Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Nat Commun ; 14(1): 5197, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37626048

ABSTRACT

Alzheimer's disease, the most common age-related neurodegenerative disease, is characterized by tau aggregation and associated with disrupted circadian rhythms and dampened clock gene expression. REV-ERBα is a core circadian clock protein which also serves as a nuclear receptor and transcriptional repressor involved in lipid metabolism and macrophage function. Global REV-ERBα deletion has been shown to promote microglial activation and mitigate amyloid plaque formation. However, the cell-autonomous effects of microglial REV-ERBα in healthy brain and in tauopathy are unexplored. Here, we show that microglial REV-ERBα deletion enhances inflammatory signaling, disrupts lipid metabolism, and causes lipid droplet (LD) accumulation specifically in male microglia. These events impair microglial tau phagocytosis, which can be partially rescued by blockage of LD formation. In vivo, microglial REV-ERBα deletion exacerbates tau aggregation and neuroinflammation in two mouse tauopathy models, specifically in male mice. These data demonstrate the importance of microglial lipid droplets in tau accumulation and reveal REV-ERBα as a therapeutically accessible, sex-dependent regulator of microglial inflammatory signaling, lipid metabolism, and tauopathy.


Subject(s)
Neurodegenerative Diseases , Tauopathies , Animals , Male , Mice , Disease Models, Animal , Inflammation/genetics , Lipid Droplets , Microglia , Tauopathies/genetics
3.
Sci Transl Med ; 12(574)2020 12 16.
Article in English | MEDLINE | ID: mdl-33328329

ABSTRACT

Regulation of glial activation and neuroinflammation are critical factors in the pathogenesis of Alzheimer's disease (AD). YKL-40, a primarily astrocytic protein encoded by the gene Chi3l1, is a widely studied cerebrospinal fluid biomarker that increases with aging and early in AD. However, the function of Chi3l1/YKL-40 in AD is unknown. In a cohort of patients with AD, we observed that a variant in the human CHI3L1 gene, which results in decreased CSF YKL-40 expression, was associated with slower AD progression. At baseline, Chi3l1 deletion in mice had no effect on astrocyte activation while modestly promoting microglial activation. In a mouse APP/PS1 model of AD, Chi3l1 deletion decreased amyloid plaque burden and increased periplaque expression of the microglial lysosomal marker CD68, suggesting that Chi3l1 may suppress glial phagocytic activation and promote amyloid accumulation. Accordingly, Chi3l1 knockdown increased phagocytosis of zymosan particles and of ß-amyloid peptide in both astrocytes and microglia in vitro. We further observed that expression of Chi3l1 is regulated by the circadian clock, as deletion of the core clock proteins BMAL1 or CLOCK/NPAS2 strongly suppresses basal Chi3l1 expression, whereas deletion of the negative clock regulators PER1/PER2 increased Chi3l1 expression. Basal Chi3l1 mRNA was nonrhythmic because of a long mRNA half-life in astrocytes. However, inflammatory induction of Chi3l1 was gated by the clock. Our findings reveal Chi3l1/YKL-40 as a modulator of glial phagocytic activation and AD pathogenesis in both mice and humans and suggest that the astrocyte circadian clock regulates inflammatory Chi3l1 induction.


Subject(s)
Alzheimer Disease , Circadian Clocks , Alzheimer Disease/genetics , Amyloid beta-Peptides , Animals , Astrocytes , Chitinase-3-Like Protein 1/genetics , Circadian Clocks/genetics , Humans , Mice , Mice, Transgenic
4.
Elife ; 92020 12 01.
Article in English | MEDLINE | ID: mdl-33258449

ABSTRACT

The circadian clock regulates various aspects of brain health including microglial and astrocyte activation. Here, we report that deletion of the master clock protein BMAL1 in mice robustly increases expression of complement genes, including C4b and C3, in the hippocampus. BMAL1 regulates expression of the transcriptional repressor REV-ERBα, and deletion of REV-ERBα causes increased expression of C4b transcript in neurons and astrocytes as well as C3 protein primarily in astrocytes. REV-ERBα deletion increased microglial phagocytosis of synapses and synapse loss in the CA3 region of the hippocampus. Finally, we observed diurnal variation in the degree of microglial synaptic phagocytosis which was antiphase to REV-ERBα expression. This daily variation in microglial synaptic phagocytosis was abrogated by global REV-ERBα deletion, which caused persistently elevated synaptic phagocytosis. This work uncovers the BMAL1-REV-ERBα axis as a regulator of complement expression and synaptic phagocytosis in the brain, linking circadian proteins to synaptic regulation.


Subject(s)
CA3 Region, Hippocampal/metabolism , Circadian Rhythm , Complement System Proteins/metabolism , Microglia/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Phagocytosis , Synapses/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Animals , CA3 Region, Hippocampal/cytology , Cells, Cultured , Complement C3/genetics , Complement C3/metabolism , Complement C4/genetics , Complement C4/metabolism , Complement System Proteins/genetics , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Signal Transduction , Time Factors , Up-Regulation
5.
J Exp Neurosci ; 13: 1179069519853233, 2019.
Article in English | MEDLINE | ID: mdl-31210735

ABSTRACT

Recently, we described a role for the circadian clock protein and nuclear receptor Rev-erbα in regulating glial activation states in the brain. Deletion of Rev-erbα resulted in microglial as well as astrocytic activation, while a Rev-erbα agonist diminished the severity of lipopolysaccharide (LPS)-induced neuroinflammation. Concomitant with this glial activation is impaired neuronal health. These findings suggest that Rev-erb proteins may play critical roles in glial biology. Pertinent ideas such as the glial cell type of most importance, the translatability of these findings to human disease, and the effect of manipulating Rev-erbs in models of neurodegeneration, need to be explored further. In this commentary, we will address the potential role of Rev-erbs in neuroinflammation related to neurodegenerative diseases and speculate on Rev-erbs as potential therapeutic targets for these conditions.

6.
Proc Natl Acad Sci U S A ; 116(11): 5102-5107, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30792350

ABSTRACT

Circadian dysfunction is a common attribute of many neurodegenerative diseases, most of which are associated with neuroinflammation. Circadian rhythm dysfunction has been associated with inflammation in the periphery, but the role of the core clock in neuroinflammation remains poorly understood. Here we demonstrate that Rev-erbα, a nuclear receptor and circadian clock component, is a mediator of microglial activation and neuroinflammation. We observed time-of-day oscillation in microglial immunoreactivity in the hippocampus, which was disrupted in Rev-erbα-/- mice. Rev-erbα deletion caused spontaneous microglial activation in the hippocampus and increased expression of proinflammatory transcripts, as well as secondary astrogliosis. Transcriptomic analysis of hippocampus from Rev-erbα-/- mice revealed a predominant inflammatory phenotype and suggested dysregulated NF-κB signaling. Primary Rev-erbα-/- microglia exhibited proinflammatory phenotypes and increased basal NF-κB activation. Chromatin immunoprecipitation revealed that Rev-erbα physically interacts with the promoter regions of several NF-κB-related genes in primary microglia. Loss of Rev-erbα in primary astrocytes had no effect on basal activation but did potentiate the inflammatory response to lipopolysaccharide (LPS). In vivo, Rev-erbα-/- mice exhibited enhanced hippocampal neuroinflammatory responses to peripheral LPS injection, while pharmacologic activation of Rev-erbs with the small molecule agonist SR9009 suppressed LPS-induced hippocampal neuroinflammation. Rev-erbα deletion influenced neuronal health, as conditioned media from Rev-erbα-deficient primary glial cultures exacerbated oxidative damage in cultured neurons. Rev-erbα-/- mice also exhibited significantly altered cortical resting-state functional connectivity, similar to that observed in neurodegenerative models. Our results reveal Rev-erbα as a pharmacologically accessible link between the circadian clock and neuroinflammation.


Subject(s)
Circadian Clocks , Inflammation/metabolism , Inflammation/pathology , Neurons/metabolism , Neurons/pathology , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Death , Gene Deletion , Gliosis/pathology , Hippocampus/pathology , Lipopolysaccharides , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , NF-kappa B/metabolism , Nerve Net/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...