Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Nat Immunol ; 24(11): 1839-1853, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37749326

ABSTRACT

The APOE4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease (AD). The contribution of microglial APOE4 to AD pathogenesis is unknown, although APOE has the most enriched gene expression in neurodegenerative microglia (MGnD). Here, we show in mice and humans a negative role of microglial APOE4 in the induction of the MGnD response to neurodegeneration. Deletion of microglial APOE4 restores the MGnD phenotype associated with neuroprotection in P301S tau transgenic mice and decreases pathology in APP/PS1 mice. MGnD-astrocyte cross-talk associated with ß-amyloid (Aß) plaque encapsulation and clearance are mediated via LGALS3 signaling following microglial APOE4 deletion. In the brains of AD donors carrying the APOE4 allele, we found a sex-dependent reciprocal induction of AD risk factors associated with suppression of MGnD genes in females, including LGALS3, compared to individuals homozygous for the APOE3 allele. Mechanistically, APOE4-mediated induction of ITGB8-transforming growth factor-ß (TGFß) signaling impairs the MGnD response via upregulation of microglial homeostatic checkpoints, including Inpp5d, in mice. Deletion of Inpp5d in microglia restores MGnD-astrocyte cross-talk and facilitates plaque clearance in APP/PS1 mice. We identify the microglial APOE4-ITGB8-TGFß pathway as a negative regulator of microglial response to AD pathology, and restoring the MGnD phenotype via blocking ITGB8-TGFß signaling provides a promising therapeutic intervention for AD.


Subject(s)
Alzheimer Disease , Female , Mice , Humans , Animals , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Microglia/metabolism , Galectin 3/genetics , Galectin 3/metabolism , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Disease Models, Animal
2.
Immunity ; 55(9): 1627-1644.e7, 2022 09 13.
Article in English | MEDLINE | ID: mdl-35977543

ABSTRACT

The apolipoprotein E4 (APOE4) allele is associated with an increased risk of Alzheimer disease and a decreased risk of glaucoma, but the underlying mechanisms remain poorly understood. Here, we found that in two mouse glaucoma models, microglia transitioned to a neurodegenerative phenotype characterized by upregulation of Apoe and Lgals3 (Galectin-3), which were also upregulated in human glaucomatous retinas. Mice with targeted deletion of Apoe in microglia or carrying the human APOE4 allele were protected from retinal ganglion cell (RGC) loss, despite elevated intraocular pressure (IOP). Similarly to Apoe-/- retinal microglia, APOE4-expressing microglia did not upregulate neurodegeneration-associated genes, including Lgals3, following IOP elevation. Genetic and pharmacologic targeting of Galectin-3 ameliorated RGC degeneration, and Galectin-3 expression was attenuated in human APOE4 glaucoma samples. These results demonstrate that impaired activation of APOE4 microglia is protective in glaucoma and that the APOE-Galectin-3 signaling can be targeted to treat this blinding disease.


Subject(s)
Apolipoprotein E4 , Glaucoma , Animals , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Apolipoprotein E4/therapeutic use , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Disease Models, Animal , Galectin 3/genetics , Galectin 3/metabolism , Galectin 3/therapeutic use , Glaucoma/drug therapy , Glaucoma/genetics , Glaucoma/metabolism , Humans , Mice , Microglia/metabolism
3.
Neuron ; 109(10): 1657-1674.e7, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33831349

ABSTRACT

The apolipoprotein E (APOE) gene is the strongest genetic risk factor for Alzheimer's disease and directly influences tauopathy and tau-mediated neurodegeneration. ApoE4 has strong deleterious effects on both parameters. In the brain, apoE is produced and secreted primarily by astrocytes and by activated microglia. The cell-specific role of each form of apoE in the setting of neurodegeneration has not been determined. We generated P301S Tau/Aldh1l1-CreERT2/apoE3flox/flox or Tau/Aldh1l1-CreERT2/apoE4flox/flox mice. At 5.5 months of age, after the onset of tau pathology, we administered tamoxifen or vehicle and compared mice at 9.5 months of age. Removing astrocytic APOE4 markedly reduced tau-mediated neurodegeneration and decreased phosphorylated tau (pTau) pathology. Single-nucleus RNA sequencing analysis revealed striking gene expression changes in all cell types, with astrocytic APOE4 removal decreasing disease-associated gene signatures in neurons, oligodendrocytes, astrocytes, and microglia. Removal of astrocytic APOE4 decreased tau-induced synaptic loss and microglial phagocytosis of synaptic elements, suggesting a key role for astrocytic apoE in synaptic degeneration.


Subject(s)
Apolipoprotein E4/metabolism , Astrocytes/metabolism , Phagocytosis , Tauopathies/metabolism , Animals , Apolipoprotein E4/deficiency , Apolipoprotein E4/genetics , Apoptosis , Humans , Mice , Mice, Inbred C57BL , Microglia/immunology , Synapses/metabolism , Synapses/pathology , Tauopathies/pathology , Transcriptome , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL