Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Front Immunol ; 14: 1267641, 2023.
Article in English | MEDLINE | ID: mdl-38283348

ABSTRACT

Introduction: Chagas disease causes a cardiac illness characterized by immunoinflammatory reactions leading to myocardial fibrosis and remodeling. The development of Chronic Chagas Cardiomyopathy (CCC) in some patients while others remain asymptomatic is not fully understood, but dysregulated inflammatory responses are implicated. The Aryl hydrocarbon receptor (AhR) plays a crucial role in regulating inflammation. Certain tryptophan (Trp) metabolites have been identified as AhR ligands with regulatory functions. Methods results and discussion: We investigated AhR expression, agonist response, ligand production, and AhR-dependent responses, such as IDO activation and regulatory T (Treg) cells induction, in two T. cruzi-infected mouse strains (B6 and Balb/c) showing different polymorphisms in AhR. Furthermore, we assessed the metabolic profile of Trp catabolites and AhR agonistic activity levels in plasma samples from patients with chronic Chagas disease (CCD) and healthy donors (HD) using a luciferase reporter assay and liquid chromatography-mass spectrophotometry (LC-MS) analysis. T. cruzi-infected B6 mice showed impaired AhR-dependent responses compared to Balb/c mice, including reduced IDO activity, kynurenine levels, Treg cell induction, CYP1A1 up-regulation, and AhR expression following agonist activation. Additionally, B6 mice exhibited no detectable AhR agonist activity in plasma and displayed lower CYP1A1 up-regulation and AhR expression upon agonist activation. Similarly, CCC patients had decreased AhR agonistic activity in plasma compared to HD patients and exhibited dysregulation in Trp metabolic pathways, resulting in altered plasma metabolite profiles. Notably, patients with severe CCC specifically showed increased N-acetylserotonin levels in their plasma. The methods and findings presented here contribute to a better understanding of CCC development mechanisms and may identify potential specific biomarkers for T. cruzi infection and the severity of associated heart disease. These insights could be valuable in designing new therapeutic strategies. Ultimately, this research aims to establish the AhR agonistic activity and Trp metabolic profile in plasma as an innovative, non-invasive predictor of prognosis for chronic Chagas disease.


Subject(s)
Chagas Cardiomyopathy , Chagas Disease , Animals , Humans , Mice , Chagas Disease/metabolism , Cytochrome P-450 CYP1A1/metabolism , Receptors, Aryl Hydrocarbon/agonists , Tryptophan/metabolism
2.
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
3.
Article in English | MEDLINE | ID: mdl-33408169

ABSTRACT

OBJECTIVE: MS is an autoimmune demyelinating disease of the CNS, which causes neurologic deficits in young adults and leads to progressive disability. The aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor, can drive anti-inflammatory functions in peripheral immune cells and also in CNS-resident cells. Laquinimod is a drug developed for the treatment of MS known to activate AHR, but the cellular targets of laquinimod are still not completely known. In this work, we analyzed the contribution of AHR activation in astrocytes to its beneficial effects in the experimental autoimmune encephalomyelitis (EAE) preclinical model of MS. METHODS: We used conditional knockout mice, in combination with genome-wide analysis of gene expression by RNA-seq and in vitro culture systems to investigate the effects of laquinimod on astrocytes. RESULTS: We found that AHR activation in astrocytes by laquinimod ameliorates EAE, a preclinical model of MS. Genome-wide RNA-seq transcriptional analyses detected anti-inflammatory effects of laquinimod in glial cells during EAE. Moreover, we established that the Delaq metabolite of laquinimod dampens proinflammatory mediator production while activating tissue-protective mechanisms in glia. CONCLUSIONS: Taken together, these findings suggest that AHR activation by clinically relevant AHR agonists may represent a novel therapeutic approach for the treatment of MS.


Subject(s)
Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Quinolones/therapeutic use , Receptors, Aryl Hydrocarbon/metabolism , Animals , Astrocytes/drug effects , Astrocytes/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Quinolones/pharmacology , Receptors, Aryl Hydrocarbon/immunology
5.
Nat Neurosci ; 23(8): 939-951, 2020 08.
Article in English | MEDLINE | ID: mdl-32690969

ABSTRACT

Zika virus (ZIKV) is a flavivirus linked to multiple birth defects including microcephaly, known as congenital ZIKV syndrome. The identification of host factors involved in ZIKV replication may guide efficacious therapeutic interventions. In genome-wide transcriptional studies, we found that ZIKV infection triggers aryl hydrocarbon receptor (AHR) activation. Specifically, ZIKV infection induces kynurenine (Kyn) production, which activates AHR, limiting the production of type I interferons (IFN-I) involved in antiviral immunity. Moreover, ZIKV-triggered AHR activation suppresses intrinsic immunity driven by the promyelocytic leukemia (PML) protein, which limits ZIKV replication. AHR inhibition suppressed the replication of multiple ZIKV strains in vitro and also suppressed replication of the related flavivirus dengue. Finally, AHR inhibition with a nanoparticle-delivered AHR antagonist or an inhibitor developed for human use limited ZIKV replication and ameliorated newborn microcephaly in a murine model. In summary, we identified AHR as a host factor for ZIKV replication and PML protein as a driver of anti-ZIKV intrinsic immunity.


Subject(s)
Receptors, Aryl Hydrocarbon/metabolism , Virus Replication , Zika Virus/metabolism , Animals , Chlorocebus aethiops , Hep G2 Cells , Humans , Vero Cells , Zika Virus Infection/metabolism
6.
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
7.
Cell Rep ; 28(12): 3120-3130.e5, 2019 Sep 17.
Article in English | MEDLINE | ID: mdl-31533035

ABSTRACT

Noradrenaline (NE), the main neurotransmitter released by sympathetic nerve terminals, is known to modulate the immune response. However, the role of the sympathetic nervous system (SNS) on the development of autoimmune diseases is still unclear. Here, we report that the SNS limits the generation of pathogenic T cells and disease development in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS). ß2-Adrenergic receptor (Adrb2) signaling limits T cell autoimmunity in EAE through a mechanism mediated by the suppression of IL-2, IFN-γ, and GM-CSF production via inducible cAMP early repressor (ICER). Accordingly, the lack of Adrb2 signaling in immune cells is sufficient to abrogate the suppressive effects of SNS activity, resulting in increased pathogenic T cell responses and EAE development. Collectively, these results uncover a suppressive role for the SNS in CNS autoimmunity while they identify potential targets for therapeutic intervention.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Immunity, Cellular , Multiple Sclerosis/immunology , Receptors, Adrenergic, beta-2/immunology , Signal Transduction/immunology , Sympathetic Nervous System/immunology , T-Lymphocytes/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Receptors, Adrenergic, beta-2/genetics , Signal Transduction/genetics , Sympathetic Nervous System/pathology , T-Lymphocytes/pathology
9.
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
10.
Nature ; 557(7707): 724-728, 2018 05.
Article in English | MEDLINE | ID: mdl-29769726

ABSTRACT

Microglia and astrocytes modulate inflammation and neurodegeneration in the central nervous system (CNS)1-3. Microglia modulate pro-inflammatory and neurotoxic activities in astrocytes, but the mechanisms involved are not completely understood4,5. Here we report that TGFα and VEGF-B produced by microglia regulate the pathogenic activities of astrocytes in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Microglia-derived TGFα acts via the ErbB1 receptor in astrocytes to limit their pathogenic activities and EAE development. Conversely, microglial VEGF-B triggers FLT-1 signalling in astrocytes and worsens EAE. VEGF-B and TGFα also participate in the microglial control of human astrocytes. Furthermore, expression of TGFα and VEGF-B in CD14+ cells correlates with the multiple sclerosis lesion stage. Finally, metabolites of dietary tryptophan produced by the commensal flora control microglial activation and TGFα and VEGF-B production, modulating the transcriptional program of astrocytes and CNS inflammation through a mechanism mediated by the aryl hydrocarbon receptor. In summary, we identified positive and negative regulators that mediate the microglial control of astrocytes. Moreover, these findings define a pathway through which microbial metabolites limit pathogenic activities of microglia and astrocytes, and suppress CNS inflammation. This pathway may guide new therapies for multiple sclerosis and other neurological disorders.


Subject(s)
Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/microbiology , Microglia/metabolism , Animals , Astrocytes/pathology , Cells, Cultured , Central Nervous System/metabolism , Central Nervous System/microbiology , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , ErbB Receptors/metabolism , Female , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Inflammation/prevention & control , Lipopolysaccharide Receptors/metabolism , Mice , Mice, Inbred C57BL , Microglia/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Receptors, Aryl Hydrocarbon/metabolism , Symbiosis , Transforming Growth Factor alpha/biosynthesis , Transforming Growth Factor alpha/metabolism , Tryptophan/deficiency , Tryptophan/metabolism , Vascular Endothelial Growth Factor B/biosynthesis , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
11.
Trends Endocrinol Metab ; 28(6): 428-436, 2017 06.
Article in English | MEDLINE | ID: mdl-28318896

ABSTRACT

Glioblastoma (GBM) progression is associated with metabolic remodeling in both glioma and immune cells, resulting in the use of aerobic glycolysis as the main source of energy and biosynthetic molecules. The transcription factor hypoxia-inducible factor (HIF)-1α drives this metabolic reorganization. Oxygen levels, as well as other factors, control the activity of HIF-1α. In addition, the ligand-activated transcription factor aryl hydrocarbon receptor (AHR) modulates tumor-specific immunity and can also participate in metabolic remodeling. AHR activity is regulated by tryptophan derivatives present in the tumor microenvironment. Thus, the tumor microenvironment and signaling via HIF-1α and AHR regulate the metabolism of gliomas and immune cells, modulating tumor-specific immunity and, consequently, tumor growth. Here, we review the roles of HIF-1α and AHR in cancer and immune cell metabolism in GBM.


Subject(s)
Glioblastoma/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Gene Expression Regulation , Glioblastoma/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Oxygen/metabolism , Receptors, Aryl Hydrocarbon/genetics
12.
Proc Natl Acad Sci U S A ; 114(8): 2012-2017, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28167760

ABSTRACT

Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of the CNS that causes disability in young adults as a result of the irreversible accumulation of neurological deficits. Although there are potent disease-modifying agents for its initial relapsing-remitting phase, these therapies show limited efficacy in secondary progressive MS (SPMS). Thus, there is an unmet clinical need for the identification of disease mechanisms and potential therapeutic approaches for SPMS. Here, we show that the sphingosine 1-phosphate receptor (S1PR) modulator fingolimod (FTY720) ameliorated chronic progressive experimental autoimmune encephalomyelitis in nonobese diabetic mice, an experimental model that resembles several aspects of SPMS, including neurodegeneration and disease progression driven by the innate immune response in the CNS. Indeed, S1PR modulation by FTY720 in murine and human astrocytes suppressed neurodegeneration-promoting mechanisms mediated by astrocytes, microglia, and CNS-infiltrating proinflammatory monocytes. Genome-wide studies showed that FTY720 suppresses transcriptional programs associated with the promotion of disease progression by astrocytes. The study of the molecular mechanisms controlling these transcriptional modules may open new avenues for the development of therapeutic strategies for progressive MS.


Subject(s)
Astrocytes/drug effects , Immunosuppressive Agents/pharmacology , Multiple Sclerosis, Chronic Progressive/drug therapy , Receptors, Lysosphingolipid/metabolism , Animals , Astrocytes/metabolism , Cell Line, Tumor , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Microglia/metabolism , Monocytes/immunology , Monocytes/metabolism , Multiple Sclerosis, Chronic Progressive/pathology , Primary Cell Culture , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , Transcriptome/drug effects
13.
Semin Immunopathol ; 39(2): 113-120, 2017 02.
Article in English | MEDLINE | ID: mdl-27646959

ABSTRACT

Deficits in immunological tolerance against self-antigens and antigens provided by the diet and commensal microbiota can result in the development of inflammatory and autoimmune disorders. Dendritic cells (DCs) are pivotal regulators of the immune response, specialized in antigen presentation to drive T cell priming and differentiation. DCs also have a tolerogenic function, participating in the enforcement of central and peripheral tolerance and the resolution of ongoing immune responses. Thus, DCs control effector and regulatory mechanisms relevant to the pathology of autoimmune disorders. In this review, we discuss recent findings regarding the control of the adaptive immune response by tolerogenic DCs. A thorough understanding of the mechanisms that control the tolerogenic DC phenotype will guide the development of novel strategies for the treatment of autoimmunity.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance , Animals , Antigen Presentation/immunology , Autoimmunity , Biomarkers , Cell Differentiation , Cell Lineage , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression , Histocompatibility Antigens Class II/metabolism , Humans , Perforin/genetics , Receptors, Cholinergic/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
14.
Semin Immunopathol ; 39(2): 165-176, 2017 02.
Article in English | MEDLINE | ID: mdl-27800584

ABSTRACT

Dendritic cells are of paramount importance bridging innate and adaptive immune responses. Depending on the context, after sensing environmental antigens, commensal microorganisms, pathogenic agents, or antigens from the diet, dendritic cells may drive either different effector adaptive immune responses or tolerance, avoiding tissue damage. Although the plasticity of the immune response and the capacity to regulate itself are considered essential to orchestrate appropriate physiological responses, it is known that the nervous system plays a relevant role controlling immune cell function. Dendritic cells present in the skin, the intestine, and lymphoid organs, besides expressing adrenergic receptors, can be reached by neurotransmitters released by sympathetic fibers innervating these tissues. These review focus on how neurotransmitters from the sympathetic nervous system can modulate dendritic cell function and how this may impact the immune response and immune-mediated disorders.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune System/cytology , Immune System/physiology , Neuroimmunomodulation , Sympathetic Nervous System/physiology , Animals , Cytokines/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/innervation , Lymphoid Tissue/immunology , Lymphoid Tissue/innervation , Lymphoid Tissue/metabolism , Norepinephrine/metabolism , Receptors, Adrenergic/metabolism , Signal Transduction , Skin/immunology , Skin/innervation , Skin/metabolism
15.
Sci Signal ; 9(433): ra61, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27330188

ABSTRACT

Type 1 diabetes (T1D) is a T cell-dependent autoimmune disease that is characterized by the destruction of insulin-producing ß cells in the pancreas. The administration to patients of ex vivo-differentiated FoxP3(+) regulatory T (Treg) cells or tolerogenic dendritic cells (DCs) that promote Treg cell differentiation is considered a potential therapy for T1D; however, cell-based therapies cannot be easily translated into clinical practice. We engineered nanoparticles (NPs) to deliver both a tolerogenic molecule, the aryl hydrocarbon receptor (AhR) ligand 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE), and the ß cell antigen proinsulin (NPITE+Ins) to induce a tolerogenic phenotype in DCs and promote Treg cell generation in vivo. NPITE+Ins administration to 8-week-old nonobese diabetic mice suppressed autoimmune diabetes. NPITE+Ins induced a tolerogenic phenotype in DCs, which was characterized by a decreased ability to activate inflammatory effector T cells and was concomitant with the increased differentiation of FoxP3(+) Treg cells. The induction of a tolerogenic phenotype in DCs by NPs was mediated by the AhR-dependent induction of Socs2, which resulted in inhibition of nuclear factor κB activation and proinflammatory cytokine production (properties of tolerogenic DCs). Together, these data suggest that NPs constitute a potential tool to reestablish tolerance in T1D and potentially other autoimmune disorders.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Suppressor of Cytokine Signaling Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Humans , Indoles/chemistry , Indoles/pharmacology , Insulin-Secreting Cells/pathology , Mice, Inbred NOD , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes, Regulatory/pathology , Thiazoles/chemistry , Thiazoles/pharmacology
16.
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
17.
Trends Immunol ; 37(7): 427-439, 2016 07.
Article in English | MEDLINE | ID: mdl-27236363

ABSTRACT

The ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1, or CD39) catalyzes the phosphohydrolysis of extracellular ATP (eATP) and ADP (eADP) released under conditions of inflammatory stress and cell injury. CD39 generates AMP, which is in turn used by the ecto-5'-nucleotidase CD73 to synthesize adenosine. These ectonucleotidases have a major impact on the dynamic equilibrium of proinflammatory eATP and ADP nucleotides versus immunosuppressive adenosine nucleosides. Indeed, CD39 plays a dominant role in the purinergic regulation of inflammation and the immune response because its expression is influenced by genetic and environmental factors. We review the specific role of CD39 in the kinetic regulation of cellular immune responses in the evolution of disease. We focus on the effects of CD39 on T cells and explore potential clinical applications in autoimmunity, chronic infections, and cancer.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Autoimmune Diseases/immunology , Immune Tolerance/immunology , Infections/immunology , Inflammation/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Adenosine Monophosphate/metabolism , Animals , Autoimmunity , Gene-Environment Interaction , Humans , Lymphocyte Activation
18.
J Immunol ; 196(2): 637-44, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26663782

ABSTRACT

Despite accumulating evidence indicating that neurotransmitters released by the sympathetic nervous system can modulate the activity of innate immune cells, we still know very little about how norepinephrine impacts signaling pathways in dendritic cells (DC) and the consequence of that in DC-driven T cell differentiation. In this article, we demonstrate that ß2-adrenergic receptor (ß2AR) activation in LPS-stimulated DC does not impair their ability to promote T cell proliferation; however, it diminishes IL-12p70 secretion, leading to a shift in the IL-12p70/IL-23 ratio. Although ß2AR stimulation in DC induces protein kinase A-dependent cAMP-responsive element-binding protein phosphorylation, the effect of changing the profile of cytokines produced upon LPS challenge occurs in a protein kinase A-independent manner and, rather, is associated with inhibition of the NF-κB and AP-1 signaling pathways. Moreover, as a consequence of the inverted IL-12p70/IL-23 ratio following ß2AR stimulation, LPS-stimulated DC promoted the generation of CD4(+) T cells that, upon TCR engagement, produced lower amounts of IFN-γ and higher levels of IL-17. These findings provide new insights into molecular and cellular mechanisms by which ß2AR stimulation in murine DC can influence the generation of adaptive immune responses and may explain some aspects of how sympathetic nervous system activity can modulate immune function.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Norepinephrine/immunology , Receptors, Adrenergic, beta-2/immunology , Signal Transduction/immunology , Animals , Blotting, Western , Cell Differentiation/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , Real-Time Polymerase Chain Reaction , Transcription Factor AP-1/immunology
19.
Trends Mol Med ; 22(1): 3-4, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26700492

ABSTRACT

Dendritic cells (DCs) regulate the generation of effector adaptive immunity and enforce peripheral tolerance. In the October 2015 issue of Immunity, Zlotnikov-Klionsky et al. present exciting data describing a population of perforin-expressing CD11c(+)DCs that limit obesity as well as metabolic and autoimmune inflammation in mice.


Subject(s)
Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Inflammation/immunology , Metabolic Syndrome/immunology , Pore Forming Cytotoxic Proteins/analysis , Animals , Female , Male
20.
Nat Med ; 21(6): 638-46, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26005855

ABSTRACT

Our understanding of the pathways that regulate lymphocyte metabolism, as well as the effects of metabolism and its products on the immune response, is still limited. We report that a metabolic program controlled by the transcription factors hypoxia inducible factor-1α (HIF1-α) and aryl hydrocarbon receptor (AHR) supports the differentiation of type 1 regulatory T cell (Tr1) cells. HIF1-α controls the early metabolic reprograming of Tr1 cells. At later time points, AHR promotes HIF1-α degradation and takes control of Tr1 cell metabolism. Extracellular ATP (eATP) and hypoxia, linked to inflammation, trigger AHR inactivation by HIF1-α and inhibit Tr1 cell differentiation. Conversely, CD39 promotes Tr1 cell differentiation by depleting eATP. CD39 also contributes to Tr1 suppressive activity by generating adenosine in cooperation with CD73 expressed by responder T cells and antigen-presenting cells. These results suggest that HIF1-α and AHR integrate immunological, metabolic and environmental signals to regulate the immune response.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity, Cellular , Inflammation/immunology , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes, Regulatory/metabolism , Adult , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Apyrase/immunology , Apyrase/metabolism , Cell Differentiation/immunology , Female , Hematopoiesis/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Inflammation/metabolism , Inflammation/pathology , Lymphocyte Activation/immunology , Mice , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...