Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Int J Mol Sci ; 25(10)2024 May 08.
Article En | MEDLINE | ID: mdl-38791146

Crohn's disease (CD) is a subtype of inflammatory bowel disease (IBD) characterized by transmural disease. The concept of transmural healing (TH) has been proposed as an indicator of deep clinical remission of CD and as a predictor of favorable treatment endpoints. Understanding the pathophysiology involved in transmural disease is critical to achieving these endpoints. However, most studies have focused on the intestinal mucosa, overlooking the contribution of the intestinal wall in Crohn's disease. Multi-omics approaches have provided new avenues for exploring the pathogenesis of Crohn's disease and identifying potential biomarkers. We aimed to use transcriptomic and proteomic technologies to compare immune and mesenchymal cell profiles and pathways in the mucosal and submucosa/wall compartments to better understand chronic refractory disease elements to achieve transmural healing. The results revealed similarities and differences in gene and protein expression profiles, metabolic mechanisms, and immune and non-immune pathways between these two compartments. Additionally, the identification of protein isoforms highlights the complex molecular mechanisms underlying this disease, such as decreased RTN4 isoforms (RTN4B2 and RTN4C) in the submucosa/wall, which may be related to the dysregulation of enteric neural processes. These findings have the potential to inform the development of novel therapeutic strategies to achieve TH.


Colon , Crohn Disease , Intestinal Mucosa , Proteomics , Humans , Crohn Disease/metabolism , Crohn Disease/pathology , Crohn Disease/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Proteomics/methods , Colon/metabolism , Colon/pathology , Transcriptome , Male , Female , Adult , Gene Expression Profiling , Biomarkers , Middle Aged , Multiomics
2.
Nature ; 611(7937): 801-809, 2022 11.
Article En | MEDLINE | ID: mdl-36266581

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.


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.
J Mol Med (Berl) ; 99(5): 663-671, 2021 05.
Article En | MEDLINE | ID: mdl-33398468

Mesenchymal stem cells (MSCs) are promising candidates for the development of cell-based drug delivery systems for autoimmune inflammatory diseases, such as multiple sclerosis (MS). Here, we investigated the effect of Ro-31-8425, an ATP-competitive kinase inhibitor, on the therapeutic properties of MSCs. Upon a simple pretreatment procedure, MSCs spontaneously took up and then gradually released significant amounts of Ro-31-8425. Ro-31-8425 (free or released by MSCs) suppressed the proliferation of CD4+ T cells in vitro following polyclonal and antigen-specific stimulation. Systemic administration of Ro-31-8425-loaded MSCs ameliorated the clinical course of experimental autoimmune encephalomyelitis (EAE), a murine model of MS, displaying a stronger suppressive effect on EAE than control MSCs or free Ro-31-8425. Ro-31-8425-MSC administration resulted in sustained levels of Ro-31-8425 in the serum of EAE mice, modulating immune cell trafficking and the autoimmune response during EAE. Collectively, these results identify MSC-based drug delivery as a potential therapeutic strategy for the treatment of autoimmune diseases. KEY MESSAGES: MSCs can spontaneously take up the ATP-competitive kinase inhibitor Ro-31-8425. Ro-31-8425-loaded MSCs gradually release Ro-31-8425 and exhibit sustained suppression of T cells. Ro-31-8425-loaded MSCs have more sustained serum levels of Ro-31-8425 than free Ro-31-8425. Ro-31-8425-loaded MSCs are more effective than MSCs and free Ro-31-8425 for EAE therapy.


Drug Delivery Systems/methods , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Enzyme Inhibitors/administration & dosage , Indoles/administration & dosage , Maleimides/administration & dosage , Mesenchymal Stem Cells/drug effects , Multiple Sclerosis/drug therapy , Transplantation, Heterologous/methods , Animals , Cell Proliferation/drug effects , Drug Liberation , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme Inhibitors/blood , Female , Humans , Immunity/drug effects , Indoles/blood , Maleimides/blood , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tissue Distribution , Treatment Outcome
5.
Nat Neurosci ; 22(5): 729-740, 2019 05.
Article En | MEDLINE | ID: mdl-30962630

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.


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
6.
Gastroenterology ; 151(6): 1100-1104, 2016 12.
Article En | MEDLINE | ID: mdl-27693323

Interleukin 10 receptor (IL10R)-deficient mice develop spontaneous colitis and, similarly, patients with loss-of-function mutations in IL10R develop severe infant-onset inflammatory bowel disease. Loss of IL10R signaling in mouse and human macrophages is associated with increased production of interleukin 1ß. We demonstrated that innate immune production of IL1ß mediates colitis in IL10R-deficient mice. Transfer of Il1r1-/- CD4+ T cells into Rag1-/-/Il10rb-/- mice reduced the severity of their colitis (compared to mice that received CD4+ T cells that express IL1R), accompanied by decreased production of interferon gamma, tumor necrosis factor-α, and IL17A. In macrophages from mice without disruption of IL10R signaling or from healthy humans (controls), incubation with IL10 reduced canonical activation of the inflammasome and production of IL1ß through transcriptional and post-translational regulation of NLRP3. Lipopolysaccharide and adenosine triphosphate stimulation of macrophages from Il10rb-/- mice or IL10R-deficient patients resulted in increased production of IL1ß. Moreover, in human IL10R-deficient macrophages, lipopolysaccharide stimulation alone triggered IL1ß secretion via non-canonical, caspase 8-dependent activation of the inflammasome. We treated 2 IL10R-deficient patients with severe and treatment-refractory infant-onset inflammatory bowel disease with the IL1-receptor antagonist anakinra. Both patients had marked clinical, endoscopic, and histologic responses after 4-7 weeks. This treatment served as successful bridge to allogeneic hematopoietic stem cell transplantation in 1 patient. Our findings indicate that loss of IL10 signaling leads to intestinal inflammation, at least in part, through increased production of IL1 by innate immune cells, leading to activation of CD4+ T cells. Agents that block IL1 signaling might be used to treat patients with inflammatory bowel disease resulting from IL10R deficiency.


Colitis/immunology , Inflammatory Bowel Diseases/drug therapy , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Receptors, Interleukin-10/genetics , Adenosine Triphosphate/pharmacology , Adult , Animals , Antirheumatic Agents/therapeutic use , CD4-Positive T-Lymphocytes , Caspase 8/metabolism , Cells, Cultured , Child, Preschool , Colitis/genetics , Colitis/metabolism , Gene Expression Regulation/drug effects , Homeodomain Proteins/genetics , Humans , Immunity, Innate , Inflammasomes/drug effects , Inflammasomes/metabolism , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interferon-gamma/metabolism , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-10/pharmacology , Interleukin-10 Receptor alpha Subunit/genetics , Interleukin-17/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides/pharmacology , Macrophages , Mice , Mice, Knockout , Mutation , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Biosynthesis/drug effects , Receptors, Interleukin-10/deficiency , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
7.
Sci Signal ; 9(433): ra61, 2016 06 21.
Article En | MEDLINE | ID: mdl-27330188

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.


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
8.
Nat Med ; 22(6): 586-97, 2016 06.
Article En | MEDLINE | ID: mdl-27158906

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.


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
9.
Cell ; 162(6): 1338-52, 2015 Sep 10.
Article En | MEDLINE | ID: mdl-26359987

Seasonal changes in disease activity have been observed in multiple sclerosis, an autoimmune disorder that affects the CNS. These epidemiological observations suggest that environmental factors influence the disease course. Here, we report that melatonin levels, whose production is modulated by seasonal variations in night length, negatively correlate with multiple sclerosis activity in humans. Treatment with melatonin ameliorates disease in an experimental model of multiple sclerosis and directly interferes with the differentiation of human and mouse T cells. Melatonin induces the expression of the repressor transcription factor Nfil3, blocking the differentiation of pathogenic Th17 cells and boosts the generation of protective Tr1 cells via Erk1/2 and the transactivation of the IL-10 promoter by ROR-α. These results suggest that melatonin is another example of how environmental-driven cues can impact T cell differentiation and have implications for autoimmune disorders such as multiple sclerosis.


Melatonin/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Light , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Recurrence , Seasons , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
10.
Nat Med ; 21(6): 638-46, 2015 Jun.
Article En | MEDLINE | ID: mdl-26005855

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.


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
11.
J Immunol ; 194(7): 3452-62, 2015 Apr 01.
Article En | MEDLINE | ID: mdl-25740944

Myeloid-derived suppressor cells (MDSCs) are key regulatory cells that control inflammation and promote tumor-immune escape. To date, no specific immunomodulatory drug has proven efficacy in targeting the expansion and/or function of these cells in different pathophysiologic settings. In this study, we identified a context-dependent effect of the nonsteroidal anti-inflammatory drug indomethacin (IND) on MDSCs, depending on whether they were derived from tumor microenvironments (TME) or from tumor-free microenvironments (TFME). Treatment of mice bearing the LP07 lung adenocarcinoma with IND inhibited the suppressive activity of splenic MDSCs, which restrained tumor growth through mechanisms involving CD8(+) T cells. The same effect was observed when MDSCs were treated with IND and conditioned media from LP07 tumor cells in vitro. However, in the absence of a tumor context, IND enhanced the intrinsic suppressive function of MDSCs and amplified their protumoral activity. In a model of autoimmune neuroinflammation, IND-treated MDSCs differentiated in TFME attenuated inflammation, whereas IND-treated MDSCs differentiated in TME aggravated clinical symptoms and delayed resolution of the disease. Mechanistically, IND reduced arginase activity as well as NO and reactive oxygen species production in MDSCs differentiated in TME but not in TFME. Moreover, expression of the C/EBP-ß transcription factor isoforms correlated with the suppressive activity of IND-treated MDSCs. Our study unveils the dual and context-dependent action of IND, a drug that serves both as an anti-inflammatory and anticancer agent, which differentially affects MDSC activity whether these cells are derived from TME or TFME. These results have broad clinical implication in cancer, chronic inflammation and autoimmunity.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cellular Microenvironment/drug effects , Cellular Microenvironment/immunology , Indomethacin/pharmacology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Autoimmunity/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Immunophenotyping , Mice , Models, Biological , Myeloid Cells/metabolism , Neoplasms/immunology , Neoplasms/pathology , Nitric Oxide/metabolism , Phenotype , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology
12.
Cancer Res ; 75(7): 1265-74, 2015 Apr 01.
Article En | MEDLINE | ID: mdl-25672979

Bidirectional cross-talk between the neuroendocrine and immune systems orchestrates immune responses in both physiologic and pathologic settings. In this study, we provide in vivo evidence of a critical role for the thyroid hormone triiodothyronine (T3) in controlling the maturation and antitumor functions of dendritic cells (DC). We used a thyroid hormone receptor (TR) ß mutant mouse (TRßPV) to establish the relevance of the T3-TRß system in vivo. In this model, TRß signaling endowed DCs with the ability to stimulate antigen-specific cytotoxic T-cell responses during tumor development. T3 binding to TRß increased DC viability and augmented DC migration to lymph nodes. Moreover, T3 stimulated the ability of DCs to cross-present antigens and to stimulate cytotoxic T-cell responses. In a B16-OVA mouse model of melanoma, vaccination with T3-stimulated DCs inhibited tumor growth and prolonged host survival, in part by promoting the generation of IFNγ-producing CD8(+) T cells. Overall, our results establish an adjuvant effect of T3-TRß signaling in DCs, suggesting an immediately translatable method to empower DC vaccination approaches for cancer immunotherapy.


Dendritic Cells/immunology , Melanoma, Experimental/immunology , Thyroid Hormone Receptors beta/metabolism , Triiodothyronine/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Cell Survival , Cross-Priming , Cytotoxicity, Immunologic , Female , Immunotherapy , Lymph Nodes/immunology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Transgenic
13.
Nat Med ; 20(10): 1147-56, 2014 Oct.
Article En | MEDLINE | ID: mdl-25216636

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.


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
14.
Nat Commun ; 5: 3753, 2014 May 06.
Article En | MEDLINE | ID: mdl-24796415

Interleukin (IL)-22 produced by innate lymphoid cells (ILCs) and CD4+ T cells plays an important role in host defence and mucosal homeostasis, thus it is important to investigate the mechanisms that regulate IL-22 production. We investigated the regulation IL-22 production by CD4+ T cells. Here we show that IL-21 triggers IL-22, but not IL-17 production by CD4+ T cells. STAT3, activated by IL-21, controls the epigenetic status of the il22 promoter and its interaction with the aryl hydrocarbon receptor (AhR). Moreover, IL-21 and AhR signalling in T cells control IL-22 production and the development of dextran sodium sulphate-induced colitis in ILC-deficient mice. Thus, we have identified IL-21 as an inducer of IL-22 production in CD4+ T cells in vitro and in vivo.


CD4-Positive T-Lymphocytes/metabolism , Interleukins/biosynthesis , Interleukins/physiology , Gene Expression Profiling , Humans , Interleukins/genetics , Interleukins/metabolism , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/metabolism , STAT3 Transcription Factor/physiology , Transcription, Genetic , Interleukin-22
15.
Immunity ; 40(5): 706-19, 2014 May 15.
Article En | MEDLINE | ID: mdl-24792912

Intact interleukin-10 receptor (IL-10R) signaling on effector and T regulatory (Treg) cells are each independently required to maintain immune tolerance. Here we show that IL-10 sensing by innate immune cells, independent of its effects on T cells, was critical for regulating mucosal homeostasis. Following wild-type (WT) CD4(+) T cell transfer, Rag2(-/-)Il10rb(-/-) mice developed severe colitis in association with profound defects in generation and function of Treg cells. Moreover, loss of IL-10R signaling impaired the generation and function of anti-inflammatory intestinal and bone-marrow-derived macrophages and their ability to secrete IL-10. Importantly, transfer of WT but not Il10rb(-/-) anti-inflammatory macrophages ameliorated colitis induction by WT CD4(+) T cells in Rag2(-/-)Il10rb(-/-) mice. Similar alterations in the generation and function of anti-inflammatory macrophages were observed in IL-10R-deficient patients with very early onset inflammatory bowel disease. Collectively, our studies define innate immune IL-10R signaling as a key factor regulating mucosal immune homeostasis in mice and humans.


Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Interleukin-10/immunology , Receptors, Interleukin-10/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-10/deficiency , Receptors, Interleukin-10/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology
16.
Cell ; 156(4): 744-58, 2014 Feb 13.
Article En | MEDLINE | ID: mdl-24529377

The clinical benefit conferred by vascular endothelial growth factors (VEGF)-targeted therapies is variable, and tumors from treated patients eventually reinitiate growth. Here, we identify a glycosylation-dependent pathway that compensates for the absence of cognate ligand and preserves angiogenesis in response to VEGF blockade. Remodeling of the endothelial cell (EC) surface glycome selectively regulated binding of galectin-1 (Gal1), which upon recognition of complex N-glycans on VEGFR2, activated VEGF-like signaling. Vessels within anti-VEGF-sensitive tumors exhibited high levels of α2-6-linked sialic acid, which prevented Gal1 binding. In contrast, anti-VEGF refractory tumors secreted increased Gal1 and their associated vasculature displayed glycosylation patterns that facilitated Gal1-EC interactions. Interruption of ß1-6GlcNAc branching in ECs or silencing of tumor-derived Gal1 converted refractory into anti-VEGF-sensitive tumors, whereas elimination of α2-6-linked sialic acid conferred resistance to anti-VEGF. Disruption of the Gal1-N-glycan axis promoted vascular remodeling, immune cell influx and tumor growth inhibition. Thus, targeting glycosylation-dependent lectin-receptor interactions may increase the efficacy of anti-VEGF treatment.


Angiogenesis Inhibitors/therapeutic use , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic , Vascular Endothelial Growth Factors/antagonists & inhibitors , Animals , Endothelial Cells/metabolism , Galectin 1/genetics , Galectin 1/metabolism , Glycosylation , Humans , Hypoxia , Mice , Receptors, Mitogen/metabolism
17.
Nat Immunol ; 14(10): 1054-63, 2013 Oct.
Article En | MEDLINE | ID: mdl-23995234

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.


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
18.
Exp Cell Res ; 319(19): 3020-34, 2013 Nov 15.
Article En | MEDLINE | ID: mdl-23973924

Toll like receptor 4 (TLR4) has been characterized for its ability to recognize bacterial endotoxin lipopolysaccharide (LPS). Considering that infections or inflammatory processes might contribute to the progression of pituitary tumors, we analyzed the TLR4 functional role by evaluating the LPS effect on lactotroph proliferation in primary cultures from experimental pituitary tumors, and examined the involvement of PI3K-Akt and NF-κB activation in this effect. In addition, the role of 17ß-estradiol as a possible modulator of LPS-induced PRL cell proliferation was further investigated. In estrogen-induced hyperplasic pituitaries, LPS triggered lactotroph cell proliferation. However, endotoxin failed to increase the number of lactotrophs taking up BrdU in normal pituitaries. Moreover, incubation with anti-TLR4 antibody significantly reduced LPS-induced lactotroph proliferation, suggesting a functional role of this receptor. As a sign of TLR4 activation, an LPS challenge increased IL-6 release in normal and tumoral cells. By flow cytometry, TLR4 baseline expression was revealed at the plasma membrane of tumoral lactotrophs, without changes noted in the percentage of double PRL/TLR4 positive cells after LPS stimulus. Increases in TLR4 intracellular expression were detected as well as rises in CD14, p-Akt and NF-κB after an LPS challenge, as assessed by western blotting. The TLR4/PRL and PRL/NF-κB co-localization was also corroborated by immunofluorescence and the involvement of PI3K/Akt signaling in lactotroph proliferation and IL-6 release was revealed through the PI3K inhibitor Ly-294002. In addition, 17ß-estradiol attenuated the LPS-evoked increase in tumoral lactotroph proliferation and IL-6 release. Collectively these results demonstrate the presence of functional TLR4 in lactotrophs from estrogen-induced hyperplasic pituitaries, which responded to the proliferative stimulation and IL-6 release induced by LPS through TLR4/CD14, with a contribution of the PI3K-Akt and NF-κB signaling pathways.


Cell Proliferation/drug effects , Lipopolysaccharides/pharmacology , Pituitary Gland/metabolism , Pituitary Neoplasms/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Hyperplasia/metabolism , Interleukin-6/metabolism , Male , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pituitary Gland/ultrastructure , Pituitary Neoplasms/immunology , Pituitary Neoplasms/ultrastructure , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction/physiology
19.
Immunity ; 37(2): 249-63, 2012 Aug 24.
Article En | MEDLINE | ID: mdl-22884314

Inflammation-mediated neurodegeneration occurs in the acute and the chronic phases of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Classically activated (M1) microglia are key players mediating this process. Here, we identified Galectin-1 (Gal1), an endogenous glycan-binding protein, as a pivotal regulator of M1 microglial activation that targets the activation of p38MAPK-, CREB-, and NF-κB-dependent signaling pathways and hierarchically suppresses downstream proinflammatory mediators, such as iNOS, TNF, and CCL2. Gal1 bound to core 2 O-glycans on CD45, favoring retention of this glycoprotein on the microglial cell surface and augmenting its phosphatase activity and inhibitory function. Gal1 was highly expressed in the acute phase of EAE, and its targeted deletion resulted in pronounced inflammation-induced neurodegeneration. Adoptive transfer of Gal1-secreting astrocytes or administration of recombinant Gal1 suppressed EAE through mechanisms involving microglial deactivation. Thus, Gal1-glycan interactions are essential in tempering microglial activation, brain inflammation, and neurodegeneration, with critical therapeutic implications for MS.


Encephalomyelitis, Autoimmune, Experimental/immunology , Galectin 1/immunology , Leukocyte Common Antigens/metabolism , Microglia/immunology , Animals , Astrocytes/metabolism , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/physiopathology , Chemokine CCL2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Galectin 1/metabolism , Galectin 1/therapeutic use , Humans , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Polysaccharides/metabolism , Protein Binding , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Am J Physiol Endocrinol Metab ; 302(10): E1189-97, 2012 May 01.
Article En | MEDLINE | ID: mdl-22354782

Considering that estradiol is a major modulator of prolactin (PRL) secretion, the aim of the present study was to analyze the role of membrane estradiol receptor-α (mERα) in the regulatory effect of this hormone on the PRL secretion induced by thyrotropin-releasing hormone (TRH) by focusing on the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway activation. Anterior pituitary cell cultures from female rats were treated with 17ß-estradiol (E(2), 10 nM) and its membrane-impermeable conjugated estradiol (E(2)-BSA, 10 nM) alone or coincubated with TRH (10 nM) for 30 min, with PRL levels being determined by RIA. Although E(2), E(2)-BSA, TRH, and E(2)/TRH differentially increased the PRL secretion, the highest levels were achieved with E(2)-BSA/TRH. ICI-182,780 did not modify the TRH-induced PRL release but significantly inhibited the PRL secretion promoted by E(2) or E(2)-BSA alone or in coincubation with TRH. The PI3K inhibitors LY-294002 and wortmannin partially inhibited the PRL release induced by E(2)-BSA, TRH, and E(2)/TRH and totally inhibited the PRL levels stimulated by E(2)-BSA/TRH, suggesting that the mER mediated the cooperative effect of E(2) on TRH-induced PRL release through the PI3K pathway. Also, the involvement of this kinase was supported by the translocation of its regulatory subunit p85α from the cytoplasm to the plasma membrane in the lactotroph cells treated with E(2)-BSA and TRH alone or in coincubation. A significant increase of phosphorylated Akt was induced by E(2)-BSA/TRH. Finally, the changes of ERα expression in the plasmalemma of pituitary cells were examined by confocal microscopy and flow cytometry, which revealed that the mobilization of intracellular ERα to the plasma membrane of lactotroph cells was only induced by E(2). These finding showed that E(2) may act as a modulator of the secretory response of lactotrophs induced by TRH through mER, with the contribution by PI3K/Akt pathway activation providing a new insight into the mechanisms underlying the nongenomic action of E(2) in the pituitary.


Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Pituitary Gland, Anterior , Prolactin/metabolism , Signal Transduction/physiology , Thyrotropin-Releasing Hormone/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Female , Membrane Proteins/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Serum Albumin, Bovine/pharmacology , Signal Transduction/drug effects
...