Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Amino Acids ; 52(3): 411-424, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32008091

ABSTRACT

We have previously shown that particle-mediated epidermal delivery (PMED) of plasmids encoding ß-galactosidase (ßGal) under control of the fascin-1 promoter (pFascin-ßGal) yielded selective production of the protein in skin dendritic cells (DCs), and suppressed Th2 responses in a mouse model of type I allergy by inducing Th1/Tc1 cells. However, intranasal challenge of mice immunized with pFascin-ßGal induced airway hyperreactivity (AHR) and neutrophilic inflammation in the lung. The tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO) has been implicated in immune suppression and tolerance induction. Here we investigated the consequences of co-application of an IDO-encoding vector on the modulatory effect of DNA vaccination by PMED using pFascin-ßGal in models of eosinophilic allergic and non-eosinophilic intrinsic airway inflammation. IDO-encoding plasmids and pFascin-ßGal or pCMV-ßGal were co-applied to abdominal skin of BALB/c mice without, before or after sensitization with ßGal protein. Immune responses in the lung were analysed after intranasal provocation and airway reactivity was determined by whole body plethysmography. Co-application of pCMV-IDO with pFascin-ßGal, but not pCMV-ßGal inhibited the Th1/Tc1 immune response after PMED. Moreover, AHR in those mice was attenuated following intranasal challenge. Therapeutic vaccination of ßGal-sensitized mice with pFascin-ßGal plus pCMV-IDO slightly suppressed airway inflammation and AHR after provocation with ßGal protein, while prophylactic vaccination was not effective. Altogether, our data suggest that only the combination of DC-restricted antigen and ubiquitous IDO expression attenuated asthma responses in mice, most probably by forming a tryptophan-depleted and kynurenine-enriched micromilieu known to affect neutrophils and T cells.


Subject(s)
Asthma/therapy , Biolistics/methods , Bronchial Hyperreactivity/therapy , Dendritic Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Vaccination/methods , Animals , Antigens/genetics , Antigens/immunology , Asthma/immunology , Bronchial Hyperreactivity/immunology , Carrier Proteins/genetics , Genetic Vectors , Mice , Mice, Inbred BALB C , Microfilament Proteins/genetics , NIH 3T3 Cells , Neutrophil Infiltration , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Transgenes , beta-Galactosidase/genetics , beta-Galactosidase/immunology
2.
Front Pharmacol ; 9: 915, 2018.
Article in English | MEDLINE | ID: mdl-30174602

ABSTRACT

Dendritic cells (DCs) are pivotal for the induction and maintenance of antigen-specific tolerance and immunity. miRNAs mediate post-transcriptional gene regulation and control in part the differentiation and stimulation-induced immunogenic function of DCs. However, the relevance of miRNAs for the induction and maintenance of a tolerogenic state of DCs has scarcely been highlighted yet. We differentiated mouse bone marrow cells to conventional/myeloid DCs or to tolerogenic antigen presenting cells (APCs) by using a glucocorticoid (dexamethasone) or interleukin-10, and assessed the miRNA expression patterns of unstimulated and LPS-stimulated cell populations by array analysis and QPCR. Differentially tolerized mouse APCs convergingly down-regulated a set of miRNA species at either state of activation as compared with the corresponding control DC population (mmu-miR-9-5p, mmu-miR-9-3p, mmu-miR-155-5p). These miRNAs were also upregulated in control DCs in response to stimulation. In contrast, miRNAs that were convergingly upregulated in both tolerized APC groups at stimulated state (mmu-miR-223-3p, mmu-miR-1224-5p) were downregulated in control DCs in response to stimulation. Overexpression of mmu-miR-223-3p in DCs was sufficient to prevent stimulation-associated acquisition of potent T cell stimulatory capacity. Overexpression of mmu-miR-223-3p in a DC line resulted in attenuated expression of known (Cflar, Rasa1, Ras) mRNA targets of this miRNA species shown to affect pathways that control DC activation. Taken together, we identified sets of miRNAs convergingly regulated in differentially tolerized APCs, which may contribute to imprint stimulation-resistant tolerogenic function as demonstrated for mmu-miR-223-3p. Knowledge of miRNAs with protolerogenic function enables immunotherapeutic approaches aimed to modulate immune responses by regulating miRNA expression.

3.
PLoS One ; 13(2): e0191927, 2018.
Article in English | MEDLINE | ID: mdl-29408931

ABSTRACT

In this study we analysed the effects of prophylactic biolistic DNA vaccination with plasmids encoding the encephalitogenic protein myelin oligodendrocyte glycoprotein (MOG) on the severity of a subsequently MOGp35-55-induced EAE and on the underlying immune response. We compared the outcome of vaccination with MOG-encoding plasmids alone or in combination with vectors encoding the regulatory cytokines IL-10 and TGF-ß1, respectively. MOG expression was restricted to skin dendritic cells (DCs) by the use of the DC-specific promoter of the fascin1 gene (pFscn-MOG). For comparison, the strong and ubiquitously active CMV promoter was employed (pCMV-MOG), which allows MOG expression in all transfected cells. Expression of IL-10 and TGF-ß1 was controlled by the CMV promoter to yield maximal synthesis (pCMV-IL10, pCMV-TGFß). Co-application of pFscn-MOG and pCMV-IL10 significantly ameliorated EAE pathology, while vaccination with pCMV-MOG plus pCMV-IL10 did not affect EAE outcome. In contrast, vaccination with either of the two MOG-encoding plasmids in combination with pCMV-TGFß significantly attenuated the clinical EAE symptoms. Mechanistically, we observed diminished infiltration of Th17 and Th1 cells as well as macrophages/DCs into the CNS, which correlated with decreased MOGp35-55-specific production of IL-17 and IFN-Ï« by spleen cells and reduced peptide-specific T cell proliferation. Our findings suggest deletion of or anergy induction in MOG-specific CD4+ T cells by the suppressive vaccination platform employed. MOG expression driven by the DC-specific fascin1 promoter yielded similar inhibitory effects on EAE progression as the ubiquitously active viral CMV promoter, when coapplying pCMV-TGFß. Our finding that pCMV-IL10 promoted tolerogenic effects only, when coapplied with pFscn-MOG, but not pCMV-MOG suggests that IL-10 affected only directly transfected DCs (pFscn-MOG), but not neighbouring DCs that engulfed MOG-containing vesicles derived from transfected keratinocytes (pCMV-MOG). Thus, due to its DC-restricted expression, the fascin1 promoter might be an interesting alternative to ubiquitously expressed promoters for vaccination strategies.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immune Tolerance , Vaccines, DNA/administration & dosage , Animals , Flow Cytometry , Mice , T-Lymphocytes, Regulatory/immunology
4.
Hum Immunol ; 77(12): 1223-1231, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27593566

ABSTRACT

Immature dendritic cells (iDCs) and tolerogenic DCs are essential for the induction and maintenance of peripheral tolerance. Tumors produce immuno-modulatory factors which imprint a pro-tolerogenic, maturation-resistant state in DCs. Here we asked for common markers of differentially tolerized human monocyte-derived DC populations. For this, PBMC-derived monocytes were differentiated to DCs in the presence of established immuno-modulators as released by tumors (IL-6, IL-10, TGF-ß, glucocorticoid [GC], prostaglandin E2 [PGE2]). Most unstimulated pro-tolerogenic DC populations commonly over-expressed some tolerance-associated markers (ILT-4, IL-10, HO-1) as compared with iDCs. These markers may contribute to imprint a pro-tolerogenic state in DCs. Furthermore, some tolerance markers were overexpressed in an immuno-modulator specific manner in DCs differentiated in the presence of TGF-ß (overexpressed tolerance markers: B7-H3, CD103, TGF-ß1), IL-10 (B7-DC, ILT-3) and PGE2 (IDO). Upon stimulation, matured control DCs (mDCs) down-regulated most pro-tolerogenic markers monitored, while some were upregulated (IkBα, IDO, B7-H1, B7-DC). In contrast, the different groups of tolerized DCs largely retained expression of pro-tolerogenic markers after stimulation. In contrast to mDCs, most groups of tolerized DCs showed impaired upregulation of CD80, and all groups retained IL-10 cytokine production after stimulation. All tolerized DC populations commonly exerted an attenuated allogenic T cell stimulatory capacity as compared with mDCs.


Subject(s)
Dendritic Cells/physiology , Immunologic Factors/metabolism , Neoplasms/immunology , T-Lymphocytes/immunology , Antigen Presentation , B7-1 Antigen/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Dinoprostone/metabolism , Glucocorticoids/metabolism , Heme Oxygenase-1/metabolism , Humans , Immune Tolerance , Lymphocyte Activation
5.
Int Immunopharmacol ; 35: 174-184, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27070502

ABSTRACT

Phosphodiesterase 4 (PDE4) inhibitors serve to prevent degradation of the intracellular second messenger cAMP, resulting in broad anti-inflammatory effects on different cell types including immune cells. Agents that elevate cAMP levels via activation of adenylate cyclase have been shown to imprint a Th17-promoting capacity in dendritic cells (DCs). Therefore, we studied the potential of therapeutically relevant PDE inhibitors to induce a pronounced Th17-skewing capacity in DCs. Here we show that mouse bone marrow-derived (BM-) DCs when treated with the PDE4 inhibitor roflumilast (ROF, trade name: Daxas) in the course of stimulation with LPS (ROF-DCs) evoked elevated IL-17 levels in cocultured allogeneic T cells. In addition, as compared with control settings, levels of IFN-γ remained unaltered, while contents of Th2 cytokines (IL-5, IL-10) were diminished. ROF enhanced expression of the Th17-promoting factor IL-23 in BM-DCs. In line, neutralizing antibodies specific for IL-23 or IL-6 when applied to DC/T cell cocultures partially inhibited the IL17-promoting effect of ROF-DCs. Furthermore, ROF-DCs displayed a markedly diminished allogeneic T cell stimulatory capacity due to enhanced production of IL-10, which was restored upon application of IL-10 specific neutralizing antibody to DC/T cell cocultures. Both the IL-17-inducing and impaired T cell stimulatory capacity of BM-DCs were mimicked by a specific activator of protein kinase A, while stimulation of EPACs (exchange proteins of activated cAMP) did not yield such effects. Taken together, our findings suggest that PDE4 inhibitors aside from their broad overall anti-inflammatory effects may enhance the Th17-polarizing capacity in DCs as an unwanted side effect.


Subject(s)
Aminopyridines/pharmacology , Anti-Inflammatory Agents/pharmacology , Benzamides/pharmacology , Dendritic Cells/drug effects , Hypersensitivity/drug therapy , Interleukin-23/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Th17 Cells/immunology , Animals , Cells, Cultured , Cyclopropanes/pharmacology , Dendritic Cells/immunology , Interleukin-10/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Th2 Cells/immunology
6.
Int Immunopharmacol ; 32: 118-124, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26824774

ABSTRACT

Many tumors are characterized by mutation-induced constitutive activation of ß-catenin which promotes tumor growth and survival. Consequently, the development of specific ß-catenin inhibitors for tumor therapy has come into the focus of drug development. ß-Catenin was also shown to contribute to the tolerance-promoting function of unstimulated dendritic cells (DCs). In response to activation, DCs acquire potent T cell stimulatory capacity and induce profound tumor antigen-specific immune responses. Here we asked for effects of pre-clinically established ß-catenin inhibitors (CCT-031374, iCRT-5, PNU-75654) on mouse bone marrow-derived (BM)DCs. All three inhibitors moderately increased surface expression of MHCII, CD80, and CD86 on unstimulated DCs, but had no enhancing effect on their capacity to stimulate the proliferation of ovalbumin (OVA) specific CD4(+) T cells. CCT-031374 interfered with upregulation of costimulators (CD40, CD86) and cytokines (IL-1ß, TNF-α, IL-6, IL-10, IL-12) by LPS-stimulated DCs. Accordingly, this DC population displayed an impaired CD4(+) T cell stimulatory activity. iCRT-5 and PNU-75654 had no detrimental effects on the immuno-phenotype of stimulated DCs. Hence, DCs treated with iCRT-5 in the course of stimulation exerted comparably strong T cell proliferation as did control DCs. In contrast, DCs stimulated in the presence of PNU-75654 induced less T cell proliferation than the control population despite enhanced uptake and processing of OVA. Our findings suggest that the differential effects of ß-catenin inhibitors on stimulated DCs reflect off target effects. Concerning potential application of ß-catenin inhibitors for tumor therapy, iCRT-5 may be most beneficial, since it did not exert detrimental effects on stimulated DCs.


Subject(s)
Antineoplastic Agents/pharmacology , Dendritic Cells/drug effects , beta Catenin/antagonists & inhibitors , Animals , Bone Marrow Cells/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/immunology , Dendritic Cells/immunology , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Phenotype
7.
Virology ; 485: 481-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26363219

ABSTRACT

Adult T-cell leukemia/lymphoma is a highly infiltrative neoplasia of CD4(+) T-lymphocytes that occurs in about 5% of carriers infected with the deltaretrovirus human T-cell leukemia virus type 1 (HTLV-1). The viral oncoprotein Tax perturbs cellular signaling pathways leading to upregulation of host cell factors, amongst them the actin-bundling protein Fascin, an invasion marker of several types of cancer. However, transcriptional regulation of Fascin by Tax is poorly understood. In this study, we identified a triple mode of transcriptional induction of Fascin by Tax, which requires (1) NF-κB-dependent promoter activation, (2) a Tax-responsive region in the Fascin promoter, and (3) a promoter-independent mechanism sensitive to the Src family kinase inhibitor PP2. Thus, Tax regulates Fascin by a multitude of signals. Beyond, using Tax-expressing and virus-transformed lymphocytes as a model system, our study is the first to identify the invasion marker Fascin as a novel target of PP2, an inhibitor of metastasis.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/metabolism , Microfilament Proteins/genetics , Promoter Regions, Genetic , Carrier Proteins/metabolism , Cell Transformation, Viral , Gene Expression Regulation/drug effects , Human T-lymphotropic virus 1/genetics , Humans , Microfilament Proteins/metabolism , Models, Biological , NF-kappa B/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/virology , Transcriptional Activation , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
8.
Immunobiology ; 220(4): 490-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25468560

ABSTRACT

The mechanisms of tolerance induction occurring in the course of allergen-specific immunotherapy have not been elucidated in full detail. Our study aimed to characterize high zone tolerance in mouse models of type I allergy and of allergic airway inflammation induced by subcutaneous sensitization of mice with high doses of the model allergen ovalbumin (OVA) without the use of adjuvant. Mice were immunized by subcutaneous injection of high doses (HD) of OVA or, for comparison, low doses (LD) of OVA in saline. HD-mice showed lower specific IgE, but augmented IgG in sera than LD-mice. Pre-treatment of mice with HD-OVA antigen-specifically inhibited IgE production subsequently induced by LD-OVA. OVA-restimulated splenocytes from HD-mice revealed hypoproliferation and impaired production of Th2-associated cytokines. HD-mice exhibited lower airway reactivity, goblet cell hyperplasia and mucus production, as well as IL-5 and IL-13 production in the lungs than LD-mice following local provocation. Recruitment of inflammatory cells into the airways was comparable, while the number of eosinophils in the bronchoalveolar lavage was substantially higher in HD-mice. Adoptive transfer of dnTC from HD-mice into naïve mice, which were subsequently sensitized with LD-OVA, suppressed IgE production in the recipients. The number of dnTC was higher in the spleens of HD-mice than LD-mice. In conclusion, our study demonstrates that subcutaneous sensitization of mice with high doses of allergen in the absence of adjuvant results in attenuated airway reactivity as compared with LD-sensitization and induces CD4(-)CD8(-) dnTC with regulatory function on IgE production.


Subject(s)
Antibody Formation/immunology , Immunoglobulin E/biosynthesis , Immunoglobulin E/immunology , Immunomodulation , Respiratory Hypersensitivity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antibody Specificity/immunology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/metabolism , Female , Gene Expression , Immunization , Immunophenotyping , Lymphocyte Activation , Mice , Ovalbumin/administration & dosage , Ovalbumin/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , Respiratory Hypersensitivity/physiopathology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
9.
J Exp Clin Cancer Res ; 33: 16, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24524692

ABSTRACT

BACKGROUND: The chaperon heat shock protein 90 (HSP90) constitutes an important target for anti-tumor therapy due to its essential role in the stabilization of oncogenes. However, HSP90 is ubiquitously active to orchestrate protein turnover, chemotherapeutics that target HSP90 may affect immune cells as a significant side effect. Therefore, we asked for potential effects of pharmacological HSP90 inhibition at a therapeutically relevant concentration on human dendritic cells (DCs) as main inducers of both cellular and humoral immune responses, and on human CD4⁺ T cells as directly activated by DCs and essential to confer B cell help. METHODS: Unstimulated human monocyte-derived DCs (MO-DCs) were treated with the prototypical HSP90 inhibitor geldanamycin (GA). Based on dose titration studies performed to assess cytotoxic effects, GA was applied at a rather low concentration, comparable to serum levels of clinically used HSP90 inhibitors. The immuno-phenotype (surface markers, cytokines), migratory capacity, allo T cell stimulatory and polarizing properties (proliferation, cytokine pattern) of GA-treated MO-DCs were assessed. Moreover, effects of GA on resting and differentially stimulated CD4⁺ T cells in terms of cytotoxicity and proliferation were analysed. RESULTS: GA induced partial activation of unstimulated MO-DCs. In contrast, when coapplied in the course of MO-DC stimulation, GA prevented the acquisition of a fully mature DC phenotype. Consequently, this MO-DC population exerted lower allo CD4⁺ T cell stimulation and cytokine production. Furthermore, GA exerted no cytotoxic effect on resting T cells, but abrogated proliferation of T cells stimulated by MO-DCs at either state of activation or by stimulatory antibodies. CONCLUSION: HSP90 inhibitors at clinically relevant concentrations may modulate adaptive immune responses both on the level of DC activation and T cell proliferation. Surprisingly, unstimulated DCs may be partially activated by that agent. However, due to the potent detrimental effects of HSP90 inhibitors on stimulated CD4⁺ T cells, as an outcome a patients T cell responses might be impaired. Therefore, HSP90 inhibitors most probably are not suitable for treatment in combination with immunotherapeutic approaches aimed to induce DC/T cell activation.


Subject(s)
Benzoquinones/pharmacology , Dendritic Cells/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Transcription Factor RelB/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Dendritic Cells/immunology , HEK293 Cells , Humans , Leukocytes, Mononuclear/physiology , Lymphocyte Activation , Phenotype , Signal Transduction , Transcription Factor RelB/genetics , Up-Regulation/drug effects
10.
Basic Res Cardiol ; 108(6): 386, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24061433

ABSTRACT

CD40 ligand (CD40L) is involved in the vascular infiltration of immune cells and pathogenesis of atherosclerosis. Additionally, T cell CD40L release causes platelet, dendritic cell and monocyte activation in thrombosis. However, the role of CD40L in angiotensin II (ATII)-driven vascular dysfunction and hypertension remains incompletely understood. We tested the hypothesis that CD40L contributes to ATII-driven vascular inflammation by promoting platelet-leukocyte activation, vascular infiltration of immune cells and by amplifying oxidative stress. C57BL/6 and CD40L-/- mice were infused with ATII (1 mg/kg/day for 7 days) using osmotic minipumps. Vascular function was recorded by isometric tension studies, and reactive oxygen species (ROS) were monitored in blood and heart by optical methods. Western blot, immunohistochemistry, FACS analysis and real-time RT-PCR were used to analyze immune cell distribution, pro-inflammatory cytokines, NAPDH oxidase subunits, T cell transcription factors and other genes of interest. ATII-treated CD40L-/- mice showed improved endothelial function, suppression of blood platelet-monocyte interaction (FACS), platelet thrombin generation (calibrated automated thrombography) and coagulation (bleeding time), as well as decreased oxidative stress in the aorta, heart and blood compared to wild-type mice. Moreover, ATII-treated CD40L-/- mice displayed decreased levels of TH1 cytokines released by splenic CD4⁺ T cells (ELISA) and lower expression levels of NOX-2, T-bet and P-selectin as well as diminished immune cell infiltration in aortic tissue compared to controls. Our results demonstrate that many ATII-induced effects on vascular dysfunction, such as vascular inflammation, oxidative stress and a pro-thrombotic state, are mediated at least in part via CD40L.


Subject(s)
Angiotensin II/metabolism , CD40 Ligand/metabolism , Endothelial Cells/metabolism , Oxidative Stress/physiology , Angiotensin II/pharmacology , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Blotting, Western , Endothelial Cells/pathology , Flow Cytometry , Immunohistochemistry , Inflammation/metabolism , Leukocytes , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation/physiology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thrombosis/metabolism , Vascular Diseases/immunology , Vascular Diseases/metabolism
11.
Cancer Immunol Immunother ; 62(8): 1315-26, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23666509

ABSTRACT

The camptothecin analogue topotecan (TPT) induces tumor cell apoptosis due to interference with topoisomerase I and is clinically used as a second-line chemotherapeutic in the treatment for metastasizing ovarian and small cell lung carcinoma. Based on the more recent finding of TPT-mediated inhibition of the transcription factor hypoxia-induced factor-1α, a hallmark of solid tumors, TPT, is currently tested in clinical trials for its suitability as a first-line chemotherapeutic for the treatment for various types of tumors. Due to the gained clinical interest in TPT and in light of its modulatory effect on signaling pathways, which are also of importance for immune cell functions, we asked for potential effects of TPT on dendritic cells (DCs), the main antigen-presenting cell population of the immune system. Here, we show that TPT at a therapeutically relevant dose partially activated monocyte-derived DCs as reflected by enhanced migratory activity, elevated expression of HLA-DR and costimulatory/maturation markers, and accordingly an increased allogenic CD4(+) T cell stimulation. In marked contrast, TPT prevented full maturation of DCs stimulated with a cocktail of proinflammatory mediators, accompanied by somewhat lower upregulation of NF-κB factors p65 and RelB.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Topotecan/pharmacology , Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Flow Cytometry , HEK293 Cells , HLA-DR Antigens/metabolism , Humans , Immunoblotting , Immunoglobulins/metabolism , Immunophenotyping , Lymphocyte Activation/drug effects , Membrane Glycoproteins/metabolism , Microscopy, Confocal , Mitogen-Activated Protein Kinases/metabolism , Monocytes/drug effects , Monocytes/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Topoisomerase I Inhibitors/pharmacology , CD83 Antigen
12.
Methods Mol Biol ; 940: 199-213, 2013.
Article in English | MEDLINE | ID: mdl-23104345

ABSTRACT

The transcriptional targeting of gene expression to selected cells by cell type-specific promoters displays a fundamental tool in gene therapy. In immunotherapy, dendritic cells (DCs) are pivotal for the elicitation of antigen-specific immune responses following gene gun-mediated biolistic transfection. Here we report on transcriptional targeting of murine skin DCs using plasmids which include the promoter of the gene of the cytoskeletal protein fascin to control antigen production. Fascin, which is mandatory for the formation of dendrites, is synthesized among the hematopoietic cells exclusively by activated DCs. The activity of the promoter of the fascin gene reflects the endogenous production of the protein, being high in mature DCs but almost absent in immature DCs or other cutaneous cells. Here we describe the analysis of transgene-specific immune responses after DC-focused biolistic transfection. In conclusion, the murine fascin promoter can be readily used to target DCs in DNA immunization approaches and thus offers new opportunities for gene therapy.


Subject(s)
Biolistics/instrumentation , Carrier Proteins/genetics , Dendritic Cells/metabolism , Microfilament Proteins/genetics , Promoter Regions, Genetic/genetics , Transfection/instrumentation , Animals , Antibody Specificity , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , DNA/administration & dosage , DNA/chemistry , DNA/genetics , Female , Gold/chemistry , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interferon-gamma/metabolism , Interleukin-5/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred BALB C , Plasmids/genetics , Spleen/immunology , Transcription, Genetic , Vaccination , beta-Galactosidase/genetics , beta-Galactosidase/immunology
13.
Methods Mol Biol ; 940: 357-70, 2013.
Article in English | MEDLINE | ID: mdl-23104354

ABSTRACT

Gene gun-mediated delivery of allergen-encoding plasmid DNA has been in focus for many years now as being a needle-free alternative to the protein-based desensitization regimen used in specific immunotherapy. Biolistic immunization with the Helios gene gun has proven to be potent in the induction of antigen-specific CD4(+) and CD8(+) T cells. Here we describe biolistic vaccination in experimental mouse models of IgE-mediated type I allergy as well as allergen-induced airway inflammation.


Subject(s)
Biolistics/instrumentation , Hypersensitivity/prevention & control , Vaccination/instrumentation , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Allergens/immunology , Animals , Antibody Specificity , Bronchoalveolar Lavage , Female , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/physiopathology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Lung/immunology , Lung/physiopathology , Mice , Mice, Inbred BALB C , Respiratory Tract Diseases/genetics , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/physiopathology , Respiratory Tract Diseases/prevention & control , Transfection/instrumentation , Vaccines, DNA/genetics , beta-Galactosidase/immunology
14.
Gene ; 472(1-2): 18-27, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21040760

ABSTRACT

Langerhans cells (LCs) represent the dendritic cell (DC) population in the epidermis. Among the set of genes induced in primary mouse LCs in response to stimulation, both isoforms of the voltage-dependent Ca²(+) channel (VDCC) regulatory subunit Cacnb3 as well as the DC maturation marker Fscn1 were upregulated most strongly. Comparable results were obtained for a recently described myeloid DC line (SP37A3). Other antigen presenting cell populations, namely, bone marrow-derived DCs, macrophages and primary B cells, showed no stimulation-associated upregulation of Cacnb3 expression. Pharmacological inhibition of Ca²(+) channel activity during the stimulation of SP37A3 cells enhanced their T cell stimulatory capacity, while selective inhibition of L-type VDCC had no effect. Both Cacnb3 isoforms, similar to Fscn1, required JNK and p38 kinase activity for stimulation-associated upregulation, and this process was inhibited by ERK and PI(3)K. The putative promoter region of Cacnb3 isoform 2, which we found to be less ubiquitously expressed than Cacnb3 isoform 1, exerted reporter activity in LC-like cell lines. Our findings suggest that Cacnb3 exerts its function in distinct activated DC populations. Further analysis of the regulatory region(s) facilitating stimulation-induced upregulation of Cacnb3 expression in these DC subsets will help to gain better insight into DC subset specific gene regulation.


Subject(s)
Calcium Channels/genetics , Dendritic Cells/metabolism , Gene Expression Regulation , Up-Regulation , Animals , Base Sequence , Calcium Channels/metabolism , Cell Differentiation/genetics , Dendritic Cells/cytology , Langerhans Cells/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Isoforms/genetics , RNA, Messenger/metabolism , Transfection
15.
Eur J Immunol ; 40(10): 2848-57, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20836156

ABSTRACT

Cylindromatosis gene (CYLD) is a ubiquitously expressed deubiquitinating enzyme, which interacts with members of the NF-κB signaling pathway and attenuates NF-κB and JNK signaling. Here, we report that DC derived from transgenic mice, which solely express a naturally occurring CYLD isoform (CYLD(ex7/8)), display a higher content of nuclear RelB and express elevated levels of NF-κB family members as well as of known NF-κB-target genes comprising costimulatory molecules and pro-inflammatory cytokines, as compared with WT DC. Accordingly, unstimulated CYLD(ex7/8) DC exhibited a significantly higher primary allogenic T-cell stimulatory capacity than WT DC and exerted no tolerogenic activity. Transduction of unstimulated CYLD(ex7/8) DC with relB-specific shRNA reduced their T-cell stimulatory capacity. Treatment with the synthetic glucocorticoid dexamethasone known to inhibit NF-κB and AP-1 activity reverted the pro-immunogenic phenotype and function of CYLD(ex7/8) DC and re-established their tolerogenic function. DC derived from CYLD knockout mice showed no functional alterations compared with WT DC. Therefore, although complete loss of CYLD may be compensated for by other endogenous NF-κB inhibitors, CYLD(ex7/8) acts in a dominant negative manner. Our findings raise the question of whether genetic defects associated with increased NF-κB activity may result in disturbed maintenance of peripheral tolerance.


Subject(s)
Dendritic Cells/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Transcription Factor AP-1/immunology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , Animals , Blotting, Western , Cell Differentiation/immunology , Dendritic Cells/enzymology , Dexamethasone/pharmacology , Female , Flow Cytometry , Glucocorticoids/pharmacology , Immune Tolerance/immunology , Isoenzymes/immunology , Mice , Mice, Knockout , Mice, Transgenic , Mutation/immunology , NF-kappa B/antagonists & inhibitors , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Transcription Factor RelB/immunology
16.
J Gene Med ; 12(3): 231-43, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20140891

ABSTRACT

BACKGROUND: Dendritic cells (DCs) constitute an attractive target for immunotherapeutic approaches. Because DCs are largely refractory to transfection with plasmid DNA, several viral transduction protocols were established. The potential side-effects of lentiviral transduction on the phenotype and activation state of DCs left unstimulated after transduction have not been assessed. There is a need to analyse these parameters as a result of the requirement of using DCs with a low activation state for therapeutic strategies intended to induce tolerance. METHODS: Lentivirally-transduced bone marrow (BM)-derived DCs (LV-DCs) in comparison with mock-transduced (Mock-DCs) and untreated DCs were analysed with regard to the induction of maturation processes on the RNA, protein and functional level. BM-DCs engineered to overexpress interleukin (IL)-10 were analysed for therapeutic potential in a mouse model of allergic contact dermatitis. RESULTS: Compared with untreated DCs, Mock-DCs and LV-DCs displayed an altered gene expression signature. Mock-DCs induced a stronger T cell proliferative response than untreated DCs. LV-DCs did not further augment the T cell proliferative response, but induced a slightly different T cell cytokine pattern compared to Mock-DCs. Accordingly, the gene promoter of the DC maturation marker fascin mediated efficient expression of the model transgene IL-10 in unstimulated-transduced BM-DCs. Nevertheless, IL-10 overexpressing BM-DCs exerted tolerogenic activity and efficiently inhibited the contact hypersensitivity response in previously hapten-sensitized mice. CONCLUSIONS: Lentiviral transduction of BM-DCs results in their partial activation. Nevertheless, the transduction of these DCs with a vector encoding the immunomodulatory cytokine IL-10 rendered them tolerogenic. Thus, lentivirally-transduced DCs expressing immunomodulatory molecules represent a promising tool for induction of tolerance.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Dermatitis, Allergic Contact/therapy , Interleukin-10/genetics , T-Lymphocytes/immunology , Animals , Female , Genetic Engineering , Immunotherapy , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Stress, Physiological , Transduction, Genetic
17.
Mol Immunol ; 47(5): 1161-70, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19945750

ABSTRACT

Dendritic cells (DCs) serve to maintain peripheral tolerance under steady state conditions. Upon triggering by activation signals they initiate strong immune responses. The activation of DCs is accompanied by a rapid upregulation of proinflammatory cytokines, which were shown in other cell types to be regulated by mechanisms at the transcriptional and posttranscriptional level. Tristetraprolin (TTP), an important RNA binding protein, is involved in the regulation of mRNA stability of such cytokines. In this study we analyzed the significance of TTP for mouse DCs, which were derived from TTP(-/-) and WT bone marrow progenitor cells (BM-DCs). Unstimulated BM-DCs of TTP(-/-) mice expressed lower levels of mRNAs encoding the costimulatory molecules CD40 and CD86 and surprisingly also the canonical TTP targets TNF-alpha and IL-10 as compared with WT DCs. On the protein level, both DC populations expressed comparable amounts of CD80 and CD86 and of either cytokine, but TTP(-/-) DCs expressed less MHCII than WT DCs. On the other hand, TTP(-/-) DCs displayed elevated expression of other TTP target mRNAs like IL-1beta, c-fos and Mkp-1. Stimulation of BM-DCs of either genotype with lipopolysaccharide resulted in a rapid upregulation to a comparable extent of all molecules monitored so far, except for c-fos mRNA. Subsequent mRNA decay analysis revealed gene-specific differences in mRNA stability, which was influenced by the presence of TTP and the activation state of the DCs. Unstimulated TTP(-/-) DCs exerted a markedly lower allogeneic T cell stimulatory potential than WT DCs. Moreover, TTP(-/-) DCs induced an altered cytokine pattern in cocultures of DCs and T cells. However, allogeneic T cells primed by unstimulated DCs of either genotype were equally refractory to restimulation and suppressed the proliferation of naive T cells to the same extent. Thus, the findings of this study lend support to the interpretation that without external stimulation antigen presenting activity in DCs in the presence of TTP is more pronounced than in its absence and that posttranscriptional regulation contributes to the control of gene expression in DCs.


Subject(s)
Dendritic Cells/immunology , RNA Stability/immunology , RNA, Messenger/immunology , RNA-Binding Proteins/immunology , Tristetraprolin/immunology , Animals , B7-2 Antigen/biosynthesis , B7-2 Antigen/immunology , CD40 Antigens/biosynthesis , CD40 Antigens/immunology , Dendritic Cells/metabolism , Dual Specificity Phosphatase 1/biosynthesis , Dual Specificity Phosphatase 1/immunology , Female , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Knockout , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/immunology , RNA Stability/drug effects , RNA Stability/genetics , RNA, Messenger/biosynthesis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Tristetraprolin/genetics , Tristetraprolin/metabolism , Up-Regulation/drug effects , Up-Regulation/immunology
18.
Int Arch Allergy Immunol ; 151(3): 214-22, 2010.
Article in English | MEDLINE | ID: mdl-19786802

ABSTRACT

BACKGROUND: The migration of dendritic cells (DCs) from the lungs to the regional lymph nodes is necessary for the development of allergic airway disease. Following activation, mast cells release a variety of stored or de novo-produced inflammatory mediators, several of them being capable of activating DCs. In this study, the role of mast cells on DC migration from the lungs to the thoracic lymph nodes was investigated in sensitized mice. METHODS: Mast cell-deficient mice (Kit(W-sh/W-sh)) and their wild-type counterparts were sensitized intraperitoneally with ovalbumine (OVA) in saline and challenged by a single intranasal administration of OVA labeled with a fluorescent dye (OVA-Alexa). RESULTS: Following challenge, the relative and absolute amount of OVA- Alexa-positive DCs was clearly increased in sensitized wild-type mice compared to nonsensitized mice. In contrast, sensitized Kit(W-sh/W-sh) showed no increase in OVA-Alexa-positive DCs compared to nonsensitized mast cell-deficient animals. In sensitized Kit(W-sh/W-sh) mice reconstituted with bone marrow-derived mast cells (BMMCs), the number of OVA- Alexa-positive DCs was comparable to that in sensitized wild-type animals. However, transfer of allergen-exposed BMMCs to sensitized mice prior to airway challenge augmented airway inflammation similarly in wild-type and mast cell-deficient mice. In line with this, sensitization with allergen-pulsed DCs induced allergic airway disease independently of mast cells. CONCLUSIONS: This study shows an interaction between mast cells and DCs following allergen challenge in sensitized hosts. However, the function of mast cells can be bypassed in models utilizing activated allergen-exposed DCs to initiate the development of allergic airway disease.


Subject(s)
Bronchial Hyperreactivity/immunology , Chemotaxis, Leukocyte/immunology , Dendritic Cells/immunology , Mast Cells/immunology , Adoptive Transfer , Allergens/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Separation , Flow Cytometry , Mice , Ovalbumin/immunology
19.
J Immunol Methods ; 343(1): 13-20, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19186183

ABSTRACT

Under steady state conditions dendritic cells (DC) exert tolerogenic function, but acquire potent immunogenic function due to strong upregulation of costimulatory molecules and proinflammatory cytokines. In numerous studies the potential of modified DC to induce tolerance or immune reactions towards a distinct antigen has been demonstrated. However, DC are refractory to transfection with plasmid DNA by non-viral methods. In this study we have tested the suitability of a newly developed electroporation device to transfect immature murine bone-marrow derived DC (BM-DC). Transfected BM-DC expressed reporter molecules at considerable extent which renders this method suitable to perform all kinds of promoter studies. While electroporation did not alter the low allostimulatory capacity of immature BM-DC, it impaired the stimulation-associated increase in allostimulatory potency of transfectants. However, stimulated transfected BM-DC pulsed with myelin oligodendrocyte protein (MOG)-derived peptide induced proliferation of MOG-reactive CD4(+) T cells as potently as did non-transfected MOG peptide-pulsed BM-DC. BM-DC transfected with an expression construct encoding MOG efficiently stimulated MOG peptide-specific T cell proliferation. Transfection of BM-DC with an IL-10 encoding expression construct resulted in high IL-10 expression and strongly diminished allogeneic T cell proliferation. Therefore, this method also allows to study functional properties of genetically altered DC.


Subject(s)
Dendritic Cells/metabolism , Electroporation/methods , Transfection/methods , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Female , Gene Expression , Genetic Vectors , Interleukin-10/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteins , Myelin-Associated Glycoprotein/metabolism , Myelin-Oligodendrocyte Glycoprotein , Plasmids , Transgenes
20.
Immunology ; 128(1 Suppl): e193-205, 2009 Sep.
Article in English | MEDLINE | ID: mdl-18800984

ABSTRACT

Gene gun-mediated biolistic DNA vaccination with beta-galactosidase (betaGal)-encoding plasmid vectors efficiently modulated antigen-induced immune responses in an animal model of type I allergy, including the inhibition of immunoglobulin E (IgE) production. Here we show that CD4(+) as well as CD8(+) T cells from mice biolistically transfected with a plasmid encoding betaGal under the control of the fascin promoter (pFascin-betaGal) are capable of inhibiting betaGal-specific IgE production after adoptive transfer into naïve recipients. Moreover, suppression of IgE production was dependent on interferon (IFN)-gamma. To analyse the modalities of activation of CD4(+) and CD8(+) T cells regarding the localization of antigen synthesis following gene gun-mediated DNA immunization, we used the fascin promoter and the keratin 5 promoter (pK5-betaGal) to direct betaGal production mainly to dendritic cells (DCs) and to keratinocytes, respectively. Gene gun-mediated DNA immunization with each vector induced considerable activation of betaGal-specific CD8(+) cytotoxic T cells. Cytokine production by re-stimulated CD4(+) T cells in draining lymph nodes and immunoglobulin isotype profiles in sera of immunized mice indicated that immunization with pFascin-betaGal induced a T helper type 1 (Th1)-biased immune response, whereas immunization with pK5-betaGal generated a mixed Th1/Th2 immune response. Nevertheless, DNA vaccination with pFascin-betaGal and pK5-betaGal, respectively, efficiently inhibited specific IgE production in the mouse model of type I allergy. In conclusion, our data show that uptake of exogenous antigen produced by keratinocytes and its presentation by untransfected DCs as well as the presentation of antigen synthesized endogenously in DCs represent equivalent pathways for efficient priming of cellular immune responses.


Subject(s)
Antigen Presentation , Biolistics , Hypersensitivity/therapy , Langerhans Cells/immunology , Vaccination/methods , Vaccines, DNA/administration & dosage , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/genetics , Cross-Priming/immunology , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Female , Genetic Vectors/immunology , Genetic Vectors/metabolism , Immunoglobulin E/blood , Immunoglobulin G/blood , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Keratin-15 , Keratin-5/immunology , Keratin-5/metabolism , Keratinocytes/immunology , Mice , Mice, Knockout , Promoter Regions, Genetic/immunology , T-Lymphocytes, Helper-Inducer/enzymology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, DNA/immunology , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...