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
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.
EMBO J ; 36(13): 1837-1853, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28559417

ABSTRACT

Genetic variants in the triggering receptor expressed on myeloid cells 2 (TREM2) increase the risk for several neurodegenerative diseases including Alzheimer's disease and frontotemporal dementia (FTD). Homozygous TREM2 missense mutations, such as p.T66M, lead to the FTD-like syndrome, but how they cause pathology is unknown. Using CRISPR/Cas9 genome editing, we generated a knock-in mouse model for the disease-associated Trem2 p.T66M mutation. Consistent with a loss-of-function mutation, we observe an intracellular accumulation of immature mutant Trem2 and reduced generation of soluble Trem2 similar to patients with the homozygous p.T66M mutation. Trem2 p.T66M knock-in mice show delayed resolution of inflammation upon in vivo lipopolysaccharide stimulation and cultured macrophages display significantly reduced phagocytic activity. Immunohistochemistry together with in vivo TSPO small animal positron emission tomography (µPET) demonstrates an age-dependent reduction in microglial activity. Surprisingly, perfusion magnetic resonance imaging and FDG-µPET imaging reveal a significant reduction in cerebral blood flow and brain glucose metabolism. Thus, we demonstrate that a TREM2 loss-of-function mutation causes brain-wide metabolic alterations pointing toward a possible function of microglia in regulating brain glucose metabolism.


Subject(s)
Brain/pathology , Frontotemporal Dementia/pathology , Glucose/metabolism , Membrane Glycoproteins/genetics , Microglia/physiology , Mutation, Missense , Perfusion , Receptors, Immunologic/genetics , Animals , Disease Models, Animal , Gene Knock-In Techniques , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mutant Proteins/genetics , Positron-Emission Tomography
3.
EMBO Rep ; 18(7): 1186-1198, 2017 07.
Article in English | MEDLINE | ID: mdl-28483841

ABSTRACT

Sequence variations in the triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to an increased risk for neurodegenerative disorders such as Alzheimer's disease and frontotemporal lobar degeneration. In the brain, TREM2 is predominantly expressed in microglia. Several disease-associated TREM2 variants result in a loss of function by reducing microglial phagocytosis, impairing lipid sensing, preventing binding of lipoproteins and affecting shielding of amyloid plaques. We here investigate the consequences of TREM2 loss of function on the microglia transcriptome. Among the differentially expressed messenger RNAs in wild-type and Trem2-/- microglia, gene clusters are identified which represent gene functions in chemotaxis, migration and mobility. Functional analyses confirm that loss of TREM2 impairs appropriate microglial responses to injury and signals that normally evoke chemotaxis on multiple levels. In an ex vivo organotypic brain slice assay, absence of TREM2 reduces the distance migrated by microglia. Moreover, migration towards defined chemo-attractants is reduced upon ablation of TREM2 and can be rescued by TREM2 re-expression. In vivo, microglia lacking TREM2 migrate less towards injected apoptotic neurons, and outgrowth of microglial processes towards sites of laser-induced focal CNS damage in the somatosensory cortex is slowed. The apparent lack of chemotactic stimulation upon depletion of TREM2 is consistent with a stable expression profile of genes characterizing the homoeostatic signature of microglia.


Subject(s)
Chemotaxis , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Microglia/physiology , Neurons/pathology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Cells, Cultured , Frontotemporal Dementia , Gene Expression Profiling , Humans , Loss of Function Mutation , Myeloid Cells , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Phagocytosis
4.
EMBO Mol Med ; 8(9): 992-1004, 2016 09.
Article in English | MEDLINE | ID: mdl-27402340

ABSTRACT

Immunotherapeutic approaches are currently the most advanced treatments for Alzheimer's disease (AD). Antibodies against amyloid ß-peptide (Aß) bind to amyloid plaques and induce their clearance by microglia via Fc receptor-mediated phagocytosis. Dysfunctions of microglia may play a pivotal role in AD pathogenesis and could result in reduced efficacy of antibody-mediated Aß clearance. Recently, heterozygous mutations in the triggering receptor expressed on myeloid cells 2 (TREM2), a microglial gene involved in phagocytosis, were genetically linked to late onset AD Loss of TREM2 reduces the ability of microglia to engulf Aß. We have now investigated whether loss of TREM2 affects the efficacy of immunotherapeutic approaches. We show that anti-Aß antibodies stimulate Aß uptake and amyloid plaque clearance in a dose-dependent manner in the presence or absence of TREM2. However, TREM2-deficient N9 microglial cell lines, macrophages as well as primary microglia showed significantly reduced uptake of antibody-bound Aß and as a consequence reduced clearance of amyloid plaques. Titration experiments revealed that reduced efficacy of amyloid plaque clearance by Trem2 knockout cells can be compensated by elevating the concentration of therapeutic antibodies.


Subject(s)
Amyloid/metabolism , Membrane Glycoproteins/deficiency , Neuroglia/immunology , Phagocytosis , Receptors, Immunologic/deficiency , Animals , Cells, Cultured , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/physiology
5.
Sci Transl Med ; 6(243): 243ra86, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-24990881

ABSTRACT

Genetic variants in the triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to Nasu-Hakola disease, Alzheimer's disease (AD), Parkinson's disease, amyotrophic lateral sclerosis, frontotemporal dementia (FTD), and FTD-like syndrome without bone involvement. TREM2 is an innate immune receptor preferentially expressed by microglia and is involved in inflammation and phagocytosis. Whether and how TREM2 missense mutations affect TREM2 function is unclear. We report that missense mutations associated with FTD and FTD-like syndrome reduce TREM2 maturation, abolish shedding by ADAM proteases, and impair the phagocytic activity of TREM2-expressing cells. As a consequence of reduced shedding, TREM2 is virtually absent in the cerebrospinal fluid (CSF) and plasma of a patient with FTD-like syndrome. A decrease in soluble TREM2 was also observed in the CSF of patients with AD and FTD, further suggesting that reduced TREM2 function may contribute to increased risk for two neurodegenerative disorders.


Subject(s)
Membrane Glycoproteins/genetics , Neurodegenerative Diseases/genetics , Phagocytosis/physiology , Receptors, Immunologic/genetics , Alzheimer Disease/genetics , Biological Transport/genetics , Biological Transport/physiology , Cell Line , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Frontotemporal Dementia/genetics , Humans , Mutation , Phagocytosis/genetics
6.
Nat Commun ; 5: 4046, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24898390

ABSTRACT

The removal of dying neurons by microglia has a key role during both development and in several diseases. To date, little is known about the cellular and molecular processes underlying neuronal engulfment in the brain. Here we took a live imaging approach to quantify neuronal cell death progression in embryonic zebrafish brains and studied the response of microglia. We show that microglia engulf dying neurons by extending cellular branches that form phagosomes at their tips. At the molecular level we found that microglia lacking the phosphatidylserine receptors BAI1 and TIM-4, are able to recognize the apoptotic targets but display distinct clearance defects. Indeed, BAI1 controls the formation of phagosomes around dying neurons and cargo transport, whereas TIM-4 is required for phagosome stabilization. Using this single-cell resolution approach we established that it is the combined activity of BAI1 and TIM-4 that allows microglia to remove dying neurons.


Subject(s)
Angiogenic Proteins/metabolism , Apoptosis/physiology , Membrane Proteins/metabolism , Microglia/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Phagocytosis/physiology , Receptors, Cell Surface/metabolism , Zebrafish Proteins/metabolism , Animals , Hepatitis A Virus Cellular Receptor 1 , Microglia/physiology , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...