Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Hum Mol Genet ; 30(3-4): 265-276, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33693650

ABSTRACT

Mutations in the WFS1 gene, encoding wolframin (WFS1), cause endoplasmic reticulum (ER) stress and are associated with a rare autosomal-recessive disorder known as Wolfram syndrome (WS). WS is clinically characterized by childhood-onset diabetes mellitus, optic atrophy, deafness, diabetes insipidus and neurological signs. We identified two novel WFS1 mutations in a patient with WS, namely, c.316-1G > A (in intron 3) and c.757A > T (in exon 7). Both mutations, located in the N-terminal region of the protein, were predicted to generate a truncated and inactive form of WFS1. We found that although the WFS1 protein was not expressed in peripheral blood mononuclear cells (PBMCs) of the proband, no constitutive ER stress activation could be detected in those cells. In contrast, WS proband's PBMCs produced very high levels of proinflammatory cytokines (i.e. TNF-α, IL-1ß, and IL-6) in the absence of any stimulus. WFS1 silencing in PBMCs from control subjects by means of small RNA interference also induced a pronounced proinflammatory cytokine profile. The same cytokines were also significantly higher in sera from the WS patient as compared to matched healthy controls. Moreover, the chronic inflammatory state was associated with a dominance of proinflammatory T helper 17 (Th17)-type cells over regulatory T (Treg) lymphocytes in the WS PBMCs. The identification of a state of systemic chronic inflammation associated with WFS1 deficiency may pave the way to innovative and personalized therapeutic interventions in WS.


Subject(s)
Inflammation , Leukocytes, Mononuclear/metabolism , Membrane Proteins/genetics , Mutation , Wolfram Syndrome/metabolism , Child , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/immunology , Sequence Analysis, DNA , Wolfram Syndrome/genetics , Wolfram Syndrome/immunology , Wolfram Syndrome/physiopathology
2.
Nat Immunol ; 12(9): 870-8, 2011 Jul 31.
Article in English | MEDLINE | ID: mdl-21804557

ABSTRACT

Regulation of tryptophan metabolism by indoleamine 2,3-dioxygenase (IDO) in dendritic cells (DCs) is a highly versatile modulator of immunity. In inflammation, interferon-γ is the main inducer of IDO for the prevention of hyperinflammatory responses, yet IDO is also responsible for self-tolerance effects in the longer term. Here we show that treatment of mouse plasmacytoid DCs (pDCs) with transforming growth factor-ß (TGF-ß) conferred regulatory effects on IDO that were mechanistically separable from its enzymic activity. We found that IDO was involved in intracellular signaling events responsible for the self-amplification and maintenance of a stably regulatory phenotype in pDCs. Thus, IDO has a tonic, nonenzymic function that contributes to TGF-ß-driven tolerance in noninflammatory contexts.


Subject(s)
Adaptive Immunity , Dendritic Cells , Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase , Signal Transduction/immunology , Transforming Growth Factor beta/immunology , Adaptive Immunity/drug effects , Animals , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/enzymology , Dendritic Cells/immunology , Humans , Hypersensitivity/immunology , Immune Tolerance/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/pharmacology , Tryptophan/metabolism
3.
EMBO Rep ; 21(12): e49756, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33159421

ABSTRACT

Knowledge of a protein's spatial dynamics at the subcellular level is key to understanding its function(s), interactions, and associated intracellular events. Indoleamine 2,3-dioxygenase 1 (IDO1) is a cytosolic enzyme that controls immune responses via tryptophan metabolism, mainly through its enzymic activity. When phosphorylated, however, IDO1 acts as a signaling molecule in plasmacytoid dendritic cells (pDCs), thus activating genomic effects, ultimately leading to long-lasting immunosuppression. Whether the two activities-namely, the catalytic and signaling functions-are spatially segregated has been unclear. We found that, under conditions favoring signaling rather than catabolic events, IDO1 shifts from the cytosol to early endosomes. The event requires interaction with class IA phosphoinositide 3-kinases (PI3Ks), which become activated, resulting in full expression of the immunoregulatory phenotype in vivo in pDCs as resulting from IDO1-dependent signaling events. Thus, IDO1's spatial dynamics meet the needs for short-acting as well as durable mechanisms of immune suppression, both under acute and chronic inflammatory conditions. These data expand the theoretical basis for an IDO1-centered therapy in inflammation and autoimmunity.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase , Phosphatidylinositol 3-Kinases , Dendritic Cells/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction
4.
Nature ; 511(7508): 184-90, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24930766

ABSTRACT

Disease tolerance is the ability of the host to reduce the effect of infection on host fitness. Analysis of disease tolerance pathways could provide new approaches for treating infections and other inflammatory diseases. Typically, an initial exposure to bacterial lipopolysaccharide (LPS) induces a state of refractoriness to further LPS challenge (endotoxin tolerance). We found that a first exposure of mice to LPS activated the ligand-operated transcription factor aryl hydrocarbon receptor (AhR) and the hepatic enzyme tryptophan 2,3-dioxygenase, which provided an activating ligand to the former, to downregulate early inflammatory gene expression. However, on LPS rechallenge, AhR engaged in long-term regulation of systemic inflammation only in the presence of indoleamine 2,3-dioxygenase 1 (IDO1). AhR-complex-associated Src kinase activity promoted IDO1 phosphorylation and signalling ability. The resulting endotoxin-tolerant state was found to protect mice against immunopathology in Gram-negative and Gram-positive infections, pointing to a role for AhR in contributing to host fitness.


Subject(s)
Disease Resistance/genetics , Disease Resistance/immunology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Bacterial Infections/immunology , Bacterial Infections/metabolism , Disease Resistance/drug effects , Endotoxemia/genetics , Endotoxemia/immunology , Endotoxemia/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/enzymology , Inflammation/genetics , Inflammation/metabolism , Kynurenine/metabolism , Lipopolysaccharides/pharmacology , Mice , Phosphorylation , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Tryptophan Oxygenase/metabolism , src-Family Kinases/metabolism
5.
J Cell Mol Med ; 21(1): 165-176, 2017 01.
Article in English | MEDLINE | ID: mdl-27696702

ABSTRACT

The enzyme indoleamine 2,3-dioxygenase 1 (IDO1) catalyses the initial, rate-limiting step in tryptophan (Trp) degradation, resulting in tryptophan starvation and the production of immunoregulatory kynurenines. IDO1's catalytic function has long been considered as the one mechanism responsible for IDO1-dependent immune suppression by dendritic cells (DCs), which are master regulators of the balance between immunity and tolerance. However, IDO1 also harbours immunoreceptor tyrosine-based inhibitory motifs, (ITIM1 and ITIM2), that, once phosphorylated, bind protein tyrosine phosphatases, (SHP-1 and SHP-2), and thus trigger an immunoregulatory signalling in DCs. This mechanism leads to sustained IDO1 expression, in a feedforward loop, which is particularly important in restraining autoimmunity and chronic inflammation. Yet, under specific conditions requiring that early and protective inflammation be unrelieved, tyrosine-phosphorylated ITIMs will instead bind the suppressor of cytokine signalling 3 (SOCS3), which drives IDO1 proteasomal degradation and shortens the enzyme half-life. To dissect any differential roles of the two IDO1's ITIMs, we generated protein mutants by replacing one or both ITIM-associated tyrosines with phospho-mimicking glutamic acid residues. Although all mutants lost their enzymic activity, the ITIM1 - but not ITIM2 mutant - did bind SHPs and conferred immunosuppressive effects on DCs, making cells capable of restraining an antigen-specific response in vivo. Conversely, the ITIM2 mutant would preferentially bind SOCS3, and IDO1's degradation was accelerated. Thus, it is the selective phosphorylation of either ITIM that controls the duration of IDO1 expression and function, in that it dictates whether enhanced tolerogenic signalling or shutdown of IDO1-dependent events will occur in a local microenvironment.


Subject(s)
Immunosuppressive Agents/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Tyrosine/immunology , Animals , Cytokines/immunology , Dendritic Cells/immunology , Female , Half-Life , Immune Tolerance/immunology , Kynurenine/immunology , Mice , Mice, Inbred C57BL , Phosphorylation/immunology , Protein Domains/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Tryptophan/immunology
6.
J Immunol ; 189(5): 2283-9, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22844124

ABSTRACT

Short synthetic oligodeoxynucleotides (ODNs) rich in CpG or GpG motifs have been considered as potential modulators of immunity in clinical settings. In this study, we show that a synthetic GpC-ODN conferred highly suppressive activity on mouse splenic plasmacytoid dendritic cells, demonstrable in vivo in a skin test assay. The underlying mechanism involved signaling by noncanonical NF-κB family members and TGF-ß-dependent expression of the immunoregulatory enzyme IDO. Unlike CpG-ODNs, the effects of GpC-ODN required TLR7/TRIF-mediated but not TLR9/MyD88-mediated events, as do sensing of viral ssRNA and the drug imiquimod. Induction of IDO by a GpC-containing ODN could also be demonstrated in human dendritic cells, allowing those cells to assist FOXP3+ T cell generation in vitro. Among potentially therapeutic ODNs, this study identifies GpC-rich sequences as novel activators of TLR7-mediated, IDO-dependent regulatory responses.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/genetics , Oligodeoxyribonucleotides/pharmacology , Animals , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/enzymology , Female , Humans , Immune Tolerance/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Interferon-beta/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , Oligodeoxyribonucleotides/genetics , Protein Structure, Tertiary , Receptors, Interleukin-1/physiology , Signal Transduction/immunology , Toll-Like Receptor 7/physiology , Transcription, Genetic/immunology , Transforming Growth Factor beta/physiology
7.
Nat Med ; 13(5): 579-86, 2007 May.
Article in English | MEDLINE | ID: mdl-17417651

ABSTRACT

Glucocorticoid-induced tumor necrosis factor receptor (GITR) on T cells and its natural ligand, GITRL, on accessory cells contribute to the control of immune homeostasis. Here we show that reverse signaling through GITRL after engagement by soluble GITR initiates the immunoregulatory pathway of tryptophan catabolism in mouse plasmacytoid dendritic cells, by means of noncanonical NF-kappaB-dependent induction of indoleamine 2,3-dioxygenase (IDO). The synthetic glucocorticoid dexamethasone administered in vivo activated IDO through the symmetric induction of GITR in CD4(+) T cells and GITRL in plasmacytoid dendritic cells. The drug exerted IDO-dependent protection in a model of allergic airway inflammation. Modulation of tryptophan catabolism via the GITR-GITRL coreceptor system might represent an effective therapeutic target in immune regulation. Induction of IDO could be an important mechanism underlying the anti-inflammatory action of corticosteroids.


Subject(s)
Dexamethasone/pharmacology , Hypersensitivity/prevention & control , Hypersensitivity/physiopathology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Receptors, Nerve Growth Factor/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Animals , Dendritic Cells/immunology , Disease Models, Animal , Enzyme Activation/drug effects , Glucocorticoid-Induced TNFR-Related Protein , Humans , Mice , Receptors, Nerve Growth Factor/drug effects , Receptors, Tumor Necrosis Factor/drug effects , Spleen/immunology , Tumor Necrosis Factors/physiology
8.
Proc Natl Acad Sci U S A ; 105(52): 20828-33, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19088199

ABSTRACT

Despite their common ability to activate intracellular signaling through CD80/CD86 molecules, cytotoxic T lymphocyte antigen 4 (CTLA-4)-Ig and CD28-Ig bias the downstream response in opposite directions, the latter promoting immunity, and CTLA-4-Ig tolerance, in dendritic cells (DCs) with opposite but flexible programs of antigen presentation. Nevertheless, in the absence of suppressor of cytokine signaling 3 (SOCS3), CD28-Ig-and the associated, dominant IL-6 response-become immunosuppressive and mimic the effect of CTLA-4-Ig, including a high functional expression of the tolerogenic enzyme indoleamine 2,3-dioxygenase (IDO). Here we show that forced SOCS3 expression antagonized CTLA-4-Ig activity in a proteasome-dependent fashion. Unrecognized by previous studies, IDO appeared to possess two tyrosine residues within two distinct putative immunoreceptor tyrosine-based inhibitory motifs, VPY(115)CEL and LLY(253)EGV. We found that SOCS3-known to interact with phosphotyrosine-containing peptides and be selectively induced by CD28-Ig/IL-6-would bind IDO and target the IDO/SOCS3 complex for ubiquitination and subsequent proteasomal degradation. This event accounted for the ability of CD28-Ig and IL-6 to convert otherwise tolerogenic, IDO-competent DCs into immunogenic cells. Thus onset of immunity in response to antigen within an early inflammatory context requires that IDO be degraded in tolerogenic DCs. In addition to identifying SOCS3 as a candidate signature for mouse DC subsets programmed to direct immunity, this study demonstrates that IDO undergoes regulatory proteolysis in response to immunogenic stimuli.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Proteasome Endopeptidase Complex/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Ubiquitination/immunology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/genetics , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD28 Antigens/genetics , CD28 Antigens/immunology , CD28 Antigens/metabolism , CTLA-4 Antigen , Dendritic Cells/cytology , Dendritic Cells/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-6/biosynthesis , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Inbred DBA , Mice, Transgenic , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Suppressor of Cytokine Signaling Proteins/genetics , Ubiquitination/genetics
9.
J Exp Med ; 200(8): 1051-62, 2004 Oct 18.
Article in English | MEDLINE | ID: mdl-15492127

ABSTRACT

Prediabetes and diabetes in nonobese diabetic (NOD) mice have been targeted by a variety of immunotherapies, including the use of a soluble form of cytotoxic T lymphocyte antigen 4 (CTLA-4) and interferon (IFN)-gamma. The cytokine, however, fails to activate tolerogenic properties in dendritic cells (DCs) from highly susceptible female mice early in prediabetes. The defect is characterized by impaired induction of immunosuppressive tryptophan catabolism, is related to transient blockade of the signal transducer and activator of transcription (STAT)1 pathway of intracellular signaling by IFN-gamma, and is caused by peroxynitrite production. Here, we show that soluble CTLA-4 imparts suppressive properties to DCs from early prediabetic NOD female mice through mechanisms that rely on autocrine signaling by IFN-gamma. Although phosphorylation of STAT1 in response to IFN-gamma is compromised in those mice, CTLA-4 obviates the defect. IFN-gamma-driven expression of tryptophan catabolism by CTLA-4-immunoglobulin is made possible through the concomitant activation of the Forkhead Box class O (FOXO) transcription factor FOXO3a, induction of the superoxide dismutase gene, and prevention of peroxynitrite formation.


Subject(s)
Dendritic Cells/metabolism , Immunoconjugates/pharmacology , Oxidative Stress , Transcription Factors/metabolism , Abatacept , Animals , Chromones/pharmacology , DNA-Binding Proteins/metabolism , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors , Interferon-gamma/pharmacology , Male , Metalloporphyrins/pharmacology , Mice , Mice, Inbred NOD , Morpholines/pharmacology , PTEN Phosphohydrolase , Peroxynitrous Acid/biosynthesis , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Tyrosine Phosphatases/genetics , STAT1 Transcription Factor , Signal Transduction , Trans-Activators/metabolism , Tumor Suppressor Proteins/genetics
10.
Trends Mol Med ; 15(2): 41-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19162548

ABSTRACT

The maintenance of self-tolerance is an integral part of the immune surveillance process, in which cytokines act as master regulators of a complex network involving multiple cell types. On such cytokines, transforming growth factor-beta (TGF-beta) exerts a suppressive control over immune reactivity, which so far appears to be mostly confined to the T-cell compartment. Recently, dendritic cells (DCs) have been found to be both an early source and a target of TGF-beta actions. In these cells, autocrine, paracrine and T-cell-derived TGF-beta activates the tolerogenic pathway of tryptophan catabolism - mediated by indoleamine 2,3-dioxygenase (IDO) - resulting in a burst of regulatory kynurenines that contribute to establishing a state of 'infectious tolerance'. Current molecular insights suggest a synergistic potential for TGF-beta and IDO in physiologically or therapeutically opposing human pathologies sustained by over-reacting immune responses.


Subject(s)
Immune Tolerance/immunology , Kynurenine/immunology , Transforming Growth Factor beta/immunology , Animals , Dendritic Cells/immunology , Humans , Immune System/physiology , Interferons/immunology , Kynurenine/chemistry , Signal Transduction/physiology , T-Lymphocytes/immunology
11.
J Immunol ; 181(8): 5194-8, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832670

ABSTRACT

CD8(-) and CD8(+) dendritic cells (DCs) are distinct subsets of mouse splenic accessory cells with opposite but flexible programs of Ag presentation, leading to immunogenic and tolerogenic responses, respectively. In this study, we show that the default tolerogenic function of CD8(+) DCs relies on autocrine TGF-beta, which sustains the activation of IDO in response to environmental stimuli. CD8(-) DCs do not produce TGF-beta, yet externally added TGF-beta induces IDO and turns those cells from immunogenic into tolerogenic cells. The acquisition of a suppressive phenotype by CD8(-) DCs correlates with activation of the PI3K/Akt and noncanonical NF-kappaB pathways. These data are the first to link TGF-beta signaling with IDO in controlling spontaneous tolerogenesis by DCs.


Subject(s)
Antigen Presentation , Autocrine Communication/immunology , Dendritic Cells/immunology , Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Transforming Growth Factor beta1/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/genetics , Autocrine Communication/drug effects , Autocrine Communication/genetics , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8 Antigens/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Activation/immunology , Immune Tolerance/drug effects , Immune Tolerance/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology
12.
Front Immunol ; 10: 1973, 2019.
Article in English | MEDLINE | ID: mdl-31481962

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzes the first step in the kynurenine pathway of tryptophan (Trp) degradation that produces several biologically active Trp metabolites. L-kynurenine (Kyn), the first byproduct by IDO1, promotes immunoregulatory effects via activation of the Aryl hydrocarbon Receptor (AhR) in dendritic cells (DCs) and T lymphocytes. We here identified the nuclear coactivator 7 (NCOA7) as a molecular target of 3-hydroxyanthranilic acid (3-HAA), a Trp metabolite produced downstream of Kyn along the kynurenine pathway. In cells overexpressing NCOA7 and AhR, the presence of 3-HAA increased the association of the two molecules and enhanced Kyn-driven, AhR-dependent gene transcription. Physiologically, conventional (cDCs) but not plasmacytoid DCs or other immune cells expressed high levels of NCOA7. In cocultures of CD4+ T cells with cDCs, the co-addition of Kyn and 3-HAA significantly increased the induction of Foxp3+ regulatory T cells and the production of immunosuppressive transforming growth factor ß in an NCOA7-dependent fashion. Thus, the co-presence of NCOA7 and the Trp metabolite 3-HAA can selectively enhance the activation of ubiquitary AhR in cDCs and consequent immunoregulatory effects. Because NCOA7 is often overexpressed and/or mutated in tumor microenvironments, our current data may provide evidence for a new immune check-point mechanism based on Trp metabolism and AhR.


Subject(s)
3-Hydroxyanthranilic Acid/metabolism , Dendritic Cells/metabolism , Nuclear Receptor Coactivators/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Dendritic Cells/immunology , Female , Humans , Kynurenine/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Nuclear Receptor Coactivators/immunology , Receptors, Aryl Hydrocarbon/immunology , T-Lymphocytes, Regulatory/immunology
13.
Cancer Res ; 66(5): 2630-8, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16510582

ABSTRACT

Neural progenitor-like cells have been isolated from bone marrow and the cells have the ability of tracking intracranial tumor. However, the capacity of the cells to deliver molecules for activating immune response against intracranial tumor and the identity of cellular and molecular factors that are involved in such immune responses have yet to be elucidated. Here, we isolated neural stem-like cells from the bone marrow of adult mice. The isolated cells were capable of producing progenies of three lineages, neurons, astrocytes, and oligodendrocytes, in vitro and tracking glioma in vivo. By genetically manipulating bone marrow-derived neural stem-like cells (BM-NSC) to express a recently discovered cytokine, interleukin (IL)-23, the cells showed protective effects in intracranial tumor-bearing C57BL/6 mice. Depletion of subpopulation lymphocytes showed that CD8(+) T cells were critical for the antitumor immunity of IL-23-expressing BM-NSCs and that CD4(+) T cells and natural killer (NK) cells participated in the activity. Furthermore, the IL-23-expressing BM-NSC-treated survivors were resistant to the same tumor rechallenge associated with enhanced IFN-gamma, but not IL-17, expression in the brain tissue. Taken together, these data suggest that IL-23-expressing BM-NSCs can effectively induce antitumor immunity against intracranial gliomas. CD8(+) T cells are critical for such antitumor activity; in addition, CD4(+) T cells and NK cells are also involved.


Subject(s)
Bone Marrow Cells/immunology , Brain Neoplasms/therapy , Glioma/therapy , Immunotherapy, Adoptive/methods , Interleukins/biosynthesis , Neurons/immunology , Stem Cells/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Glioma/immunology , Glioma/metabolism , Glioma/pathology , Interleukin-23 , Interleukin-23 Subunit p19 , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , NIH 3T3 Cells , Neurons/cytology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocytes, Cytotoxic/immunology , Transgenes
14.
JCI Insight ; 3(6)2018 03 22.
Article in English | MEDLINE | ID: mdl-29563329

ABSTRACT

A defect in indoleamine 2,3-dioxygenase 1 (IDO1), which is responsible for immunoregulatory tryptophan catabolism, impairs development of immune tolerance to autoantigens in NOD mice, a model for human autoimmune type 1 diabetes (T1D). Whether IDO1 function is also defective in T1D is still unknown. We investigated IDO1 function in sera and peripheral blood mononuclear cells (PBMCs) from children with T1D and matched controls. These children were further included in a discovery study to identify SNPs in IDO1 that might modify the risk of T1D. T1D in children was characterized by a remarkable defect in IDO1 function. A common haplotype, associated with dysfunctional IDO1, increased the risk of developing T1D in the discovery and also confirmation studies. In T1D patients sharing such a common IDO1 haplotype, incubation of PBMCs in vitro with tocilizumab (TCZ) - an IL-6 receptor blocker - would, however, rescue IDO1 activity. In an experimental setting with diabetic NOD mice, TCZ was found to restore normoglycemia via IDO1-dependent mechanisms. Thus, functional SNPs of IDO1 are associated with defective tryptophan catabolism in human T1D, and maneuvers aimed at restoring IDO1 function would be therapeutically effective in at least a subgroup of T1D pediatric patients.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Tryptophan/metabolism , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Child , Cytokines/metabolism , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic , Genetic Association Studies , Humans , Immune Tolerance , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Multivariate Analysis , Polymorphism, Single Nucleotide , Receptors, Interleukin-6/drug effects
15.
Cytokine Growth Factor Rev ; 35: 37-45, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28545736

ABSTRACT

Indoleamine 2,3-dioxygenases (IDOs) - belonging in the heme dioxygenase family and degrading tryptophan - are responsible for the de novo synthesis of nicotinamide adenine dinucleotide (NAD+). As such, they are expressed by a variety of invertebrate and vertebrate species. In mammals, IDO1 has remarkably evolved to expand its functions, so to become a prominent homeostatic regulator, capable of modulating infection and immunity in multiple ways, including local tryptophan deprivation, production of biologically active tryptophan catabolites, and non-enzymatic cell-signaling activity. Much like IDO1, arginase 1 (Arg1) is an immunoregulatory enzyme that catalyzes the degradation of arginine. Here, we discuss the possible role of amino-acid degradation as related to the evolution of the immune systems and how the functions of those enzymes are linked by an entwined pathway selected by phylogenesis to meet the newly arising needs imposed by an evolving environment.


Subject(s)
Amino Acids/metabolism , Dendritic Cells/immunology , Animals , Arginase/metabolism , Dendritic Cells/enzymology , Gene Expression Regulation , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/immunology , Interleukin-4/immunology , Mice , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction , Transforming Growth Factor beta1/immunology , Tryptophan/metabolism
16.
Front Immunol ; 8: 428, 2017.
Article in English | MEDLINE | ID: mdl-28450863

ABSTRACT

Bortezomib (BTZ) is a first-in-class proteasome inhibitor approved for the therapy of multiple myeloma that also displays unique regulatory activities on immune cells. The enzyme indoleamine 2,3-dioxygenase 1 (IDO1) is a tryptophan metabolizing enzyme exerting potent immunoregulatory effects when expressed in dendritic cells (DCs), the most potent antigen-presenting cells capable of promoting either immunity or tolerance. We previously demonstrated that, in inflammatory conditions, IDO1 is subjected to proteasomal degradation in DCs, turning these cells from immunoregulatory to immunostimulatory. In non-obese diabetic (NOD) mice, an experimental model of autoimmune diabetes, we also identified an IDO1 defect such that the DCs do not develop tolerance toward pancreatic islet autoantigens. We found that BTZ rescues IDO1 protein expression in vitro in a particular subset of DCs, i.e., plasmacytoid DCs (pDCs) from NOD mice. When administered in vivo to prediabetic mice, the drug prevented diabetes onset through IDO1- and pDC-dependent mechanisms. Although the drug showed no therapeutic activity when administered alone to overtly diabetic mice, its combination with otherwise suboptimal dosages of autoimmune-preventive anti-CD3 antibody resulted in disease reversal in 70% diabetic mice, a therapeutic effect similar to that afforded by full-dosage anti-CD3. Thus, our data indicate a potential for BTZ in the immunotherapy of autoimmune diabetes and further underline the importance of IDO1-mediated immune regulation in such disease.

17.
Transpl Immunol ; 17(1): 58-60, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17157218

ABSTRACT

Tryptophan catabolism is a tolerogenic effector system in regulatory T cell function, yet the general mechanisms whereby tryptophan catabolism affects T cell responses remain unclear. We provide evidence that its effects include the emergence of a regulatory phenotype in naive CD4(+)CD25(-) cells via the general control non-depressing 2 (GCN2) protein kinase mediated induction of the forkhead transcription factor Foxp3. These cells are capable of effective control of diabetogenic T cells in vivo.


Subject(s)
Autoimmunity , T-Lymphocytes, Regulatory/immunology , Tryptophan/immunology , Tryptophan/metabolism , Animals , Dendritic Cells/immunology , Female , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Immune Tolerance , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Mice, Inbred DBA , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Models, Immunological , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism
18.
Neuropharmacology ; 102: 59-71, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26522434

ABSTRACT

Metabotropic glutamate receptor 4 (mGluR4) possesses immune modulatory properties in vivo, such that a positive allosteric modulator (PAM) of the receptor confers protection on mice with relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE). ADX88178 is a newly-developed, one such mGluR4 modulator with high selectivity, potency, and optimized pharmacokinetics. Here we found that application of ADX88178 in the RR-EAE model system converted disease into a form of mild-yet chronic-neuroinflammation that remained stable for over two months after discontinuing drug treatment. In vitro, ADX88178 modulated the cytokine secretion profile of dendritic cells (DCs), increasing production of tolerogenic IL-10 and TGF-ß. The in vitro effects required activation of a Gi-independent, alternative signaling pathway that involved phosphatidylinositol-3-kinase (PI3K), Src kinase, and the signaling activity of indoleamine 2,3-dioxygenase 1 (IDO1). A PI3K inhibitor as well as small interfering RNA targeting Ido1-but not pertussis toxin, which affects Gi protein-dependent responses-abrogated the tolerogenic effects of ADX88178-conditioned DCs in vivo. Thus our data indicate that, in DCs, highly selective and potent mGluR4 PAMs such as ADX88178 may activate a Gi-independent, long-lived regulatory pathway that could be therapeutically exploited in chronic autoimmune diseases such as multiple sclerosis.


Subject(s)
Dendritic Cells/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Allosteric Regulation/physiology , Animals , Dendritic Cells/metabolism , Female , Mice , Phosphatidylinositol 3-Kinases/metabolism , Pyrimidines/pharmacology , RNA, Small Interfering , Thiazoles/pharmacology
19.
Crit Rev Immunol ; 22(5-6): 373-90, 2002.
Article in English | MEDLINE | ID: mdl-12803317

ABSTRACT

Recent evidence from different experimental systems has demonstrated that autocrine activation of antigen-presenting cells (APCs) may be important at the initiation of an immune response, and that a specific set of cytokines may meet the dual needs of activating APCs and priming and/or maintaining the antigen-specific T-cell response. Composite factors with p40, including IL-12 (p40/p35) and IL-23 (p40/p19), may be two such immunoregulatory cytokines, their effects encompassing actions on both myeloid APCs and T cells. However, although both cytokines enhance the Th1 costimulatory functions of APCs, and IL-23 does induce IL-12 from APCs, their effects, which in part overlap, can be differentiated from one another. This review summarizes recent data on the actions of IL-12 and IL-23 on dendritic cells and macrophages at the interface between innate and adaptive immunity.


Subject(s)
Dendritic Cells/immunology , Immunity, Cellular/immunology , Immunity, Innate/immunology , Interleukin-12/immunology , Interleukins/immunology , Macrophages/immunology , Animals , Autocrine Communication/immunology , Dendritic Cells/metabolism , Humans , Interleukin-12/metabolism , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukins/metabolism , Macrophages/metabolism , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism , Signal Transduction/immunology
20.
Nat Commun ; 4: 1852, 2013.
Article in English | MEDLINE | ID: mdl-23673637

ABSTRACT

CpG-rich oligodeoxynucleotides activate the immune system, leading to innate and acquired immune responses. The immune-stimulatory effects of CpG-rich oligodeoxynucleotides are being exploited as a therapeutic approach. Here we show that at high doses, CpG-rich oligodeoxynucleotides promote an opposite, tolerogenic response in mouse plasmacytoid dendritic cells in vivo and in a human in vitro model. Unveiling a previously undescribed role for TRIF and TRAF6 proteins in Toll-like receptor 9 (TLR9) signalling, we demonstrate that physical association of TLR9, TRIF and TRAF6 leads to activation of noncanonical NF-κB signalling and the induction of IRF3- and TGF-ß-dependent immune-suppressive tryptophan catabolism. In vivo, the TLR9-TRIF circuit--but not MyD88 signalling--was required for CpG protection against allergic inflammation. Our findings may be relevant to an increased understanding of the complexity of Toll-like receptor signalling and optimal exploitation of CpG-rich oligodeoxynucleotides as immune modulators.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Immune Tolerance/drug effects , Oligodeoxyribonucleotides/pharmacology , Signal Transduction/immunology , Toll-Like Receptor 9/metabolism , Animals , Antigens, CD/metabolism , Aspergillosis, Allergic Bronchopulmonary/immunology , Aspergillosis, Allergic Bronchopulmonary/pathology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Immunomodulation/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon Regulatory Factors/metabolism , Interferon Type I/metabolism , Interleukin-23/genetics , Interleukin-23/metabolism , Membrane Glycoproteins/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Pneumonia/immunology , Pneumonia/pathology , Signal Transduction/drug effects , Skin Tests , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL