Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(35): e2211310119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994674

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). Astrocytes are the most abundant glial cells in the CNS, and their dysfunction contributes to the pathogenesis of MS and its animal model, experimental autoimmune encephalomyelitis (EAE). Recent advances highlight the pivotal role of cellular metabolism in programming immune responses. However, the underlying immunometabolic mechanisms that drive astrocyte pathogenicity remain elusive. Nicotinamide adenine dinucleotide (NAD+) is a vital coenzyme involved in cellular redox reactions and a substrate for NAD+-dependent enzymes. Cellular NAD+ levels are dynamically controlled by synthesis and degradation, and dysregulation of this balance has been associated with inflammation and disease. Here, we demonstrate that cell-autonomous generation of NAD+ via the salvage pathway regulates astrocyte immune function. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT), a key enzyme in the salvage pathway, results in depletion of NAD+, inhibits oxidative phosphorylation, and limits astrocyte inflammatory potential. We identified CD38 as the main NADase up-regulated in reactive mouse and human astrocytes in models of neuroinflammation and MS. Genetic or pharmacological blockade of astrocyte CD38 activity augmented NAD+ levels, suppressed proinflammatory transcriptional reprogramming, impaired chemotactic potential to inflammatory monocytes, and ameliorated EAE. We found that CD38 activity is mediated via calcineurin/NFAT signaling in mouse and human reactive astrocytes. Thus, NAMPT-NAD+-CD38 circuitry in astrocytes controls their ability to meet their energy demands and drives the expression of proinflammatory transcriptional modules, contributing to CNS pathology in EAE and, potentially, MS. Our results identify candidate therapeutic targets in MS.


Subject(s)
ADP-ribosyl Cyclase 1 , Astrocytes , Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , NAD , ADP-ribosyl Cyclase 1/metabolism , Animals , Astrocytes/immunology , Astrocytes/metabolism , Autoimmunity , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Mice , Multiple Sclerosis/immunology , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism
2.
Brain ; 145(9): 3288-3307, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35899587

ABSTRACT

Malignant brain tumours are the cause of a disproportionate level of morbidity and mortality among cancer patients, an unfortunate statistic that has remained constant for decades. Despite considerable advances in the molecular characterization of these tumours, targeting the cancer cells has yet to produce significant advances in treatment. An alternative strategy is to target cells in the glioblastoma microenvironment, such as tumour-associated astrocytes. Astrocytes control multiple processes in health and disease, ranging from maintaining the brain's metabolic homeostasis, to modulating neuroinflammation. However, their role in glioblastoma pathogenicity is not well understood. Here we report that depletion of reactive astrocytes regresses glioblastoma and prolongs mouse survival. Analysis of the tumour-associated astrocyte translatome revealed astrocytes initiate transcriptional programmes that shape the immune and metabolic compartments in the glioma microenvironment. Specifically, their expression of CCL2 and CSF1 governs the recruitment of tumour-associated macrophages and promotes a pro-tumourigenic macrophage phenotype. Concomitantly, we demonstrate that astrocyte-derived cholesterol is key to glioma cell survival, and that targeting astrocytic cholesterol efflux, via ABCA1, halts tumour progression. In summary, astrocytes control glioblastoma pathogenicity by reprogramming the immunological properties of the tumour microenvironment and supporting the non-oncogenic metabolic dependency of glioblastoma on cholesterol. These findings suggest that targeting astrocyte immunometabolic signalling may be useful in treating this uniformly lethal brain tumour.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Animals , Astrocytes/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Glioma/genetics , Mice , Tumor Microenvironment , Virulence
3.
Sci Rep ; 11(1): 14644, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34282238

ABSTRACT

Inhibition of extracellular glutamate (Glu) release decreases proliferation and invasion, induces apoptosis, and inhibits melanoma metastatic abilities. Previous studies have shown that Blood-glutamate scavenging (BGS), a novel treatment approach, has been found to be beneficial in attenuating glioblastoma progression by reducing brain Glu levels. Therefore, in this study we evaluated the ability of BGS treatment to inhibit brain metastatic melanoma progression in-vivo. RET melanoma cells were implanted in C56BL/6J mice to induce brain melanoma tumors followed by treatment with BGS or vehicle administered for fourteen days. Bioluminescent imaging was conducted to evaluate tumor growth, and plasma/CSF Glu levels were monitored throughout. Immunofluorescence staining of Ki67 and 53BP1 was used to analyze tumor cell proliferation and DNA double-strand breaks. In addition, we analyzed CD8, CD68, CD206, p-STAT1 and iNOS expression to evaluate alterations in tumor micro-environment and anti-tumor immune response due to treatment. Our results show that BGS treatment reduces CSF Glu concentration and consequently melanoma growth in-vivo by decreasing tumor cell proliferation and increasing pro-apoptotic signaling in C56BL/6J mice. Furthermore, BGS treatment supported CD8+ cell recruitment and CD68+ macrophage invasion. These findings suggest that BGS can be of potential therapeutic relevance in the treatment of metastatic melanoma.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/administration & dosage , Brain Neoplasms/drug therapy , Glutamic Acid/metabolism , Melanoma/drug therapy , Oxaloacetic Acid/administration & dosage , Animals , Apoptosis/drug effects , Aspartate Aminotransferase, Cytoplasmic/pharmacology , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Therapy, Combination , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/secondary , Humans , Melanoma/pathology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Oxaloacetic Acid/pharmacology , Recombinant Proteins/administration & dosage , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
4.
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.
Nat Neurosci ; 22(5): 729-740, 2019 05.
Article in English | MEDLINE | ID: mdl-30962630

ABSTRACT

Tumor-associated macrophages (TAMs) play an important role in the immune response to cancer, but the mechanisms by which the tumor microenvironment controls TAMs and T cell immunity are not completely understood. Here we report that kynurenine produced by glioblastoma cells activates aryl hydrocarbon receptor (AHR) in TAMs to modulate their function and T cell immunity. AHR promotes CCR2 expression, driving TAM recruitment in response to CCL2. AHR also drives the expression of KLF4 and suppresses NF-κB activation in TAMs. Finally, AHR drives the expression of the ectonucleotidase CD39 in TAMs, which promotes CD8+ T cell dysfunction by producing adenosine in cooperation with CD73. In humans, the expression of AHR and CD39 was highest in grade 4 glioma, and high AHR expression was associated with poor prognosis. In summary, AHR and CD39 expressed in TAMs participate in the regulation of the immune response in glioblastoma and constitute potential targets for immunotherapy.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Brain Neoplasms/immunology , Glioblastoma/immunology , Kynurenine/metabolism , Macrophages/metabolism , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes/metabolism , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Disease Progression , Glioblastoma/metabolism , Humans , Kruppel-Like Factor 4 , Lipopolysaccharide Receptors/metabolism , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/metabolism , STAT1 Transcription Factor , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , Tumor Microenvironment
7.
Nat Methods ; 16(4): 327-332, 2019 04.
Article in English | MEDLINE | ID: mdl-30886410

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) is a rich resource of cellular heterogeneity, opening new avenues in the study of complex tissues. We introduce Cell Population Mapping (CPM), a deconvolution algorithm in which reference scRNA-seq profiles are leveraged to infer the composition of cell types and states from bulk transcriptome data ('scBio' CRAN R-package). Analysis of individual variations in lungs of influenza-virus-infected mice reveals that the relationship between cell abundance and clinical symptoms is a cell-state-specific property that varies gradually along the continuum of cell-activation states. The gradual change is confirmed in subsequent experiments and is further explained by a mathematical model in which clinical outcomes relate to cell-state dynamics along the activation process. Our results demonstrate the power of CPM in reconstructing the continuous spectrum of cell states within heterogeneous tissues.


Subject(s)
Computational Biology , Genomics , Sequence Analysis, RNA , Single-Cell Analysis , Algorithms , Animals , Cell Separation , Female , Fibroblasts/metabolism , Flow Cytometry , Gene Expression Profiling , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Lung/virology , Markov Chains , Mice , Mice, Inbred C57BL , Orthomyxoviridae , Phagocytes/metabolism , Reference Values , Software , Transcriptome
8.
PLoS Biol ; 17(3): e2006859, 2019 03.
Article in English | MEDLINE | ID: mdl-30921319

ABSTRACT

Brain metastases are prevalent in various types of cancer and are often terminal, given the low efficacy of available therapies. Therefore, preventing them is of utmost clinical relevance, and prophylactic treatments are perhaps the most efficient strategy. Here, we show that systemic prophylactic administration of a toll-like receptor (TLR) 9 agonist, CpG-C, is effective against brain metastases. Acute and chronic systemic administration of CpG-C reduced tumor cell seeding and growth in the brain in three tumor models in mice, including metastasis of human and mouse lung cancer, and spontaneous melanoma-derived brain metastasis. Studying mechanisms underlying the therapeutic effects of CpG-C, we found that in the brain, unlike in the periphery, natural killer (NK) cells and monocytes are not involved in controlling metastasis. Next, we demonstrated that the systemically administered CpG-C is taken up by endothelial cells, astrocytes, and microglia, without affecting blood-brain barrier (BBB) integrity and tumor brain extravasation. In vitro assays pointed to microglia, but not astrocytes, as mediators of CpG- C effects through increased tumor killing and phagocytosis, mediated by direct microglia-tumor contact. In vivo, CpG-C-activated microglia displayed elevated mRNA expression levels of apoptosis-inducing and phagocytosis-related genes. Intravital imaging showed that CpG-C-activated microglia cells contact, kill, and phagocytize tumor cells in the early stages of tumor brain invasion more than nonactivated microglia. Blocking in vivo activation of microglia with minocycline, and depletion of microglia with a colony-stimulating factor 1 inhibitor, indicated that microglia mediate the antitumor effects of CpG-C. Overall, the results suggest prophylactic CpG-C treatment as a new intervention against brain metastasis, through an essential activation of microglia.


Subject(s)
Brain Neoplasms/complications , Brain Neoplasms/metabolism , Microglia/metabolism , Microglia/pathology , Oligodeoxyribonucleotides/therapeutic use , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Colony-Stimulating Factors/antagonists & inhibitors , Colony-Stimulating Factors/metabolism , Female , Humans , Lung Neoplasms/complications , Lung Neoplasms/metabolism , Male , Melanoma/complications , Melanoma/metabolism , Mice , Minocycline/metabolism , Phagocytosis/drug effects , Signal Transduction/drug effects
9.
Nat Commun ; 9(1): 4578, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385785

ABSTRACT

Previous studies have reported that microglia depletion leads to impairment of synapse formation and these cells rapidly repopulate from CNS progenitors. However, the impact of microglia depletion and repopulation in the long-term state of the CNS environment has not been characterized. Here, we report that acute and synchronous microglia depletion and subsequent repopulation induces gray matter microgliosis, neuronal death in the somatosensory cortex and ataxia-like behavior. We find a type 1 interferon inflammatory signature in degenerating somatosensory cortex from microglia-depleted mice. Transcriptomic and mass cytometry analysis of repopulated microglia demonstrates an interferon regulatory factor 7-driven activation state. Minocycline and anti-IFNAR1 antibody treatment attenuate the CNS type 1 interferon-driven inflammation, restore microglia homeostasis and reduce ataxic behavior. Neither microglia depletion nor repopulation impact neuropathology or T-cell responses during experimental autoimmune encephalomyelitis. Together, we found that acute microglia ablation induces a type 1 interferon activation state of gray matter microglia associated with acute neurodegeneration.


Subject(s)
Cell Death/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Gray Matter/immunology , Interferon Type I/immunology , Microglia/immunology , Neurons/immunology , Somatosensory Cortex/immunology , Animals , Anti-Bacterial Agents/pharmacology , Ataxia/immunology , Ataxia/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Flow Cytometry , Gene Expression Profiling , Gray Matter/pathology , Homeostasis , Immunohistochemistry , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Mice , Microscopy, Confocal , Minocycline/pharmacology , Neurons/pathology , Receptor, Interferon alpha-beta/antagonists & inhibitors , Rotarod Performance Test , Somatosensory Cortex/pathology
10.
Cell Syst ; 6(6): 679-691.e4, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29886109

ABSTRACT

The influenza virus is a major cause of morbidity and mortality worldwide. Yet, both the impact of intracellular viral replication and the variation in host response across different cell types remain uncharacterized. Here we used single-cell RNA sequencing to investigate the heterogeneity in the response of lung tissue cells to in vivo influenza infection. Analysis of viral and host transcriptomes in the same single cell enabled us to resolve the cellular heterogeneity of bystander (exposed but uninfected) as compared with infected cells. We reveal that all major immune and non-immune cell types manifest substantial fractions of infected cells, albeit at low viral transcriptome loads relative to epithelial cells. We show that all cell types respond primarily with a robust generic transcriptional response, and we demonstrate novel markers specific for influenza-infected as opposed to bystander cells. These findings open new avenues for targeted therapy aimed exclusively at infected cells.


Subject(s)
Host-Pathogen Interactions/genetics , Influenza, Human/genetics , Orthomyxoviridae/genetics , Animals , Base Sequence/genetics , Cell Line , Epithelial Cells/immunology , Female , Gene Expression Profiling/methods , Host-Pathogen Interactions/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Lung/metabolism , Mice , Mice, Inbred C57BL , Orthomyxoviridae/metabolism , Orthomyxoviridae Infections/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome/genetics , Virus Replication
11.
Science ; 353(6299): 603-8, 2016 Aug 05.
Article in English | MEDLINE | ID: mdl-27493188

ABSTRACT

Mutations in the optineurin (OPTN) gene have been implicated in both familial and sporadic amyotrophic lateral sclerosis (ALS). However, the role of this protein in the central nervous system (CNS) and how it may contribute to ALS pathology are unclear. Here, we found that optineurin actively suppressed receptor-interacting kinase 1 (RIPK1)-dependent signaling by regulating its turnover. Loss of OPTN led to progressive dysmyelination and axonal degeneration through engagement of necroptotic machinery in the CNS, including RIPK1, RIPK3, and mixed lineage kinase domain-like protein (MLKL). Furthermore, RIPK1- and RIPK3-mediated axonal pathology was commonly observed in SOD1(G93A) transgenic mice and pathological samples from human ALS patients. Thus, RIPK1 and RIPK3 play a critical role in mediating progressive axonal degeneration. Furthermore, inhibiting RIPK1 kinase may provide an axonal protective strategy for the treatment of ALS and other human degenerative diseases characterized by axonal degeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Apoptosis , Axons/pathology , Nerve Degeneration/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Transcription Factor TFIIIA/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Apoptosis/genetics , Cell Cycle Proteins , Humans , Inflammation/genetics , Inflammation/pathology , Membrane Transport Proteins , Mice , Mice, Transgenic , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Suppression, Genetic , Transcription Factor TFIIIA/genetics
12.
Brain ; 139(Pt 7): 1939-57, 2016 07.
Article in English | MEDLINE | ID: mdl-27246324

ABSTRACT

SEE WINGER AND ZAMVIL DOI101093/BRAIN/AWW121 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE: The innate immune system plays a central role in the chronic central nervous system inflammation that drives neurological disability in progressive forms of multiple sclerosis, for which there are no effective treatments. The mucosal immune system is a unique tolerogenic organ that provides a physiological approach for the induction of regulatory T cells. Here we report that nasal administration of CD3-specific antibody ameliorates disease in a progressive animal model of multiple sclerosis. This effect is IL-10-dependent and is mediated by the induction of regulatory T cells that share a similar transcriptional profile to Tr1 regulatory cells and that suppress the astrocyte inflammatory transcriptional program. Treatment results in an attenuated inflammatory milieu in the central nervous system, decreased microglia activation, reduced recruitment of peripheral monocytes, stabilization of the blood-brain barrier and less neurodegeneration. These findings suggest a new therapeutic approach for the treatment of progressive forms of multiple sclerosis and potentially other types of chronic central nervous system inflammation.


Subject(s)
Astrocytes/immunology , CD3 Complex/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunologic Factors/pharmacology , Interleukin-10/immunology , Muromonab-CD3/pharmacology , T-Lymphocytes, Regulatory/immunology , Administration, Intranasal , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Immunologic Factors/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Muromonab-CD3/administration & dosage , Pneumonia, Pneumococcal/immunology
13.
Nat Med ; 22(6): 586-97, 2016 06.
Article in English | MEDLINE | ID: mdl-27158906

ABSTRACT

Astrocytes have important roles in the central nervous system (CNS) during health and disease. Through genome-wide analyses we detected a transcriptional response to type I interferons (IFN-Is) in astrocytes during experimental CNS autoimmunity and also in CNS lesions from patients with multiple sclerosis (MS). IFN-I signaling in astrocytes reduces inflammation and experimental autoimmune encephalomyelitis (EAE) disease scores via the ligand-activated transcription factor aryl hydrocarbon receptor (AHR) and the suppressor of cytokine signaling 2 (SOCS2). The anti-inflammatory effects of nasally administered interferon (IFN)-ß are partly mediated by AHR. Dietary tryptophan is metabolized by the gut microbiota into AHR agonists that have an effect on astrocytes to limit CNS inflammation. EAE scores were increased following ampicillin treatment during the recovery phase, and CNS inflammation was reduced in antibiotic-treated mice by supplementation with the tryptophan metabolites indole, indoxyl-3-sulfate, indole-3-propionic acid and indole-3-aldehyde, or the bacterial enzyme tryptophanase. In individuals with MS, the circulating levels of AHR agonists were decreased. These findings suggest that IFN-Is produced in the CNS function in combination with metabolites derived from dietary tryptophan by the gut flora to activate AHR signaling in astrocytes and suppress CNS inflammation.


Subject(s)
Astrocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Gastrointestinal Microbiome , Interferon Type I/immunology , Multiple Sclerosis/immunology , Receptors, Aryl Hydrocarbon/immunology , T-Lymphocytes/immunology , Tryptophan/metabolism , Animals , Case-Control Studies , Cell Proliferation , Central Nervous System/immunology , Central Nervous System/metabolism , Chemokine CCL2/metabolism , Chromatin Immunoprecipitation , Chromatography, High Pressure Liquid , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Knockdown Techniques , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunoblotting , Indican/urine , Indoles/metabolism , Inflammation , Interferon-beta/pharmacology , Limosilactobacillus reuteri , Mice , Mice, Knockout , Multiple Sclerosis/metabolism , Myxovirus Resistance Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Optical Imaging , Polymerase Chain Reaction , Receptor, Interferon alpha-beta/genetics , Receptors, Aryl Hydrocarbon/metabolism , STAT1 Transcription Factor/metabolism , Serotonin , Suppressor of Cytokine Signaling Proteins , Tryptophanase/metabolism
14.
Nat Med ; 20(10): 1147-56, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25216636

ABSTRACT

Astrocytes have complex roles in health and disease, thus it is important to study the pathways that regulate their function. Here we report that lactosylceramide (LacCer) synthesized by ß-1,4-galactosyltransferase 6 (B4GALT6) is upregulated in the central nervous system (CNS) of mice during chronic experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). LacCer acts in an autocrine manner to control astrocyte transcriptional programs that promote neurodegeneration. In addition, LacCer in astrocytes controls the recruitment and activation of microglia and CNS-infiltrating monocytes in a non-cell autonomous manner by regulating production of the chemokine CCL2 and granulocyte-macrophage colony-stimulating factor (GM-CSF), respectively. We also detected high B4GALT6 gene expression and LacCer concentrations in CNS MS lesions. Inhibition of LacCer synthesis in mice suppressed local CNS innate immunity and neurodegeneration in EAE and interfered with the activation of human astrocytes in vitro. Thus, B4GALT6 regulates astrocyte activation and is a potential therapeutic target for MS and other neuroinflammatory disorders.


Subject(s)
Astrocytes/immunology , Astrocytes/metabolism , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Galactosyltransferases/metabolism , Glycolipids/metabolism , Animals , Antigens, CD/metabolism , Central Nervous System/pathology , Chemokine CCL2/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Galactosyltransferases/genetics , Gene Knockdown Techniques , Glial Fibrillary Acidic Protein , Humans , Immunity, Innate , Lactosylceramides/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Up-Regulation
15.
Nat Immunol ; 14(10): 1054-63, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23995234

ABSTRACT

Dendritic cells (DCs) control the balance between effector T cells and regulatory T cells in vivo. Hence, the study of DCs might identify mechanisms of disease pathogenesis and guide new therapeutic approaches for disorders mediated by the immune system. We found that interleukin 27 (IL-27) signaling in mouse DCs limited the generation of effector cells of the TH1 and TH17 subsets of helper T cells and the development of experimental autoimmune encephalomyelitis (EAE). The effects of IL-27 were mediated at least in part through induction of the immunoregulatory molecule CD39 in DCs. IL-27-induced CD39 decreased the extracellular concentration of ATP and downregulated nucleotide-dependent activation of the NLRP3 inflammasome. Finally, therapeutic vaccination with IL-27-conditioned DCs suppressed established relapsing-remitting EAE. Thus, IL-27 signaling in DCs limited pathogenic T cell responses and the development of autoimmunity.


Subject(s)
Antigens, CD/genetics , Apyrase/genetics , Autoimmunity , Dendritic Cells/drug effects , Dendritic Cells/immunology , Interleukin-17/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Animals , Antibodies/immunology , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigens, CD/metabolism , Apyrase/metabolism , Autoantibodies/immunology , Autoimmunity/drug effects , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression , Gene Expression Regulation/drug effects , Immune Tolerance/immunology , Mice , Mice, Knockout , Myelin Sheath/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, Interleukin , Signal Transduction , T-Lymphocyte Subsets/cytology , Transcription, Genetic/drug effects
16.
Glia ; 61(6): 985-1002, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536313

ABSTRACT

Transforming growth factor ß1 (TGF-ß1) is a pleiotropic cytokine expressed throughout the CNS. Previous studies demonstrated that TGF-ß1 contributes to maintain neuronal survival, but mechanistically this effect is not well understood. We generated a CNS-specific TGF-ß1-deficient mouse model to investigate the functional consequences of TGF-ß1-deficiency in the adult mouse brain. We found that depletion of TGF-ß1 in the CNS resulted in a loss of the astrocyte glutamate transporter (GluT) proteins GLT-1 (EAAT2) and GLAST (EAAT1) and decreased glutamate uptake in the mouse hippocampus. Treatment with TGF-ß1 induced the expression of GLAST and GLT-1 in cultured astrocytes and enhanced astroglial glutamate uptake. Similar to GLT-1-deficient mice, CNS-TGF-ß1-deficient mice had reduced brain weight and neuronal loss in the CA1 hippocampal region. CNS-TGF-ß1-deficient mice showed GluN2B-dependent aberrant synaptic plasticity in the CA1 area of the hippocampus similar to the glutamate transport inhibitor DL-TBOA and these mice were highly sensitive to excitotoxic injury. In addition, hippocampal neurons from TGF-ß1-deficient mice had elevated GluN2B-mediated calcium signals in response to extrasynaptic glutamate receptor stimulation, whereas cells treated with TGF-ß1 exhibited reduced GluN2B-mediated calcium signals. In summary, our study demonstrates a previously unrecognized function of TGF-ß1 in the CNS to control extracellular glutamate homeostasis and GluN2B-mediated calcium responses in the mouse hippocampus.


Subject(s)
Calcium/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Apoptosis/physiology , Dendritic Spines/metabolism , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Hippocampus/metabolism , Mice , Mice, Knockout , Synaptic Transmission/physiology , Transforming Growth Factor beta1/genetics
17.
Immunol Rev ; 248(1): 170-87, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22725961

ABSTRACT

Demyelinating diseases such as multiple sclerosis are chronic inflammatory autoimmune diseases with a heterogeneous clinical presentation and course. Both the adaptive and the innate immune systems have been suggested to contribute to their pathogenesis and recovery. In this review, we discuss the role of the innate immune system in mediating demyelinating diseases. In particular, we provide an overview of the anti-inflammatory or pro-inflammatory functions of dendritic cells, mast cells, natural killer (NK) cells, NK-T cells, γδ T cells, microglial cells, and astrocytes. We emphasize the interaction of astroctyes with the immune system and how this interaction relates to the demyelinating pathologies. Given the pivotal role of the innate immune system, it is possible that targeting these cells may provide an effective therapeutic approach for demyelinating diseases.


Subject(s)
Demyelinating Diseases/immunology , Immunity, Innate , Adaptive Immunity , Animals , Astrocytes/immunology , Astrocytes/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Demyelinating Diseases/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mononuclear Phagocyte System/immunology , Mononuclear Phagocyte System/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
18.
Neuro Oncol ; 14(8): 1037-49, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22700727

ABSTRACT

Gliomas are the most frequent primary tumors of the brain, and for highly malignant gliomas there is no successful treatment. The tumor microenvironment contains large numbers of infiltrating microglia and macrophages (MM). There is increasing evidence that the tumor-associated MM support glioma expansion. CD38 is a multifunctional ectoenzyme that uses nicotinamide adenine dinucleotide as a substrate to generate second messengers. Previously we showed that CD38 deficiency modulates microglial "activation" and impaired recovery from head trauma by a microglia-associated mechanism. In view of the supportive role of MM in glioma progression and the role of CD38 in microglia activation, we hypothesize that deficiency of CD38 in the tumor microenvironment would inhibit glioma progression. Using the syngeneic GL261 model of glioma progression in wild-type and CD38 null mice, we show here that CD38 deficiency significantly attenuates glioma expansion and prolongs the life span of the glioma-bearing mice. The CD38 deficiency effect was associated with increased cell death and decreased metalloproteinase-12 expression in the tumor mass, as well as modulation of the tumor-induced MM properties, as indicated by a reduction in the expression of the MM marker F4/80 and matrix metalloproteinases. Our results thus suggest that CD38 participates in the tumor-supporting action of MM and that targeting CD38 might be a potential therapeutic approach for glioma treatment.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Brain Neoplasms/pathology , Glioma/pathology , Macrophages/pathology , Membrane Glycoproteins/metabolism , Microglia/pathology , Tumor Microenvironment/immunology , ADP-ribosyl Cyclase 1/deficiency , Animals , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cell Separation , Disease Models, Animal , Disease Progression , Flow Cytometry , Glioma/immunology , Glioma/metabolism , Immunoblotting , Immunohistochemistry , Macrophages/immunology , Male , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Reverse Transcriptase Polymerase Chain Reaction
19.
Glia ; 59(3): 397-412, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21264947

ABSTRACT

Apoptosis is a controlled cell-death process mediated inter alia by proteins of the Bcl-2 family. Some proteins previously shown to promote the apoptotic process were found to have nonapoptotic functions as well. Microglia, the resident immune cells of the central nervous system, respond to brain derangements by becoming activated to contend with the brain damage. Activated microglia can also undergo activation-induced cell death. Previous studies have addressed the role of core apoptotic proteins in the death process, but whether these proteins also play a role or not in the activation process is not been reported. Here we explore the effect of the BH3-only protein Bid on the immunological features of microglia and macrophages. Our results showed that Bid regulates both the phagocytotic activities and the inflammatory profiles of these cells. Deficiency of Bid attenuated the phagocytotic activity of primary microglia and peritoneal macrophages. It also changed the expression profile of distinct inflammation-related genes in lipopolysaccharide-activated microglia and peritoneal macrophages in vitro and in an in vivo sepsis-like paradigm. Notably, similar changes followed downregulation of Bid in the N9 microglial cell line. Cell death could not be detected in any of the systems examined. Our findings demonstrate that Bid can regulate the immunological profiles of activated microglial and macrophages, via a novel nonapoptotic activity. In view of the critical role of these cells in various pathologies, including acute and chronic brain insults, our findings suggest that impairments in Bid expression may contribute to these pathologies also via a nonapoptotic activity.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/physiology , Macrophages/immunology , Microglia/immunology , Nerve Degeneration/immunology , Animals , Apoptosis/immunology , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/genetics , Cell Line , Cells, Cultured , Female , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Lipopolysaccharides/toxicity , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Phagocytosis/immunology , Signal Transduction/immunology
20.
J Neurotrauma ; 26(9): 1521-33, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19257806

ABSTRACT

Traumatic brain injury (TBI) is a major cause of death and disability worldwide. It causes progressive tissue atrophy and consequent neurological dysfunctions. TBI is accompanied by neuroinflammation, a process mediated largely by microglia. CD38 is an ectoenzyme that promotes transmembrane signaling via the synthesis of potent calcium mobilizing agents or via its receptor activity. CD38 is expressed in the brain in various cell types including microglia. In previous studies, we showed that CD38 regulates microglial activation and response to chemokines. In view of the important role of neuroinflammation in TBI and the effects of CD38 on microglial responses, the present study examines the role of CD38 in the recovery of mice from closed head injury (CHI), a model of focal TBI. For this purpose, CD38-deficient and wild-type (WT) mice were subjected to a similar severity of CHI and the effect of the injury on neurobehavioral and cognitive functions was assessed by the Neurological Severity Score (NSS) and the Object Recognition Test, at various time points post-injury. The results show that recovery after CHI (as indicated by the NSS) was significantly lower in CD38-deficient mice than in WT mice and that the object recognition performance after injury was significantly impaired in injured CD38-deficient mice than in WT mice. In addition, we also observed that the amount of activated microglia/macrophages at the injury site was significantly lower in CD38-deficient mice compared with WT mice. Taken together, our findings indicate that CD38 plays a beneficial role in the recovery of mice from CHI and that this effect is mediated, at least in part, via the effect of CD38 on microglia responses.


Subject(s)
ADP-ribosyl Cyclase 1/physiology , Brain Injuries/genetics , Brain Injuries/pathology , ADP-ribosyl Cyclase 1/genetics , Animals , Astrocytes/pathology , Behavior, Animal , Brain Injuries/psychology , Head Injuries, Closed/genetics , Head Injuries, Closed/pathology , Head Injuries, Closed/psychology , Image Processing, Computer-Assisted , Immunohistochemistry , Macrophages/pathology , Memory/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Microglia/pathology , Neurons/pathology , Recognition, Psychology/physiology , Recovery of Function , Trauma Severity Indices
SELECTION OF CITATIONS
SEARCH DETAIL
...