Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Glia ; 72(7): 1340-1355, 2024 Jul.
Article En | MEDLINE | ID: mdl-38597386

Several in vivo studies have shown that systemic inflammation, mimicked by LPS, triggers an inflammatory response in the CNS, driven by microglia, characterized by an increase in inflammatory cytokines and associated sickness behavior. However, most studies induce relatively high systemic inflammation, not directly compared with the more common low-grade inflammatory events experienced in humans during the life course. Using mice, we investigated the effects of low-grade systemic inflammation during an otherwise healthy early life, and how this may precondition the onset and severity of Alzheimer's disease (AD)-like pathology. Our results indicate that low-grade systemic inflammation induces sub-threshold brain inflammation and promotes microglial proliferation driven by the CSF1R pathway, contrary to the effects caused by high systemic inflammation. In addition, repeated systemic challenges with low-grade LPS induce disease-associated microglia. Finally, using an inducible model of AD-like pathology (Line 102 mice), we observed that preconditioning with repeated doses of low-grade systemic inflammation, prior to APP induction, promotes a detrimental effect later in life, leading to an increase in Aß accumulation and disease-associated microglia. These results support the notion that episodic low-grade systemic inflammation has the potential to influence the onset and severity of age-related neurological disorders, such as AD.


Alzheimer Disease , Inflammation , Lipopolysaccharides , Mice, Inbred C57BL , Mice, Transgenic , Microglia , Animals , Microglia/metabolism , Microglia/pathology , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Inflammation/pathology , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Disease Models, Animal , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Male , Female , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Brain/pathology , Brain/metabolism , Amyloid beta-Peptides/metabolism , Cytokines/metabolism
2.
Cell Rep ; 35(10): 109228, 2021 06 08.
Article En | MEDLINE | ID: mdl-34107254

The sustained proliferation of microglia is a key hallmark of Alzheimer's disease (AD), accelerating its progression. Here, we aim to understand the long-term impact of the early and prolonged microglial proliferation observed in AD, hypothesizing that extensive and repeated cycling would engender a distinct transcriptional and phenotypic trajectory. We show that the early and sustained microglial proliferation seen in an AD-like model promotes replicative senescence, characterized by increased ßgal activity, a senescence-associated transcriptional signature, and telomere shortening, correlating with the appearance of disease-associated microglia (DAM) and senescent microglial profiles in human post-mortem AD cases. The prevention of early microglial proliferation hinders the development of senescence and DAM, impairing the accumulation of Aß, as well as associated neuritic and synaptic damage. Overall, our results indicate that excessive microglial proliferation leads to the generation of senescent DAM, which contributes to early Aß pathology in AD.


Amyloid beta-Peptides/genetics , Cellular Senescence/genetics , Microglia/metabolism , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic
3.
Cell Rep ; 18(2): 391-405, 2017 01 10.
Article En | MEDLINE | ID: mdl-28076784

Microglia play key roles in brain development, homeostasis, and function, and it is widely assumed that the adult population is long lived and maintained by self-renewal. However, the precise temporal and spatial dynamics of the microglial population are unknown. We show in mice and humans that the turnover of microglia is remarkably fast, allowing the whole population to be renewed several times during a lifetime. The number of microglial cells remains steady from late postnatal stages until aging and is maintained by the spatial and temporal coupling of proliferation and apoptosis, as shown by pulse-chase studies, chronic in vivo imaging of microglia, and the use of mouse models of dysregulated apoptosis. Our results reveal that the microglial population is constantly and rapidly remodeled, expanding our understanding of its role in the maintenance of brain homeostasis.


Aging/physiology , Apoptosis , Brain/cytology , Microglia/cytology , Animals , Cell Count , Cell Proliferation , Gene Expression Profiling , Homeostasis , Humans , Mice , Microglia/metabolism , Monocytes/cytology , Monocytes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors
4.
Sci Rep ; 6: 25663, 2016 05 13.
Article En | MEDLINE | ID: mdl-27174644

Inflammation is a common neuropathological feature in several neurological disorders, including amyotrophic lateral sclerosis (ALS). We have studied the contribution of CSF1R signalling to inflammation in ALS, as a pathway previously reported to control the expansion and activation of microglial cells. We found that microglial cell proliferation in the spinal cord of SOD1(G93A) transgenic mice correlates with the expression of CSF1R and its ligand CSF1. Administration of GW2580, a selective CSF1R inhibitor, reduced microglial cell proliferation in SOD1(G93A) mice, indicating the importance of CSF1-CSF1R signalling in microgliosis in ALS. Moreover, GW2580 treatment slowed disease progression, attenuated motoneuron cell death and extended survival of SOD1(G93A) mice. Electrophysiological assessment revealed that GW2580 treatment protected skeletal muscle from denervation prior to its effects on microglial cells. We found that macrophages invaded the peripheral nerve of ALS mice before CSF1R-induced microgliosis occurred. Interestingly, treatment with GW2580 attenuated the influx of macrophages into the nerve, which was partly caused by the monocytopenia induced by CSF1R inhibition. Overall, our findings provide evidence that CSF1R signalling regulates inflammation in the central and peripheral nervous system in ALS, supporting therapeutic targeting of CSF1R in this disease.


Amyotrophic Lateral Sclerosis/metabolism , Macrophages/metabolism , Microglia/metabolism , Peripheral Nerves/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Anisoles/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Disease Progression , Gliosis/genetics , Gliosis/metabolism , Inflammation/genetics , Inflammation/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Motor Neurons/metabolism , Pyrimidines/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
5.
Glia ; 64(5): 826-39, 2016 May.
Article En | MEDLINE | ID: mdl-26847266

The past decade has witnessed a revolution in our understanding of microglia. These immune cells were shown to actively remodel neuronal circuits, leading to propose new pathogenic mechanisms. To study microglial implication in the loss of synapses, the best pathological correlate of cognitive decline across chronic stress, aging, and diseases, we recently conducted ultrastructural analyses. Our work uncovered the existence of a new microglial phenotype that is rarely present under steady state conditions, in hippocampus, cerebral cortex, amygdala, and hypothalamus, but becomes abundant during chronic stress, aging, fractalkine signaling deficiency (CX3 CR1 knockout mice), and Alzheimer's disease pathology (APP-PS1 mice). Even though these cells display ultrastructural features of microglia, they are strikingly distinct from the other phenotypes described so far at the ultrastructural level. They exhibit several signs of oxidative stress, including a condensed, electron-dense cytoplasm and nucleoplasm making them as "dark" as mitochondria, accompanied by a pronounced remodeling of their nuclear chromatin. Dark microglia appear to be much more active than the normal microglia, reaching for synaptic clefts, while extensively encircling axon terminals and dendritic spines with their highly ramified and thin processes. They stain for the myeloid cell markers IBA1 and GFP (in CX3 CR1-GFP mice), and strongly express CD11b and microglia-specific 4D4 in their processes encircling synaptic elements, and TREM2 when they associate with amyloid plaques. Overall, these findings suggest that dark microglia, a new phenotype that we identified based on their unique properties, could play a significant role in the pathological remodeling of neuronal circuits, especially at synapses.


Aging/pathology , Alzheimer Disease/pathology , Cerebral Cortex/pathology , Microglia/pathology , Stress, Psychological/pathology , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Antigens, CD/metabolism , CX3C Chemokine Receptor 1 , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-NH Group Donors , Phenotype , Presenilin-1/genetics , Presenilin-1/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Stress, Psychological/genetics
6.
Brain ; 139(Pt 3): 891-907, 2016 Mar.
Article En | MEDLINE | ID: mdl-26747862

The proliferation and activation of microglial cells is a hallmark of several neurodegenerative conditions. This mechanism is regulated by the activation of the colony-stimulating factor 1 receptor (CSF1R), thus providing a target that may prevent the progression of conditions such as Alzheimer's disease. However, the study of microglial proliferation in Alzheimer's disease and validation of the efficacy of CSF1R-inhibiting strategies have not yet been reported. In this study we found increased proliferation of microglial cells in human Alzheimer's disease, in line with an increased upregulation of the CSF1R-dependent pro-mitogenic cascade, correlating with disease severity. Using a transgenic model of Alzheimer's-like pathology (APPswe, PSEN1dE9; APP/PS1 mice) we define a CSF1R-dependent progressive increase in microglial proliferation, in the proximity of amyloid-ß plaques. Prolonged inhibition of CSF1R in APP/PS1 mice by an orally available tyrosine kinase inhibitor (GW2580) resulted in the blockade of microglial proliferation and the shifting of the microglial inflammatory profile to an anti-inflammatory phenotype. Pharmacological targeting of CSF1R in APP/PS1 mice resulted in an improved performance in memory and behavioural tasks and a prevention of synaptic degeneration, although these changes were not correlated with a change in the number of amyloid-ß plaques. Our results provide the first proof of the efficacy of CSF1R inhibition in models of Alzheimer's disease, and validate the application of a therapeutic strategy aimed at modifying CSF1R activation as a promising approach to tackle microglial activation and the progression of Alzheimer's disease.


Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Cell Proliferation/drug effects , Disease Progression , Drug Delivery Systems , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Alzheimer Disease/metabolism , Animals , Anisoles/administration & dosage , Cell Proliferation/physiology , Drug Delivery Systems/methods , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/prevention & control , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
7.
Brain Behav Immun ; 55: 179-190, 2016 07.
Article En | MEDLINE | ID: mdl-26541819

Neurogenesis is altered in neurodegenerative disorders, partly regulated by inflammatory factors. We have investigated whether microglia, the innate immune brain cells, regulate hippocampal neurogenesis in neurodegeneration. Using the ME7 model of prion disease we applied gain- or loss-of CSF1R function, as means to stimulate or inhibit microglial proliferation, respectively, to dissect the contribution of these cells to neurogenesis. We found that increased hippocampal neurogenesis correlates with the expansion of the microglia population. The selective inhibition of microglial proliferation caused a reduction in neurogenesis and a restoration of normal neuronal differentiation, supporting a pro-neurogenic role for microglia. Using a gene screening strategy, we identified TGFß as a molecule controlling the microglial pro-neurogenic response in chronic neurodegeneration, supported by loss-of-function mechanistic experiments. By the selective targeting of microglial proliferation we have been able to uncover a pro-neurogenic role for microglia in chronic neurodegeneration, suggesting promising therapeutic targets to normalise the neurogenic niche during neurodegeneration.


Hippocampus/physiology , Microglia/physiology , Neurogenesis/physiology , Prion Diseases/physiopathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Transforming Growth Factor beta/physiology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL
...