otub1 fused with a green fluorescent protein (GFP) tag (AAV–Otub1) or expressing GFP (AAV–GFP) driven by neuron-specific Syn promoter were generated using a previously described plasmid (Addgene, plasmid #26976). Mutant Otub1 C91A and N-terminal truncation (N-T) were generated by molecular cloning starting from the WT-Otub1 construct. AAV viruses were produced in HEK-AAV cells using AAV-DJ8 kit (Cell Biolabs, Inc.), and subsequently purified and concentrated by iodixanol-based ultracentrifugation. AAV titer was tested using a QuickTiter™ AAV Quantitation Kit (Cell Biolabs, Inc). For P0 injection, each mouse was injected into the lateral ventricles of both cerebral hemispheres with 4.2 × 109 total viral particles per side, TauP301S transgenic mice were euthanized, and brains were dissected as described previously following transcardial flushing and analyzed at 2 months postinjection [64, 65, 72].

Immunoprecipitation and Western blot analysis

To detect Tau ubiquitination, primary neurons were infected with AAV–Otub1 (wild type; catalytically dead mutant C91A; N-terminal truncation) or AAV–GFP, five days after infection, cells were washed with phosphate-buffered saline (PBS) and lysed in TGN lysis buffer (50 mM Tris HCl, pH 7.5, 200 mM NaCl, 50 mM sodium β-glycerophosphate, 1% Tween 20, 0.2% NP40) containing phosphatase inhibitor (PhosSTOP™; Roche) and protease inhibitor cocktails (cOmplete™, Mini, EDTA-free; Roche) at 4 °C for 30 min on a wheel rotor, and spun at 12,000×g for 15 min. Following preclearing without antibodies at 4 °C for 1 h, the supernatants were incubated with specific antibodies for 1 h at 4 °C, followed by incubation with protein A-Sepharose beads at 4 °C for 45 min. Following stringent washing with TGN buffer and PBS, immunoprecipitated proteins were analyzed by immunoblotting.
For Western blot analysis, cells were washed twice with PBS and extracted for 30 min at 4 °C with Triton lysis buffer (1% Triton X-100, 50 mM Tris, 150 mM NaCl, pH 7.6) containing protease and phosphatase inhibitors, and centrifuged at 12,000×g for 15 min at 4 °C to remove insoluble material. Protein content was determined by BCA Protein Assay kit (Thermo Fisher Scientific, Waltham, MA, USA). Samples (10 μg) were separated using precast 8% Tris–glycine gels or 4–12% Bis–Tris gels (MOPS running; Invitrogen) and transferred to polyvinylidene difluoride membranes. Immunoblotting was performed using the indicated primary antibodies with corresponding secondary antibodies, and developed using ECL kit (PerkinElmer, Waltham, MA, USA).

Tau aggregation assay

Tau PFFs (synthetic preformed fibrils), referred to as “Tau seeds,” were generated as described previously [24, 65, 72]. Truncated human Tau fragments bearing a proaggregation mutant (Tau-P301L) containing the four-repeat domain [K18; Q244-E372 (4RTau)], N-terminally Myc-tagged were produced in Escherichia coli (TEBU-Bio). Tau-PFFs were obtained by incubation of Tau fragments (66 μM) at 37 °C for 5 days in presence of heparin (133 μM) in 100 mM ammonium acetate buffer (pH 7.0), spun down (100,000×g, 1 h, 4 °C), resuspended to 333 μM, and sonicated before use.
In vitro Tau aggregation assay in HEK293 cells: PFF-induced Tau aggregation in vitro was performed essentially as described previously [24, 65, 72]. Sonicated Tau seeds were diluted in 100 mM ammonium acetate buffer (pH 7.0) to 10 µM solution and sonicated with 8 pulses/30% amplitude and added to the cells using BioPORTER® (AMS Biotechnology, Milton, UK) according to previously described protocol. To detect Tau aggregation, cells were fixed with 4% paraformaldehyde (PFA) containing 2% sucrose and 1% Triton X-100 for 15 min to remove soluble proteins. After washing with PBS, aggregated Tau-GFP was analyzed microscopically. To detect the effect of Otub1, USP5, and USP9x overexpression or knockdown on Tau aggregation, stably Tau-expressing QBI-293 cells were transfected with plasmid and empty vector or siRNA in 24-well plates. At 24 h after transfection, the growth medium was replaced with OptiMEM, and sonicated Tau seeds (10 μM) were added to BioPORTER® single-use tubes and added to cells. Three days after seeding, Tau-GFP aggregation was measured by microscopy.
In vitro Tau aggregation assay in primary neurons was performed as described previously [65]. Tau seeds (10 nM) were added at DIV3 and DIV6 to primary cortical neuronal cultures (PNC) from P0 heterozygous TauP301S pups. To detect the effect of Otub1 on Tau aggregation in primary neurons, AAV–Otub1 and AAV–GFP infections were carried out at DIV3, Tau seeds (10 nM) were added at DIV6 and DIV9, and cells were fixed at DIV12 with 4% PFA containing 2% sucrose and 1% Triton X-100 for 15 min to remove soluble proteins. After washing with PBS, aggregated Tau-GFP was detected under microscope.

Immunofluorescence microscopy assay

At indicated times after infection, cells were washed by phosphate-buffered saline (PBS) for three times and fixed with 4% paraformaldehyde in PBS for 15 min at room temperature (RT). For stringent extraction to detect aggregated Tau, 1% Triton X-100 (4% PFA, 2% sucrose) was used. Cells were briefly permeabilized in PBS/0.2% Triton X-100, then blocked in blocking solution (PBS containing 10% fetal calf serum and 0.1% Triton X-100). Primary and secondary antibody incubations were performed in blocking solution overnight at 4 °C or 1 h at RT using the indicated antibodies and goat anti-mouse IgG1 or goat anti-rabbit IgG1 secondary antibody coupled to Alexa® 488, Alexa® 568 or Alexa® 647. Cells were visualized using a digital inverted fluorescence microscope (EVOS-xl auto microscope).

Tau interactome mapping

Total mouse brain homogenates were used for coimmunoprecipitation in combination with three different polyclonal anti-Tau antibodies and control antibodies that were cross-linked to Sepharose beads. Mass spectrometry procedures were performed essentially as previously reported [4]. A detailed description of the Tau interactome mapping procedure is provided in the Supplemental Experimental Procedures.

Results

Tau interactome mapping identifies Tau-interacting proteins in mouse brain, including Otub1 as a potential Tau modifier

To identify Tau-interacting proteins with Tau-modifying potential, we performed Tau interactome mapping of endogenous Tau in mouse brain (Fig. 1) using a proteomic approach [3, 5]. Hereto, iTRAQ-based quantitative mass spectrometry was used in combination with differential coimmunoprecipitation of endogenous Tau from mouse brain. The use of isobaric isotope labels allows side-by-side quantitative comparison of up to four different samples [76] (Fig. 1a). Four Tau immunoprecipitation experiments were performed using three different anti-Tau antibodies and two different control non-Tau-capturing antibodies for differential analysis (Fig. 1a). Statistical tools were applied to score proteins for selective coprecipitation with Tau. Retrieval of several well-characterized Tau-interacting proteins validated the experimental approach (Fig. 1b). These included several tubulin isoforms [41, 75, 79], different components of the dynactin complex [47], enzymes modifying Tau phosphorylation, and members of the heat shock protein family, i.e., Hsc70 (Hspa8), Hsp70 (Hspa4), and Hsp110 (Hsp110) [18, 30, 57]. In addition, we identified many new putative Tau interactors, based on their significant enrichment in anti-Tau immunoprecipitation compared with control immunoprecipitations (Tables S1, S2). The full Tau interactome is presented in Supplementary Tables S1–S4. Gene ontology and STRING analysis were performed (Fig. S1; Table S3), and candidate interactors grouped according to their function (Fig. 1c; Table S4) to yield new insights into the pathophysiological role of Tau. Within this project, we focused on proteins involved in regulation of Tau ubiquitination and its degradation by the ubiquitin proteasome system, generally considered to be involved in degradation of small soluble proteins. We focused particularly on Tau deubiquitination and identified three major candidate deubiquitinating enzymes, including Otub1, USP5, and USP9x, with Otub1 as the strongest Tau interactor according to statistical analysis.

Otub1 increases Tau stability and increases Tau aggregation in a well-characterized cellular Tau aggregation model

To analyze whether Tau-interacting proteins, identified by the Tau interactome mapping, exerted modifying effects on Tau aggregation, we used a well-validated cellular Tau seeding model [24, 65, 72]. We analyzed the effect of the three identified candidate DUBs on Tau aggregation in vitro. In this assay, QBI cells stably expressing the longest human Tau isoform (2N4R), with the aggregation-prone P301L mutation and GFP tagged, were used in combination with preformed preaggregated synthetic Tau seeds consisting of Tau fragments harboring P301L mutant (myc-K18/P301L) to induce monomeric Tau aggregation. In this assay, addition of Tau seeds induces robust aggregation of monomeric human Tau, as assessed by cytological analysis following permeabilization with 1% Triton X-100 to remove soluble forms of Tau while retaining aggregated hyperphosphorylated Tau (Fig. 2a), as previously demonstrated and confirmed biochemically (Fig. S2) [65, 72]. Expression of the candidate deubiquitinases Otub1, USP5, and USP9x in this assay demonstrated significantly increased Tau aggregation following expression of Otub1 (Fig. 2a; Fig. S3), not observed following expression of USP5 or USP9x (Fig. S3). Next, we knocked down Otub1 in the Tau seeding assay, using two siRNAs of Otub1 with different knockdown efficiency. Endogenous Otub1 depletion significantly inhibited Tau aggregation, with the level of inhibition correlating with the level of knockdown efficiency, while use of a control siRNA did not affect levels of Otub1 nor aggregation efficiency (Fig. 2b). Combined, these data indicate that Otub1 affects Tau aggregation in a well-characterized Tau-seeded Tau aggregation model.
Since Otub1 is a deubiquitinase, known to regulate the stability of many crucial proteins in different cellular processes [23, 26, 32, 44, 66], we assessed whether Otub1 affected Tau stability by measuring Tau half-life following addition of the protein synthesis inhibitor cycloheximide (CHX). Knockdown of Otub1 increased Tau turnover, correlating with the decrease in expression of Otub1 obtained by the different siRNAs (Fig. 2c). Taken together, our data indicate that Otub1, a deubiquitinating enzyme, is a novel positive regulator of Tau aggregation and Tau stability in vitro, in a nonneuronal cell line.

Otub1 increases total Tau concentration and Tau phosphorylation in primary neurons

We next analyzed the modulatory effect of Otub1 on Tau in primary neurons, as relevant cell type for the study of Tauopathies and AD. Hereto, primary cortical neurons, derived from TauP301S transgenic pups (PS19) [65, 81], were infected with AAV–Otub1–GFP or AAV–GFP driving neuron-specific expression. Immunocytological analysis with AT8 antibody against phosphorylated Tau (Ser202/Thr205) demonstrated that Otub1 expression drastically increased phospho-Tau (p-Tau Ser202/Thr205) in primary neurons compared with GFP expression (Fig. 3a). Biochemical analysis confirmed a sharp increase of phosphorylated Tau at Ser202/Thr205 in Otub1-infected primary neurons (Fig. 3b). Otub1 also significantly increased total Tau protein level compared with GFP control (Fig. 3b). This increase was, however, less robust than the increase in AT8-positive Tau, as reflected by the ratio of AT8/total Tau. Taken together, we conclude that overexpressed Otub1 in primary neurons expressing TauP301S increased total Tau and induced a robust increase in AT8-positive Tau.

Otub1 enhances Tau oligomerization and Tau-seeded Tau aggregation in primary neurons

We next analyzed the effect of Otub1 in a validated Tau aggregation assay in primary neurons [65, 81]. Addition of Tau seeds allows bypass of the lag phase of Tau aggregation, resulting in recruitment of soluble Tau into insoluble Tau aggregates, reflected in a robust detergent-resistant AT8 (phosphorylated Tau) signal throughout soma and neurites (Fig. 4a, Fig. S2a). We used this well-characterized assay to analyze a potential effect of Otub1 on Tau aggregation in TauP301S cortical primary neurons using AAV-driven expression of Otub1 or GFP for control. Otub1 expression strikingly enhanced detergent-resistant AT8-positive Tau accumulation compared with GFP-infected neurons (Fig. 4b), indicating that Otub1 increased Tau-seeded Tau aggregation in primary neurons, corroborating the results obtained in a nonneuronal cell line.
To measure the effect of Otub1 expression on oligomeric Tau forms, we performed Western blotting analysis in nonreducing conditions, following AAV-driven expression of Otub1 and GFP, in absence of Tau seeding. This revealed a significant increase of oligomeric Tau forms in AAV–Otub1-infected neurons compared with AAV–GFP-infected neurons (Fig. 4c). In addition, we found robust induction of a (homo- or heterotypic) Tau complex (Tauc), which was strongly detected in AAV–Otub1-infected neurons but absent in AAV–GFP-infected neurons, indicating clearcut Otub1-induced modulation of Tau in neurons. The induction of oligomeric Tau forms was further confirmed using dot-blot analysis with T22 antibody (Fig. 4d), revealing a significant increase in oligomeric Tau following expression of Otub1.
Taken together, our data demonstrate that Otub1, identified as a Tau-interacting protein in the Tau interactome mapping, modulates Tau, by increasing Tau-seeded Tau aggregation, by formation of a Otub1-induced Tau complex Tauc, and by increasing the concentration of oligomeric Tau forms in primary neurons.

Otub1 but not Otub1-C91A impairs Tau degradation by removing Lys48-linked polyubiquitin chains from Tau in primary neurons

Previous studies have shown that Otub1 has dual functions in regulating both ubiquitin assembly and disassembly [77, 78]. Otub1, as an isopeptidase, can remove Lys48-linked polyubiquitin chains from its substrates [17, 48, 74], but can also serve as an E2 enzyme inhibitor to impede Lys63-specific polyubiquitin chain formation in a noncanonical manner [61]. Since Otub1 interacts with Tau and promotes phosphorylated and aggregated Tau levels in primary neurons, we hypothesized that Otub1 could act as a Tau deubiquitinase, interfering with pathological Tau degradation and hence Tau aggregation. To test this assumption experimentally, we analyzed the ubiquitination status of Tau by immunoprecipitation. Following AAV-driven expression of Otub1 or GFP in primary neurons, Tau proteins were pulled down, and Lys48- and Lys63-linked polyubiquitination of Tau was measured by linkage-specific ubiquitin antibodies. To enrich ubiquitinated proteins, proteasome inhibitor was added to the medium 6 h before harvesting. Otub1 displayed strong preference for disassembling Lys48-linked polyubiquitin chains, while leaving Lys63-linked polyubiquitin chains unaffected (Fig. 5a). Analysis of a catalytically dead mutant with a C91A mutation revealed no effect on Lys48-linked polyubiquitination of Tau. Conversely, AAV-driven expression of N-terminally truncated Otub1 significantly decreased Lys48-linked polyubiquitin chains of Tau (Fig. 5b), demonstrating that the N-terminal domain of Otub1, which is crucial for its noncanonical role, is not involved in regulation of Tau ubiquitination.
To confirm the effect of Lys48-linked polyubiquitination change on Tau degradation, TauP301S neurons were infected with AAV–Otub1–GFP or AAV–GFP, and subsequently treated with protein synthesis inhibitor cycloheximide (CHX) for different durations up to 36 h to quantify the rate of Tau turnover using Western blot. We found that Tau degradation was significantly impaired in primary neurons infected with Otub1 WT, but not with catalytically dead mutant C91A, compared with GFP control. These results indicate that Otub1 regulates Tau degradation, dependent on its catalytic activity (Fig. 5c).
We further analyzed whether catalytically dead Otub1-C91A could affect Tau phosphorylation in primary neurons expressing TauP301S. AAV-driven expression of Otub1-C91A did not increase AT8-positive Tau, in contrast to expression of wild-type Otub1 and the N-terminally truncated form of Otub1. This was demonstrated by immunofluorescence staining (Fig. 6a) and confirmed by biochemical analysis (Fig. 6b).
Taken together, our data prove that Otub1 can remove Lys48-linked polyubiquitin chains from Tau in a catalytic activity-dependent manner, implicating the canonical pathway. Consequently, Otub1-dependent deubiquitination inhibits Tau degradation through the proteasome pathway and prolongs its half-life, leading to accumulation of pathological forms of Tau.

Otub1 promotes Tau phosphorylation and oligomerization in vivo in Tau transgenic mice

To investigate the effect of Otub1 on Tau in vivo, AAV–Otub1 and AAV–GFP were bilaterally intraventricularly injected into TauP301S transgenic pups (P0), resulting in widespread expression of Otub1 and GFP in cortical neurons at 2 months of age (Fig. 7a). Immunostaining with AT8 antibody revealed abundant AT8-positive neurons in AAV–Otub1-injected mice, absent in AAV–GFP-infected mice (Fig. 7a), at 2 months postinjection. Biochemical analysis further confirmed increased AT8-positive Tau by Otub1 expression (Fig. 7b), with AT8 phosphorylated Tau more robustly increased than total Tau, as reflected by the AT8/total Tau ratio. Cortex homogenates from TauP301S transgenic mice injected with AAV–Otub1 and control mice injected with AAV–GFP were analyzed under nonreducing condition. This revealed a significant increase of monomeric Tau and oligomeric Tau in AAV–Otub1-injected mice compared with AAV–GFP-injected mice (Fig. 7c). Similarly as in primary neurons, Tauc (a homo- or heterotypic Tau complex) was robustly induced in AAV–Otub1- but not AAV–GFP-injected mice, strongly confirming Otub1-induced modulation of Tau in brain (Fig. 7c). To further confirm the increased concentration of oligomeric Tau forms following Otub1 expression, dot-blot analysis using oligomeric Tau-specific antibody T22 was performed. This revealed significantly increased oligomeric Tau in AAV–Otub1-injected mice compared with AAV–GFP-injected control cases (Fig. 7d).

The effect of Otub1 on Tau phosphorylation and oligomerization in vivo is dependent on its catalytic function

To further assess the requirement for catalytically active Otub1 and the presence of the catalytically conserved C91 of Otub1, for accumulation of pathological Tau forms in vivo, we performed P0 injections of AAV–Otub1-C91A or AAV–Otub1-N-terminal truncated mutant, and AAV–Otub1-WT in TauP301S mice. Immunostaining and biochemical analysis, 2 months postinjection, unambiguously revealed that the catalytically dead mutant C91A did not affect AT8-stained Tau in vivo, while N-terminal truncated mutant of Otub1 increased AT8-positive Tau in vivo, similarly as wild-type Otub1 (Fig. 8).
To further analyze the requirement for catalytically active Otub1 for induction of oligomeric Tau forms, brain extracts of AAV-infected TauP301S mice were biochemically analyzed 2 months postinjection, under nonreducing condition. This revealed significantly increased oligomeric Tau forms, following expression of wild-type Otub1, but not following expression of GFP or of the catalytically inactive form of Otub1 (C91A) (Fig. 8c). This was further confirmed by dot-blot analysis using T22 antibody recognizing oligomeric Tau forms (Fig. 8d). These data indicate that expression of catalytically active Otub1 is required for accumulation of phosphorylated Tau and oligomeric Tau forms.
Noteworthy, to analyze whether endogenous Otub1 is modulated in conditions of accumulating pathological Tau forms, we took advantage of our microarray analysis performed for a different project in which we analyzed time-dependent changes in gene expression in hippocampus following Tau-seeded Tau aggregation in Tau transgenic mice. Interestingly, Otub1 expression is gradually downregulated with increasing Tau-seeded Tau aggregation (Fig. S4a), which could be considered as a protective mechanism. Furthermore, analysis of hippocampal gene expression in aging mice, using publicly available data (NCBI database GDS2082) [73], indicates a significant increase in Otub1 expression with aging (Fig. S4b). The latter suggests that increased expression of Otub1 with aging might contribute to increased risk for accumulation of pathological Tau forms with aging.

Discussion

Accumulation of aberrantly folded proteins is common to many neurodegenerative disorders, including AD, making (dys)regulation of proteostasis not only a potentially implicated pathogenetic mechanism, but most importantly an attractive therapeutic target. In AD, symptom progression strongly correlates with progression of Tau pathology, with early, soluble, oligomeric forms of Tau generally considered as toxic culprits. Aberrant protein degradation occurs through either the ALS or UPS, with the former mainly involved in degradation of large insoluble protein aggregates, while the latter is preferentially involved in clearance of smaller soluble forms, being the focus of this work. In view of the importance of the UPS in AD, Tau-ubiquitinating enzymes have been identified, while Tau deubiquitination has received less attention. Starting from a Tau interactome mapping, we identified here for the first time Otub1 as a novel Tau deubiquitinase, affecting accumulation of pathological forms of Tau in vitro and in vivo.
Here, we present a proteome-wide screening approach to identify Tau-interacting proteins, as a basis to gain insight into Tau pathophysiology and to identify Tau modifiers with therapeutic potential. Proteome-wide screening approaches are increasingly used—in analogy to genome-wide screening approaches—to gain novel insights from an unbiased starting point for drug discovery and fundamental science [3, 10, 27, 58, 62]. We used an iTRAQ-based approach to identify proteins interacting with endogenous Tau from mouse brain. The validity of our approach is reflected in the identification of several well-characterized Tau-interacting proteins and the current identification of Otub1 as a Tau deubiquitinase. The presented Tau interactome provides important information, extending beyond the current work, providing the basis for novel insights into the pathological role of Tau and to identify novel Tau-directed targets.
Here, we validated this approach and identified for the first time Otub1 as a novel Tau deubiquitinase, based on the Tau interactome map. Accumulating evidence implicates dysregulated proteostasis and a dysregulated UPS in AD, as highlighted in the “Introduction.” Different types of polyubiquitination of Tau, including Lys48 and Lys63 linked, have been identified in AD patients by mass spectrometry and immunological analysis [13, 4951, 56]. Interestingly, we here identified Otub1 as a Tau deubiquitinase which decreases the levels of Lys48- but not Lys63-linked polyubiquitin chains on Tau, increasing Tau stability in neurons. Our finding is in line with previous reports showing that Otub1 has preference for Lys48-linked polyubiquitination [17, 48, 74] and functions as a regulator of protein turnover [23, 26, 32, 44, 66]. Mutation of the catalytically conserved cysteine (C91) abolished its role in Tau polyubiquitination, further confirming the crucial role of its catalytic capacity for its involvement in the regulation of Lys48 polyubiquitination on Tau. Otub1 has also been found to have a noncanonical role in DNA damage response by impeding Lys63 polyubiquitin chain formation [53, 77, 78], but no effect of Otub1 on Lys63 polyubiquitination of Tau nor implication of the N-terminal part of Otub1 was demonstrated.
Notably, the effect of polyubiquitination modification is determinant for the fate of its substrate. Lys48 polyubiquitin chains act as a proteasomal degradative signal, while Lys63-linked ubiquitin modification has been proposed to contribute to biogenesis of inclusions and clearance of larger aggregates by autophagy [36, 55, 68, 80]. Although soluble Tau forms as well as large insoluble aggregated Tau forms can be polyubiquitinated [13, 49, 50], higher-order protein aggregations are unlikely to pass through the narrow proteasome opening, and they are normally sequestrated into inclusions or aggresomes and cleared through the ALS. Recently, several lines of evidence have indicated selective autophagy using Lys63-linked polyubiquitin chains as a cargo recognition signal [36, 55, 68, 80]. In our experiments, we found that Otub1 only influences Lys48 polyubiquitination but not Lys63-linked polyubiquitin chains of Tau, and therefore would be rather involved in clearance of smaller soluble forms of Tau through the proteasome.
These findings are further strengthened by our assessment of the effect of Otub1 on Tau metabolism. We demonstrate that Otub1-dependent Tau deubiquitination is linked to accumulation of Tau phosphorylated at the pathologically relevant AT8 epitope—used for Braak staging—and accumulation of oligomeric soluble forms of Tau, which have been considered to be toxic culprits. Hence, overexpressed Otub1, which regulates Lys48-linked polyubiquitination of Tau, promotes accumulation of abnormally phosphorylated Tau and oligomeric Tau, both in Tau transgenic mice and in primary neurons. These findings are in line with previously published findings. Depletion of CHIP in Tau mice leads to significant accumulation of ubiquitin-negative, and phosphorylated soluble Tau species [16]. In a nonneuronal cell line which overexpresses the longest human Tau isoform, proteasome inhibitor treatment stabilizes phosphorylated and aggregated Tau species which arise from Tau phosphatase PPA inactivation and normally decay within 24 h [21]. In a triple transgenic mouse model, amyloid β-peptide (Aβ) immunotherapy leads to clearance of early soluble Tau forms by the UPS, but not of late Tau aggregates [54]. Along the same vein, molecular chaperon FKBP51, together with Hsp90, increase Tau oligomer formation by inhibiting Tau degradation through the proteasome in a mouse model [8]. In line with these reports, our work corroborates regulation of the early pathological Tau forms by the UPS, including deubiquitination as an important modulator.
Our data are important in view of the fact that early oligomeric forms of Tau are generally considered to be toxic culprits in AD and related Tauopathies. We have previously shown in a Tau seeding assay in in vitro and in vivo models that neuronal dysfunction correlated strongly with early pathological forms of Tau rather than with full-blown mature NFTs [65]. Previous studies using elegant approaches have indicated that the Tau aggregation process and early pathological forms rather than full-blown NFTs correlate with neuronal dysfunction [6, 11, 12, 15, 33, 37, 39, 40, 59, 60, 67]. The detrimental role of Tau oligomers in neuronal function and behavior was further demonstrated using acute injection of oligomeric Tau and immunization with anti-Tau oligomeric antibodies [11, 12, 39, 40]. Taken together, these data highlight the importance of targeting early oligomeric Tau forms as pathogenetic culprits, and hence the importance of the demonstrated effect of Otub1 on soluble oligomeric forms of Tau.
Interestingly, we show that the conserved catalytic cysteine (C91) of Otub1 is crucial for accumulation of hyperphosphorylated Tau and oligomeric Tau species. This finding is not only important as an internal control but also in the context of drug development, providing a druggable target. It must be noted that upstream modulators of Otub1, mechanisms regulating Otub1 activity, expression and probably the binding of Otub1 to chaperone molecules must be considered as potential therapeutic targets as well. Regulation of key proteins via recruitment of distinct E3 ligases or DUBs mediated by specific adaptor proteins is common in many cellular pathways. While the mechanisms upstream of Otub1 remain unknown, they may provide additional therapeutic targets. Inhibition of deubiquitinases as therapeutic targets is attracting increasing attention for different diseases, including cancer. Our findings identifying Otub1 as a Tau deubiquitinase affecting pathological forms of Tau in vitro and in vivo may therefore yield new perspectives for therapy.
Taken together, in this work we present a Tau interactome map, which yields a basis for novel insights into Tau biology and its pathological role, validated by and extending beyond our current work. We furthermore provide novel insight into the link between the UPS and AD and open new avenues for development of small-molecule inhibitors specifically targeting accumulation of pathological forms of Tau in AD and related Tauopathies.

Acknowledgements

This work was supported by the Belgian Fonds National pour la Recherche Scientifique–Fonds de la Recherche Scientifique (FNRS-FRS; Qualified Researcher, Impulse Financing, Research Credits), by Interuniversity Attraction Poles Programme–Belgian State–Belgian Science Policy, by the Belgian Fonds de la Recherche Scientifique Médicale, by the Queen Elisabeth Medical Foundation of Belgium (QEMF-FMRE), by the Stichting Alzheimer Onderzoek (SAO), by the Institute for the Promotion of Innovation by Science and Technology (IWT) in Flanders (IWT O&O, currently FWO), Belgium, and by Stellar funding of Janssen Research Foundation. We would like to thank Markus Boesche, Carola Doce, Frank Fischer, and Melanie Jundt for their technical expertise.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
3.
Zurück zum Zitat Bantscheff M, Eberhard D, Abraham Y, Bastuck S, Boesche M, Hobson S, Mathieson T, Perrin J, Raida M, Rau C, Reader V, Sweetman G, Bauer A, Bouwmeester T, Hopf C, Kruse U, Neubauer G, Ramsden N, Rick J, Kuster B, Drewes G (2007) Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat Biotechnol 25:1035–1044. doi:10.1038/nbt1328 CrossRefPubMed Bantscheff M, Eberhard D, Abraham Y, Bastuck S, Boesche M, Hobson S, Mathieson T, Perrin J, Raida M, Rau C, Reader V, Sweetman G, Bauer A, Bouwmeester T, Hopf C, Kruse U, Neubauer G, Ramsden N, Rick J, Kuster B, Drewes G (2007) Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat Biotechnol 25:1035–1044. doi:10.​1038/​nbt1328 CrossRefPubMed
4.
Zurück zum Zitat Bantscheff M, Hopf C, Savitski MM, Dittmann A, Grandi P, Michon AM, Schlegl J, Abraham Y, Becher I, Bergamini G, Boesche M, Delling M, Dumpelfeld B, Eberhard D, Huthmacher C, Mathieson T, Poeckel D, Reader V, Strunk K, Sweetman G, Kruse U, Neubauer G, Ramsden NG, Drewes G (2011) Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes. Nat Biotechnol 29:255–265. doi:10.1038/nbt.1759 CrossRefPubMed Bantscheff M, Hopf C, Savitski MM, Dittmann A, Grandi P, Michon AM, Schlegl J, Abraham Y, Becher I, Bergamini G, Boesche M, Delling M, Dumpelfeld B, Eberhard D, Huthmacher C, Mathieson T, Poeckel D, Reader V, Strunk K, Sweetman G, Kruse U, Neubauer G, Ramsden NG, Drewes G (2011) Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes. Nat Biotechnol 29:255–265. doi:10.​1038/​nbt.​1759 CrossRefPubMed
6.
Zurück zum Zitat Berger Z, Roder H, Hanna A, Carlson A, Rangachari V, Yue M, Wszolek Z, Ashe K, Knight J, Dickson D, Andorfer C, Rosenberry TL, Lewis J, Hutton M, Janus C (2007) Accumulation of pathological tau species and memory loss in a conditional model of tauopathy. J Neurosci 27:3650–3662. doi:10.1523/JNEUROSCI.0587-07.2007 CrossRefPubMed Berger Z, Roder H, Hanna A, Carlson A, Rangachari V, Yue M, Wszolek Z, Ashe K, Knight J, Dickson D, Andorfer C, Rosenberry TL, Lewis J, Hutton M, Janus C (2007) Accumulation of pathological tau species and memory loss in a conditional model of tauopathy. J Neurosci 27:3650–3662. doi:10.​1523/​JNEUROSCI.​0587-07.​2007 CrossRefPubMed
7.
Zurück zum Zitat Berke SJ, Paulson HL (2003) Protein aggregation and the ubiquitin proteasome pathway: gaining the UPPer hand on neurodegeneration. Curr Opin Genet Dev 13:253–261CrossRefPubMed Berke SJ, Paulson HL (2003) Protein aggregation and the ubiquitin proteasome pathway: gaining the UPPer hand on neurodegeneration. Curr Opin Genet Dev 13:253–261CrossRefPubMed
8.
Zurück zum Zitat Blair LJ, Nordhues BA, Hill SE, Scaglione KM, O’Leary JC 3rd, Fontaine SN, Breydo L, Zhang B, Li P, Wang L, Cotman C, Paulson HL, Muschol M, Uversky VN, Klengel T, Binder EB, Kayed R, Golde TE, Berchtold N, Dickey CA (2013) Accelerated neurodegeneration through chaperone-mediated oligomerization of tau. J Clin Invest 123:4158–4169. doi:10.1172/JCI69003 CrossRefPubMedPubMedCentral Blair LJ, Nordhues BA, Hill SE, Scaglione KM, O’Leary JC 3rd, Fontaine SN, Breydo L, Zhang B, Li P, Wang L, Cotman C, Paulson HL, Muschol M, Uversky VN, Klengel T, Binder EB, Kayed R, Golde TE, Berchtold N, Dickey CA (2013) Accelerated neurodegeneration through chaperone-mediated oligomerization of tau. J Clin Invest 123:4158–4169. doi:10.​1172/​JCI69003 CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Braak H, Braak E (1991) Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 82:239–259CrossRefPubMed Braak H, Braak E (1991) Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 82:239–259CrossRefPubMed
10.
Zurück zum Zitat Brajenovic M, Joberty G, Kuster B, Bouwmeester T, Drewes G (2004) Comprehensive proteomic analysis of human Par protein complexes reveals an interconnected protein network. J Biol Chem 279:12804–12811. doi:10.1074/jbc.M312171200 CrossRefPubMed Brajenovic M, Joberty G, Kuster B, Bouwmeester T, Drewes G (2004) Comprehensive proteomic analysis of human Par protein complexes reveals an interconnected protein network. J Biol Chem 279:12804–12811. doi:10.​1074/​jbc.​M312171200 CrossRefPubMed
11.
Zurück zum Zitat Castillo-Carranza DL, Gerson JE, Sengupta U, Guerrero-Munoz MJ, Lasagna-Reeves CA, Kayed R (2014) Specific targeting of tau oligomers in Htau mice prevents cognitive impairment and tau toxicity following injection with brain-derived tau oligomeric seeds. J Alzheimers Dis 40(Suppl 1):S97–S111. doi:10.3233/JAD-132477 CrossRefPubMed Castillo-Carranza DL, Gerson JE, Sengupta U, Guerrero-Munoz MJ, Lasagna-Reeves CA, Kayed R (2014) Specific targeting of tau oligomers in Htau mice prevents cognitive impairment and tau toxicity following injection with brain-derived tau oligomeric seeds. J Alzheimers Dis 40(Suppl 1):S97–S111. doi:10.​3233/​JAD-132477 CrossRefPubMed
12.
Zurück zum Zitat Castillo-Carranza DL, Sengupta U, Guerrero-Munoz MJ, Lasagna-Reeves CA, Gerson JE, Singh G, Estes DM, Barrett AD, Dineley KT, Jackson GR, Kayed R (2014) Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J Neurosci 34:4260–4272. doi:10.1523/JNEUROSCI.3192-13.2014 CrossRefPubMed Castillo-Carranza DL, Sengupta U, Guerrero-Munoz MJ, Lasagna-Reeves CA, Gerson JE, Singh G, Estes DM, Barrett AD, Dineley KT, Jackson GR, Kayed R (2014) Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J Neurosci 34:4260–4272. doi:10.​1523/​JNEUROSCI.​3192-13.​2014 CrossRefPubMed
13.
Zurück zum Zitat Cripps D, Thomas SN, Jeng Y, Yang F, Davies P, Yang AJ (2006) Alzheimer disease-specific conformation of hyperphosphorylated paired helical filament-Tau is polyubiquitinated through Lys-48, Lys-11, and Lys-6 ubiquitin conjugation. J Biol Chem 281:10825–10838. doi:10.1074/jbc.M512786200 CrossRefPubMed Cripps D, Thomas SN, Jeng Y, Yang F, Davies P, Yang AJ (2006) Alzheimer disease-specific conformation of hyperphosphorylated paired helical filament-Tau is polyubiquitinated through Lys-48, Lys-11, and Lys-6 ubiquitin conjugation. J Biol Chem 281:10825–10838. doi:10.​1074/​jbc.​M512786200 CrossRefPubMed
14.
Zurück zum Zitat David DC, Layfield R, Serpell L, Narain Y, Goedert M, Spillantini MG (2002) Proteasomal degradation of tau protein. J Neurochem 83:176–185CrossRefPubMed David DC, Layfield R, Serpell L, Narain Y, Goedert M, Spillantini MG (2002) Proteasomal degradation of tau protein. J Neurochem 83:176–185CrossRefPubMed
16.
Zurück zum Zitat Dickey CA, Yue M, Lin WL, Dickson DW, Dunmore JH, Lee WC, Zehr C, West G, Cao S, Clark AM, Caldwell GA, Caldwell KA, Eckman C, Patterson C, Hutton M, Petrucelli L (2006) Deletion of the ubiquitin ligase CHIP leads to the accumulation, but not the aggregation, of both endogenous phospho- and caspase-3-cleaved tau species. J Neurosci 26:6985–6996. doi:10.1523/JNEUROSCI.0746-06.2006 CrossRefPubMed Dickey CA, Yue M, Lin WL, Dickson DW, Dunmore JH, Lee WC, Zehr C, West G, Cao S, Clark AM, Caldwell GA, Caldwell KA, Eckman C, Patterson C, Hutton M, Petrucelli L (2006) Deletion of the ubiquitin ligase CHIP leads to the accumulation, but not the aggregation, of both endogenous phospho- and caspase-3-cleaved tau species. J Neurosci 26:6985–6996. doi:10.​1523/​JNEUROSCI.​0746-06.​2006 CrossRefPubMed
17.
Zurück zum Zitat Edelmann MJ, Iphofer A, Akutsu M, Altun M, di Gleria K, Kramer HB, Fiebiger E, Dhe-Paganon S, Kessler BM (2009) Structural basis and specificity of human otubain 1-mediated deubiquitination. Biochem J 418:379–390. doi:10.1042/BJ20081318 CrossRefPubMed Edelmann MJ, Iphofer A, Akutsu M, Altun M, di Gleria K, Kramer HB, Fiebiger E, Dhe-Paganon S, Kessler BM (2009) Structural basis and specificity of human otubain 1-mediated deubiquitination. Biochem J 418:379–390. doi:10.​1042/​BJ20081318 CrossRefPubMed
20.
Zurück zum Zitat Giannakopoulos P, Herrmann FR, Bussiere T, Bouras C, Kovari E, Perl DP, Morrison JH, Gold G, Hof PR (2003) Tangle and neuron numbers, but not amyloid load, predict cognitive status in Alzheimer’s disease. Neurology 60:1495–1500CrossRefPubMed Giannakopoulos P, Herrmann FR, Bussiere T, Bouras C, Kovari E, Perl DP, Morrison JH, Gold G, Hof PR (2003) Tangle and neuron numbers, but not amyloid load, predict cognitive status in Alzheimer’s disease. Neurology 60:1495–1500CrossRefPubMed
21.
Zurück zum Zitat Goldbaum O, Oppermann M, Handschuh M, Dabir D, Zhang B, Forman MS, Trojanowski JQ, Lee VM, Richter-Landsberg C (2003) Proteasome inhibition stabilizes tau inclusions in oligodendroglial cells that occur after treatment with okadaic acid. J Neurosci 23:8872–8880PubMed Goldbaum O, Oppermann M, Handschuh M, Dabir D, Zhang B, Forman MS, Trojanowski JQ, Lee VM, Richter-Landsberg C (2003) Proteasome inhibition stabilizes tau inclusions in oligodendroglial cells that occur after treatment with okadaic acid. J Neurosci 23:8872–8880PubMed
25.
Zurück zum Zitat Han DH, Na HK, Choi WH, Lee JH, Kim YK, Won C, Lee SH, Kim KP, Kuret J, Min DH, Lee MJ (2014) Direct cellular delivery of human proteasomes to delay tau aggregation. Nat Commun 5:5633. doi:10.1038/ncomms6633 CrossRefPubMed Han DH, Na HK, Choi WH, Lee JH, Kim YK, Won C, Lee SH, Kim KP, Kuret J, Min DH, Lee MJ (2014) Direct cellular delivery of human proteasomes to delay tau aggregation. Nat Commun 5:5633. doi:10.​1038/​ncomms6633 CrossRefPubMed
28.
Zurück zum Zitat Ihara Y, Morishima-Kawashima M, Nixon R (2012) The ubiquitin-proteasome system and the autophagic-lysosomal system in Alzheimer disease. Cold Spring Harb Perspect Med 2. doi:10.1101/cshperspect.a006361 Ihara Y, Morishima-Kawashima M, Nixon R (2012) The ubiquitin-proteasome system and the autophagic-lysosomal system in Alzheimer disease. Cold Spring Harb Perspect Med 2. doi:10.​1101/​cshperspect.​a006361
29.
Zurück zum Zitat Irmler M, Gentier RJ, Dennissen FJ, Schulz H, Bolle I, Holter SM, Kallnik M, Cheng JJ, Klingenspor M, Rozman J, Ehrhardt N, Hermes DJ, Gailus-Durner V, Fuchs H, Hrabe de Angelis M, Meyer HE, Hopkins DA, Van Leeuwen FW, Beckers J (2012) Long-term proteasomal inhibition in transgenic mice by UBB(+1) expression results in dysfunction of central respiration control reminiscent of brainstem neuropathology in Alzheimer patients. Acta Neuropathol 124:187–197. doi:10.1007/s00401-012-1003-7 CrossRefPubMedPubMedCentral Irmler M, Gentier RJ, Dennissen FJ, Schulz H, Bolle I, Holter SM, Kallnik M, Cheng JJ, Klingenspor M, Rozman J, Ehrhardt N, Hermes DJ, Gailus-Durner V, Fuchs H, Hrabe de Angelis M, Meyer HE, Hopkins DA, Van Leeuwen FW, Beckers J (2012) Long-term proteasomal inhibition in transgenic mice by UBB(+1) expression results in dysfunction of central respiration control reminiscent of brainstem neuropathology in Alzheimer patients. Acta Neuropathol 124:187–197. doi:10.​1007/​s00401-012-1003-7 CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Jinwal UK, O’Leary JC 3rd, Borysov SI, Jones JR, Li Q, Koren J 3rd, Abisambra JF, Vestal GD, Lawson LY, Johnson AG, Blair LJ, Jin Y, Miyata Y, Gestwicki JE, Dickey CA (2010) Hsc70 rapidly engages tau after microtubule destabilization. J Biol Chem 285:16798–16805. doi:10.1074/jbc.M110.113753 CrossRefPubMedPubMedCentral Jinwal UK, O’Leary JC 3rd, Borysov SI, Jones JR, Li Q, Koren J 3rd, Abisambra JF, Vestal GD, Lawson LY, Johnson AG, Blair LJ, Jin Y, Miyata Y, Gestwicki JE, Dickey CA (2010) Hsc70 rapidly engages tau after microtubule destabilization. J Biol Chem 285:16798–16805. doi:10.​1074/​jbc.​M110.​113753 CrossRefPubMedPubMedCentral
31.
32.
Zurück zum Zitat Karunarathna U, Kongsema M, Zona S, Gong C, Cabrera E, Gomes AR, Man EP, Khongkow P, Tsang JW, Khoo US, Medema RH, Freire R, Lam EW (2016) OTUB1 inhibits the ubiquitination and degradation of FOXM1 in breast cancer and epirubicin resistance. Oncogene 35:1433–1444. doi:10.1038/onc.2015.208 CrossRefPubMed Karunarathna U, Kongsema M, Zona S, Gong C, Cabrera E, Gomes AR, Man EP, Khongkow P, Tsang JW, Khoo US, Medema RH, Freire R, Lam EW (2016) OTUB1 inhibits the ubiquitination and degradation of FOXM1 in breast cancer and epirubicin resistance. Oncogene 35:1433–1444. doi:10.​1038/​onc.​2015.​208 CrossRefPubMed
34.
Zurück zum Zitat Keck S, Nitsch R, Grune T, Ullrich O (2003) Proteasome inhibition by paired helical filament-tau in brains of patients with Alzheimer’s disease. J Neurochem 85:115–122CrossRefPubMed Keck S, Nitsch R, Grune T, Ullrich O (2003) Proteasome inhibition by paired helical filament-tau in brains of patients with Alzheimer’s disease. J Neurochem 85:115–122CrossRefPubMed
35.
39.
Zurück zum Zitat Lasagna-Reeves CA, Castillo-Carranza DL, Guerrero-Muoz MJ, Jackson GR, Kayed R (2010) Preparation and characterization of neurotoxic tau oligomers. Biochemistry 49:10039–10041. doi:10.1021/bi1016233 CrossRefPubMed Lasagna-Reeves CA, Castillo-Carranza DL, Guerrero-Muoz MJ, Jackson GR, Kayed R (2010) Preparation and characterization of neurotoxic tau oligomers. Biochemistry 49:10039–10041. doi:10.​1021/​bi1016233 CrossRefPubMed
41.
Zurück zum Zitat Lee G, Neve RL, Kosik KS (1989) The microtubule binding domain of tau protein. Neuron 2:1615–1624CrossRefPubMed Lee G, Neve RL, Kosik KS (1989) The microtubule binding domain of tau protein. Neuron 2:1615–1624CrossRefPubMed
42.
Zurück zum Zitat Leroy E, Boyer R, Auburger G, Leube B, Ulm G, Mezey E, Harta G, Brownstein MJ, Jonnalagada S, Chernova T, Dehejia A, Lavedan C, Gasser T, Steinbach PJ, Wilkinson KD, Polymeropoulos MH (1998) The ubiquitin pathway in Parkinson’s disease. Nature 395:451–452. doi:10.1038/26652 CrossRefPubMed Leroy E, Boyer R, Auburger G, Leube B, Ulm G, Mezey E, Harta G, Brownstein MJ, Jonnalagada S, Chernova T, Dehejia A, Lavedan C, Gasser T, Steinbach PJ, Wilkinson KD, Polymeropoulos MH (1998) The ubiquitin pathway in Parkinson’s disease. Nature 395:451–452. doi:10.​1038/​26652 CrossRefPubMed
43.
Zurück zum Zitat Levine B, Klionsky DJ (2004) Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell 6:463–477CrossRefPubMed Levine B, Klionsky DJ (2004) Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell 6:463–477CrossRefPubMed
44.
Zurück zum Zitat Li S, Zheng H, Mao AP, Zhong B, Li Y, Liu Y, Gao Y, Ran Y, Tien P, Shu HB (2010) Regulation of virus-triggered signaling by OTUB1- and OTUB2-mediated deubiquitination of TRAF3 and TRAF6. J Biol Chem 285:4291–4297. doi:10.1074/jbc.M109.074971 CrossRefPubMed Li S, Zheng H, Mao AP, Zhong B, Li Y, Liu Y, Gao Y, Ran Y, Tien P, Shu HB (2010) Regulation of virus-triggered signaling by OTUB1- and OTUB2-mediated deubiquitination of TRAF3 and TRAF6. J Biol Chem 285:4291–4297. doi:10.​1074/​jbc.​M109.​074971 CrossRefPubMed
45.
Zurück zum Zitat Lindsten K, de Vrij FM, Verhoef LG, Fischer DF, van Leeuwen FW, Hol EM, Masucci MG, Dantuma NP (2002) Mutant ubiquitin found in neurodegenerative disorders is a ubiquitin fusion degradation substrate that blocks proteasomal degradation. J Cell Biol 157:417–427. doi:10.1083/jcb.200111034 CrossRefPubMedPubMedCentral Lindsten K, de Vrij FM, Verhoef LG, Fischer DF, van Leeuwen FW, Hol EM, Masucci MG, Dantuma NP (2002) Mutant ubiquitin found in neurodegenerative disorders is a ubiquitin fusion degradation substrate that blocks proteasomal degradation. J Cell Biol 157:417–427. doi:10.​1083/​jcb.​200111034 CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Luo HB, Xia YY, Shu XJ, Liu ZC, Feng Y, Liu XH, Yu G, Yin G, Xiong YS, Zeng K, Jiang J, Ye K, Wang XC, Wang JZ (2014) SUMOylation at K340 inhibits tau degradation through deregulating its phosphorylation and ubiquitination. Proc Natl Acad Sci USA 111:16586–16591. doi:10.1073/pnas.1417548111 CrossRefPubMedPubMedCentral Luo HB, Xia YY, Shu XJ, Liu ZC, Feng Y, Liu XH, Yu G, Yin G, Xiong YS, Zeng K, Jiang J, Ye K, Wang XC, Wang JZ (2014) SUMOylation at K340 inhibits tau degradation through deregulating its phosphorylation and ubiquitination. Proc Natl Acad Sci USA 111:16586–16591. doi:10.​1073/​pnas.​1417548111 CrossRefPubMedPubMedCentral
48.
49.
Zurück zum Zitat Mori H, Kondo J, Ihara Y (1987) Ubiquitin is a component of paired helical filaments in Alzheimer’s disease. Science 235:1641–1644CrossRefPubMed Mori H, Kondo J, Ihara Y (1987) Ubiquitin is a component of paired helical filaments in Alzheimer’s disease. Science 235:1641–1644CrossRefPubMed
50.
Zurück zum Zitat Morishima-Kawashima M, Hasegawa M, Takio K, Suzuki M, Titani K, Ihara Y (1993) Ubiquitin is conjugated with amino-terminally processed tau in paired helical filaments. Neuron 10:1151–1160CrossRefPubMed Morishima-Kawashima M, Hasegawa M, Takio K, Suzuki M, Titani K, Ihara Y (1993) Ubiquitin is conjugated with amino-terminally processed tau in paired helical filaments. Neuron 10:1151–1160CrossRefPubMed
51.
Zurück zum Zitat Morris M, Knudsen GM, Maeda S, Trinidad JC, Ioanoviciu A, Burlingame AL, Mucke L (2015) Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat Neurosci 18:1183–1189. doi:10.1038/nn.4067 CrossRefPubMed Morris M, Knudsen GM, Maeda S, Trinidad JC, Ioanoviciu A, Burlingame AL, Mucke L (2015) Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat Neurosci 18:1183–1189. doi:10.​1038/​nn.​4067 CrossRefPubMed
52.
Zurück zum Zitat Myeku N, Clelland CL, Emrani S, Kukushkin NV, Yu WH, Goldberg AL, Duff KE (2016) Tau-driven 26S proteasome impairment and cognitive dysfunction can be prevented early in disease by activating cAMP-PKA signaling. Nat Med 22:46–53. doi:10.1038/nm.4011 CrossRefPubMed Myeku N, Clelland CL, Emrani S, Kukushkin NV, Yu WH, Goldberg AL, Duff KE (2016) Tau-driven 26S proteasome impairment and cognitive dysfunction can be prevented early in disease by activating cAMP-PKA signaling. Nat Med 22:46–53. doi:10.​1038/​nm.​4011 CrossRefPubMed
53.
Zurück zum Zitat Nakada S, Tai I, Panier S, Al-Hakim A, Iemura S, Juang YC, O’Donnell L, Kumakubo A, Munro M, Sicheri F, Gingras AC, Natsume T, Suda T, Durocher D (2010) Non-canonical inhibition of DNA damage-dependent ubiquitination by OTUB1. Nature 466:941–946. doi:10.1038/nature09297 CrossRefPubMed Nakada S, Tai I, Panier S, Al-Hakim A, Iemura S, Juang YC, O’Donnell L, Kumakubo A, Munro M, Sicheri F, Gingras AC, Natsume T, Suda T, Durocher D (2010) Non-canonical inhibition of DNA damage-dependent ubiquitination by OTUB1. Nature 466:941–946. doi:10.​1038/​nature09297 CrossRefPubMed
55.
Zurück zum Zitat Olzmann JA, Chin LS (2008) Parkin-mediated K63-linked polyubiquitination: a signal for targeting misfolded proteins to the aggresome-autophagy pathway. Autophagy 4:85–87CrossRefPubMed Olzmann JA, Chin LS (2008) Parkin-mediated K63-linked polyubiquitination: a signal for targeting misfolded proteins to the aggresome-autophagy pathway. Autophagy 4:85–87CrossRefPubMed
57.
Zurück zum Zitat Petrucelli L, Dickson D, Kehoe K, Taylor J, Snyder H, Grover A, De Lucia M, McGowan E, Lewis J, Prihar G, Kim J, Dillmann WH, Browne SE, Hall A, Voellmy R, Tsuboi Y, Dawson TM, Wolozin B, Hardy J, Hutton M (2004) CHIP and Hsp70 regulate tau ubiquitination, degradation and aggregation. Hum Mol Genet 13:703–714. doi:10.1093/hmg/ddh083 CrossRefPubMed Petrucelli L, Dickson D, Kehoe K, Taylor J, Snyder H, Grover A, De Lucia M, McGowan E, Lewis J, Prihar G, Kim J, Dillmann WH, Browne SE, Hall A, Voellmy R, Tsuboi Y, Dawson TM, Wolozin B, Hardy J, Hutton M (2004) CHIP and Hsp70 regulate tau ubiquitination, degradation and aggregation. Hum Mol Genet 13:703–714. doi:10.​1093/​hmg/​ddh083 CrossRefPubMed
58.
Zurück zum Zitat Reinhard FB, Eberhard D, Werner T, Franken H, Childs D, Doce C, Savitski MF, Huber W, Bantscheff M, Savitski MM, Drewes G (2015) Thermal proteome profiling monitors ligand interactions with cellular membrane proteins. Nat Methods 12:1129–1131. doi:10.1038/nmeth.3652 CrossRefPubMed Reinhard FB, Eberhard D, Werner T, Franken H, Childs D, Doce C, Savitski MF, Huber W, Bantscheff M, Savitski MM, Drewes G (2015) Thermal proteome profiling monitors ligand interactions with cellular membrane proteins. Nat Methods 12:1129–1131. doi:10.​1038/​nmeth.​3652 CrossRefPubMed
59.
Zurück zum Zitat Roberson ED, Scearce-Levie K, Palop JJ, Yan F, Cheng IH, Wu T, Gerstein H, Yu GQ, Mucke L (2007) Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science 316:750–754. doi:10.1126/science.1141736 CrossRefPubMed Roberson ED, Scearce-Levie K, Palop JJ, Yan F, Cheng IH, Wu T, Gerstein H, Yu GQ, Mucke L (2007) Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science 316:750–754. doi:10.​1126/​science.​1141736 CrossRefPubMed
60.
Zurück zum Zitat Santacruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, Guimaraes A, DeTure M, Ramsden M, McGowan E, Forster C, Yue M, Orne J, Janus C, Mariash A, Kuskowski M, Hyman B, Hutton M, Ashe KH (2005) Tau suppression in a neurodegenerative mouse model improves memory function. Science 309:476–481. doi:10.1126/science.1113694 CrossRefPubMedPubMedCentral Santacruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, Guimaraes A, DeTure M, Ramsden M, McGowan E, Forster C, Yue M, Orne J, Janus C, Mariash A, Kuskowski M, Hyman B, Hutton M, Ashe KH (2005) Tau suppression in a neurodegenerative mouse model improves memory function. Science 309:476–481. doi:10.​1126/​science.​1113694 CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Sato Y, Yamagata A, Goto-Ito S, Kubota K, Miyamoto R, Nakada S, Fukai S (2012) Molecular basis of Lys-63-linked polyubiquitination inhibition by the interaction between human deubiquitinating enzyme OTUB1 and ubiquitin-conjugating enzyme UBC13. J Biol Chem 287:25860–25868. doi:10.1074/jbc.M112.364752 CrossRefPubMedPubMedCentral Sato Y, Yamagata A, Goto-Ito S, Kubota K, Miyamoto R, Nakada S, Fukai S (2012) Molecular basis of Lys-63-linked polyubiquitination inhibition by the interaction between human deubiquitinating enzyme OTUB1 and ubiquitin-conjugating enzyme UBC13. J Biol Chem 287:25860–25868. doi:10.​1074/​jbc.​M112.​364752 CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Savitski MM, Reinhard FB, Franken H, Werner T, Savitski MF, Eberhard D, Martinez Molina D, Jafari R, Dovega RB, Klaeger S, Kuster B, Nordlund P, Bantscheff M, Drewes G (2014) Tracking cancer drugs in living cells by thermal profiling of the proteome. Science 346:1255784. doi:10.1126/science.1255784 CrossRefPubMed Savitski MM, Reinhard FB, Franken H, Werner T, Savitski MF, Eberhard D, Martinez Molina D, Jafari R, Dovega RB, Klaeger S, Kuster B, Nordlund P, Bantscheff M, Drewes G (2014) Tracking cancer drugs in living cells by thermal profiling of the proteome. Science 346:1255784. doi:10.​1126/​science.​1255784 CrossRefPubMed
64.
Zurück zum Zitat Stancu IC, Ris L, Vasconcelos B, Marinangeli C, Goeminne L, Laporte V, Haylani LE, Couturier J, Schakman O, Gailly P, Pierrot N, Kienlen-Campard P, Octave JN, Dewachter I (2014) Tauopathy contributes to synaptic and cognitive deficits in a murine model for Alzheimer’s disease. FASEB J 28:2620–2631. doi:10.1096/fj.13-246702 CrossRefPubMed Stancu IC, Ris L, Vasconcelos B, Marinangeli C, Goeminne L, Laporte V, Haylani LE, Couturier J, Schakman O, Gailly P, Pierrot N, Kienlen-Campard P, Octave JN, Dewachter I (2014) Tauopathy contributes to synaptic and cognitive deficits in a murine model for Alzheimer’s disease. FASEB J 28:2620–2631. doi:10.​1096/​fj.​13-246702 CrossRefPubMed
65.
Zurück zum Zitat Stancu IC, Vasconcelos B, Ris L, Wang P, Villers A, Peeraer E, Buist A, Terwel D, Baatsen P, Oyelami T, Pierrot N, Casteels C, Bormans G, Kienlen-Campard P, Octave JN, Moechars D, Dewachter I (2015) Templated misfolding of Tau by prion-like seeding along neuronal connections impairs neuronal network function and associated behavioral outcomes in Tau transgenic mice. Acta Neuropathol 129:875–894. doi:10.1007/s00401-015-1413-4 CrossRefPubMedPubMedCentral Stancu IC, Vasconcelos B, Ris L, Wang P, Villers A, Peeraer E, Buist A, Terwel D, Baatsen P, Oyelami T, Pierrot N, Casteels C, Bormans G, Kienlen-Campard P, Octave JN, Moechars D, Dewachter I (2015) Templated misfolding of Tau by prion-like seeding along neuronal connections impairs neuronal network function and associated behavioral outcomes in Tau transgenic mice. Acta Neuropathol 129:875–894. doi:10.​1007/​s00401-015-1413-4 CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Sydow A, Van der Jeugd A, Zheng F, Ahmed T, Balschun D, Petrova O, Drexler D, Zhou L, Rune G, Mandelkow E, D’Hooge R, Alzheimer C, Mandelkow EM (2011) Tau-induced defects in synaptic plasticity, learning, and memory are reversible in transgenic mice after switching off the toxic Tau mutant. J Neurosci 31:2511–2525. doi:10.1523/JNEUROSCI.5245-10.2011 CrossRefPubMed Sydow A, Van der Jeugd A, Zheng F, Ahmed T, Balschun D, Petrova O, Drexler D, Zhou L, Rune G, Mandelkow E, D’Hooge R, Alzheimer C, Mandelkow EM (2011) Tau-induced defects in synaptic plasticity, learning, and memory are reversible in transgenic mice after switching off the toxic Tau mutant. J Neurosci 31:2511–2525. doi:10.​1523/​JNEUROSCI.​5245-10.​2011 CrossRefPubMed
68.
Zurück zum Zitat Tan JM, Wong ES, Kirkpatrick DS, Pletnikova O, Ko HS, Tay SP, Ho MW, Troncoso J, Gygi SP, Lee MK, Dawson VL, Dawson TM, Lim KL (2008) Lysine 63-linked ubiquitination promotes the formation and autophagic clearance of protein inclusions associated with neurodegenerative diseases. Hum Mol Genet 17:431–439. doi:10.1093/hmg/ddm320 CrossRefPubMed Tan JM, Wong ES, Kirkpatrick DS, Pletnikova O, Ko HS, Tay SP, Ho MW, Troncoso J, Gygi SP, Lee MK, Dawson VL, Dawson TM, Lim KL (2008) Lysine 63-linked ubiquitination promotes the formation and autophagic clearance of protein inclusions associated with neurodegenerative diseases. Hum Mol Genet 17:431–439. doi:10.​1093/​hmg/​ddm320 CrossRefPubMed
69.
Zurück zum Zitat Thal DR, Rub U, Orantes M, Braak H (2002) Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology 58:1791–1800CrossRefPubMed Thal DR, Rub U, Orantes M, Braak H (2002) Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology 58:1791–1800CrossRefPubMed
70.
Zurück zum Zitat Van der Jeugd A, Hochgrafe K, Ahmed T, Decker JM, Sydow A, Hofmann A, Wu D, Messing L, Balschun D, D’Hooge R, Mandelkow EM (2012) Cognitive defects are reversible in inducible mice expressing pro-aggregant full-length human Tau. Acta Neuropathol 123:787–805. doi:10.1007/s00401-012-0987-3 CrossRefPubMedPubMedCentral Van der Jeugd A, Hochgrafe K, Ahmed T, Decker JM, Sydow A, Hofmann A, Wu D, Messing L, Balschun D, D’Hooge R, Mandelkow EM (2012) Cognitive defects are reversible in inducible mice expressing pro-aggregant full-length human Tau. Acta Neuropathol 123:787–805. doi:10.​1007/​s00401-012-0987-3 CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat van Leeuwen FW, de Kleijn DP, van den Hurk HH, Neubauer A, Sonnemans MA, Sluijs JA, Koycu S, Ramdjielal RD, Salehi A, Martens GJ, Grosveld FG, Peter J, Burbach H, Hol EM (1998) Frameshift mutants of beta amyloid precursor protein and ubiquitin-B in Alzheimer’s and Down patients. Science 279:242–247CrossRefPubMed van Leeuwen FW, de Kleijn DP, van den Hurk HH, Neubauer A, Sonnemans MA, Sluijs JA, Koycu S, Ramdjielal RD, Salehi A, Martens GJ, Grosveld FG, Peter J, Burbach H, Hol EM (1998) Frameshift mutants of beta amyloid precursor protein and ubiquitin-B in Alzheimer’s and Down patients. Science 279:242–247CrossRefPubMed
72.
Zurück zum Zitat Vasconcelos B, Stancu IC, Buist A, Bird M, Wang P, Vanoosthuyse A, Van Kolen K, Verheyen A, Kienlen-Campard P, Octave JN, Baatsen P, Moechars D, Dewachter I (2016) Heterotypic seeding of Tau fibrillization by pre-aggregated Abeta provides potent seeds for prion-like seeding and propagation of Tau-pathology in vivo. Acta Neuropathol 131:549–569. doi:10.1007/s00401-015-1525-x CrossRefPubMedPubMedCentral Vasconcelos B, Stancu IC, Buist A, Bird M, Wang P, Vanoosthuyse A, Van Kolen K, Verheyen A, Kienlen-Campard P, Octave JN, Baatsen P, Moechars D, Dewachter I (2016) Heterotypic seeding of Tau fibrillization by pre-aggregated Abeta provides potent seeds for prion-like seeding and propagation of Tau-pathology in vivo. Acta Neuropathol 131:549–569. doi:10.​1007/​s00401-015-1525-x CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW (1975) A protein factor essential for microtubule assembly. Proc Natl Acad Sci USA 72:1858–1862CrossRefPubMedPubMedCentral Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW (1975) A protein factor essential for microtubule assembly. Proc Natl Acad Sci USA 72:1858–1862CrossRefPubMedPubMedCentral
79.
Zurück zum Zitat Witman GB, Cleveland DW, Weingarten MD, Kirschner MW (1976) Tubulin requires tau for growth onto microtubule initiating sites. Proc Natl Acad Sci USA 73:4070–4074CrossRefPubMedPubMedCentral Witman GB, Cleveland DW, Weingarten MD, Kirschner MW (1976) Tubulin requires tau for growth onto microtubule initiating sites. Proc Natl Acad Sci USA 73:4070–4074CrossRefPubMedPubMedCentral
80.
Zurück zum Zitat Wooten MW, Geetha T, Babu JR, Seibenhener ML, Peng J, Cox N, Diaz-Meco MT, Moscat J (2008) Essential role of sequestosome 1/p62 in regulating accumulation of Lys63-ubiquitinated proteins. J Biol Chem 283:6783–6789. doi:10.1074/jbc.M709496200 CrossRefPubMed Wooten MW, Geetha T, Babu JR, Seibenhener ML, Peng J, Cox N, Diaz-Meco MT, Moscat J (2008) Essential role of sequestosome 1/p62 in regulating accumulation of Lys63-ubiquitinated proteins. J Biol Chem 283:6783–6789. doi:10.​1074/​jbc.​M709496200 CrossRefPubMed
Metadaten
Titel
Tau interactome mapping based identification of Otub1 as Tau deubiquitinase involved in accumulation of pathological Tau forms in vitro and in vivo
verfasst von
Peng Wang
Gerard Joberty
Arjan Buist
Alexandre Vanoosthuyse
Ilie-Cosmin Stancu
Bruno Vasconcelos
Nathalie Pierrot
Maria Faelth-Savitski
Pascal Kienlen-Campard
Jean-Noël Octave
Marcus Bantscheff
Gerard Drewes
Diederik Moechars
Ilse Dewachter
Publikationsdatum
12.01.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Acta Neuropathologica / Ausgabe 5/2017
Print ISSN: 0001-6322
Elektronische ISSN: 1432-0533
DOI
https://doi.org/10.1007/s00401-016-1663-9

Weitere Artikel der Ausgabe 5/2017

Acta Neuropathologica 5/2017 Zur Ausgabe

ACKNOWLEDGEMENT TO REFEREES

Acknowledgement to referees

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

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