Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Immunol ; 48(9): 1588-1591, 2018 09.
Article in English | MEDLINE | ID: mdl-30007005

ABSTRACT

Dimethyl fumarate (DMF) promotes an IL-17Alow IFN-γlow IL-4+ CD4+ T cell phenotype. Adoptive transfer of in vitro DMF-treated myelin peptide-reactive IL-17Alow IFN-γlow IL-4+ CD4+ T cells prior to immunization for EAE reduces the severity of encephalomyelitis. This beneficial effect of transferred DMF-treated CD4+ T cells requires an early in vivo recall.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dimethyl Fumarate/pharmacology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immunosuppressive Agents/pharmacology , Interferon-gamma/immunology , Interleukin-4/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/transplantation , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-4/biosynthesis , Mice , Th1 Cells/immunology , Th2 Cells/immunology
2.
J Dermatol Sci ; 87(3): 268-277, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28655472

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is a common inflammatory skin disease with dysfunction of the skin barrier, an abnormal immune response and frequent allergies to environmental antigens like food antigens. Clinical observations suggest that certain diets can influence the course of AD. OBJECTIVE: Here we compared the phenotype of food allergen-specific T cells activated through skin or gut allergen exposure to transfer skin inflammation into naïve recipients upon epicutaenous allergen challenge. METHODS: Ovalbumin (OVA) TCR-transgenic mice were treated epicutaneously with OVA or were fed OVA. CD4+ T cells from skin lymph nodes or mesenteric lymph nodes were transferred into naïve BALB/c mice, which were challenged with OVA epicutaneously. Skin inflammation was determined by histological parameters. In addition, we analyzed the phenotype of the immune response in lymphoid tissues and in skin tissue. RESULTS: TCR-transgenic T cells activated through epicutaneous or oral OVA exposure both migrate to skin lymph nodes after adoptive transfer and epicutaenous OVA exposure. AD-like skin inflammation could only be induced by the transfer of epicutaneously primed OVA T cells. Analysis of the immune phenotype demonstrated an IL-22/IL-17A-dominated immune phenotype of skin-pathogenic T cells. CONCLUSION: IL-22 seems to be the critical cytokine for the development of AD and is induced in this model by epicutaneous sensitization with OVA.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dermatitis, Atopic/immunology , Food Hypersensitivity/immunology , Interleukins/immunology , Skin/immunology , Allergens/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Humans , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/metabolism , Intestines/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Vaccination/methods , Interleukin-22
3.
Sci Rep ; 7: 44482, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28290522

ABSTRACT

The nutritional curcumin (CUR) is beneficial in cell-mediated autoimmune diseases. The molecular mechanisms underlying this food-mediated silencing of inflammatory immune responses are poorly understood. By investigating antigen-specific immune responses we found that dietary CUR impairs the differentiation of Th1/Th17 cells in vivo during encephalomyelitis and instead promoted Th2 cells. In contrast, feeding CUR had no inhibitory effect on ovalbumin-induced airway inflammation. Mechanistically, we found that CUR induces an anti-inflammatory phenotype in dendritic cells (DC) with enhanced STAT3 phosphorylation and suppressed expression of Il12b and Il23a. On the molecular level CUR readily induced NRF2-sensitive heme oxygenase 1 (HO-1) mRNA and protein in LPS-activated DC. HO-1 enhanced STAT3 phosphorylation, which enriched to Il12b and Il23a loci and negatively regulated their transcription. These findings demonstrate the underlying mechanism through which a nutritional can interfere with the immune response. CUR silences IL-23/Th17-mediated pathology by enhancing HO-1/STAT3 interaction in DC.


Subject(s)
Autoimmune Diseases/drug therapy , Curcumin/administration & dosage , Heme Oxygenase-1/genetics , Inflammation/drug therapy , Interleukin-23/genetics , Membrane Proteins/genetics , STAT3 Transcription Factor/genetics , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Dendritic Cells/drug effects , Encephalomyelitis, Autoimmune, Experimental , Immunity, Cellular/drug effects , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Mice , Ovalbumin/toxicity , Phosphorylation , Th17 Cells/drug effects , Th2 Cells/drug effects , Th2 Cells/immunology
4.
J Immunol ; 195(5): 2216-23, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26232431

ABSTRACT

Small interfering RNA (siRNA)-based therapies allow targeted correction of molecular defects in distinct cell populations. Although efficient in multiple cell populations, dendritic cells (DCs) seem to resist siRNA delivery. Using fluorescence labeling and radiolabeling, we show that cholesterol modification enables siRNA uptake by DCs in vitro and in vivo. Delivery of cholesterol-modified p40 siRNA selectively abolished p40 transcription and suppressed TLR-triggered p40 production by DCs. During immunization with peptide in CFA, cholesterol-modified p40 siRNA generated p40-deficient, IL-10-producing DCs that prevented IL-17/Th17 and IFN-γ/Th1 responses. Only cholesterol-modified p40-siRNA established protective immunity against experimental autoimmune encephalomyelitis and suppressed IFN-γ and IL-17 expression by CNS-infiltrating mononuclear cells without inducing regulatory T cells. Because cholesterol-modified siRNA can thus modify selected DC functions in vivo, it is intriguing for targeted immune therapy of allergic, autoimmune, or neoplastic diseases.


Subject(s)
Cholesterol/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-12 Subunit p40/immunology , RNA, Small Interfering/immunology , Animals , Cholesterol/chemistry , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Flow Cytometry , Gene Expression/immunology , Immunization/methods , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-12 Subunit p40/blood , Interleukin-12 Subunit p40/genetics , Mice, Inbred Strains , Molecular Structure , RNA, Small Interfering/genetics , RNAi Therapeutics/methods , Reverse Transcriptase Polymerase Chain Reaction , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Treatment Outcome
5.
J Immunol ; 192(8): 3530-9, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24639357

ABSTRACT

Sulforaphane (SFN), an isothiocyanate, is part of an important group of naturally occurring small molecules with anti-inflammatory properties. The published reports are best conceivable with an inhibition of T cell function, but the mode of action remains unknown. We therefore analyzed the effect of SFN on T cell-mediated autoimmune disease. Feeding mice with SFN protected from severe experimental autoimmune encephalomyelitis. Disease amelioration was associated with reduced IL-17 and IFN-γ expression in draining lymph nodes. In vitro, SFN treatment of T cells did not directly alter T cell cytokine secretion. In contrast, SFN treatment of dendritic cells (DCs) inhibited TLR4-induced IL-12 and IL-23 production, and severely suppressed Th1 and Th17 development of T cells primed by SFN-treated DCs. SFN regulated the activity of the TLR4-induced transcription factor NF-κB, without affecting the degradation of its inhibitor IκB-α. Instead, SFN treatment of DCs resulted in strong expression of the stress response protein heme oxygenase-1 (HO-1), which interacts with and thereby inhibits NF-κB p65. Consistent with these findings, HO-1 bound to p65 and subsequently inhibited the p65 activity at the IL23a and IL12b promoters. Importantly, SFN suppressed Il23a and Il12b expression in vivo and silenced Th17/Th1 responses within the CNS. Thus, our data show that SFN improves Th17/Th1-mediated autoimmune disease by inducing HO-1 and inhibiting NF-κB p65-regulated IL-23 and IL-12 expression.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Interleukin-12/metabolism , Interleukin-23/metabolism , Isothiocyanates/pharmacology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Autoimmune Diseases/prevention & control , Cell Differentiation/drug effects , Cluster Analysis , Cytokines/biosynthesis , DNA/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Heme Oxygenase-1/metabolism , Interleukin-12/genetics , Interleukin-23/genetics , Isothiocyanates/administration & dosage , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , NF-kappa B/metabolism , Phenotype , Protein Binding/drug effects , Sulfoxides , T-Lymphocyte Subsets/drug effects , Th1 Cells/cytology , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism
6.
J Exp Med ; 208(11): 2291-303, 2011 Oct 24.
Article in English | MEDLINE | ID: mdl-21987655

ABSTRACT

Fumarates improve multiple sclerosis (MS) and psoriasis, two diseases in which both IL-12 and IL-23 promote pathogenic T helper (Th) cell differentiation. However, both diseases show opposing responses to most established therapies. First, we show in humans that fumarate treatment induces IL-4-producing Th2 cells in vivo and generates type II dendritic cells (DCs) that produce IL-10 instead of IL-12 and IL-23. In mice, fumarates also generate type II DCs that induce IL-4-producing Th2 cells in vitro and in vivo and protect mice from experimental autoimmune encephalomyelitis. Type II DCs result from fumarate-induced glutathione (GSH) depletion, followed by increased hemoxygenase-1 (HO-1) expression and impaired STAT1 phosphorylation. Induced HO-1 is cleaved, whereupon the N-terminal fragment of HO-1 translocates into the nucleus and interacts with AP-1 and NF-κB sites of the IL-23p19 promoter. This interaction prevents IL-23p19 transcription without affecting IL-12p35, whereas STAT1 inactivation prevents IL-12p35 transcription without affecting IL-23p19. As a consequence, GSH depletion by small molecules such as fumarates induces type II DCs in mice and in humans that ameliorate inflammatory autoimmune diseases. This therapeutic approach improves Th1- and Th17-mediated autoimmune diseases such as psoriasis and MS by interfering with IL-12 and IL-23 production.


Subject(s)
Dendritic Cells/immunology , Fumarates/immunology , Fumarates/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Psoriasis/drug therapy , Psoriasis/immunology , Animals , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Heme Oxygenase-1/metabolism , Humans , Interleukin-12/immunology , Interleukin-23/immunology , Macrophages/immunology , Mice , NIH 3T3 Cells , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcription Factor RelA/metabolism
7.
Mol Plant Pathol ; 6(3): 299-314, 2005 May 01.
Article in English | MEDLINE | ID: mdl-20565658

ABSTRACT

SUMMARY Tobacco pathogenesis-related (PR) genes of group 1 are induced during pathogen defence (hypersensitive response, HR, and systemic acquired resistance, SAR), after exogenous application of salicylic acid (SA), and by developmental cues. Likewise, SA enhances transcripts for Arabidopsis NIMIN-1 and NIMIN-2, which interact with NPR1/NIM1, a key regulator of SAR. To further illuminate gene activation during pathogen defence, reporter gene expression from the NIMIN-1 and NIMIN-2 promoters was analysed in transgenic tobacco plants in direct comparison to PR-1 gene expression. NIMIN[GUS] chimeric genes were highly sensitive to SA, whereas NIMIN[GUS], unlike PR1a[GUS], expression was only weak in necrotic tissue exhibiting HR. Furthermore, PR-1a, but not NIMIN, promoter constructs were activated systemically in response to local cell death elicited by expression of the proapoptotic Bax gene. Conversely, NIMIN-1[GUS] expression was completely suppressed during pathogen defence in plants depleted from SA, whereas PR-1 proteins still accumulated in necrotic tissue. These findings demonstrate that SA-dependent gene activation can be uncoupled from cell death-induced gene activation. Whereas PR-1a induction during the HR and SAR responses is mediated by HR-associated signals and SA, activation of the NIMIN-1 and NIMIN-2 promoters in infected tobacco relies on SA, but not on cell death signals.

SELECTION OF CITATIONS
SEARCH DETAIL
...