Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Immunol ; 192(5): 2202-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24489100

ABSTRACT

Although multiple dendritic cell (DC) subsets have the potential to induce Th17 differentiation in vitro, the key DC that is critical in Th17 induction and Th17-mediated disease remains moot. In this study, we revealed that CCR2(+) monocyte-derived DCs (moDCs), but not conventional DCs, were critical for in vivo Th17 induction and autoimmune inflammation. Functional comparison in vitro indicated that moDCs are the most potent type of Th17-inducing DCs compared with conventional DCs and plasmacytoid DCs. Furthermore, we demonstrated that the importance of GM-CSF in Th17 induction and Th17-mediated disease is its endowment of moDCs to induce Th17 differentiation in vivo, although it has little effect on moDC numbers. Our findings identify the in vivo cellular targets that can be selectively manipulated to ameliorate Th17-mediated inflammatory diseases, as well as the mechanism of GM-CSF antagonism in such diseases.


Subject(s)
Autoimmune Diseases/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Monocytes/immunology , Th17 Cells/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Cell Differentiation/genetics , Dendritic Cells/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Knockout , Monocytes/cytology , Th17 Cells/cytology
2.
J Immunol ; 192(2): 572-80, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24337380

ABSTRACT

CD8(+) T cells are critical in human type 1 diabetes and in the NOD mouse. In this study, we elucidated the natural history of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-specific CD8(+) T cells in NOD diabetes using MHC-tetramer technology. IGRP206-214-specific T cells in the peripheral lymphoid tissue increased with age, and their numbers correlated with insulitis progression. IGRP206-214-specific T cells in the peripheral lymphoid tissue expressed markers of chronic Ag stimulation, and their numbers were stable after diagnosis of diabetes, consistent with their memory phenotype. IGRP206-214-specific T cells in NOD mice expand, acquire the phenotype of effector-memory T cells in the islets, and emigrate to the peripheral lymphoid tissue. Our observations suggest that enumeration of effector-memory T cells of multiple autoantigen specificities in the periphery of type 1 diabetic subjects could be a reliable reporter for progression of islet pathology.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Immunologic Memory/immunology , Islets of Langerhans/immunology , Animals , Autoantigens/immunology , Diabetes Mellitus, Type 1/pathology , Glucose-6-Phosphatase/immunology , Islets of Langerhans/pathology , Lymphocytes/immunology , Mice , Mice, Inbred NOD
3.
Immunol Cell Biol ; 92(7): 640-4, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24777314

ABSTRACT

CD8(+) T cells are prominent in autoimmune diabetes of both humans and non-obese diabetic (NOD) mice. For example, CD8(+) T cells against islet-specific glucose 6-phosphatase catalytic subunit-related protein (IGRP) can be detected readily in older NOD mice. It has been suggested that the enumeration of islet-specific CD8(+) T cells in the peripheral blood may be a predictive biomarker for autoimmune type 1 diabetes (T1D). Here, we determined the natural history of the functional endogenous IGRP(206-214)-specific cytotoxic T lymphocytes (CTLs) in NOD mice with regard to age (3- to 15-week-old pre-diabetic mice and diabetic mice) and sex. We demonstrated that in vivo IGRP(206-214)-specific CTLs significantly increased after 12 weeks of age and in vivo cytotoxicity in female NOD mice was significantly higher than in male NOD mice. To determine the in vivo IGRP(206-214)-specific CTL frequency without killing the mice, we performed splenectomies on a cohort of mice after injecting IGRP(206-214)-coated targets and then followed their diabetes progression. We found that CTL frequency correlated with future of disease onset. Thus, our data support that IGRP(206-214)-specific CTLs may be a potent biomarker for T1D.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Epitopes, T-Lymphocyte/immunology , Glucose-6-Phosphatase/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Age Factors , Animals , Cytotoxicity, Immunologic , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/chemistry , Female , Glucose-6-Phosphatase/chemistry , Male , Mice , Mice, Inbred NOD , Peptides/chemistry , Prognosis
4.
J Immunol ; 187(4): 1566-77, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21742968

ABSTRACT

Defects in the Bcl-2-regulated apoptotic pathway inhibit the deletion of self-reactive T cells. What is unresolved, however, is the nature and fate of such self-reactive T cells escaping deletion. In this study, we report that mice with such defects contained increased numbers of CD25(low)Foxp3(+) cells in the thymus and peripheral lymph tissues. The increased CD25(low)Foxp3(+) population contained a large fraction of cells bearing self-reactive TCRs, evident from a prominent increase in self-superantigen-specific Foxp3(+)Vß5(+)CD4(+) T cells in BALB/c Bim(-/-) mice compared with control animals. The survival rate of the expanded CD25(low)Foxp3(+) cells was similar to that of CD25(high)Foxp3(+) CD4 T cells in vitro and in vivo. IL-2R stimulation, but not TCR ligation, upregulated CD25 on CD25(low)Foxp3(+)CD4(+) T cells in vitro and in vivo. The expanded CD25(low)Foxp3(+)CD4(+) T cells from Bim(-/-) mice were anergic but also had weaker regulatory function than CD25(high)Foxp3(+) CD4(+) T cells from the same mice. Analysis of Bim(-/-) mice that also lacked Fas showed that the peripheral homeostasis of this expanded population was in part regulated by this death receptor. In conclusion, these results show that self-reactive T cell escapes from thymic deletion in mice defective in the Bcl-2-regulated apoptotic pathway upregulate Foxp3 and become unresponsive upon encountering self-Ag without necessarily gaining potent regulatory function. This clonal functional diversion may help to curtail autoaggressiveness of escaped self-reactive CD4(+) T cells and thereby safeguard immunological tolerance.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Clonal Anergy , Proto-Oncogene Proteins c-bcl-2/immunology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Bcl-2-Like Protein 11 , Cell Survival/genetics , Cell Survival/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/immunology
5.
Sci Rep ; 12(1): 7443, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35523930

ABSTRACT

Dietary antigens affect the adaptive immunity of the host by inducing regulatory T cells and IgE-producing B cells. However, their roles in innate immune compartments such as innate lymphoid cells (ILCs) and intestinal epithelial cells (IECs) are unclear. Here, using antigen-free (AF) mice, which are germ-free (GF) mice fed with amino-acid-based diet, we found dietary proteins suppress the development of GATA-3-expressing ILC2s independent of the adaptive immune cells. These cells produce more type 2 cytokines and upregulated proliferation and activation markers such as Ki-67, CD69, and CD25. With this, AF mice had increased expressions of tuft cell-specific transcripts such as Il25, Il33, Dclk1, Trpm5, and Pou2f3 in IECs. Accordingly, expanded ILC2s upregulated IL-17RB, a receptor of IL-25, and their proliferation was blocked by IL-25 neutralizing or IL-17RB blocking antibodies. These results suggest a new dialogue between dietary antigens, IECs, and ILCs in which dietary antigens suppress ILC2 activation and proliferation by restraining homeostatic IL-25 production, potentially limiting type 2 immunity by food antigens.


Subject(s)
Immunity, Innate , Lymphocytes , Animals , Cell Proliferation , Cytokines , Diet , Mice
6.
Eur J Immunol ; 40(12): 3499-509, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21108470

ABSTRACT

The autoimmune regulator (AIRE) promotes "promiscuous" expression of tissue-restricted antigens (TRA) in thymic medullary epithelial cells to facilitate thymic deletion of autoreactive T-cells. Here, we show that AIRE-deficient mice showed an earlier development of myelin oligonucleotide glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). To determine the outcome of ectopic Aire expression, we used a retroviral transduction system to over-express Aire in vitro, in cell lines and in bone marrow (BM). In the cell lines that included those of thymic medullary and dendritic cell origin, ectopically expressed Aire variably promoted expression of TRA including Mog and Ins2 (proII) autoantigens associated, respectively, with the autoimmune diseases multiple sclerosis and type 1 diabetes. BM chimeras generated from BM transduced with a retrovirus encoding Aire displayed elevated levels of Mog and Ins2 expression in thymus and spleen. Following induction of EAE with MOG(35-55), transplanted mice displayed significant delay in the onset of EAE compared with control mice. To our knowledge, this is the first example showing that in vivo ectopic expression of AIRE can modulate TRA expression and alter autoimmune disease development.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Encephalomyelitis, Autoimmune, Experimental/immunology , Epithelial Cells/metabolism , Transcription Factors/metabolism , Animals , Antigen Presentation/genetics , Autoantigens/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Line , Cloning, Molecular , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/therapy , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Thymus Gland/pathology , Transcription Factors/genetics , Transcription Factors/immunology , Transgenes/genetics , AIRE Protein
7.
Mol Pharm ; 8(5): 1488-94, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21732672

ABSTRACT

A key function of the immune system is to protect us from foreign pathogens such as viruses, bacteria, fungi and multicellular parasites. However, it is also important in many other aspects of human health such as cancer surveillance, tissue transplantation, allergy and autoimmune disease. Autoimmunity can be defined as a chronic immune response that targets self-antigens leading to tissue pathology and clinical disease. Autoimmune diseases, as a group of diseases that include type 1 diabetes, multiple sclerosis, rheumatoid arthritis and systemic lupus erythematosus, have no effective cures, and treatment is often based on long-term broad-spectrum immunosuppressive regimes. While a number of strategies aimed at providing disease specific treatments are being explored, one avenue of study involves the use of hematopoietic stem cells to promote tolerance. In this manuscript, we will review the literature in this area but in particular examine the relatively new experimental field of gene therapy and hematopoietic stem cell transplantation as a molecular therapeutic strategy to combat autoimmune disease.


Subject(s)
Autoimmune Diseases/therapy , Gene Transfer Techniques , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Immunosuppression Therapy/methods , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Cells, Cultured/transplantation , Combined Modality Therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Encephalomyelitis, Autoimmune, Experimental/therapy , Hematopoietic Stem Cells/immunology , Humans , Mice
8.
Front Immunol ; 11: 1897, 2020.
Article in English | MEDLINE | ID: mdl-32849649

ABSTRACT

Retinal dehydrogenase (RALDH) enzymatic activities catalyze the conversion of vitamin A to its metabolite Retinoic acid (RA) in intestinal dendritic cells (DCs) and promote immunological tolerance. However, precise understanding of the exogenous factors that act as initial trigger of RALDH activity in these cells is still evolving. By using germ-free (GF) mice raised on an antigen free (AF) elemental diet, we find that certain components in diet are critically required to establish optimal RALDH expression and activity, most prominently in small intestinal CD103+CD11b+ DCs (siLP-DCs) right from the beginning of their lives. Surprisingly, systematic screens using modified diets devoid of individual dietary components indicate that proteins, starch and minerals are dispensable for this activity. On the other hand, in depth comparison between subtle differences in dietary composition among different dietary regimes reveal that adequate glucose concentration in diet is a critical determinant for establishing RALDH activity specifically in siLP-DCs. Consequently, pre-treatment of siLP-DCs, and not mesenteric lymph node derived MLNDCs with glucose, results in significant enhancement in the in vitro generation of induced Regulatory T (iTreg) cells. Our findings reveal previously underappreciated role of dietary glucose concentration in establishing regulatory properties in intestinal DCs, thereby extending a potential therapeutic module against intestinal inflammation.


Subject(s)
Antigens, CD/metabolism , CD11b Antigen/metabolism , Dendritic Cells/drug effects , Dietary Sugars/administration & dosage , Glucose/administration & dosage , Integrin alpha Chains/metabolism , Intestine, Small/drug effects , Retinal Dehydrogenase/metabolism , Animal Feed , Animals , Antigens, CD/immunology , CD11b Antigen/immunology , Cells, Cultured , Coculture Techniques , Dendritic Cells/enzymology , Dendritic Cells/immunology , Integrin alpha Chains/immunology , Intestine, Small/enzymology , Intestine, Small/immunology , Mice, Inbred C57BL , Retinal Dehydrogenase/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
9.
Cancer Immunol Res ; 7(3): 498-509, 2019 03.
Article in English | MEDLINE | ID: mdl-30728152

ABSTRACT

GM-CSF as an adjuvant has been shown to promote antitumor immunity in mice and humans; however, the underlying mechanism of GM-CSF-induced antitumor immunity remains incompletely understood. In this study, we demonstrate that GM-CSF potentiates the efficacy of cancer vaccines through IL9-producing Th (Th9) cells. GM-CSF selectively enhanced Th9 cell differentiation by regulating the COX2-PGE2 pathway while inhibiting the differentiation of induced regulatory T (iTreg) cells in vitro and in vivo GM-CSF-activated monocyte-derived dendritic cells converted tumor-specific naïve Th cells into Th9 cells, and delayed tumor growth by inducing antitumor CTLs in an IL9-dependent manner. Our findings reveal a mechanism for the adjuvanticity of GM-CSF and provide a rationale for the use of GM-CSF in cancer vaccines.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-9/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Helper-Inducer/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/immunology , Cancer Vaccines/immunology , Cell Differentiation/drug effects , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Dendritic Cells/immunology , Dinoprostone/metabolism , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunotherapy , Interleukin-9/metabolism , Lymphocyte Activation/drug effects , Mice , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
10.
Sci Adv ; 5(5): eaaw1507, 2019 05.
Article in English | MEDLINE | ID: mdl-31131325

ABSTRACT

Immunoglobulin E (IgE), a key mediator in allergic diseases, is spontaneously elevated in mice with disrupted commensal microbiota such as germ-free (GF) and antibiotics-treated mice. However, the underlying mechanisms for aberrant IgE elevation are still unclear. Here, we demonstrate that food antigens drive spontaneous IgE elevation in GF and antibiotics-treated mice by generating T helper 2 (TH2)-skewed T follicular helper (TFH) cells in gut-associated lymphoid tissues (GALTs). In these mice, depriving contact with food antigens results in defective IgE elevation as well as impaired generation of TFH cells and IgE-producing cells in GALT. Food antigen-driven TFH cells in GF mice are mostly generated in early life, especially during the weaning period. We also reveal that food antigen-driven TFH cells in GF mice are actively depleted by colonization with commensal microbiota. Thus, our findings provide a possible explanation for why the perturbation of commensal microbiota in early life increases the occurrence of allergic diseases.


Subject(s)
Antigens/immunology , Food Hypersensitivity/immunology , Gastrointestinal Microbiome/immunology , Immunoglobulin E/immunology , Allergens/immunology , Animals , Bone Marrow Cells/metabolism , CD4-Positive T-Lymphocytes/cytology , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Germ-Free Life , Immune System , Immunoglobulin E/blood , Lymph Nodes/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Symbiosis
11.
Arthritis Rheumatol ; 67(3): 797-808, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25418983

ABSTRACT

OBJECTIVE: Interferon-α (IFNα)-producing plasmacytoid dendritic cells (PDCs) are implicated in the pathogenesis of systemic lupus erythematosus (SLE). IFNα-related genes are highlighted among SLE susceptibility alleles and are characteristically expressed in the blood of patients with SLE, while in mouse models of lupus, PDC numbers and IFNα production are increased. This study was undertaken to investigate the effects of inhibitors that selectively target different antiapoptotic molecules on the survival of PDCs. METHODS: PDC numbers, in vitro survival, and expression of antiapoptotic molecules were evaluated in lupus-prone (NZB × NZW)F1 (NZB/NZW) mice. The impact of Bcl-2 antagonists and glucocorticoids on PDCs was evaluated in vitro and in vivo. IFNα production by NZB/NZW mice was evaluated before and after treatment with Bcl-2 antagonists. RESULTS: PDCs, but not lymphoid tissue-resident conventional DCs, largely relied on the antiapoptotic protein Bcl-2 for survival. The enlarged PDC compartment in NZB/NZW mice was associated with selectively prolonged survival and increased Bcl-2 transcription. Functionally, this resulted in enhanced production of IFNα. Bcl-2 inhibitors selectively killed mouse and human PDCs, including PDCs from SLE patients, but not conventional DCs, dampened IFNα production by PDCs, and synergized with glucocorticoids to kill activated PDCs. CONCLUSION: Enhanced PDC survival is a likely contributing factor to enhanced IFNα production by lupus PDCs. Bcl-2 antagonists potently and selectively kill PDCs and reduce IFNα production. Thus, we believe that they are attractive candidates for treating PDC-associated diseases.


Subject(s)
Biphenyl Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Dendritic Cells/drug effects , Interferon-alpha/metabolism , Lupus Erythematosus, Systemic/immunology , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Annexin A5/metabolism , Antibodies, Antinuclear/blood , Cell Survival , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/pathology , Flow Cytometry , Glucocorticoids/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Transgenic , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Real-Time Polymerase Chain Reaction
12.
J Exp Med ; 212(6): 927-38, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26008898

ABSTRACT

Gain-of-function mutations that activate the innate immune system can cause systemic autoinflammatory diseases associated with increased IL-1ß production. This cytokine is activated identically to IL-18 by an intracellular protein complex known as the inflammasome; however, IL-18 has not yet been specifically implicated in the pathogenesis of hereditary autoinflammatory disorders. We have now identified an autoinflammatory disease in mice driven by IL-18, but not IL-1ß, resulting from an inactivating mutation of the actin-depolymerizing cofactor Wdr1. This perturbation of actin polymerization leads to systemic autoinflammation that is reduced when IL-18 is deleted but not when IL-1 signaling is removed. Remarkably, inflammasome activation in mature macrophages is unaltered, but IL-18 production from monocytes is greatly exaggerated, and depletion of monocytes in vivo prevents the disease. Small-molecule inhibition of actin polymerization can remove potential danger signals from the system and prevents monocyte IL-18 production. Finally, we show that the inflammasome sensor of actin dynamics in this system requires caspase-1, apoptosis-associated speck-like protein containing a caspase recruitment domain, and the innate immune receptor pyrin. Previously, perturbation of actin polymerization by pathogens was shown to activate the pyrin inflammasome, so our data now extend this guard hypothesis to host-regulated actin-dependent processes and autoinflammatory disease.


Subject(s)
Actins/physiology , Cytoskeletal Proteins/metabolism , Hereditary Autoinflammatory Diseases/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Microfilament Proteins/metabolism , Actins/chemistry , Animals , Bone Marrow Cells/cytology , Caspase 1/metabolism , Caspases/metabolism , Clodronic Acid/chemistry , Crosses, Genetic , Culture Media, Conditioned/chemistry , Enzyme-Linked Immunosorbent Assay , Interleukin-18/metabolism , Lipopolysaccharides/metabolism , Liposomes/chemistry , Liver/embryology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Monocytes/cytology , Pyrin , Signal Transduction
13.
Nat Commun ; 6: 6442, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25778803

ABSTRACT

Intracellular nucleotide binding and oligomerization domain (NOD) receptors recognize antigens including bacterial peptidoglycans and initiate immune responses by triggering the production of pro-inflammatory cytokines through activating NF-κB and MAP kinases. Receptor interacting protein kinase 2 (RIPK2) is critical for NOD-mediated NF-κB activation and cytokine production. Here we develop and characterize a selective RIPK2 kinase inhibitor, WEHI-345, which delays RIPK2 ubiquitylation and NF-κB activation downstream of NOD engagement. Despite only delaying NF-κB activation on NOD stimulation, WEHI-345 prevents cytokine production in vitro and in vivo and ameliorates experimental autoimmune encephalomyelitis in mice. Our study highlights the importance of the kinase activity of RIPK2 for proper immune responses and demonstrates the therapeutic potential of inhibiting RIPK2 in NOD-driven inflammatory diseases.


Subject(s)
Cytokines/metabolism , Inflammation/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/chemistry , Animals , Chromatography, Liquid , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Humans , Immune System , Inhibitory Concentration 50 , Interferon-gamma/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , NF-kappa B/metabolism , Protein Binding , Protein Conformation , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Recombinant Proteins/metabolism , Signal Transduction , Tandem Mass Spectrometry , Ubiquitin/metabolism
14.
PLoS One ; 9(3): e91126, 2014.
Article in English | MEDLINE | ID: mdl-24637385

ABSTRACT

Migratory CD103+ and lymphoid-resident CD8+ dendritic cells (DCs) share many attributes, such as dependence on the same transcription factors, cross-presenting ability and expression of certain surface molecules, such that it has been proposed they belong to a common sub-lineage. The functional diversity of the two DC types is nevertheless incompletely understood. Here we reveal that upon skin infection with herpes simplex virus, migratory CD103+ DCs from draining lymph nodes were more potent at inducing Th17 cytokine production by CD4+ T cells than CD8+ DCs. This superior capacity to drive Th17 responses was also evident in CD103+ DCs from uninfected mice. Their differential potency to induce Th17 differentiation was reflected by higher production of IL-1ß and IL-6 by CD103+ DCs compared with CD8+ DCs upon stimulation. The two types of DCs from isolated lymph nodes also differ in expression of certain pattern recognition receptors. Furthermore, elevated levels of GM-CSF, typical of those found in inflammation, substantially increased the pool size of CD103+ DCs in lymph nodes and skin. We argue that varied levels of GM-CSF may explain the contrasting reports regarding the positive role of GM-CSF in regulating development of CD103+ DCs. Together, we find that these two developmentally closely-related DC subsets display functional differences and that GM-CSF has differential effect on the two types of DCs.


Subject(s)
Antigens, CD/metabolism , CD8 Antigens/metabolism , Dendritic Cells/metabolism , Inflammation/metabolism , Integrin alpha Chains/metabolism , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/virology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunophenotyping , Inflammation/immunology , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Pattern Recognition/metabolism , Simplexvirus/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Toll-Like Receptors/metabolism
15.
Autoimmunity ; 44(3): 177-87, 2011 May.
Article in English | MEDLINE | ID: mdl-20883147

ABSTRACT

Haematopoietic stem cell (HSC) transfer coupled with gene therapy is a powerful approach to treating fatal diseases such as X-linked severe combined immunodeficiency. This ability to isolate and genetically manipulate HSCs also offers a strategy for inducing immune tolerance through ectopic expression of autoantigens. We have previously shown that retroviral transduction of bone marrow (BM) with vectors encoding the autoantigen, myelin oligodendrocyte glycoprotein (MOG), can prevent the induction of experimental autoimmune encephalomyelitis (EAE). However, ubiquitous cellular expression of autoantigen driven by retroviral promoters may not be the best approach for clinical translation and a targeted expression approach may be more acceptable. As BM-derived dendritic cells (DCs) play a major role in tolerance induction, we asked whether targeted expression of MOG, a target autoantigen in EAE, to DCs can promote tolerance induction and influence the development of EAE. Self-inactivating retroviral vectors incorporating the mouse CD11c promoter were generated and used to transduce mouse BM cells. Transplantation of gene-modified cells into irradiated recipients resulted in the generation of chimeric mice with transgene expression limited to DCs. Notably, chimeric mice transplanted with MOG-expressing BM cells manifest a significant delay in the development of EAE suggesting that targeted antigen expression to tolerogenic cell types may be a feasible approach to inducing antigen-specific tolerance.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/immunology , CD11c Antigen/genetics , Cell Line , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gene Order , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/metabolism , Mice , Mice, Inbred C57BL , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Retroviridae/genetics , Retroviridae/metabolism
16.
Discov Med ; 9(49): 512-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20587340

ABSTRACT

Autoimmune diseases such as multiple sclerosis, type 1 diabetes, systemic sclerosis, and rheumatoid arthritis affect approximately 5% of the population and are characterized by a destructive immune response directed to self-tissues. Treatments are often designed to dampen the immune system and are therefore associated with unwanted side effects. A major challenge is to find a cure that does not compromise normal immune function. From our understanding of how the immune system develops, it is clear that mechanisms designed to eliminate or maintain control over self-reactive clones are critical for normal health. These key concepts form the crux of many experimental strategies currently aimed at abrogating the autoimmune response. In this review, we focus on the strategy of harnessing the bone marrow compartment through genetic manipulation directed at promoting ectopic autoantigen expression. Our experience with this strategy is presented in the context of reports in the literature and we argue for the potential benefit of translating this approach to the treatment of human autoimmune disease.


Subject(s)
Autoimmune Diseases/therapy , Bone Marrow/immunology , Animals , Autoimmune Diseases/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL