Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Hosp Med ; 18(8): 748-752, 2023 08.
Article in English | MEDLINE | ID: mdl-37338114
2.
Neuron ; 111(15): 2383-2398.e7, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37315555

ABSTRACT

The circadian clock protein BMAL1 modulates glial activation and amyloid-beta deposition in mice. However, the effects of BMAL1 on other aspects of neurodegenerative pathology are unknown. Here, we show that global post-natal deletion of Bmal1 in mouse tauopathy or alpha-synucleinopathy models unexpectedly suppresses both tau and alpha-synuclein (αSyn) aggregation and related pathology. Astrocyte-specific Bmal1 deletion is sufficient to prevent both αSyn and tau pathology in vivo and induces astrocyte activation and the expression of Bag3, a chaperone critical for macroautophagy. Astrocyte Bmal1 deletion enhances phagocytosis of αSyn and tau in a Bag3-dependent manner, and astrocyte Bag3 overexpression is sufficient to mitigate αSyn spreading in vivo. In humans, BAG3 is increased in patients with AD and is highly expressed in disease-associated astrocytes (DAAs). Our results suggest that early activation of astrocytes via Bmal1 deletion induces Bag3 to protect against tau and αSyn pathologies, providing new insights into astrocyte-specific therapies for neurodegeneration.


Subject(s)
Synucleinopathies , Tauopathies , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Amyloid beta-Peptides/metabolism , Apoptosis Regulatory Proteins/metabolism , ARNTL Transcription Factors/genetics , Astrocytes/metabolism , Synucleinopathies/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Tauopathies/metabolism
3.
Mol Neurodegener ; 14(1): 11, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30813965

ABSTRACT

BACKGROUND: Alzheimer's disease is characterized by two main neuropathological hallmarks: extracellular plaques of amyloid-ß (Aß) protein and intracellular aggregates of tau protein. Although tau is normally a soluble monomer that bind microtubules, in disease it forms insoluble, hyperphosphorylated aggregates in the cell body. Aside from its role in AD, tau is also involved in several other neurodegenerative disorders collectively called tauopathies, such as progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), some forms of frontotemporal dementia, and argyrophilic grain disease (AGD). The prion hypothesis suggests that after an initial trigger event, misfolded forms of tau are released into the extracellular space, where they spread through different brain regions, enter cells, and seeding previously normal forms. Thus understanding mechanisms regulating the clearance of extracellular tau from the CNS is important. The discovery of a true lymphatic system in the dura and its potential role in mediating Aß pathology prompted us to investigate its role in regulating extracellular tau clearance. METHODS: To study clearance of extracellular tau from the brain, we conjugated monomeric human tau with a near-infrared dye cypate, and injected this labeled tau in the parenchyma of both wild-type and K14-VEGFR3-Ig transgenic mice, which lack a functional CNS lymphatic system. Following injection we performed longitudinal imaging using fluorescence molecular tomography (FMT) and quantified fluorescence to calculate clearance of tau from the brain. To complement this, we also measured tau clearance to the periphery by measuring plasma tau in both groups of mice. RESULTS: Our results show that a significantly higher amount of tau is retained in the brains of K14-VEGFR3-Ig vs. wild type mice at 48 and 72 h post-injection and its subsequent clearance to the periphery is delayed. We found that clearance of reference tracer human serum albumin (HSA) was also significantly delayed in the K14-VEGFR3-Ig mice. CONCLUSIONS: The dural lymphatic system appears to play an important role in clearance of extracellular tau, since tau clearance is impaired in the absence of functional lymphatics. Based on our baseline characterization of extracellular tau clearance, future studies are warranted to look at the interaction between tau pathology and efficiency of lymphatic function.


Subject(s)
Dura Mater , Glymphatic System , Lymphatic Vessels , tau Proteins/metabolism , Animals , Humans , Mice , Mice, Transgenic
4.
J Exp Med ; 215(5): 1267-1268, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29685951

ABSTRACT

In this issue, Chiang et al. (https://doi.org/10.1084/jem.20171484) make a notable contribution to Alzheimer disease (AD) therapeutics in a thorough and rigorous study demonstrating superior efficacy of dual therapy against Aß in a mouse model of amyloid ß deposition.


Subject(s)
Alzheimer Disease , Amyloidosis , Amyloid beta-Peptides , Animals , Brain , Cognition , Disease Models, Animal , Mice , TOR Serine-Threonine Kinases
5.
Sci Transl Med ; 9(386)2017 04 19.
Article in English | MEDLINE | ID: mdl-28424326

ABSTRACT

Tauopathies are a group of disorders in which the cytosolic protein tau aggregates and accumulates in cells within the brain, resulting in neurodegeneration. A promising treatment being explored for tauopathies is passive immunization with anti-tau antibodies. We previously found that administration of an anti-tau antibody to human tau transgenic mice increased the concentration of plasma tau. We further explored the effects of administering an anti-tau antibody on plasma tau. After peripheral administration of an anti-tau antibody to human patients with tauopathy and to mice expressing human tau in the central nervous system, there was a dose-dependent increase in plasma tau. In mouse plasma, we found that tau had a short half-life of 8 min that increased to more than 3 hours after administration of anti-tau antibody. As tau transgenic mice accumulated insoluble tau in the brain, brain soluble and interstitial fluid tau decreased. Administration of anti-tau antibody to tau transgenic mice that had decreased brain soluble tau and interstitial fluid tau resulted in an increase in plasma tau, but this increase was less than that observed in tau transgenic mice without these brain changes. Tau transgenic mice subjected to acute neuronal injury using 3-nitropropionic acid showed increased interstitial fluid tau and plasma tau. These data suggest that peripheral administration of an anti-tau antibody results in increased plasma tau, which correlates with the concentration of extracellular and soluble tau in the brain.


Subject(s)
Antibodies/pharmacology , Tauopathies/blood , Tauopathies/metabolism , tau Proteins/blood , tau Proteins/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Mice , Mice, Transgenic , Nitro Compounds/toxicity , Propionates/toxicity
6.
Mol Neurodegener ; 10: 55, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26502977

ABSTRACT

BACKGROUND: Intracellular accumulation of tau as neurofibrillary tangles (NFTs) is the hallmark of Alzheimer's disease (AD) as well as in other tauopathies. Tau is present not only in the cytoplasm but also in the extracellular space such as cerebrospinal fluid (CSF) and brain interstitial fluid (ISF). Although clearance is one critical parameter leading to such intracellular/extracellular tau accumulation, in vivo turnover of tau has not been well characterized. The current study has attempted to precisely determine in vivo turnover rates of tau utilizing tet-off regulatable mice. In particular, we assessed intracellular tau and extracellular tau, soluble tau, insoluble tau and phosphorylated tau at certain sites utilizing a combination of in vivo microdialysis, biochemical analysis and specific ELISAs recognizing each species. To examine the effect of a tauopathy-associated mutation on tau clearance, half-lives of various tau species were compared between the mice with a FTDP-17 mutation that induces ß-sheet formation, ΔK280 mutation (pro-aggregant mice) and control mice with additional ß-sheet breaking mutations (anti-aggregant mice). RESULTS: Here we report that tau is metabolized at much slower turnover rates in vivo than in cell culture. We found that insoluble tau in pro-aggregant mice had a significantly slower half-life (t1/2 = ~34.2 days) than soluble tau (t1/2 = ~9.7 days). In contrast, soluble tau phosphorylated in the proline rich region was cleared faster than total soluble tau. When comparing pro-aggregant mice to anti-agregant mice, turnover rates of soluble tau species were not significantly different. CONCLUSIONS: The current study provides a comprehensive description of in vivo turnover of various tau species present in mice that express human tau. The turnover rate of soluble tau was not significantly altered between pro-aggregant mice and anti-aggregant mice. This suggests that altered conformation by ΔK280 does not have a major impact on clearance pathways for soluble tau. In contrast, different tau species displayed different half-lives. Turnover was significantly delayed for insoluble tau whereas it was accelerated for soluble tau phosphorylated in the proline rich region. These differences in susceptibilities to clearance suggest that aggregation and phosphorylation influences tau clearance which may be important in tau pathogenesis.


Subject(s)
Brain/metabolism , Frontotemporal Dementia/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Animals , Disease Models, Animal , Memory/physiology , Mice , Mice, Transgenic , Mutation/genetics , Tauopathies/genetics
7.
J Exp Med ; 211(3): 387-93, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24534188

ABSTRACT

Tau is primarily a cytoplasmic protein that stabilizes microtubules. However, it is also found in the extracellular space of the brain at appreciable concentrations. Although its presence there may be relevant to the intercellular spread of tau pathology, the cellular mechanisms regulating tau release into the extracellular space are not well understood. To test this in the context of neuronal networks in vivo, we used in vivo microdialysis. Increasing neuronal activity rapidly increased the steady-state levels of extracellular tau in vivo. Importantly, presynaptic glutamate release is sufficient to drive tau release. Although tau release occurred within hours in response to neuronal activity, the elimination rate of tau from the extracellular compartment and the brain is slow (half-life of ∼11 d). The in vivo results provide one mechanism underlying neuronal tau release and may link trans-synaptic spread of tau pathology with synaptic activity itself.


Subject(s)
Brain/metabolism , Extracellular Space/metabolism , Neurons/metabolism , Synaptic Transmission/physiology , Tauopathies/physiopathology , tau Proteins/metabolism , Analysis of Variance , Animals , Electroencephalography , Enzyme-Linked Immunosorbent Assay , Female , Glutamic Acid/metabolism , Half-Life , Kinetics , Luciferases , Male , Mice , Mice, Transgenic , Microdialysis , Tetrodotoxin
SELECTION OF CITATIONS
SEARCH DETAIL
...