Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
EMBO J ; 41(18): e10242, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35993331

ABSTRACT

Microtubule-associated protein tau is a central factor in Alzheimer's disease and other tauopathies. However, the physiological functions of tau are unclear. Here, we used proximity-labelling proteomics to chart tau interactomes in primary neurons and mouse brains in vivo. Tau interactors map onto pathways of cytoskeletal, synaptic vesicle and postsynaptic receptor regulation and show significant enrichment for Parkinson's, Alzheimer's and prion disease. We find that tau interacts with and dose-dependently reduces the activity of N-ethylmaleimide sensitive fusion protein (NSF), a vesicular ATPase essential for AMPA-type glutamate receptor (AMPAR) trafficking. Tau-deficient (tau-/- ) neurons showed mislocalised expression of NSF and enhanced synaptic AMPAR surface levels, reversible through the expression of human tau or inhibition of NSF. Consequently, enhanced AMPAR-mediated associative and object recognition memory in tau-/- mice is suppressed by both hippocampal tau and infusion with an NSF-inhibiting peptide. Pathologic mutant tau from mouse models or Alzheimer's disease significantly enhances NSF inhibition. Our results map neuronal tau interactomes and delineate a functional link of tau with NSF in plasticity-associated AMPAR-trafficking and memory.


Subject(s)
Alzheimer Disease , Receptors, AMPA , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Hippocampus/metabolism , Humans , Memory , Mice , N-Ethylmaleimide-Sensitive Proteins/genetics , N-Ethylmaleimide-Sensitive Proteins/metabolism , Neurons/metabolism , Protein Transport , Receptors, AMPA/genetics , Receptors, AMPA/metabolism
2.
Sci Adv ; 8(27): eabl8809, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35857446

ABSTRACT

Hyperphosphorylation of the neuronal tau protein is a hallmark of neurodegenerative tauopathies such as Alzheimer's disease. A central unanswered question is why tau becomes progressively hyperphosphorylated. Here, we show that tau phosphorylation is governed by interdependence- a mechanistic link between initial site-specific and subsequent multi-site phosphorylation. Systematic assessment of site interdependence identified distinct residues (threonine-50, threonine-69, and threonine-181) as master sites that determine propagation of phosphorylation at multiple epitopes. CRISPR point mutation and expression of human tau in Alzheimer's mice showed that site interdependence governs physiologic and amyloid-associated multi-site phosphorylation and cognitive deficits, respectively. Combined targeting of master sites and p38α, the most central tau kinase linked to interdependence, synergistically ablated hyperphosphorylation. In summary, our work delineates how complex tau phosphorylation arises to inform therapeutic and biomarker design for tauopathies.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Humans , Mice , Phosphorylation , Tauopathies/genetics , Tauopathies/metabolism , Threonine/metabolism , tau Proteins/genetics , tau Proteins/metabolism
3.
Neurobiol Dis ; 171: 105783, 2022 09.
Article in English | MEDLINE | ID: mdl-35675895

ABSTRACT

Increasing evidence suggests that kynurenine pathway (KP) dyshomeostasis may promote disease progression in dementia. Studies in Alzheimer's disease (AD) patients confirm KP dyshomeostasis in plasma and cerebrospinal fluid (CSF) which correlates with amyloid-ß and tau pathology. Herein, we performed the first comprehensive study assessing baseline levels of KP metabolites in participants enrolling in the Australian Imaging Biomarkers Flagship Study of Aging. Our purpose was to test the hypothesis that changes in KP metabolites may be biomarkers of dementia processes that are largely silent. We used a cross-sectional analytical approach to assess non-progressors (N = 73); cognitively normal (CN) or mild cognitive impairment (MCI) participants at baseline and throughout the study, and progressors (N = 166); CN or MCI at baseline but progressing to either MCI or AD during the study. Significant KP changes in progressors included increased 3-hydroxyanthranilic acid (3-HAA) and 3-hydroxyanthranilic acid/anthranilic acid (3-HAA/AA) ratio, the latter having the largest effect on the odds of an individual being a progressor (OR 35.3; 95% CI between 14 and 104). 3-HAA levels were hence surprisingly bi-phasic, high in progressors but low in non-progressors or participants who had already transitioned to MCI or dementia. This is a new, unexpected and interesting result, as most studies of the KP in neurodegenerative disease show reduced 3-HAA/AA ratio after diagnosis. The neuroprotective metabolite picolinic acid was also significantly decreased while the neurotoxic metabolite 3-hydroxykynurenine increased in progressors. These results were significant even after adjustment for confounders. Considering the magnitude of the OR to predict change in cognition, it is important that these findings are replicated in other populations. Independent validation of our findings may confirm the utility of 3-HAA/AA ratio to predict change in cognition leading to dementia in clinical settings.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Neurodegenerative Diseases , 3-Hydroxyanthranilic Acid , Alzheimer Disease/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Australia , Biomarkers , Cognitive Dysfunction/cerebrospinal fluid , Cross-Sectional Studies , Disease Progression , Humans , Kynurenine , Peptide Fragments/cerebrospinal fluid , tau Proteins/cerebrospinal fluid
4.
Neuroscience ; 484: 53-65, 2022 02 21.
Article in English | MEDLINE | ID: mdl-35031398

ABSTRACT

Persistent improvement of cognitive deficits in Alzheimer's disease (AD), a common form of dementia, is an unattained therapeutic objective. Gene therapy holds promise for treatment of familial and sporadic forms of AD. p38γ, a member of the p38 mitogen-activated protein (MAP) kinase family, inhibits amyloid-ß toxicity through regulation of tau phosphorylation. We recently showed that a gene delivery approach increasing p38γ resulted in markedly better learning and memory performance in mouse models of AD at advanced stages of amyloid-ß- and tau-mediated cognitive impairment. Notably, low-to-moderate expression of p38γ had beneficial outcomes on cognition. The impact of high levels of p38γ on neuronal function remain unclear. Therefore, we addressed the outcomes of high levels of active p38γ on brain function, by direct injection of p38γ-encoding adeno-associated virus (AAV) into the forebrain of aged mice of an APP transgenic AD mouse model. While motor function in p38γ-expressing APP transgenic mice 2 months post-injection was comparable to control treated APP mice, their activity was markedly reduced in the open field test and included frequent bouts of immobility. Moreover, their learning and memory function was markedly impaired compared to control-treated aged APP mice. These results suggest that high neuronal levels of active p38γ emphasize a stress kinase role of p38γ, perturbing circuit function in motivation, navigation, and spatial learning. Overall, this work shows excessive neuronal p38γ levels can aggravate circuit dysfunction and advises adjustable expression systems will be required for sustainable AD gene therapy based on p38γ activity.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Prosencephalon/metabolism , tau Proteins/genetics , tau Proteins/metabolism
5.
Biochem Cell Biol ; 99(5): 606-616, 2021 10.
Article in English | MEDLINE | ID: mdl-33794133

ABSTRACT

The microtubule-associated protein tau is a key factor in neurodegenerative proteinopathies and is predominantly found in neuronal axons. However, somatodendritic localization of tau occurs in a subset of pathological and physiological tau. Dendritic tau can localize to post-synapses where it interacts with proteins of the post-synaptic density (PSD) protein PSD-95, a membrane-associated guanylate kinase (MAGUK) scaffold factor for organization of protein complexes within the PSD, to mediate downstream signals. However, the molecular details of this interaction remain unclear. Here, we used interaction mapping in cultured cells to demonstrate that tau interacts with the guanylate kinase (GUK) domain in the C-terminal region of PSD-95. The PSD-95 GUK domain is required for a complex with full-length human tau. Mapping the interaction of the MAGUK core with tau revealed that the microtubule binding repeats 2 and 3 and the proline-rich region contributes to this interaction, while the N- and C-terminal regions of tau inhibit interaction. These results reveal the intramolecular determinants of the protein complex of tau and PSD-95 and increase our understanding of tau interactions regulating neurotoxic signaling at the molecular level.


Subject(s)
Disks Large Homolog 4 Protein/metabolism , Guanylate Kinases/metabolism , Microtubules/metabolism , Proline/metabolism , tau Proteins/metabolism , Cells, Cultured , HEK293 Cells , Humans , Protein Binding
6.
Behav Brain Res ; 397: 112943, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33017638

ABSTRACT

Frontotemporal dementia (FTD) and Alzheimer's disease (AD) exhibit intracellular inclusions [neurofibrillary tangles (NFT's)] of microtubule-associated protein tau that contributes to neuronal dysfunction and death. Mutations in the microtubule-associated protein tau (MAPT) gene leads to tau hyperphosphorylation and promotes NFT formation. The TAU58/2 transgenic mouse model expresses mutant human tau (P301S mutation) and exhibits behavioural abnormalities relevant to dementia in early adulthood. Here we comprehensively determined the behavioural phenotype of TAU58/2 transgenic female mice at 14 months of age using test paradigms relevant to FTD and AD. TAU58/2 females showed a significant motor deficit and lower bodyweight compared to WT littermates. Transgenic females failed to habituate to the test arena in the light-dark test. Interestingly, transgenics did not exhibit an anxiolytic-like phenotype and intermediate-term spatial learning in the cheeseboard test was intact. However, a significant learning deficit was detected in the 1st trial across test days indicating impaired long-term spatial memory. In addition, the preference for a previously rewarded location was absent in transgenic females during probe trial testing. Finally, TAU58/2 mice had a defective acoustic startle response and impaired sensorimotor gating. In conclusion TAU58/2 mice exhibit several behavioural deficits that resemble those observed in human FTD and AD. Additionally, we observed a novel startle response deficit in these mice. At 14 months of age, TAU58/2 females represent a later disease stage and are therefore a potentially useful model to test efficacy of therapeutics to reverse or ameliorate behavioural deficits in post-onset tauopapthy-related neurodegenerative disorders.


Subject(s)
Behavior, Animal/physiology , Dementia/physiopathology , Disease Models, Animal , Reflex, Startle/physiology , Tauopathies/physiopathology , tau Proteins/genetics , Age Factors , Alzheimer Disease/physiopathology , Animals , Female , Frontotemporal Dementia/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , Mutant Proteins , Phenotype
7.
Front Mol Neurosci ; 13: 570586, 2020.
Article in English | MEDLINE | ID: mdl-33013322

ABSTRACT

Mitogen-activated protein (MAP) kinases are a central component in signaling networks in a multitude of mammalian cell types. This review covers recent advances on specific functions of p38 MAP kinases in cells of the central nervous system. Unique and specific functions of the four mammalian p38 kinases are found in all major cell types in the brain. Mechanisms of p38 activation and downstream phosphorylation substrates in these different contexts are outlined and how they contribute to functions of p38 in physiological and under disease conditions. Results in different model organisms demonstrated that p38 kinases are involved in cognitive functions, including functions related to anxiety, addiction behavior, neurotoxicity, neurodegeneration, and decision making. Finally, the role of p38 kinases in psychiatric and neurological conditions and the current progress on therapeutic inhibitors targeting p38 kinases are covered and implicate p38 kinases in a multitude of CNS-related physiological and disease states.

8.
Sci Rep ; 10(1): 13845, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796905

ABSTRACT

Antibodies have been explored extensively as a potential therapeutic for Alzheimer's disease, where amyloid-ß (Aß) peptides and the tau protein deposit in patient brains. While the major focus of antibody-based therapy development was on Aß, arguably with limited success in clinical trials, targeting tau has become an emerging strategy, possibly extending therapies to dementias with isolated tau pathology. Interestingly, low titres of autoantibodies to pathological tau have been described in humans and transgenic mouse models, but their pathophysiological relevance remained elusive. Here, we used two independent approaches to deplete the B-cell lineage and hence antibody formation in human P301S mutant tau transgenic mice, TAU58/2. TAU58/2 mice were either crossed with the B-cell-deficient Ighm knockout line (muMT-/-) or treated with anti-CD20 antibodies that target B-cell precursors. In both models, B-cell depletion significantly reduced astrocytosis in TAU58/2 mice. Only when B-cells were absent throughout life, in TAU58/2.muMT-/- mice, were spatial learning deficits moderately aggravated while motor performance improved as compared to B-cell-competent TAU58/2 mice. This was associated with changes in brain region-specific tau solubility. No other relevant behavioural or neuropathological changes were observed in TAU58/2 mice in the absence of B-cells/antibodies. Taken together, our data suggests that the presence of antibodies throughout life contributes to astrocytosis in TAU58/2 mice and limits learning deficits, while other deficits and neuropathological changes appear to be independent of the presence of B-cells/antibodies.


Subject(s)
Autoantibodies , B-Lymphocytes/immunology , Gliosis/genetics , Gliosis/immunology , Learning Disabilities/genetics , Learning Disabilities/immunology , tau Proteins/genetics , tau Proteins/immunology , Animals , Brain/metabolism , Disease Models, Animal , Humans , Mice, Transgenic , Mutation , tau Proteins/metabolism
9.
Acta Neuropathol ; 140(3): 279-294, 2020 09.
Article in English | MEDLINE | ID: mdl-32725265

ABSTRACT

Hyperphosphorylation of the neuronal tau protein contributes to Alzheimer's disease (AD) by promoting tau pathology and neuronal and cognitive deficits. In contrast, we have previously shown that site-specific tau phosphorylation can inhibit toxic signals induced by amyloid-ß (Aß) in mouse models. The post-synaptic mitogen-activated protein (MAP) kinase p38γ mediates this site-specific phosphorylation on tau at Threonine-205 (T205). Using a gene therapeutic approach, we draw on this neuroprotective mechanism to improve memory in two Aß-dependent mouse models of AD at stages when advanced memory deficits are present. Increasing activity of post-synaptic kinase p38γ that targets T205 in tau reduced memory deficits in symptomatic Aß-induced AD models. Reconstitution experiments with wildtype human tau or phosphorylation-deficient tauT205A showed that T205 modification is critical for downstream effects of p38γ that prevent memory impairment in APP-transgenic mice. Furthermore, genome editing of the T205 codon in the murine Mapt gene showed that this single side chain in endogenous tau critically modulates memory deficits in APP-transgenic Alzheimer's mice. Ablating the protective effect of p38γ activity by genetic p38γ deletion in a tau transgenic mouse model that expresses non-pathogenic tau rendered tau toxic and resulted in impaired memory function in the absence of human Aß. Thus, we propose that modulating neuronal p38γ activity serves as an intrinsic tau-dependent therapeutic approach to augment compromised cognition in advanced dementia.


Subject(s)
Alzheimer Disease/metabolism , Cognition Disorders/metabolism , Memory Disorders/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Cognition Disorders/genetics , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Disease Models, Animal , Memory/physiology , Memory Disorders/genetics , Mice , Mice, Transgenic
10.
Pharmacol Biochem Behav ; 196: 172970, 2020 09.
Article in English | MEDLINE | ID: mdl-32562718

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive decline, motor impairments, and accumulation of hallmark proteins, amyloid-beta (Aß) and tau. Traditionally, transgenic mouse models for AD have focused on Aß pathology, however, recently a number of tauopathy transgenic models have been developed, including the TAU58/2 transgenic model. Cannabidiol (CBD), a non-toxic constituent of the Cannabis sativa plant, has been shown to prevent and reverse cognitive deficits in Aß transgenic mouse models of AD. Importantly, the therapeutic properties of CBD on the behavioural phenotype of tauopathy mouse models have not been investigated. We assessed the impact of chronic CBD treatment (i.e. 50 mg/kg CBD i.p. administration starting 3 weeks prior to behavioural assessments) on disease-relevant behaviours of 4-month-old TAU58/2 transgenic males in paradigms for anxiety, motor functions, and cognition. TAU58/2 transgenic males demonstrated reduced body weight, anxiety and impaired motor functions. Furthermore, they demonstrated increased freezing in fear conditioning compared to wild type-like animals. Interestingly, both sociability and social recognition memory were intact in AD transgenic mice. Chronic CBD treatment did not affect behavioural changes in transgenic males. In summary, 4-month-old TAU58/2 transgenic males exhibited no deficits in social recognition memory, suggesting that motor deficits and changes in anxiety at this age do not impact on social domains. The moderate increase in fear-associated memory needs further investigation but could be related to differences in fear extinction. Future investigations will need to clarify CBD's therapeutic potential for reversing motor deficits in TAU58/2 transgenic mice by considering alternative CBD treatment designs including changed CBD dosing.


Subject(s)
Cannabidiol/administration & dosage , tau Proteins/genetics , Animals , Anxiety/genetics , Behavior, Animal , Body Weight , Extinction, Psychological , Male , Mice , Mice, Transgenic
11.
Brain ; 143(6): 1889-1904, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32375177

ABSTRACT

Hyperphosphorylation and deposition of tau in the brain characterizes frontotemporal dementia and Alzheimer's disease. Disease-associated mutations in the tau-encoding MAPT gene have enabled the generation of transgenic mouse models that recapitulate aspects of human neurodegenerative diseases, including tau hyperphosphorylation and neurofibrillary tangle formation. Here, we characterized the effects of transgenic P301S mutant human tau expression on neuronal network function in the murine hippocampus. Onset of progressive spatial learning deficits in P301S tau transgenic TAU58/2 mice were paralleled by long-term potentiation deficits and neuronal network aberrations during electrophysiological and EEG recordings. Gene-expression profiling just prior to onset of apparent deficits in TAU58/2 mice revealed a signature of immediate early genes that is consistent with neuronal network hypersynchronicity. We found that the increased immediate early gene activity was confined to neurons harbouring tau pathology, providing a cellular link between aberrant tau and network dysfunction. Taken together, our data suggest that tau pathology drives neuronal network dysfunction through hyperexcitation of individual, pathology-harbouring neurons, thereby contributing to memory deficits.


Subject(s)
Tauopathies/genetics , tau Proteins/genetics , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/pathology , Disease Models, Animal , Frontotemporal Dementia/genetics , Hippocampus/metabolism , Long-Term Potentiation/genetics , Male , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Phosphorylation , Tauopathies/physiopathology
12.
Protein Sci ; 29(5): 1196-1210, 2020 05.
Article in English | MEDLINE | ID: mdl-32189389

ABSTRACT

Mitogen-activated protein (MAP) kinase signaling is central to multiple cellular responses and processes. MAP kinase p38α is the best characterized member of the p38 MAP kinase family. Upstream factors and downstream targets of p38α have been identified in the past by conventional methods such as coimmunoprecipitation. However, a complete picture of its interaction partners and substrates in cells is lacking. Here, we employ a proximity-dependent labeling approach using biotinylation tagging to map the interactome of p38α in cultured 293T cells. Fusing the advanced biotin ligase BioID2 to the N-terminus of p38α, we used mass spectrometry to identify 37 biotin-labeled proteins that putatively interact with p38α. Gene ontology analysis confirms known upstream and downstream factors in the p38 MAP kinase cascade (e.g., MKK3, MAPKAPK2, TAB2, and c-jun). We furthermore identify a cluster of zinc finger (ZnF) domain-containing proteins that is significantly enriched among proximity-labeled interactors and is involved in gene transcription and DNA damage response. Fluorescence imaging and coimmunoprecipitation with overexpressed p38α in cells supports an interaction of p38α with ZnF protein XPA, a key factor in the DNA damage response, that is promoted by UV irradiation. These results define an extensive network of interactions of p38α in cells and new direct molecular targets of MAP kinase p38α in gene regulation and the DNA damage response.


Subject(s)
Biotinylation/methods , Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism , Carbon-Nitrogen Ligases/analysis , Carbon-Nitrogen Ligases/genetics , Escherichia coli Proteins/analysis , Escherichia coli Proteins/genetics , HEK293 Cells , Humans , Mass Spectrometry , Repressor Proteins/analysis , Repressor Proteins/genetics , Signal Transduction/genetics , Ultraviolet Rays , p38 Mitogen-Activated Protein Kinases/genetics
13.
Neuroscience ; 431: 166-175, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32058066

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease characterised by progressive cognitive decline and the accumulation of two hallmark proteins, amyloid-beta (Aß) and tau. Traditionally, transgenic mouse models for AD have generally focused on Aß pathology, however, in recent years a number of tauopathy transgenic mouse models have been developed, including the TAU58/2 mouse model. These mice develop tau pathology and neurofibrillary tangles from 2 months of age and show motor impairments and alterations in the behavioural response to elevated plus maze (EPM) testing. The cognitive and social phenotype of this model has not yet been assessed comprehensively. Furthermore, the behavioural changes seen in the EPM have previously been linked to both anxiety and disinhibitory phenotypes. Thus, this study assessed 4-month-old TAU58/2 males comprehensively for disinhibitory and social behaviours, social recognition memory, and sensorimotor gating. TAU58/2 males demonstrated reduced exploration and anxiety-like behaviours but no changes to disinhibitory behaviours, reduced sociability in the social preference test and impaired acoustic startle and prepulse inhibition. Aggressive and socio-positive behaviours were not affected except a reduction in the occurrence of nosing and anogenital sniffing. Our study identified new phenotypic characteristics of young adult male TAU58/2 transgenic mice and clarified the nature of changes detected in the behavioural response of these mice to EPM testing. Social withdrawal and inappropriate social behaviours are common symptoms in both AD and FTD patients and impaired sensorimotor gating is seen in moderate-late stage AD, emphasising the relevance of the TAU58/2 model to these diseases.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Tauopathies , Alzheimer Disease/genetics , Animals , Disease Models, Animal , Humans , Male , Mice , Mice, Transgenic , Neurofibrillary Tangles , Tauopathies/genetics , tau Proteins/genetics
14.
J Neurosci ; 39(48): 9645-9659, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31641049

ABSTRACT

Sphingosine 1-phosphate (S1P) is a potent vasculoprotective and neuroprotective signaling lipid, synthesized primarily by sphingosine kinase 2 (SK2) in the brain. We have reported pronounced loss of S1P and SK2 activity early in Alzheimer's disease (AD) pathogenesis, and an inverse correlation between hippocampal S1P levels and age in females, leading us to speculate that loss of S1P is a sensitizing influence for AD. Paradoxically, SK2 was reported to mediate amyloid ß (Aß) formation from amyloid precursor protein (APP) in vitro To determine whether loss of S1P sensitizes to Aß-mediated neurodegeneration, we investigated whether SK2 deficiency worsens pathology and memory in male J20 (PDGFB-APPSwInd) mice. SK2 deficiency greatly reduced Aß content in J20 mice, associated with significant improvements in epileptiform activity and cross-frequency coupling measured by hippocampal electroencephalography. However, several key measures of APPSwInd-dependent neurodegeneration were enhanced on the SK2-null background, despite reduced Aß burden. These included hippocampal volume loss, oligodendrocyte attrition and myelin loss, and impaired performance in Y-maze and social novelty memory tests. Inhibition of the endosomal cholesterol exporter NPC1 greatly reduced sphingosine phosphorylation in glial cells, linking loss of SK2 activity and S1P in AD to perturbed endosomal lipid metabolism. Our findings establish SK2 as an important endogenous regulator of both APP processing to Aß, and oligodendrocyte survival, in vivo These results urge greater consideration of the roles played by oligodendrocyte dysfunction and altered membrane lipid metabolic flux as drivers of neurodegeneration in AD.SIGNIFICANCE STATEMENT Genetic, neuropathological, and functional studies implicate both Aß and altered lipid metabolism and/or signaling as key pathogenic drivers of Alzheimer's disease. In this study, we first demonstrate that the enzyme SK2, which generates the signaling lipid S1P, is required for Aß formation from APP in vivo Second, we establish a new role for SK2 in the protection of oligodendrocytes and myelin. Loss of SK2 sensitizes to Aß-mediated neurodegeneration by attenuating oligodendrocyte survival and promoting hippocampal atrophy, despite reduced Aß burden. Our findings support a model in which Aß-independent sensitizing influences such as loss of neuroprotective S1P are more important drivers of neurodegeneration than gross Aß concentration or plaque density.


Subject(s)
Alzheimer Disease/metabolism , Demyelinating Diseases/metabolism , Disease Models, Animal , Hippocampus/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Plaque, Amyloid/metabolism , Alzheimer Disease/pathology , Animals , Demyelinating Diseases/pathology , Demyelinating Diseases/prevention & control , Female , Hippocampus/pathology , Male , Mice , Mice, Transgenic , Neuroprotection/physiology , Organ Culture Techniques , Organ Size/physiology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Plaque, Amyloid/pathology
15.
J Biol Chem ; 294(38): 14149-14162, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31366728

ABSTRACT

The microtubule-associated protein tau undergoes aberrant modification resulting in insoluble brain deposits in various neurodegenerative diseases, including frontotemporal dementia (FTD), progressive supranuclear palsy, and corticobasal degeneration. Tau aggregates can form in different cell types of the central nervous system (CNS) but are most prevalent in neurons. We have previously recapitulated aspects of human FTD in mouse models by overexpressing mutant human tau in CNS neurons, including a P301S tau variant in TAU58/2 mice, characterized by early-onset and progressive behavioral deficits and FTD-like neuropathology. The molecular mechanisms underlying the functional deficits of TAU58/2 mice remain mostly elusive. Here, we employed functional genomics (i.e. RNAseq) to determine differentially expressed genes in young and aged TAU58/2 mice to identify alterations in cellular processes that may contribute to neuropathy. We identified genes in cortical brain samples differentially regulated between young and old TAU58/2 mice relative to nontransgenic littermates and by comparative analysis with a dataset of CNS cell type-specific genes expressed in nontransgenic mice. Most differentially-regulated genes had known or putative roles in neurons and included presynaptic and excitatory genes. Specifically, we observed changes in presynaptic factors, glutamatergic signaling, and protein scaffolding. Moreover, in the aged mice, expression levels of several genes whose expression was annotated to occur in other brain cell types were altered. Immunoblotting and immunostaining of brain samples from the TAU58/2 mice confirmed altered expression and localization of identified and network-linked proteins. Our results have revealed genes dysregulated by progressive tau accumulation in an FTD mouse model.


Subject(s)
Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/genetics , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Central Nervous System/metabolism , Disease Models, Animal , Frontotemporal Dementia/genetics , Gene Expression Regulation/genetics , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Sequence Analysis, RNA/methods , Tauopathies/physiopathology , tau Proteins/metabolism
16.
Sci Rep ; 8(1): 14296, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250211

ABSTRACT

Modulation of behavioural responses by neuronal signalling pathways remains incompletely understood. Signalling via mitogen-activated protein (MAP) kinase cascades regulates multiple neuronal functions. Here, we show that neuronal p38α, a MAP kinase of the p38 kinase family, has a critical and specific role in modulating anxiety-related behaviour in mice. Neuron-specific p38α-knockout mice show increased levels of anxiety in behaviour tests, yet no other behavioural, cognitive or motor deficits. Using CRISPR-mediated deletion of p38α in cells, we show that p38α inhibits c-Jun N-terminal kinase (JNK) activity, a function that is specific to p38α over other p38 kinases. Consistently, brains of neuron-specific p38α-knockout mice show increased JNK activity. Inhibiting JNK using a specific blood-brain barrier-permeable inhibitor reduces JNK activity in brains of p38α-knockout mice to physiological levels and reverts anxiety behaviour. Thus, our results suggest that neuronal p38α negatively regulates JNK activity that is required for specific modulation of anxiety-related behaviour.


Subject(s)
Anxiety/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Behavior, Animal , Enzyme Activation , Mice, Inbred C57BL , Mice, Knockout
17.
Neuron ; 99(1): 13-27, 2018 07 11.
Article in English | MEDLINE | ID: mdl-30001506

ABSTRACT

The microtubule-associated protein tau and amyloid-ß (Aß) are key players in Alzheimer's disease (AD). Aß and tau are linked in a molecular pathway at the post-synapse with tau-dependent synaptic dysfunction being a major pathomechanism in AD. Recent work on site-specific modification of dendritic and more specifically post-synaptic tau has revealed new endogenous functions of tau that limits synaptic Aß toxicity. Thus, molecular studies opened a new perspective on tau, placing it at the center of neurotoxic and neuroprotective signaling at the post-synapse. Here, we review recent advances on tau in the dendritic compartments, with implications for understanding and treatment of AD and related neurological conditions.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Dendrites/metabolism , Synapses/metabolism , tau Proteins/metabolism , Humans , Neurofibrillary Tangles , Neurons/metabolism , Plaque, Amyloid/metabolism
18.
J Alzheimers Dis ; 62(2): 571-578, 2018.
Article in English | MEDLINE | ID: mdl-29480201

ABSTRACT

Alzheimer's disease and other dementias present with tau pathology. Several mouse lines with knockout of the tau-encoding Mapt gene have been reported, yet findings often differed between lines and sites. Here, we report a new tau knockout strain (tauΔex1), generated by CRISPR/Cas9-mediated genome editing of intron -1/exon 1 of Mapt in C57Bl/6J mice. TauΔex1 mice had no overt phenotype, but, in line with previous models, they showed a significantly reduced susceptibility to excitotoxic seizures, with normal memory formation in young mice. This new in vivo resource will be made freely available to the research community.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Mice, Knockout , tau Proteins/genetics , Animals , Exons , Mice , Mice, Inbred C57BL
19.
J Biol Chem ; 293(10): 3710-3719, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29382714

ABSTRACT

Compared with other mammalian species, humans are particularly susceptible to tau-mediated neurodegenerative disorders. Differential interactions of the tau protein with other proteins are critical for mediating tau's physiological functions as well as tau-associated pathological processes. Primate tau harbors an 11-amino acid-long motif in its N-terminal region (residues 18-28), which is not present in non-primate species and whose function is unknown. Here, we used deletion mutagenesis to remove this sequence region from the longest human tau isoform, followed by glutathione S-transferase (GST) pulldown assays paired with isobaric tags for relative and absolute quantitation (iTRAQ) multiplex labeling, a quantitative method to measure protein abundance by mass spectrometry. Using this method, we found that the primate-specific N-terminal tau motif differentially mediates interactions with neuronal proteins. Among these binding partners are proteins involved in synaptic transmission (synapsin-1 and synaptotagmin-1) and signaling proteins of the 14-3-3 family. Furthermore, we identified an interaction of tau with a member of the annexin family (annexin A5) that was linked to the 11-residue motif. These results suggest that primate Tau has evolved specific residues that differentially regulate protein-protein interactions compared with tau proteins from other non-primate mammalian species. Our findings provide in vitro insights into tau's interactions with other proteins that may be relevant to human disease.


Subject(s)
Nerve Tissue Proteins/metabolism , tau Proteins/metabolism , Amino Acid Sequence , Animals , Cerebral Cortex/metabolism , Computational Biology , Conserved Sequence , Gene Deletion , Gene Ontology , HEK293 Cells , Humans , Immunoprecipitation , Mice, Knockout , Mutagenesis, Site-Directed , Nerve Tissue Proteins/chemistry , Neurons/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Primates , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , tau Proteins/chemistry , tau Proteins/genetics
20.
Nat Commun ; 8(1): 473, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28883427

ABSTRACT

Neuronal excitotoxicity induced by aberrant excitation of glutamatergic receptors contributes to brain damage in stroke. Here we show that tau-deficient (tau-/-) mice are profoundly protected from excitotoxic brain damage and neurological deficits following experimental stroke, using a middle cerebral artery occlusion with reperfusion model. Mechanistically, we show that this protection is due to site-specific inhibition of glutamate-induced and Ras/ERK-mediated toxicity by accumulation of Ras-inhibiting SynGAP1, which resides in a post-synaptic complex with tau. Accordingly, reducing SynGAP1 levels in tau-/- mice abolished the protection from pharmacologically induced excitotoxicity and middle cerebral artery occlusion-induced brain damage. Conversely, over-expression of SynGAP1 prevented excitotoxic ERK activation in wild-type neurons. Our findings suggest that tau mediates excitotoxic Ras/ERK signaling by controlling post-synaptic compartmentalization of SynGAP1.Excitotoxicity contributes to neuronal injury following stroke. Here the authors show that tau promotes excitotoxicity by a post-synaptic mechanism, involving site-specific control of ERK activation, in a mouse model of stroke.


Subject(s)
Brain Injuries/genetics , Disease Models, Animal , Stroke/genetics , tau Proteins/genetics , Animals , Brain Injuries/etiology , Brain Injuries/metabolism , Cells, Cultured , Gene Expression Profiling/methods , Humans , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Signal Transduction/genetics , Stroke/etiology , Stroke/metabolism , Synaptosomes/metabolism , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , tau Proteins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...