Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 115
1.
Front Immunol ; 12: 634441, 2021.
Article En | MEDLINE | ID: mdl-33717175

Birdshot Uveitis (BU) is a blinding inflammatory eye condition that only affects HLA-A29-positive individuals. Genetic association studies linked ERAP2 with BU, an aminopeptidase which trims peptides before their presentation by HLA class I at the cell surface, which suggests that ERAP2-dependent peptide presentation by HLA-A29 drives the pathogenesis of BU. However, it remains poorly understood whether the effects of ERAP2 on the HLA-A29 peptidome are distinct from its effect on other HLA allotypes. To address this, we focused on the effects of ERAP2 on the immunopeptidome in patient-derived antigen presenting cells. Using complementary HLA-A29-based and pan-class I immunopurifications, isotope-labeled naturally processed and presented HLA-bound peptides were sequenced by mass spectrometry. We show that the effects of ERAP2 on the N-terminus of ligands of HLA-A29 are shared across endogenous HLA allotypes, but discover and replicate that one peptide motif generated in the presence of ERAP2 is specifically bound by HLA-A29. This motif can be found in the amino acid sequence of putative autoantigens. We further show evidence for internal sequence specificity for ERAP2 imprinted in the immunopeptidome. These results reveal that ERAP2 can generate an HLA-A29-specific antigen repertoire, which supports that antigen presentation is a key disease pathway in BU.


Aminopeptidases/metabolism , Antigen-Presenting Cells/enzymology , Autoantigens/metabolism , Autoimmunity , Birdshot Chorioretinopathy/enzymology , HLA-A Antigens/metabolism , Aged, 80 and over , Amino Acid Motifs , Aminopeptidases/genetics , Antigen-Presenting Cells/immunology , Autoantigens/genetics , Autoantigens/immunology , Birdshot Chorioretinopathy/diagnosis , Birdshot Chorioretinopathy/genetics , Birdshot Chorioretinopathy/immunology , Cell Line , Female , HLA-A Antigens/genetics , HLA-A Antigens/immunology , Humans
2.
J Invest Dermatol ; 138(3): 679-687, 2018 03.
Article En | MEDLINE | ID: mdl-29054599

The enzyme indoleamine 2,3-dioxygenase (IDO) is emerging as a facilitator of cancer development through its effects on cancer-associated inflammation. Recent studies report a significant improvement of the response rates in melanoma patients to PD-1 antibodies when IDO inhibitors were added to the regimen. Data on IDO expression in primary human melanomas are, however, incomplete and conflicting. Here, we show that the level of IDO expression in primary human melanoma cells significantly correlates with Breslow thickness (P = 0.003), the presence of tumor-infiltrating lymphocytes (P = 0.029), and the intensity of the peritumoral inflammatory infiltrate (P = 0.001). The expression of IDO in melanoma cells predicted independently of Breslow thickness and tumor stage (P = 0.04). We further show that CD11c+ dendritic cells and CD68+ macrophages in the microenvironment of melanomas express IDO. The level of IDO expression in antigen-presenting cells correlated positively to peritumoral inflammation (P = 0.001) but not to tumor-infiltrating lymphocytes. Significant negative correlation with progression-free survival was found for patients for whom antigen-presenting cells were very strongly IDO positive. These results suggest that IDO induction within melanoma cells may directly reflect tumor progression, whereas IDO in antigen-presenting cells may determine immune surveillance with impact on local and systemic tolerance.


Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Melanoma/enzymology , Skin Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Antigen-Presenting Cells/enzymology , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Macrophages/enzymology , Male , Melanoma/immunology , Melanoma/mortality , Melanoma/pathology , Middle Aged , Prognosis , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Tumor Microenvironment
3.
Am J Physiol Renal Physiol ; 312(4): F661-F670, 2017 04 01.
Article En | MEDLINE | ID: mdl-28122713

The protein dipeptidyl peptidase 4 (DPP4) is a target in diabetes management and reduction of associated cardiovascular risk. Inhibition of the enzymatic function and genetic deletion of DPP4 is associated with tremendous benefits to the heart, vasculature, adipose tissue, and the kidney in rodent models of obesity, diabetes and hypertension, and associated complications. The recently concluded, "Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mellitus-Thrombolysis in Myocardial Infarction 53" trial revealed a reduction in proteinuria in chronic kidney disease patients (stages 1-3). These results have spurred immense interest in the nonenzymatic and enzymatic role of DPP4 in the kidney. DPP4 is expressed predominantly in the glomeruli and S1-S3 segments of the nephron and to a lesser extent in other segments. DPP4 is known to facilitate absorption of cleaved dipeptides and regulate the function of the sodium/hydrogen exchanger-3 in the proximal tubules. DPP4, also known as CD26, has an important role in costimulation of lymphocytes via caveolin-1 on antigen-presenting cells in peripheral blood. Herein, we present our perspectives for the ongoing interest in the role of DPP4 in the kidney.


Diabetes Mellitus/enzymology , Diabetic Nephropathies/enzymology , Dipeptidyl Peptidase 4/metabolism , Hypertension/enzymology , Kidney/enzymology , Renal Insufficiency, Chronic/enzymology , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Caveolin 1/metabolism , Diabetes Mellitus/drug therapy , Diabetes Mellitus/immunology , Diabetes Mellitus/physiopathology , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/immunology , Diabetic Nephropathies/physiopathology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Disease Progression , Humans , Hypertension/drug therapy , Hypertension/immunology , Hypertension/physiopathology , Kidney/drug effects , Kidney/immunology , Kidney/physiopathology , Lymphocyte Activation , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/physiopathology , Signal Transduction , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism
4.
J Immunol Methods ; 432: 87-94, 2016 May.
Article En | MEDLINE | ID: mdl-26899824

Cathepsin S (CTSS) is a eukaryotic protease mostly expressed in professional antigen presenting cells (APCs). Since CTSS activity regulation plays a role in the pathogenesis of various autoimmune diseases like multiple sclerosis, atherosclerosis, Sjögren's syndrome and psoriasis as well as in cancer progression, there is an ongoing interest in the reliable detection of cathepsin S activity. Various applications have been invented for specific detection of this enzyme. However, most of them have only been shown to be suitable for human samples, do not deliver quantitative results or the experimental procedure requires technical equipment that is not commonly available in a standard laboratory. We have tested a fluorogen substrate, Mca-GRWPPMGLPWE-Lys(Dnp)-DArg-NH2, that has been described to specifically detect CTSS activities in human APCs for its potential use for mouse samples. We have modified the protocol and thereby offer a cheap, easy, reproducible and quick activity assay to detect CTSS activities in mouse APCs. Since most of basic research on CTSS is performed in mice, this method closes a gap and offers a possibility for reliable and quantitative CTSS activity detection that can be performed in almost every laboratory.


Antigen-Presenting Cells/enzymology , Cathepsins/metabolism , Escherichia coli Infections/enzymology , Fluorescent Dyes/metabolism , Peptides/metabolism , Spectrometry, Fluorescence , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/microbiology , Cathepsins/antagonists & inhibitors , Cathepsins/deficiency , Cathepsins/genetics , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Hydrolysis , Leucine/analogs & derivatives , Leucine/pharmacology , Mice, Knockout , Reproducibility of Results , Substrate Specificity , Time Factors
5.
Mol Immunol ; 68(2 Pt A): 124-8, 2015 Dec.
Article En | MEDLINE | ID: mdl-26116226

Gamma-interferon-inducible lysosomal thiol reductase (GILT) is the only enzyme known to catalyze disulfide bond reduction in the endocytic pathway. GILT facilitates the presentation of a subset of epitopes from disulfide bond-containing antigens. Enhanced presentation of MHC class II-restricted epitopes alters central tolerance and modulates CD4+ T cell-mediated autoimmunity. Improved cross-presentation of viral epitopes results in improved cross-priming of viral-specific CD8+ T cells. GILT regulates the cellular redox state. In GILT-/- cells, there is a shift from the reduced to the oxidized form of glutathione, resulting in mitochondrial autophagy, decreased superoxide dismutase 2, and elevated superoxide levels. GILT expression diminishes cellular activation, including decreased phosphorylated ERK1/2, and decreases cellular proliferation. GILT enhances the activity of bacterial hemolysins, such as listeriolysin O, and increases bacterial replication and infection. GILT expression in cancer cells is associated with improved patient survival. These diverse roles of GILT are discussed.


Antigen Presentation , Antigen-Presenting Cells/immunology , Histocompatibility Antigens Class II/immunology , Lysosomes/immunology , Oxidoreductases Acting on Sulfur Group Donors/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/enzymology , Antigens, Viral/genetics , Antigens, Viral/immunology , Autophagy , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Disulfides/chemistry , Disulfides/immunology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Glutathione/immunology , Glutathione/metabolism , Histocompatibility Antigens Class II/genetics , Humans , Lysosomes/enzymology , Mitochondria/immunology , Mitochondria/metabolism , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/immunology
6.
J Leukoc Biol ; 94(6): 1113-21, 2013 Dec.
Article En | MEDLINE | ID: mdl-23883517

AMPK is a serine/threonine kinase that regulates energy homeostasis and metabolic stress in eukaryotes. Previous work from our laboratory, as well as by others, has provided evidence that AMPKα1 acts as a negative regulator of TLR-induced inflammatory function. Herein, we demonstrate that AMPKα1-deficient macrophages and DCs exhibit heightened inflammatory function and an enhanced capacity for antigen presentation favoring the promotion of Th1 and Th17 responses. Macrophages and DCs generated from AMPKα1-deficient mice produced higher levels of proinflammatory cytokines and decreased production of the anti-inflammatory cytokine IL-10 in response to TLR and CD40 stimulation as compared with WT cells. In assays of antigen presentation, AMPKα1 deficiency in the myeloid APC and T cell populations contributed to enhanced IL-17 and IFN-γ production. Focusing on the CD154-CD40 interaction, we found that CD40 stimulation resulted in increased phosphorylation of ERK1/2, p38, and NF-κB p65 and decreased activation of the anti-inflammatory Akt -GSK3ß-CREB pathway in DCs deficient for AMPKα1. Our data demonstrate that AMPKα1 serves to attenuate LPS and CD40-mediated proinflammatory activity of myeloid APCs and that AMPKα1 activity in both APC and T cells contributes to T cell functional polarization during antigen presentation.


AMP-Activated Protein Kinases/immunology , Antigen Presentation , Antigen-Presenting Cells/immunology , CD40 Antigens/immunology , MAP Kinase Signaling System/immunology , Myeloid Cells/immunology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/enzymology , CD40 Antigens/genetics , CD40 Antigens/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/enzymology , Th1 Cells/cytology , Th1 Cells/enzymology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/enzymology , Th17 Cells/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transcription Factor RelA/metabolism
7.
Autoimmunity ; 46(7): 439-45, 2013 Nov.
Article En | MEDLINE | ID: mdl-23763593

Proteasome generates peptides for presentation to CD8+ T-cells by antigen presenting cells (APCs) and peripheral cells. Inflammation alters proteasome to immunoproteasome, producing a different pool of peptides. The effect of immunoproteasome expression in APCs and in peripheral cells on CD8+ T-cell self-tolerance was evaluated. Splenocytes (SP) were used as a source of APCs and CD8+ T-cells. Renal epithelial cells (RC) from the same mouse strain were used as peripheral cells. Lipopolysaccharide (LPS) was used for increasing immunoproteasome expression, whereas transduction with lentiviral particles encoding short-hairpin RNA targeting LMP7 (LnLMP7) were used for decreasing the expression of the LMP7 subunit of the immunoproteasome in SP and/or RC. LMP7 expression was tested with Western blotting, while the cytotoxicity of isolated CD8+ T-cells against RC with LDH release assay. LPS increased LMP7 expression in SP, while did not affect its expression in RC. LnLMP7 decreased LMP7 expression in both LPS-treated and untreated SP and RC. CD8+ T-cell auto-reactivity increased in case of LPS-treated APCs, further enhanced in the event of both APCs and RC treated with LPS. Transduction of APCs or RC with LnLMP7 decreased CD8+ T-cell auto-reactivity, which was further decreased in case of concurrent transduction of both APCs and RC. In conclusion, in controlled in vitro conditions, exposure to LPS of APCs and peripheral cells induces CD8+ T-cell auto-reactivity. Over-expression of the LMP7 subunit of the immunoproteasome in APCs due to LPS exposure, as well as LMP7 expression in peripheral cells, are required for CD8+ T-cell auto-reactivity.


Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Epithelial Cells/immunology , Lipopolysaccharides/pharmacology , Proteasome Endopeptidase Complex/physiology , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/metabolism , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Kidney/cytology , Kidney/immunology , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Primary Cell Culture , Proteasome Endopeptidase Complex/biosynthesis , Proteasome Endopeptidase Complex/genetics , Protein Subunits/biosynthesis , Protein Subunits/physiology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Transduction, Genetic
8.
Antioxid Redox Signal ; 18(12): 1463-74, 2013 Apr 20.
Article En | MEDLINE | ID: mdl-22900704

SIGNIFICANCE: An unexpected finding, revealed by positional cloning of genetic polymorphisms controlling models for rheumatoid arthritis, exposed a new function of Ncf1 and NADPH oxidase (NOX) 2 controlled oxidative burst. RECENT ADVANCES: A decreased capacity to produce ROS due to a natural polymorphism was found to be the major factor leading to more severe arthritis and increased T cell-dependent autoimmunity. CRITICAL ISSUES: In the vein of this finding, we here review a possible new role of ROS in regulating inflammatory cell and autoreactive T cell activity. It is postulated that peroxide is an immunologic transmitter secreted by antigen-presenting cells that downregulate the responses by autoreactive T cells. FUTURE DIRECTIONS: This may operate at different levels of T cell selection and activation: during negative selection in the thymus, priming of T cells in draining lymph nodes, and while interacting with macrophages in peripheral target tissues.


Hydrogen Peroxide/metabolism , Immunologic Factors/metabolism , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Arthritis, Rheumatoid/immunology , Humans , Lymphoid Tissue/immunology , Macrophages/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Respiratory Burst , Thymus Gland/immunology
9.
Blood ; 121(7): 1145-56, 2013 Feb 14.
Article En | MEDLINE | ID: mdl-23243279

Antigen presenting cells (APCs) that express a catalytically inactive version of the deubiquitylase YOD1 (YOD1-C160S) present exogenous antigens more efficiently to CD8(+) T cells, both in vitro and in vivo. Compared with controls, immunization of YOD1-C160S mice led to greater expansion of specific CD8(+) T cells and showed improved control of infection with a recombinant -herpes virus, MHV-68, engineered to express SIINFEKL peptide, the ligand for the ovalbumin-specific TCR transgenic OT-I cells. Enhanced expansion of specific CD8(+) T cells was likewise observed on infection of YOD1-C160S mice with a recombinant influenza A virus expressing SIINFEKL. YOD1-C160S APCs retained antigen longer than did control APCs. Enhanced crosspresentation by YOD1-C160S APCs was transporter associated with antigen processing (TAP1)-independent but sensitive to inclusion of inhibitors of acidification and of the proteasome. The activity of deubiquitylating enzymes may thus help control antigenspecific CD8(+) T-cell responses during immunization.


Cross-Priming , Mutation, Missense , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Adoptive Transfer , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Brefeldin A/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/drug effects , Cross-Priming/genetics , Female , Hydrogen-Ion Concentration , Immunization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Peptide Fragments/immunology , Rhadinovirus/immunology , Rhadinovirus/pathogenicity , Ubiquitin Thiolesterase/metabolism
10.
Biochim Biophys Acta ; 1824(1): 105-12, 2012 Jan.
Article En | MEDLINE | ID: mdl-21664991

Cathepsin E is an intracellular aspartic proteinase of the pepsin superfamily, which is predominantly expressed in certain cell types, including the immune system cells and rapidly regenerating gastric mucosal and epidermal keratinocytes. The intracellular localization of this protein varies with different cell types. The endosomal localization is primarily found in antigen-presenting cells and gastric cells. The membrane association is observed with certain cell types such as erythrocytes, osteoclasts, gastric parietal cells and renal proximal tubule cells. This enzyme is also found in the endoplasmic reticulum, Golgi complex and cytosolic compartments in various cell types. In addition to its intracellular localization, cathepsin E occurs in the culture medium of activated phagocytes and cancer cells as the catalytically active enzyme. Its strategic expression and localization thus suggests the association of this enzyme with specific biological functions of the individual cell types. Recent genetic and pharmacological studies have particularly suggested that cathepsin E plays an important role in host defense against cancer cells and invading microorganisms. This review focuses emerging roles of cathepsin E in immune system cells and skin keratinocytes, and in host defense against cancer cells. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.


Cathepsin E/physiology , Immune System/enzymology , Immune System/physiology , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/physiology , Cathepsin E/genetics , Cathepsin E/metabolism , Humans , Immune System/metabolism , Models, Biological , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/metabolism , Skin/enzymology , Skin/immunology , Skin/metabolism , Skin Physiological Phenomena/genetics , Skin Physiological Phenomena/immunology
11.
J Immunol ; 187(4): 1986-97, 2011 Aug 15.
Article En | MEDLINE | ID: mdl-21746963

Cytosolic phospholipase A(2)α (cPLA(2)α) is the rate-limiting enzyme for release of arachidonic acid, which is converted primarily to PGs via the cyclooxygenase 1 and 2 pathways and to leukotrienes via the 5-lipoxygenase pathway. We used adoptive transfer and relapsing-remitting forms of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, in two different strains of mice (SJL or C57BL/6) to demonstrate that blockade of cPLA(2)α with a highly specific small-molecule inhibitor during the tissue-damage effector phase abrogates the clinical manifestation of disease. Using the adoptive transfer model in SJL mice, we demonstrated that the blockade of cPLA(2)α during the effector phase of disease was more efficacious in ameliorating the disease pathogenesis than the blockade of each of the downstream enzymes, cyclooxygenase-1/2 and 5-lipooxygenase. Similarly, blockade of cPLA(2)α was highly efficacious in ameliorating disease pathogenesis during the effector phase of EAE in the adoptive transfer model of EAE in C57BL/6 mice. Investigation of the mechanism of action indicates that cPLA(2)α inhibitors act on APCs to diminish their ability to induce Ag-specific effector T cell proliferation and proinflammatory cytokine production. Furthermore, cPLA(2)α inhibitors may prevent activation of CNS-resident microglia and may increase oligodendrocyte survival. Finally, in a relapsing-remitting model of EAE in SJL mice, therapeutic administration of a cPLA(2)α inhibitor, starting from the peak of disease or during remission, completely protected the mice from subsequent relapses.


Antigen-Presenting Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Enzyme Inhibitors/pharmacology , Group IV Phospholipases A2/antagonists & inhibitors , Multiple Sclerosis/prevention & control , Adoptive Transfer , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/pathology , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/immunology , Arachidonate 5-Lipoxygenase/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Cyclooxygenase 1/genetics , Cyclooxygenase 1/immunology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2 , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Group IV Phospholipases A2/genetics , Group IV Phospholipases A2/immunology , Group IV Phospholipases A2/metabolism , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Microglia/enzymology , Microglia/immunology , Microglia/pathology , Multiple Sclerosis/enzymology , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Oligodendroglia/enzymology , Oligodendroglia/immunology , Oligodendroglia/pathology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
12.
Life Sci ; 89(5-6): 176-81, 2011 Aug 01.
Article En | MEDLINE | ID: mdl-21683717

AIMS: To investigate whether sulfatides modulate indoleamine 2,3-dioxygenase (IDO)1, a fine-tuned enzymatic mechanism for controlling immune responses, gene expression/function in antigen presenting cells (APC). The relationship between structure and activity (SAR) of newly synthesized sulfatide isoforms (C16:0, C18:0, C22:0, C24:1) was also evaluated. MAIN METHODS: CD1d-transfected THP-1 human cells were used as APC and treated with increasing concentrations (0.01-10µΜ) of each compound for an appropriate period of time. The gene expression and the enzymatic activity of IDO1 were examined using reverse transcription-polymerase chain reaction (RT-PCR) and high performance liquid chromatography (HPLC). Compound-untreated cells were taken as negative, while 1000U/ml interferon (IFN)-γ-treated cells as positive controls. KEY FINDINGS: Not all sulfatides induced the same effect: the basal IDO1 expression was significantly reduced (-48 ± 3% at 0.01µΜ) by C16:0 sulfatide, while it was increased by C18:0 or C24:1 sulfatide (+87 ± 7% and +50 ± 5% at 1µΜ, respectively) over negative controls; C22:0 sulfatide resulted ineffective at all concentrations tested. These effects functionally correlated with changes in IDO1 activity: l-kynurenine contents in the culture media were significantly reduced by C16:0 sulfatide (-29 ± 4% at 0.01µM), while it was increased by C18:0 or C24:1 sulfatide (+61 ± 8% and +48 ± 4% at 1µM, respectively) over negative controls. C22:0 sulfatide resulted ineffective at all concentration tested. SIGNIFICANCE: The overall data demonstrate that specific sulfatide isoforms differently modulate IDO1 in APC. The sulfatide-induced effects are structurally dependent on the length/saturation of their fatty acid chain.


Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Sulfoglycosphingolipids/pharmacology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/enzymology , DNA/metabolism , Enzyme-Linked Immunosorbent Assay , Fatty Acids/chemistry , Gene Expression Regulation, Enzymologic/drug effects , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Isomerism , NF-kappa B/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship , Sulfoglycosphingolipids/chemical synthesis , Sulfoglycosphingolipids/chemistry , Tryptophan/metabolism
13.
Nat Immunol ; 12(7): 647-54, 2011 May 22.
Article En | MEDLINE | ID: mdl-21602810

Polarization of the T cell microtubule-organizing center (MTOC) toward the antigen-presenting cell (APC) is driven by the accumulation of diacylglycerol (DAG) at the immunological synapse (IS). The mechanisms that couple DAG to the MTOC are not known. By single-cell photoactivation of the T cell antigen receptor (TCR), we found that three distinct isoforms of protein kinase C (PKC) were recruited by DAG to the IS in two steps. PKC-ɛ and PKC-η accumulated first in a broad region of membrane, whereas PKC-θ arrived later in a smaller zone. Functional experiments indicated that PKC-θ was required for MTOC reorientation and that PKC-ɛ and PKC-η operated redundantly to promote the recruitment of PKC-θ and subsequent polarization responses. Our results establish a previously uncharacterized role for PKC proteins in T cell polarity.


Cell Polarity/immunology , Cytoskeleton/enzymology , Isoenzymes/immunology , Protein Kinase C-epsilon/immunology , Protein Kinase C/immunology , T-Lymphocytes/enzymology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Cytoskeleton/immunology , Diglycerides/immunology , Immunological Synapses/enzymology , Immunological Synapses/immunology , Mice , Mice, Transgenic , Protein Kinase C-theta , Receptors, Antigen, T-Cell/immunology , Single-Cell Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology
14.
Cancer Biol Ther ; 11(10): 866-73, 2011 May 15.
Article En | MEDLINE | ID: mdl-21372637

Indolamine2, 3-dioxygenase (IDO) is expressed in tumor antigen presenting cells (APCs) and plays an important role in tumor immune tolerance. Inhibiting its activity may break tumor immune tolerance and thus promote therapeutic effects. Thus, a specific inhibitor of IDO, 1-methyl-tryptophan (1-MT), is being used more and more frequently in anti-tumor studies. However, IDO also maintains systemic immune balance by suppressing abnormal immune responses. Therefore, targeting IDO in tumor-associated APCs in a way that does not disrupt immune balance warrants further investigation. In this study, we developed a new tumor vaccine, FAPτ-MT, which was produced by conjugating 1-MT to a tumor associated antigen, fibroblast activation protein α (FAPα). The results in vitro confirmed that 1-MT could be dissociated from the FAPτ-MT vaccine and inhibit intracellular IDO activity. In an FAPα-positive tumor model, the FAPτ-MT vaccine elicited an anti-tumor response which was similar to systemic treatment with the FAPτ vaccine plus 1-MT. Most importantly, administration of the FAPτ-MT vaccine did not lead to pregnancy failiure in mice carrying allogeneic fetuses. These findings that FAPτ-MT breaks tumor immune tolerance as a local IDO inhibitor, suggest that conjugation of 1-MT to a tumor antigen peptide is a potentially effective clinical cancer immunotherapy.


Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Cancer Vaccines/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Neoplasms/enzymology , Neoplasms/immunology , Animals , Cell Line, Tumor , Endopeptidases , Female , Fertility/immunology , Gelatinases/immunology , Immune Tolerance , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Macrophages/immunology , Male , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pregnancy , Serine Endopeptidases/immunology , Xenograft Model Antitumor Assays
15.
Nat Immunol ; 12(5): 416-24, 2011 May.
Article En | MEDLINE | ID: mdl-21441935

The molecular mechanisms involved in the full activation of innate immunity achieved through Toll-like receptors (TLRs) remain to be fully elucidated. In addition to their classical antigen-presenting function, major histocompatibility complex (MHC) class II molecules might mediate reverse signaling. Here we report that deficiency in MHC class II attenuated the TLR-triggered production of proinflammatory cytokines and type I interferon in macrophages and dendritic cells, which protected mice from endotoxin shock. Intracellular MHC class II molecules interacted with the tyrosine kinase Btk via the costimulatory molecule CD40 and maintained Btk activation, but cell surface MHC class II molecules did not. Then, Btk interacted with the adaptor molecules MyD88 and TRIF and thereby promoted TLR signaling. Therefore, intracellular MHC class II molecules can act as adaptors, promoting full activation of TLR-triggered innate immune responses.


Histocompatibility Antigens Class II/immunology , Immunity, Innate/immunology , Protein-Tyrosine Kinases/metabolism , Toll-Like Receptors/immunology , Adaptor Proteins, Vesicular Transport/immunology , Agammaglobulinaemia Tyrosine Kinase , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , CD40 Antigens/immunology , Cell Line , Cytokines/blood , Cytokines/immunology , Enzyme Activation , Immunoblotting , Interferon-gamma/blood , Interferon-gamma/immunology , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Protein-Tyrosine Kinases/immunology , Sepsis/immunology , Specific Pathogen-Free Organisms
16.
Tissue Antigens ; 77(2): 136-42, 2011 Feb.
Article En | MEDLINE | ID: mdl-21092022

Immunomodulatory properties of IDO1 relate to tryptophan catabolism. The degradation of tryptophan by IDO1 leads to suppression of T cell responses. Recently, another enzyme with IDO-like activity, indoleamine 2,3-dioxygenase-like-protein 1 (INDOL1, IDO2), has been described in both mice and humans. In order to study the gene expression of IDO1 and IDO2, we have developed a quantitative PCR (qPCR) assay. In an exploratory application to the study of the differential expression of IDO1 and IDO2 by professional antigen-presenting cells and MSCs (mesenchymal stromal cells) under the influence of interferon-γ (IFN-γ) and T-lymphocyte conditioned media (TCM), substantial differences were observed. IDO expression measured by qPCR was valid and reliable in the cell types investigated. Further studies are needed to delineate factors driving IDO expression in MSCs.


Antigen-Presenting Cells/enzymology , Dendritic Cells/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Mesoderm/enzymology , Stromal Cells/enzymology , Animals , Antigen-Presenting Cells/drug effects , Antiviral Agents/pharmacology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Dendritic Cells/drug effects , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/pharmacology , Mesoderm/cytology , Mesoderm/drug effects , Mice , Polymerase Chain Reaction , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stromal Cells/drug effects , T-Lymphocytes/metabolism
17.
Transplant Proc ; 42(9): 3489-96, 2010 Nov.
Article En | MEDLINE | ID: mdl-21094802

BACKGROUND: Indoleamine 2,3-dioxygenase (IDO) is a tryptophan-degrading enzyme that suppresses T-lymphocyte activity. Costimulation blockade through CTLA4lg increases IDO in antigen-presenting cells. The suppressive effect of IDO is thought to be mediated by Foxp3+CD4+CD25+ regulatory T-cells (Tregs). OBJECTIVE: In this descriptive study, we evaluated the percentage of IDO-expressing peripheral cell subpopulations as well as Tregs in 27 stable kidney transplant recipients receiving either belatacept (LEA29Y), a daughter compound of abatacept (CTLA4lg; n = 19) or cyclosporine (n = 8). METHODS: Blood samples were obtained at 24 ± 2 months (belatacept) and 23 ± 6 months (cyclosporine) of treatment. Intracellular IDO was analyzed by flow cytometry in CD14+, CD11c+, CD16+, CD56+, and CD8+ cell subpopulations. Tregs were assessed by intracellular Foxp3 detection in CD4+CD25+ cells. CD3+, CD4+, CD8+, CD20+, CD68+, IDO+, and Foxp3+ cells were evaluated by immunohistochemistry on graft biopsies obtained preimplantation, at 12 months posttransplant, and in subjects with dysfunction during the first 12 months. RESULTS: Only percentages of CD16+/IDO+-expressing peripheral monocytes were significantly increased among the group receiving belatacept. No differences were observed in peripheral Tregs between the groups. In contrast, higher percentages of Tregs, CD4+, CD8+, and CD68+ cells were noted in dysfunction and at 12 months vs baseline among graft biopsies in subjects receiving belatacept, and also among dysfunction cohorts of belatacept vs Cyclosporine treatment. CONCLUSION: Patients receiving belatacept showed greater amounts of peripheral blood CD16+/IDO+ cells and Tregs on graft biopsies than those under cyclosporine treatment.


Antigen-Presenting Cells/drug effects , Cyclosporine/administration & dosage , Immunoconjugates/administration & dosage , Immunosuppressive Agents/administration & dosage , Indoleamine-Pyrrole 2,3,-Dioxygenase/blood , Kidney Transplantation , Kidney/drug effects , Receptors, IgG/blood , T-Lymphocytes, Regulatory/drug effects , Abatacept , Adult , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Biopsy , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , GPI-Linked Proteins/blood , Humans , Immunohistochemistry , Kidney/immunology , Kidney/pathology , Kidney Transplantation/adverse effects , Male , Middle Aged , T-Lymphocytes, Regulatory/immunology , Time Factors , Treatment Outcome
18.
Mol Immunol ; 47(4): 658-65, 2010 Jan.
Article En | MEDLINE | ID: mdl-19910052

Contributions from multiple cathepsins within endosomal antigen processing compartments are necessary to process antigenic proteins into antigenic peptides. Cysteine and aspartyl cathepsins have been known to digest antigenic proteins. A role for the serine protease, cathepsin G (CatG), in this process has been described only recently, although CatG has long been known to be a granule-associated proteolytic enzyme of neutrophils. In line with a role for this enzyme in antigen presentation, CatG is found in endocytic compartments of a variety of antigen presenting cells. CatG is found in primary human monocytes, B cells, myeloid dendritic cells 1 (mDC1), mDC2, plasmacytoid DC (pDC), and murine microglia, but is not expressed in B cell lines or monocyte-derived DC. Purified CatG can be internalized into endocytic compartments in CatG non-expressing cells, widening the range of cells where this enzyme may play a role in antigen processing. Functional assays have implicated CatG as a critical enzyme in processing of several antigens and autoantigens. In this review, historical and recent data on CatG expression, distribution, function and involvement in disease will be summarized and discussed, with a focus on its role in antigen presentation and immune-related events.


Antigen Presentation/immunology , Cathepsin G/immunology , Amino Acid Sequence , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Cathepsin G/chemistry , Cathepsin G/genetics , Humans , Models, Immunological , Molecular Sequence Data , Neutrophils/enzymology , Neutrophils/immunology , Substrate Specificity/immunology
19.
Nat Rev Immunol ; 9(12): 871-82, 2009 Dec.
Article En | MEDLINE | ID: mdl-19935806

The cellular endolysosomal compartment is dynamic, complex and incompletely understood. Its organelles and constituents vary between different cell types, but endolysosomal proteases are key components of this compartment in all cells. In immune cells, these proteases function in pathogen recognition and elimination, signal processing and cell homeostasis, and they are regulated by dedicated inhibitors. Pathogens can produce analogous proteases to subvert the host immune response. The balance in activity between a protease and its inhibitor can tune the immune response or cause damage as a result of mislocalized proteolysis. In this Review, we highlight recent developments in this area and emphasize the importance of studying the role of endolysosomal proteases, and their natural inhibitors, in the initiation and regulation of immune responses.


Lysosomes/enzymology , Peptide Hydrolases/immunology , Protease Inhibitors/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Host-Pathogen Interactions/immunology , Humans , Lymphocytes/enzymology , Lymphocytes/immunology , Lysosomes/immunology
20.
J Immunol ; 182(12): 7548-57, 2009 Jun 15.
Article En | MEDLINE | ID: mdl-19494278

Many cancers are known to produce high amounts of PGE(2), which is involved in both tumor progression and tumor-induced immune dysfunction. The key enzyme responsible for the biological inactivation of PGE(2) in tissue is NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase (15-PGDH). It is well established that cancer cells frequently show down-regulated expression of 15-PGDH, which plays a major role in catabolism of the PGE(2). Here we demonstrate that tumor-infiltrated CD11b cells are also deficient for the 15-PGDH gene. Targeted adenovirus-mediated delivery of 15-PGDH gene resulted in substantial inhibition of tumor growth in mice with implanted CT-26 colon carcinomas. PGDH-mediated antitumor effect was associated with attenuated tumor-induced immune suppression and substantially reduced secretion of immunosuppressive mediators and cytokines such as PGE(2), IL-10, IL-13, and IL-6 by intratumoral CD11b cells. We show also that introduction of 15-PGDH gene in tumor tissue is sufficient to redirect the differentiation of intratumoral CD11b cells from immunosuppressive M2-oriented F4/80(+) tumor-associated macrophages (TAM) into M1-oriented CD11c(+) MHC class II-positive myeloid APCs. Notably, the administration of the 15-PGDH gene alone demonstrated a significant therapeutic effect promoting tumor eradication and long-term survival in 70% of mice with preestablished tumors. Surviving mice acquired antitumor T cell-mediated immune response. This study for the first time demonstrates an important role of the 15-PGDH in regulation of local antitumor immune response and highlights the potential to be implemented to enhance the efficacy of cancer therapy and immunotherapy.


CD11b Antigen/immunology , Hydroxyprostaglandin Dehydrogenases/metabolism , Myeloid Cells/enzymology , Myeloid Cells/immunology , Neoplasms/enzymology , Neoplasms/immunology , Tumor Escape/immunology , Adenoviridae/genetics , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Lymph Nodes/enzymology , Lymph Nodes/immunology , Male , Mice , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/pathology , Survival Rate
...