Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Immunity ; 47(3): 566-581.e9, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28930663

ABSTRACT

Microglia play a pivotal role in the maintenance of brain homeostasis but lose homeostatic function during neurodegenerative disorders. We identified a specific apolipoprotein E (APOE)-dependent molecular signature in microglia from models of amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and Alzheimer's disease (AD) and in microglia surrounding neuritic ß-amyloid (Aß)-plaques in the brains of people with AD. The APOE pathway mediated a switch from a homeostatic to a neurodegenerative microglia phenotype after phagocytosis of apoptotic neurons. TREM2 (triggering receptor expressed on myeloid cells 2) induced APOE signaling, and targeting the TREM2-APOE pathway restored the homeostatic signature of microglia in ALS and AD mouse models and prevented neuronal loss in an acute model of neurodegeneration. APOE-mediated neurodegenerative microglia had lost their tolerogenic function. Our work identifies the TREM2-APOE pathway as a major regulator of microglial functional phenotype in neurodegenerative diseases and serves as a novel target that could aid in the restoration of homeostatic microglia.


Subject(s)
Apolipoproteins E/metabolism , Membrane Glycoproteins/metabolism , Microglia/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Transcriptome , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis/genetics , Apoptosis/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cluster Analysis , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Targeting , Humans , Immune Tolerance , Mice , Mice, Knockout , Mice, Transgenic , Microglia/immunology , Monocytes/immunology , Monocytes/metabolism , Neurodegenerative Diseases/immunology , Neurons/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Phenotype , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Transforming Growth Factor beta/metabolism
2.
Ann Neurol ; 77(1): 75-99, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25381879

ABSTRACT

OBJECTIVE: To investigate miR-155 in the SOD1 mouse model and human sporadic and familial amyotrophic lateral sclerosis (ALS). METHODS: NanoString microRNA, microglia and immune gene profiles, protein mass spectrometry, and RNA-seq analyses were measured in spinal cord microglia, splenic monocytes, and spinal cord tissue from SOD1 mice and in spinal cord tissue of familial and sporadic ALS. miR-155 was targeted by genetic ablation or by peripheral or centrally administered anti-miR-155 inhibitor in SOD1 mice. RESULTS: In SOD1 mice, we found loss of the molecular signature that characterizes homeostatic microglia and increased expression of miR-155. There was loss of the microglial molecules P2ry12, Tmem119, Olfml3, transcription factors Egr1, Atf3, Jun, Fos, and Mafb, and the upstream regulators Csf1r, Tgfb1, and Tgfbr1, which are essential for microglial survival. Microglia biological functions were suppressed including phagocytosis. Genetic ablation of miR-155 increased survival in SOD1 mice by 51 days in females and 27 days in males and restored the abnormal microglia and monocyte molecular signatures. Disease severity in SOD1 males was associated with early upregulation of inflammatory genes, including Apoe in microglia. Treatment of adult microglia with apolipoprotein E suppressed the M0-homeostatic unique microglia signature and induced an M1-like phenotype. miR-155 expression was increased in the spinal cord of both familial and sporadic ALS. Dysregulated proteins that we identified in human ALS spinal cord were restored in SOD1(G93A) /miR-155(-/-) mice. Intraventricular anti-miR-155 treatment derepressed microglial miR-155 targeted genes, and peripheral anti-miR-155 treatment prolonged survival. INTERPRETATION: We found overexpression of miR-155 in the SOD1 mouse and in both sporadic and familial human ALS. Targeting miR-155 in SOD1 mice restores dysfunctional microglia and ameliorates disease. These findings identify miR-155 as a therapeutic target for the treatment of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Gene Expression Regulation/genetics , MicroRNAs/metabolism , Spinal Cord/pathology , Superoxide Dismutase/genetics , Aged , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Animals , Apolipoproteins E/pharmacology , Apolipoproteins E/therapeutic use , Cells, Cultured , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Hippocampus/cytology , Humans , Male , Mice , Mice, Transgenic , MicroRNAs/chemistry , MicroRNAs/genetics , Microglia/drug effects , Microglia/metabolism , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Neurons/drug effects , Neurons/metabolism , Oligoribonucleotides, Antisense/therapeutic use , Phagocytosis/drug effects , Phagocytosis/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
3.
Brain Pathol ; 27(5): 590-602, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27558169

ABSTRACT

Prion diseases are fatal transmissible diseases, where conversion of the endogenous prion protein (PrPC ) into a misfolded isoform (PrPSc ) leads to neurodegeneration. Microglia, the immune cells of the brain, are activated in neurodegenerative disorders including prion diseases; however, their impact on prion disease pathophysiology is unclear with both beneficial PrPSc -clearing and detrimental potentially neurotoxic effects. Moreover, monocytes entering the brain from the periphery during disease course might add to disease pathophysiology. Here, the degree of microglia activation in the brain of prion infected mice with and without an additional intraperitoneal retrovirus infection was studied. Peripheral murine retrovirus infection leads to activation of parenchymal microglia without recruitment of monocytes. This activation correlated with transient clearance or delay in accumulation of infectious prions specifically from the brain at early time points in the diseases course. Microglia expression profiling showed upregulation of genes involved in protein degradation coinciding with prion clearance. This enforces a concept where microglia act beneficial in prion disease if adequately activated. Once microglia activation has ceased, prion disease reemerges leading to disease kinetics undistinguishable from the situation in prion-only infected mice. This might be caused by the loss of microglial homeostatic function at clinical prion disease.


Subject(s)
Brain/immunology , Microglia/immunology , Prion Diseases/immunology , Prions/immunology , Retroviridae Infections/immunology , Animals , Infectious Disease Incubation Period , Mice , Microglia/metabolism , Monocytes/immunology , Prion Diseases/complications , Proteolysis , Retroviridae Infections/complications
4.
Nat Neurosci ; 17(1): 131-43, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24316888

ABSTRACT

Microglia are myeloid cells of the CNS that participate both in normal CNS function and in disease. We investigated the molecular signature of microglia and identified 239 genes and 8 microRNAs that were uniquely or highly expressed in microglia versus myeloid and other immune cells. Of the 239 genes, 106 were enriched in microglia as compared with astrocytes, oligodendrocytes and neurons. This microglia signature was not observed in microglial lines or in monocytes recruited to the CNS, and was also observed in human microglia. We found that TGF-ß was required for the in vitro development of microglia that express the microglial molecular signature characteristic of adult microglia and that microglia were absent in the CNS of TGF-ß1-deficient mice. Our results identify a unique microglial signature that is dependent on TGF-ß signaling and provide insights into microglial biology and the possibility of targeting microglia for the treatment of CNS disease.


Subject(s)
Central Nervous System/cytology , Microglia/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta1/metabolism , Analysis of Variance , Animals , Animals, Newborn , Antigens, CD/metabolism , Cells, Cultured , Chromatography, Ion Exchange , Embryo, Mammalian , Female , Flow Cytometry , Gene Expression Regulation, Developmental/physiology , Humans , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Microglia/classification , Neurons/metabolism , Receptors, Purinergic P2Y12/metabolism , Tissue Array Analysis , Transforming Growth Factor beta1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL