Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Annu Rev Immunol ; 39: 251-277, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33556248

ABSTRACT

The immune system of the central nervous system (CNS) consists primarily of innate immune cells. These are highly specialized macrophages found either in the parenchyma, called microglia, or at the CNS interfaces, such as leptomeningeal, perivascular, and choroid plexus macrophages. While they were primarily thought of as phagocytes, their function extends well beyond simple removal of cell debris during development and diseases. Brain-resident innate immune cells were found to be plastic, long-lived, and host to an outstanding number of risk genes for multiple pathologies. As a result, they are now considered the most suitable targets for modulating CNS diseases. Additionally, recent single-cell technologies enhanced our molecular understanding of their origins, fates, interactomes, and functional cell statesduring health and perturbation. Here, we review the current state of our understanding and challenges of the myeloid cell biology in the CNS and treatment options for related diseases.


Subject(s)
Central Nervous System , Microglia , Animals , Brain , Humans , Macrophages , Myeloid Cells
2.
Cell ; 186(7): 1309-1327, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001498

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of the central nervous system afflicting nearly three million individuals worldwide. Neuroimmune interactions between glial, neural, and immune cells play important roles in MS pathology and offer potential targets for therapeutic intervention. Here, we review underlying risk factors, mechanisms of MS pathogenesis, available disease modifying therapies, and examine the value of emerging technologies, which may address unmet clinical needs and identify novel therapeutic targets.


Subject(s)
Multiple Sclerosis , Humans , Multiple Sclerosis/drug therapy , Central Nervous System , Neuroglia , Cell Physiological Phenomena , Inflammation/pathology
3.
Nat Immunol ; 25(3): 432-447, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38409259

ABSTRACT

Central nervous system (CNS)-resident cells such as microglia, oligodendrocytes and astrocytes are gaining increasing attention in respect to their contribution to CNS pathologies including multiple sclerosis (MS). Several studies have demonstrated the involvement of pro-inflammatory glial subsets in the pathogenesis and propagation of inflammatory events in MS and its animal models. However, it has only recently become clear that the underlying heterogeneity of astrocytes and microglia can not only drive inflammation, but also lead to its resolution through direct and indirect mechanisms. Failure of these tissue-protective mechanisms may potentiate disease and increase the risk of conversion to progressive stages of MS, for which currently available therapies are limited. Using proteomic analyses of cerebrospinal fluid specimens from patients with MS in combination with experimental studies, we here identify Heparin-binding EGF-like growth factor (HB-EGF) as a central mediator of tissue-protective and anti-inflammatory effects important for the recovery from acute inflammatory lesions in CNS autoimmunity. Hypoxic conditions drive the rapid upregulation of HB-EGF by astrocytes during early CNS inflammation, while pro-inflammatory conditions suppress trophic HB-EGF signaling through epigenetic modifications. Finally, we demonstrate both anti-inflammatory and tissue-protective effects of HB-EGF in a broad variety of cell types in vitro and use intranasal administration of HB-EGF in acute and post-acute stages of autoimmune neuroinflammation to attenuate disease in a preclinical mouse model of MS. Altogether, we identify astrocyte-derived HB-EGF and its epigenetic regulation as a modulator of autoimmune CNS inflammation and potential therapeutic target in MS.


Subject(s)
Astrocytes , Multiple Sclerosis , Animals , Humans , Mice , Anti-Inflammatory Agents , Disease Models, Animal , Epigenesis, Genetic , Heparin-binding EGF-like Growth Factor/genetics , Inflammation , Proteomics
4.
Cell ; 176(3): 581-596.e18, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30661753

ABSTRACT

Genome-wide studies have identified genetic variants linked to neurologic diseases. Environmental factors also play important roles, but no methods are available for their comprehensive investigation. We developed an approach that combines genomic data, screens in a novel zebrafish model, computational modeling, perturbation studies, and multiple sclerosis (MS) patient samples to evaluate the effects of environmental exposure on CNS inflammation. We found that the herbicide linuron amplifies astrocyte pro-inflammatory activities by activating signaling via sigma receptor 1, inositol-requiring enzyme-1α (IRE1α), and X-box binding protein 1 (XBP1). Indeed, astrocyte-specific shRNA- and CRISPR/Cas9-driven gene inactivation combined with RNA-seq, ATAC-seq, ChIP-seq, and study of patient samples suggest that IRE1α-XBP1 signaling promotes CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, MS. In summary, these studies define environmental mechanisms that control astrocyte pathogenic activities and establish a multidisciplinary approach for the systematic investigation of the effects of environmental exposure in neurologic disorders.


Subject(s)
Astrocytes/metabolism , Central Nervous System/metabolism , Animals , Central Nervous System/immunology , Computational Biology/methods , Encephalomyelitis, Autoimmune, Experimental/immunology , Endoribonucleases/metabolism , Environment , Environmental Exposure/adverse effects , Genome , Genomics , Humans , Inflammation/metabolism , Linuron/adverse effects , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Protein Serine-Threonine Kinases/metabolism , Receptors, sigma/drug effects , Receptors, sigma/metabolism , Signal Transduction , X-Box Binding Protein 1/metabolism , Zebrafish
5.
Cell ; 179(7): 1483-1498.e22, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31813625

ABSTRACT

Metabolism has been shown to control peripheral immunity, but little is known about its role in central nervous system (CNS) inflammation. Through a combination of proteomic, metabolomic, transcriptomic, and perturbation studies, we found that sphingolipid metabolism in astrocytes triggers the interaction of the C2 domain in cytosolic phospholipase A2 (cPLA2) with the CARD domain in mitochondrial antiviral signaling protein (MAVS), boosting NF-κB-driven transcriptional programs that promote CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, multiple sclerosis. cPLA2 recruitment to MAVS also disrupts MAVS-hexokinase 2 (HK2) interactions, decreasing HK enzymatic activity and the production of lactate involved in the metabolic support of neurons. Miglustat, a drug used to treat Gaucher and Niemann-Pick disease, suppresses astrocyte pathogenic activities and ameliorates EAE. Collectively, these findings define a novel immunometabolic mechanism that drives pro-inflammatory astrocyte activities, outlines a new role for MAVS in CNS inflammation, and identifies candidate targets for therapeutic intervention.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Phospholipases A2, Secretory/metabolism , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , 1-Deoxynojirimycin/therapeutic use , Adaptor Proteins, Signal Transducing/genetics , Animals , Astrocytes/drug effects , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Hexokinase/metabolism , Humans , Lactic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phospholipases A2, Secretory/genetics
6.
Immunity ; 56(9): 1983-1985, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37703827

ABSTRACT

Many mechanisms by which stress mediates its effects within the central nervous system still remain unknown. Byun, Kim, Kim et al. find that early-life stress triggers corticosterone release to drive astrocyte-dependent synapse elimination and altered behavior. Thus, this work defines a steroid-sensitive astrocyte transcriptional circuit controlling behavior, highlighting how the study of CNS immunoregulation may shed light on behavior.


Subject(s)
Astrocytes , Synapses , Central Nervous System , Steroids
7.
Nature ; 627(8005): 865-872, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38509377

ABSTRACT

Disease-associated astrocyte subsets contribute to the pathology of neurologic diseases, including multiple sclerosis and experimental autoimmune encephalomyelitis1-8 (EAE), an experimental model for multiple sclerosis. However, little is known about the stability of these astrocyte subsets and their ability to integrate past stimulation events. Here we report the identification of an epigenetically controlled memory astrocyte subset that exhibits exacerbated pro-inflammatory responses upon rechallenge. Specifically, using a combination of single-cell RNA sequencing, assay for transposase-accessible chromatin with sequencing, chromatin immunoprecipitation with sequencing, focused interrogation of cells by nucleic acid detection and sequencing, and cell-specific in vivo CRISPR-Cas9-based genetic perturbation studies we established that astrocyte memory is controlled by the metabolic enzyme ATP-citrate lyase (ACLY), which produces acetyl coenzyme A (acetyl-CoA) that is used by histone acetyltransferase p300 to control chromatin accessibility. The number of ACLY+p300+ memory astrocytes is increased in acute and chronic EAE models, and their genetic inactivation ameliorated EAE. We also detected the pro-inflammatory memory phenotype in human astrocytes in vitro; single-cell RNA sequencing and immunohistochemistry studies detected increased numbers of ACLY+p300+ astrocytes in chronic multiple sclerosis lesions. In summary, these studies define an epigenetically controlled memory astrocyte subset that promotes CNS pathology in EAE and, potentially, multiple sclerosis. These findings may guide novel therapeutic approaches for multiple sclerosis and other neurologic diseases.


Subject(s)
Astrocytes , Encephalomyelitis, Autoimmune, Experimental , Epigenetic Memory , Multiple Sclerosis , Animals , Female , Humans , Male , Mice , Acetyl Coenzyme A/metabolism , Astrocytes/enzymology , Astrocytes/metabolism , Astrocytes/pathology , ATP Citrate (pro-S)-Lyase/metabolism , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation Sequencing , CRISPR-Cas Systems , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/enzymology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Multiple Sclerosis/enzymology , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Single-Cell Gene Expression Analysis , Transposases/metabolism
8.
Nature ; 614(7947): 318-325, 2023 02.
Article in English | MEDLINE | ID: mdl-36599978

ABSTRACT

Rare CD4 T cells that contain HIV under antiretroviral therapy represent an important barrier to HIV cure1-3, but the infeasibility of isolating and characterizing these cells in their natural state has led to uncertainty about whether they possess distinctive attributes that HIV cure-directed therapies might exploit. Here we address this challenge using a microfluidic technology that isolates the transcriptomes of HIV-infected cells based solely on the detection of HIV DNA. HIV-DNA+ memory CD4 T cells in the blood from people receiving antiretroviral therapy showed inhibition of six transcriptomic pathways, including death receptor signalling, necroptosis signalling and antiproliferative Gα12/13 signalling. Moreover, two groups of genes identified by network co-expression analysis were significantly associated with HIV-DNA+ cells. These genes (n = 145) accounted for just 0.81% of the measured transcriptome and included negative regulators of HIV transcription that were higher in HIV-DNA+ cells, positive regulators of HIV transcription that were lower in HIV-DNA+ cells, and other genes involved in RNA processing, negative regulation of mRNA translation, and regulation of cell state and fate. These findings reveal that HIV-infected memory CD4 T cells under antiretroviral therapy are a distinctive population with host gene expression patterns that favour HIV silencing, cell survival and cell proliferation, with important implications for the development of HIV cure strategies.


Subject(s)
CD4-Positive T-Lymphocytes , Gene Expression Regulation, Viral , HIV Infections , HIV-1 , Virus Latency , Humans , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA, Viral/isolation & purification , Gene Expression Regulation, Viral/drug effects , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/genetics , HIV-1/isolation & purification , HIV-1/pathogenicity , Immunologic Memory , Microfluidics , Necroptosis/drug effects , Signal Transduction/drug effects , Transcriptome/drug effects , Virus Latency/drug effects , Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use
9.
Nature ; 614(7947): 326-333, 2023 02.
Article in English | MEDLINE | ID: mdl-36599367

ABSTRACT

Multiple sclerosis is a chronic inflammatory disease of the central nervous system1. Astrocytes are heterogeneous glial cells that are resident in the central nervous system and participate in the pathogenesis of multiple sclerosis and its model experimental autoimmune encephalomyelitis2,3. However, few unique surface markers are available for the isolation of astrocyte subsets, preventing their analysis and the identification of candidate therapeutic targets; these limitations are further amplified by the rarity of pathogenic astrocytes. Here, to address these challenges, we developed focused interrogation of cells by nucleic acid detection and sequencing (FIND-seq), a high-throughput microfluidic cytometry method that combines encapsulation of cells in droplets, PCR-based detection of target nucleic acids and droplet sorting to enable in-depth transcriptomic analyses of cells of interest at single-cell resolution. We applied FIND-seq to study the regulation of astrocytes characterized by the splicing-driven activation of the transcription factor XBP1, which promotes disease pathology in multiple sclerosis and experimental autoimmune encephalomyelitis4. Using FIND-seq in combination with conditional-knockout mice, in vivo CRISPR-Cas9-driven genetic perturbation studies and bulk and single-cell RNA sequencing analyses of samples from mouse experimental autoimmune encephalomyelitis and humans with multiple sclerosis, we identified a new role for the nuclear receptor NR3C2 and its corepressor NCOR2 in limiting XBP1-driven pathogenic astrocyte responses. In summary, we used FIND-seq to identify a therapeutically targetable mechanism that limits XBP1-driven pathogenic astrocyte responses. FIND-seq enables the investigation of previously inaccessible cells, including rare cell subsets defined by unique gene expression signatures or other nucleic acid markers.


Subject(s)
Astrocytes , Encephalomyelitis, Autoimmune, Experimental , Microfluidics , Multiple Sclerosis , Nucleic Acids , Single-Cell Gene Expression Analysis , Animals , Humans , Mice , Astrocytes/metabolism , Astrocytes/pathology , Gene Expression Regulation , Mice, Knockout , Multiple Sclerosis/pathology , Microfluidics/methods , Single-Cell Gene Expression Analysis/methods , Nucleic Acids/analysis , Gene Editing
10.
Nature ; 620(7975): 881-889, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37558878

ABSTRACT

Dendritic cells (DCs) have a role in the development and activation of self-reactive pathogenic T cells1,2. Genetic variants that are associated with the function of DCs have been linked to autoimmune disorders3,4, and DCs are therefore attractive therapeutic targets for such diseases. However, developing DC-targeted therapies for autoimmunity requires identification of the mechanisms that regulate DC function. Here, using single-cell and bulk transcriptional and metabolic analyses in combination with cell-specific gene perturbation studies, we identify a regulatory loop of negative feedback that operates in DCs to limit immunopathology. Specifically, we find that lactate, produced by activated DCs and other immune cells, boosts the expression of NDUFA4L2 through a mechanism mediated by hypoxia-inducible factor 1α (HIF-1α). NDUFA4L2 limits the production of mitochondrial reactive oxygen species that activate XBP1-driven transcriptional modules in DCs that are involved in the control of pathogenic autoimmune T cells. We also engineer a probiotic that produces lactate and suppresses T cell autoimmunity through the activation of HIF-1α-NDUFA4L2 signalling in DCs. In summary, we identify an immunometabolic pathway that regulates DC function, and develop a synthetic probiotic for its therapeutic activation.


Subject(s)
Autoimmune Diseases , Central Nervous System , Dendritic Cells , Hypoxia-Inducible Factor 1, alpha Subunit , Lactic Acid , Humans , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/prevention & control , Autoimmunity , Central Nervous System/cytology , Central Nervous System/immunology , Central Nervous System/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactic Acid/metabolism , Probiotics/therapeutic use , Reactive Oxygen Species/metabolism , T-Lymphocytes/immunology , Feedback, Physiological , Lactase/genetics , Lactase/metabolism , Single-Cell Analysis
11.
Nature ; 611(7937): 801-809, 2022 11.
Article in English | MEDLINE | ID: mdl-36266581

ABSTRACT

Genome-wide association studies have identified risk loci linked to inflammatory bowel disease (IBD)1-a complex chronic inflammatory disorder of the gastrointestinal tract. The increasing prevalence of IBD in industrialized countries and the augmented disease risk observed in migrants who move into areas of higher disease prevalence suggest that environmental factors are also important determinants of IBD susceptibility and severity2. However, the identification of environmental factors relevant to IBD and the mechanisms by which they influence disease has been hampered by the lack of platforms for their systematic investigation. Here we describe an integrated systems approach, combining publicly available databases, zebrafish chemical screens, machine learning and mouse preclinical models to identify environmental factors that control intestinal inflammation. This approach established that the herbicide propyzamide increases inflammation in the small and large intestine. Moreover, we show that an AHR-NF-κB-C/EBPß signalling axis operates in T cells and dendritic cells to promote intestinal inflammation, and is targeted by propyzamide. In conclusion, we developed a pipeline for the identification of environmental factors and mechanisms of pathogenesis in IBD and, potentially, other inflammatory diseases.


Subject(s)
Environment , Herbicides , Inflammation , Inflammatory Bowel Diseases , Intestines , Animals , Mice , Inflammation/chemically induced , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Zebrafish , Machine Learning , Databases, Factual , Disease Models, Animal , Intestines/drug effects , Intestines/immunology , Intestines/metabolism , Intestines/pathology , NF-kappa B , CCAAT-Enhancer-Binding Protein-beta , Receptors, Aryl Hydrocarbon , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Herbicides/adverse effects
12.
Nature ; 590(7846): 473-479, 2021 02.
Article in English | MEDLINE | ID: mdl-33408417

ABSTRACT

Astrocytes are glial cells that are abundant in the central nervous system (CNS) and that have important homeostatic and disease-promoting functions1. However, little is known about the homeostatic anti-inflammatory activities of astrocytes and their regulation. Here, using high-throughput flow cytometry screening, single-cell RNA sequencing and CRISPR-Cas9-based cell-specific in vivo genetic perturbations in mice, we identify a subset of astrocytes that expresses the lysosomal protein LAMP12 and the death receptor ligand TRAIL3. LAMP1+TRAIL+ astrocytes limit inflammation in the CNS by inducing T cell apoptosis through TRAIL-DR5 signalling. In homeostatic conditions, the expression of TRAIL in astrocytes is driven by interferon-γ (IFNγ) produced by meningeal natural killer (NK) cells, in which IFNγ expression is modulated by the gut microbiome. TRAIL expression in astrocytes is repressed by molecules produced by T cells and microglia in the context of inflammation. Altogether, we show that LAMP1+TRAIL+ astrocytes limit CNS inflammation by inducing T cell apoptosis, and that this astrocyte subset is maintained by meningeal IFNγ+ NK cells that are licensed by the microbiome.


Subject(s)
Astrocytes/immunology , Gastrointestinal Microbiome/immunology , Inflammation/prevention & control , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Lysosomal Membrane Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis , Astrocytes/metabolism , Biomarkers , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Homeostasis , Humans , Inflammation/immunology , Meninges/cytology , Meninges/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/cytology , T-Lymphocytes/immunology
13.
Cell ; 145(7): 1142-55, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21664664

ABSTRACT

Neurogenesis and gliogenesis continue in discrete regions of the adult mammalian brain. A fundamental question remains whether cell genesis occurs from distinct lineage-restricted progenitors or from self-renewing and multipotent neural stem cells in the adult brain. Here, we developed a genetic marking strategy for lineage tracing of individual, quiescent, and nestin-expressing radial glia-like (RGL) precursors in the adult mouse dentate gyrus. Clonal analysis identified multiple modes of RGL activation, including asymmetric and symmetric self-renewal. Long-term lineage tracing in vivo revealed a significant percentage of clones that contained RGL(s), neurons, and astrocytes, indicating capacity of individual RGLs for both self-renewal and multilineage differentiation. Furthermore, conditional Pten deletion in RGLs initially promotes their activation and symmetric self-renewal but ultimately leads to terminal astrocytic differentiation and RGL depletion in the adult hippocampus. Our study identifies RGLs as self-renewing and multipotent neural stem cells and provides novel insights into in vivo properties of adult neural stem cells.


Subject(s)
Adult Stem Cells/cytology , Hippocampus/cytology , Multipotent Stem Cells/cytology , Neural Stem Cells/cytology , Neurogenesis , Animals , Dentate Gyrus/cytology , Intermediate Filament Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nestin
14.
Nature ; 578(7796): 593-599, 2020 02.
Article in English | MEDLINE | ID: mdl-32051591

ABSTRACT

Multiple sclerosis is a chronic inflammatory disease of the CNS1. Astrocytes contribute to the pathogenesis of multiple sclerosis2, but little is known about the heterogeneity of astrocytes and its regulation. Here we report the analysis of astrocytes in multiple sclerosis and its preclinical model experimental autoimmune encephalomyelitis (EAE) by single-cell RNA sequencing in combination with cell-specific Ribotag RNA profiling, assay for transposase-accessible chromatin with sequencing (ATAC-seq), chromatin immunoprecipitation with sequencing (ChIP-seq), genome-wide analysis of DNA methylation and in vivo CRISPR-Cas9-based genetic perturbations. We identified astrocytes in EAE and multiple sclerosis that were characterized by decreased expression of NRF2 and increased expression of MAFG, which cooperates with MAT2α to promote DNA methylation and represses antioxidant and anti-inflammatory transcriptional programs. Granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling in astrocytes drives the expression of MAFG and MAT2α and pro-inflammatory transcriptional modules, contributing to CNS pathology in EAE and, potentially, multiple sclerosis. Our results identify candidate therapeutic targets in multiple sclerosis.


Subject(s)
Astrocytes/pathology , Central Nervous System/pathology , Inflammation/pathology , MafG Transcription Factor/genetics , Repressor Proteins/genetics , Animals , Antioxidants/metabolism , Astrocytes/metabolism , Central Nervous System/metabolism , DNA Methylation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation/genetics , Male , Methionine Adenosyltransferase/genetics , Mice , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , NF-E2-Related Factor 2/genetics , Sequence Analysis, RNA , Signal Transduction , Transcription, Genetic
15.
Nature ; 586(7829): 417-423, 2020 10.
Article in English | MEDLINE | ID: mdl-32999463

ABSTRACT

Microglia, the brain's resident macrophages, help to regulate brain function by removing dying neurons, pruning non-functional synapses, and producing ligands that support neuronal survival1. Here we show that microglia are also critical modulators of neuronal activity and associated behavioural responses in mice. Microglia respond to neuronal activation by suppressing neuronal activity, and ablation of microglia amplifies and synchronizes the activity of neurons, leading to seizures. Suppression of neuronal activation by microglia occurs in a highly region-specific fashion and depends on the ability of microglia to sense and catabolize extracellular ATP, which is released upon neuronal activation by neurons and astrocytes. ATP triggers the recruitment of microglial protrusions and is converted by the microglial ATP/ADP hydrolysing ectoenzyme CD39 into AMP; AMP is then converted into adenosine by CD73, which is expressed on microglia as well as other brain cells. Microglial sensing of ATP, the ensuing microglia-dependent production of adenosine, and the adenosine-mediated suppression of neuronal responses via the adenosine receptor A1R are essential for the regulation of neuronal activity and animal behaviour. Our findings suggest that this microglia-driven negative feedback mechanism operates similarly to inhibitory neurons and is essential for protecting the brain from excessive activation in health and disease.


Subject(s)
Feedback, Physiological , Microglia/physiology , Neural Inhibition , Neurons/physiology , 5'-Nucleotidase/metabolism , Action Potentials , Adenosine/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Calcium/metabolism , Corpus Striatum/cytology , Corpus Striatum/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Neural Inhibition/genetics , Receptor, Adenosine A1/metabolism , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Time Factors
16.
Nature ; 627(8003): 277-279, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38448528
18.
J Neurochem ; 158(1): 25-35, 2021 07.
Article in English | MEDLINE | ID: mdl-32402091

ABSTRACT

Sphingolipids are a class of lipids highly enriched in the central nervous system (CNS), which shows great diversity and complexity, and has been implicated in CNS development and function. Alterations in sphingolipid metabolism have been described in multiple diseases, including those affecting the central nervous system (CNS). In this review, we discuss the role of sphingolipid metabolism in neurodegeneration, evaluating its direct roles in neuron development and health, and also in the induction of neurotoxic activities in CNS-resident astrocytes and microglia in the context of neurologic diseases such as multiple sclerosis and Alzheimer's disease. Finally, we focus on the metabolism of gangliosides and sphingosine-1-phosphate, its contribution to the pathogenesis of neurologic diseases, and its potential as a candidate target for the therapeutic modulation of neurodegeneration.


Subject(s)
Neurodegenerative Diseases/metabolism , Sphingolipids/metabolism , Animals , Central Nervous System Diseases/metabolism , Gangliosides/metabolism , Humans , Lipid Metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism
19.
PLoS Genet ; 13(4): e1006712, 2017 04.
Article in English | MEDLINE | ID: mdl-28379965

ABSTRACT

Somatosensory information from the periphery is routed to the spinal cord through centrally-projecting sensory axons that cross into the central nervous system (CNS) via the dorsal root entry zone (DREZ). The glial cells that ensheath these axons ensure rapid propagation of this information. Despite the importance of this glial-axon arrangement, how this afferent nerve is assembled during development is unknown. Using in vivo, time-lapse imaging we show that as centrally-projecting pioneer axons from dorsal root ganglia (DRG) enter the spinal cord, they initiate expression of the cytokine TNFalpha. This induction coincides with ensheathment of these axons by associated glia via a TNF receptor 2 (TNFR2)-mediated process. This work identifies a signaling cascade that mediates peripheral glial-axon interactions and it functions to ensure that DRG afferent projections are ensheathed after pioneer axons complete their navigation, which promotes efficient somatosensory neural function.


Subject(s)
Neuroglia/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Spinal Nerve Roots/metabolism , Tumor Necrosis Factor-alpha/genetics , Animals , Astrocytes/metabolism , Axons/metabolism , Central Nervous System/growth & development , Central Nervous System/metabolism , Ganglia, Spinal , Gene Expression Regulation, Developmental , Mice , Neuroglia/cytology , Neurons, Afferent/metabolism , Peripheral Nervous System/growth & development , Peripheral Nervous System/metabolism , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Signal Transduction , Spinal Cord/growth & development , Spinal Cord/metabolism , Spinal Nerve Roots/growth & development , Tumor Necrosis Factor-alpha/biosynthesis , Zebrafish/genetics , Zebrafish/growth & development
20.
J Biol Chem ; 292(30): 12383-12389, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28615443

ABSTRACT

Genetic and environmental factors contribute to the development of immune-mediated diseases. Although numerous genetic factors contributing to autoimmunity have been identified in recent years, our knowledge on environmental factors contributing to the pathogenesis of autoimmune diseases and the mechanisms involved is still limited. In this context, the diet, microbiome, geographical location, as well as environmental pollutants have been shown to modulate autoimmune disease development. These environmental factors interact with cellular components of the immune system in distinct and defined ways and can influence immune responses at the transcriptional and protein level. Moreover, endogenous metabolites generated from basic cellular processes such as glycolysis and oxidative phosphorylation also contribute to the shaping of the immune response. In this minireview, we highlight recent progress in our understanding of the modulation of the immune response by the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor whose activity is regulated by small molecules provided by diet, commensal flora, environmental pollutants, and metabolism. We focus on the role of AhR in integrating signals from the diet and the intestinal flora to modulate ongoing inflammation in the central nervous system, and we also discuss the potential therapeutic value of AhR agonists for multiple sclerosis and other autoimmune diseases.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmunity/immunology , Inflammation/immunology , Inflammation/pathology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Autoimmune Diseases/genetics , Humans , Inflammation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL