Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
2.
Proc Natl Acad Sci U S A ; 117(9): 5006-5015, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071246

ABSTRACT

Multiple G protein-coupled receptors (GPCRs) are targets in the treatment of dementia, and the arrestins are common to their signaling. ß-Arrestin2 was significantly increased in brains of patients with frontotemporal lobar degeneration (FTLD-tau), a disease second to Alzheimer's as a cause of dementia. Genetic loss and overexpression experiments using genetically encoded reporters and defined mutant constructs in vitro, and in cell lines, primary neurons, and tau P301S mice crossed with ß-arrestin2-/- mice, show that ß-arrestin2 stabilizes pathogenic tau and promotes tau aggregation. Cell and mouse models of FTLD showed this to be maladaptive, fueling a positive feedback cycle of enhanced neuronal tau via non-GPCR mechanisms. Genetic ablation of ß-arrestin2 markedly ablates tau pathology and rescues synaptic plasticity defects in tau P301S transgenic mice. Atomic force microscopy and cellular studies revealed that oligomerized, but not monomeric, ß-arrestin2 increases tau by inhibiting self-interaction of the autophagy cargo receptor p62/SQSTM1, impeding p62 autophagy flux. Hence, reduction of oligomerized ß-arrestin2 with virus encoding ß-arrestin2 mutants acting as dominant-negatives markedly reduces tau-laden neurofibrillary tangles in FTLD mice in vivo. Reducing ß-arrestin2 oligomeric status represents a new strategy to alleviate tau pathology in FTLD and related tauopathies.


Subject(s)
Frontotemporal Dementia/pathology , beta-Arrestin 2/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Autophagy , Brain/metabolism , Brain/pathology , Disease Models, Animal , Frontotemporal Dementia/metabolism , Frontotemporal Lobar Degeneration/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Transcriptome , beta-Arrestin 2/genetics
3.
Commun Biol ; 2: 112, 2019.
Article in English | MEDLINE | ID: mdl-30911686

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common form of dementia. While the accumulation of Aß is pivotal to the etiology of AD, both the microtubule-associated protein tau (MAPT) and the F-actin severing protein cofilin are necessary for the deleterious effects of Aß. However, the molecular link between tau and cofilin remains unclear. In this study, we found that cofilin competes with tau for direct microtubule binding in vitro, in cells, and in vivo, which inhibits tau-induced microtubule assembly. Genetic reduction of cofilin mitigates tauopathy and synaptic defects in Tau-P301S mice and movement deficits in tau transgenic C. elegans. The pathogenic effects of cofilin are selectively mediated by activated cofilin, as active but not inactive cofilin selectively interacts with tubulin, destabilizes microtubules, and promotes tauopathy. These results therefore indicate that activated cofilin plays an essential intermediary role in neurotoxic signaling that promotes tauopathy.


Subject(s)
Actin Depolymerizing Factors/genetics , Microtubules/metabolism , Tauopathies/etiology , Tauopathies/metabolism , Transcriptional Activation , tau Proteins/genetics , tau Proteins/metabolism , Actin Depolymerizing Factors/metabolism , Animals , Caenorhabditis elegans , Disease Models, Animal , Mice , Mice, Knockout , Neurons/metabolism , Protein Binding , Tubulin/metabolism
4.
Nat Commun ; 8: 15558, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28585542

ABSTRACT

Although multiple CHCHD10 mutations are associated with the spectrum of familial and sporadic frontotemporal dementia-amyotrophic lateral sclerosis (FTD-ALS) diseases, neither the normal function of endogenous CHCHD10 nor its role in the pathological milieu (that is, TDP-43 pathology) of FTD/ALS have been investigated. In this study, we made a series of observations utilizing Caenorhabditis elegans models, mammalian cell lines, primary neurons and mouse brains, demonstrating that CHCHD10 normally exerts a protective role in mitochondrial and synaptic integrity as well as in the retention of nuclear TDP-43, whereas FTD/ALS-associated mutations (R15L and S59L) exhibit loss of function phenotypes in C. elegans genetic complementation assays and dominant negative activities in mammalian systems, resulting in mitochondrial/synaptic damage and cytoplasmic TDP-43 accumulation. As such, our results provide a pathological link between CHCHD10-associated mitochondrial/synaptic dysfunction and cytoplasmic TDP-43 inclusions.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Caenorhabditis elegans/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/genetics , Mitochondrial Proteins/genetics , Synapses/metabolism , Animals , Cell Line , Cytoplasm/metabolism , Genetic Complementation Test , HEK293 Cells , Humans , Mice , Microscopy, Fluorescence , Mitochondria/metabolism , Mutation , NIH 3T3 Cells , Neurons/metabolism , Phenotype , Protein Binding , Protein Domains , RNA, Small Interfering/metabolism , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...