Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cells ; 11(14)2022 07 13.
Article in English | MEDLINE | ID: mdl-35883631

ABSTRACT

Heterodimeric ß2 integrin surface receptors (CD11a-d/CD18) are specifically expressed by leukocytes that contribute to pathogen uptake, cell migration, immunological synapse formation and cell signaling. In humans, the loss of CD18 expression results in leukocyte adhesion deficiency syndrome (LAD-)1, largely characterized by recurrent severe infections. All available mouse models display the constitutive and ubiquitous knockout of either α or the common ß2 (CD18) subunit, which hampers the analysis of the cell type-specific role of ß2 integrins in vivo. To overcome this limitation, we generated a CD18 gene floxed mouse strain. Offspring generated from crossing with CD11c-Cre mice displayed the efficient knockdown of ß2 integrins, specifically in dendritic cells (DCs). Stimulated ß2-integrin-deficient splenic DCs showed enhanced cytokine production and the concomitantly elevated activity of signal transducers and activators of transcription (STAT) 1, 3 and 5, as well as the impaired expression of suppressor of cytokine signaling (SOCS) 2-6 as assessed in bone marrow-derived (BM) DCs. Paradoxically, these BMDCs also showed the attenuated expression of genes involved in inflammatory signaling. In line, in experimental autoimmune encephalomyelitis mice with a conditional DC-specific ß2 integrin knockdown presented with a delayed onset and milder course of disease, associated with lower frequencies of T helper cell populations (Th)1/Th17 in the inflamed spinal cord. Altogether, our mouse model may prove to be a valuable tool to study the leukocyte-specific functions of ß2 integrins in vivo.


Subject(s)
CD18 Antigens , Dendritic Cells , Encephalomyelitis, Autoimmune, Experimental , Inflammation , Animals , CD18 Antigens/genetics , CD18 Antigens/metabolism , Cytokines/metabolism , Dendritic Cells/metabolism , Encephalomyelitis , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression , Inflammation/genetics , Leukocyte-Adhesion Deficiency Syndrome , Mice
2.
Cancers (Basel) ; 14(11)2022 May 31.
Article in English | MEDLINE | ID: mdl-35681718

ABSTRACT

BACKGROUND: Stimulated dendritic cells (DCs), which constitute the most potent population of antigen-presenting cells (APCs), express the actin-bundling protein Fascin-1 (Fscn1). In tumor cells, de novo expression of Fscn1 correlates with their invasive and metastatic properties. Therefore, Fscn1 inhibitors have been developed to serve as antitumor agents. In this study, we were interested in better understanding the impact of Fscn1 inhibitors on DCs. METHODS: In parallel settings, murine spleen cells and bone-marrow-derived DCs (BMDCs) were stimulated with lipopolysaccharide in the presence of Fscn1 inhibitors (NP-G2-044 and BDP-13176). An analysis of surface expression of costimulatory and coinhibitory receptors, as well as cytokine production, was performed by flow cytometry. Cytoskeletal alterations were assessed by confocal microscopy. The effects on the interactions of BMDCs with antigen-specific T cells were monitored by time lapse microscopy. The T-cell stimulatory and polarizing capacity of BMDCs were measured in proliferation assays and cytokine studies. RESULTS: Administration of Fscn1 inhibitors diminished Fscn1 expression and the formation of dendritic processes by stimulated BMDCs and elevated CD273 (PD-L2) expression. Fscn1 inhibition attenuated the interaction of DCs with antigen-specific T cells and concomitant T-cell proliferation. CONCLUSIONS: Systemic administration of Fscn1 inhibitors for tumor therapy may also modulate DC-induced antitumor immune responses.

3.
ACS Nano ; 16(3): 4426-4443, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35103463

ABSTRACT

The generation of specific humoral and cellular immune responses plays a pivotal role in the development of effective vaccines against tumors. Especially the presence of antigen-specific, cytotoxic T cells influences the outcome of therapeutic cancer vaccinations. Different strategies, ranging from delivering antigen-encoding mRNAs to peptides or full antigens, are accessible but often suffer from insufficient immunogenicity and require immune-boosting adjuvants as well as carrier platforms to ensure stability and adequate retention. Here, we introduce a pH-responsive nanogel platform as a two-component antitumor vaccine that is safe for intravenous application and elicits robust immune responses in vitro and in vivo. The underlying chemical design allows for straightforward covalent attachment of a model antigen (ovalbumin) and an immune adjuvant (imidazoquinoline-type TLR7/8 agonist) onto the same nanocarrier system. In addition to eliciting antigen-specific T and B cell responses that outperform mixtures of individual components, our two-component nanovaccine leads in prophylactic and therapeutic studies to an antigen-specific growth reduction of different tumors expressing ovalbumin intracellularly or on their surface. Regarding the versatile opportunities for functionalization, our nanogels are promising for the development of highly customized and potent nanovaccines.


Subject(s)
Cancer Vaccines , Neoplasms , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Adjuvants, Immunologic , Animals , Antigens , Immunity, Cellular , Mice , Mice, Inbred C57BL , Nanogels , Neoplasms/therapy , Ovalbumin , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists
4.
ACS Nano ; 15(9): 15191-15209, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34431291

ABSTRACT

Despite considerable progress in the design of multifunctionalized nanoparticles (NPs) that selectively target specific cell types, their systemic application often results in unwanted liver accumulation. The exact mechanisms for this general observation are still unclear. Here we asked whether the number of cell-targeting antibodies per NP determines the extent of NP liver accumulation and also addressed the mechanisms by which antibody-coated NPs are retained in the liver. We used polysarcosine-based peptobrushes (PBs), which in an unmodified form remain in the circulation for >24 h due to the absence of a protein corona formation and low unspecific cell binding, and conjugated them with specific average numbers (2, 6, and 12) of antibodies specific for the dendritic cell (DC) surface receptor, DEC205. We assessed the time-dependent biodistribution of PB-antibody conjugates by in vivo imaging and flow cytometry. We observed that PB-antibody conjugates were trapped in the liver and that the extent of liver accumulation strongly increased with the number of attached antibodies. PB-antibody conjugates were selectively captured in the liver via Fc receptors (FcR) on liver sinusoidal endothelial cells, since systemic administration of FcR-blocking agents or the use of F(ab')2 fragments prevented liver accumulation. Cumulatively, our study demonstrates that liver endothelial cells play a yet scarcely acknowledged role in liver entrapment of antibody-coated NPs and that low antibody numbers on NPs and the use of F(ab')2 antibody fragments are both sufficient for cell type-specific targeting of secondary lymphoid organs and necessary to minimize unwanted liver accumulation.


Subject(s)
Nanoparticles , Receptors, Fc , Endothelial Cells , Liver , Tissue Distribution
5.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799879

ABSTRACT

The development of nanocarriers (NC) for biomedical applications has gained large interest due to their potential to co-deliver drugs in a cell-type-targeting manner. However, depending on their surface characteristics, NC accumulate serum factors, termed protein corona, which may affect their cellular binding. We have previously shown that NC coated with carbohydrates to enable biocompatibility triggered the lectin-dependent complement pathway, resulting in enhanced binding to B cells via complement receptor (CR)1/2. Here we show that such NC also engaged all types of splenic leukocytes known to express CR3 at a high rate when NC were pre-incubated with native mouse serum resulting in complement opsonization. By focusing on dendritic cells (DC) as an important antigen-presenting cell type, we show that CR3 was essential for binding/uptake of complement-opsonized NC, whereas CR4, which in mouse is specifically expressed by DC, played no role. Further, a minor B cell subpopulation (B-1), which is important for first-line pathogen responses, and co-expressed CR1/2 and CR3, in general, engaged NC to a much higher extent than normal B cells. Here, we identified CR-1/2 as necessary for binding of complement-opsonized NC, whereas CR3 was dispensable. Interestingly, the binding of complement-opsonized NC to both DC and B-1 cells affected the expression of activation markers. Our findings may have important implications for the design of nano-vaccines against infectious diseases, which codeliver pathogen-specific protein antigen and adjuvant, aimed to induce a broad adaptive cellular and humoral immune response by inducing cytotoxic T lymphocytes that kill infected cells and pathogen-neutralizing antibodies, respectively. Decoration of nano-vaccines either with carbohydrates to trigger complement activation in vivo or with active complement may result in concomitant targeting of DC and B cells and thereby may strongly enhance the extent of dual cellular/humoral immune responses.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , CD11b Antigen/immunology , Complement System Proteins/immunology , Dendritic Cells/immunology , Receptors, Complement/immunology , Animals , B-Lymphocyte Subsets/metabolism , B-Lymphocytes/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cells, Cultured , Complement Activation/immunology , Complement System Proteins/metabolism , Dendritic Cells/metabolism , Dextrans/chemistry , Drug Carriers/chemistry , Humans , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Nanoparticles/chemistry , Opsonin Proteins/immunology , Opsonin Proteins/metabolism , Phagocytosis/immunology , Receptors, Complement/metabolism
6.
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
7.
J Immunol Res ; 2019: 4726532, 2019.
Article in English | MEDLINE | ID: mdl-31511826

ABSTRACT

The KH-type splicing regulatory protein (KSRP) is a RNA-binding protein, which regulates the stability of many mRNAs encoding immune-relevant proteins. As KSRP regulates innate immune responses, for instance by the modulation of type I interferon mRNA stability, we were interested whether knockdown of the protein (KSRP-/-) interferes with T cell activation and polarization. Polyclonally stimulated KSRP-/- CD4+ T cells proliferated at a higher extent and higher frequency and expressed the activation marker CD25 more than wild-type T cells. In supernatants of stimulated KSRP-/- CD4+ T cells, levels of IL-5, IL-9, IL-10, and IL-13 were observed to be increased compared to those of the control group. KSRP-/- CD8+ T cells showed no altered proliferative capacity upon polyclonal stimulation, but supernatants contained lower levels of interferon-γ. Similar changes in the cytokine expression patterns were also detected in T cells derived from KSRP-/- mice undergoing arthritis induction indicative of a pathophysiological role of KSRP-dependent T cell polarization. We demonstrated the direct binding of KSRP to the 3' untranslated region of IL-13, IL-10, and IFN-γ mRNA in in vitro experiments. Moreover, since IL-4 mRNA decay was reduced in KSRP-/- CD4+ T cells, we identify KSRP as a negative regulator of IL-4 expression. These data indicate that overexpression of IL-4, which constitutes the primary inducer of Th2 polarization, may cause the Th2 bias of polyclonally stimulated KSRP-/- CD4+ T cells. This is the first report demonstrating that KSRP is involved in the regulation of T cell responses. We present strong evidence that T cells derived from KSRP-/- mice favor Th2-driven immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/genetics , Gene Expression Regulation , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , 3' Untranslated Regions , Animals , Cytokines/metabolism , Humans , Mice , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Trans-Activators/genetics
8.
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.

9.
PLoS One ; 13(2): e0192541, 2018.
Article in English | MEDLINE | ID: mdl-29425215

ABSTRACT

The murine Friend virus (FV) retrovirus model has been widely used to study anti-viral immune responses, and virus-induced cancer. Here we analyzed FV immune evasion mechanisms on the level of dendritic cells (DC) essential for the induction of primary adaptive immune responses. Comparative quantitative proteome analysis of FV-infected DC (FV-DC) of different genotypes (BALB/c, C57BL/6) and non-infected DC revealed numerous genotype-independently regulated proteins rergulating metabolic activity, cytoskeletal rearrangements, and antigen processing/presentation. These alterations may promote virion production in FV-DC. Stimulation of FV-DC with LPS resulted in strongly enhanced IL-10 production which was partially responsible for their attenuated T cell (CD4+, CD8+) stimulatory capacity. Stimulated FV-DC induced less IFN-γ production in T cells required for cellular anti-viral responses, but more T helper cell type 2 (Th2)-associated cytokines (IL-4, IL-5, IL-13). We conclude that FV reprograms DC to promote viral spreading and immune deviation by imprinting a largely maturation-resistant, Th2-biased immunophenotype.


Subject(s)
Dendritic Cells/immunology , Friend murine leukemia virus/physiology , Immunity, Cellular/physiology , Th2 Cells/immunology , Animals , Cytokines/metabolism , Dendritic Cells/metabolism , Immunophenotyping , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteome
10.
Mol Immunol ; 87: 207-216, 2017 07.
Article in English | MEDLINE | ID: mdl-28511090

ABSTRACT

The KH type splicing regulatory protein (KSRP) is a nucleic acid binding protein, which negatively regulates the stability and/or translatability of many mRNA species encoding immune-relevant proteins. As KSRP is expressed in immune cells including T and B cells, neutrophils, macrophages and dendritic cells, we wanted to analyze its importance for the development of autoimmune diseases. We chose collagen antibody-induced arthritis (CAIA) as an appropriate autoimmune disease mouse model in which neutrophils and macrophages constitute the main effector cell populations. We compared arthritis induction in wild type (WT) and KSRP-/- mice and paws were taken for histological sections and qPCR analysis. Furthermore, we determined the frequencies of spleen immune cells by flow cytometry. Cytokine levels in spleen cell supernatants were determined by cytometric bead array analyses (CBA). After CAIA induction we unexpectedly observed in WT animals much stronger swelling of the paws than in KSRP-/- mice. In accordance, histological staining of paw sections of KSRP-/- animals revealed much lower frequencies of infiltrating immune cells in the joints compared to WT animals. Furthermore, CAIA-treatment resulted in reduced expression of several inflammatory factors (like CXCL-1, iNOS, TNF-α and S100A8) as well as immune cell marker genes (e.g. LFA-1, CD68, Ly6G) in the joints of KSRP-/- mice. Spleen cells of KSRP-/- mice showed lower frequencies of myeloid cells. On cytokine level IFN-γ production was increased in spleen cells of KSRP-/- mice compared to WT samples. These data surprisingly suggest that the absence of KSRP protects against the induction of inflammatory arthritis.


Subject(s)
Antibodies/metabolism , Arthritis, Experimental/genetics , Collagen/metabolism , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Ly/metabolism , Arthritis, Experimental/metabolism , Calgranulin A/metabolism , Chemokine CXCL1/metabolism , Cytokines/metabolism , Inflammation/genetics , Inflammation/metabolism , Interferon-gamma/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
Nanomedicine (Lond) ; 11(20): 2647-2662, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27628310

ABSTRACT

AIM: We wanted to assess the potency of a trifunctional nanoparticle (NP) that targeted and activated CD8+ dendritic cells (DC) and delivered an antigen to induce antitumor responses. MATERIALS & METHODS: The DC targeting and activating properties of ferrous NPs conjugated with immunostimulatory CpG-oligonucleotides, anti-DEC205 antibody and ovalbumin (OVA) as a model antigen to induce antigen-specific T-cell responses and antitumor responses were analyzed. RESULTS: OVA-loaded NP conjugated with immunostimulatory CpG-oligonucleotides and anti-DEC205 antibody efficiently targeted and activated CD8+ DC in vivo, and induced strong OVA-specific T-cell activation. Vaccination of B16/OVA tumor-burdened mice with this NP formulation resulted in tumor growth arrest. CONCLUSION: CD8+ DC-targeting trifunctional nanocarriers bear significant potential for antitumor immunotherapy.


Subject(s)
CD8 Antigens/metabolism , Dendritic Cells/immunology , Magnetite Nanoparticles/chemistry , Melanoma, Experimental/therapy , Oligonucleotides/immunology , Ovalbumin/immunology , Animals , Antibodies/chemistry , Antibodies/immunology , Antigens, CD/immunology , Cell Proliferation , CpG Islands , Dendritic Cells/metabolism , Dextrans/chemistry , Fluorescent Dyes/chemistry , Humans , Immunotherapy , Lectins, C-Type/immunology , Lymphocyte Activation , Magnetite Nanoparticles/therapeutic use , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Minor Histocompatibility Antigens/immunology , Oligonucleotides/chemistry , Receptors, Cell Surface/immunology , Surface Properties , Tumor Burden , Vaccination
12.
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
13.
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
14.
Macromol Biosci ; 16(1): 106-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26222986

ABSTRACT

Polylysine-b-p[HPMA] block copolymers containing a redox-responsive disulfide bond between both blocks are synthesized by RAFT polymerization of pentafluorphenyl-methacrylate with a macro-CTA from Nϵ-benzyloxycarbonyl (Cbz) protected polylysine (synthesized by NCA polymerization). This polylysine-b-p[PFMA] precursor block copolymer is converted to polylysine(Cbz)-b-p[HPMA] by postpolymerization modification with 2-hydroxypropylamine. After removal of the Cbz protecting group, cationic polylysine-b-p[HPMA] copolymers with a biosplittable disulfide moiety became available, which can be used as polymeric transfection vectors. These disulfide linked polylysine-S-S-b-p[HPMA] block copolymers show low cytotoxicity and increased transfection efficiencies (HEK-293T cells) compared to analogous blockcopolymers without disulfide group making them interesting for the transfection of sensitive immune cells.


Subject(s)
Polymers/chemistry , Transfection/methods , HEK293 Cells , Humans , Polylysine/analogs & derivatives , Polylysine/chemical synthesis , Polymerization , Polymers/chemical synthesis , Polymethacrylic Acids/chemical synthesis
15.
Macromol Biosci ; 15(8): 1159-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25974845

ABSTRACT

Herein we describe the synthesis of poly-L-lysine-b-poly[N-(2-hydroxypropyl)-metha-crylamide)] (poly[HPMA]) block copolymers by combination of solid phase peptide synthesis or polymerization of α-amino acid-N-carboxy-anhydrides (NCA-polymerization) with the reversible addition-fragmentation chain transfer polymerization (RAFT). In the presence of p-DNA, these polymers form polyplex micelles with a size of 100-200 nm in diameter (monitored by SDS-PAGE and FCS). Primary in vitro studies with HEK-293T cells reveal their cellular uptake (FACS studies and CLSM) and proof successful transfection with efficiencies depending on the length of polylysine. Moreover, these polyplexes display minimal toxicity (MTT-assay and FACS-measurements) featuring a p[HPMA] corona for efficient extracellular shielding and the potential ligation with antibodies.


Subject(s)
DNA/drug effects , Methacrylates/chemistry , Polylysine/chemistry , Polymerization , DNA/chemistry , HEK293 Cells/drug effects , Humans , Methacrylates/adverse effects , Methacrylates/pharmacology , Polylysine/adverse effects , Polylysine/pharmacology , Transfection
16.
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
17.
J Immunol ; 193(2): 485-95, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24929002

ABSTRACT

Signaling via the Wnt/ß-catenin pathway plays crucial roles in embryogenesis and homeostasis of adult tissues. In the lung, the canonical Wnt/ß-catenin pathway has been implicated in remodeling processes, development of emphysema, and fibrosis. However, its relevance for the modulation of allergic responses in the lung remains unclear. Using genetically modified mice with lung-specific inducible (doxycycline) Wnt-1 expression (CCSP-rtTA × tetO-Wnt1), the impact of Wnt on the development of allergic airway disease was analyzed. Overexpression of Wnt during the allergen challenge phase attenuated the development of airway inflammation in an acute model, as well as in a more therapeutic model of secondary challenge. These findings were further supported by treatment of allergen-sensitized mice with LiCl during challenge. Similar to Wnt, LiCl prevented the degradation of ß-catenin and, thus, attenuated allergic airway inflammation and hyperresponsiveness. Migration studies revealed that lung-specific expression of Wnt reduced the migration of Ag-loaded dendritic cells (DCs) into the draining lymph nodes following allergen challenge. Administration of in vitro allergen-loaded DCs overcame Wnt-mediated suppression of airway inflammation. Furthermore, in vitro studies confirmed that DC-dependent T cell activation is impaired by blocking ß-catenin degradation. These results demonstrate an important role for the canonical Wnt/ß-catenin pathway in the DC-mediated regulation of allergic responses in the lung.


Subject(s)
Respiratory Hypersensitivity/immunology , Signal Transduction/immunology , Wnt1 Protein/immunology , beta Catenin/immunology , Adjuvants, Immunologic/pharmacology , Animals , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Doxycycline/pharmacology , Flow Cytometry , Lithium Chloride/immunology , Lithium Chloride/pharmacology , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Ovalbumin/pharmacology , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/prevention & control , Signal Transduction/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , beta Catenin/metabolism
18.
PLoS One ; 8(12): e80904, 2013.
Article in English | MEDLINE | ID: mdl-24339889

ABSTRACT

Dendritic cells (DCs) constitute an attractive target for specific delivery of nanovaccines for immunotherapeutic applications. Here we tested nano-sized dextran (DEX) particles to serve as a DC-addressing nanocarrier platform. Non-functionalized DEX particles had no immunomodulatory effect on bone marrow (BM)-derived murine DCs in vitro. However, when adsorbed with ovalbumine (OVA), DEX particles were efficiently engulfed by BM-DCs in a mannose receptor-dependent manner. A DEX-based nanovaccine containing OVA and lipopolysaccharide (LPS) as a DC stimulus induced strong OVA peptide-specific CD4(+) and CD8(+) T cell proliferation both in vitro and upon systemic application in mice, as well as a robust OVA-specific humoral immune response (IgG1>IgG2a) in vivo. Accordingly, this nanovaccine also raised both a more pronounced delayed-type hypersensitivity response and a stronger induction of cytotoxic CD8(+) T cells than obtained upon administration of OVA and LPS in soluble form. Therefore, DEX-based nanoparticles constitute a potent, versatile and easy to prepare nanovaccine platform for immunotherapeutic approaches.


Subject(s)
Dendritic Cells/immunology , Dextrans/immunology , Immunity, Cellular , Immunity, Humoral , Nanoparticles , Vaccines/chemistry , Vaccines/immunology , Adsorption , Animals , Bone Marrow Cells/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Dendritic Cells/cytology , Dextrans/chemistry , Lectins, C-Type/metabolism , Lipopolysaccharides/chemistry , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Ovalbumin/chemistry , Receptors, Cell Surface/metabolism , Th2 Cells/immunology
19.
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
20.
J Allergy Clin Immunol ; 117(1): 196-203, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16387606

ABSTRACT

BACKGROUND: Allergen gene transfer represents an alternative approach to specific immunotherapy with allergen extracts. Gene gun-mediated DNA immunization with plasmid vectors expressing a transgene under control of the promoter of the fascin gene (pFascin) allows for antigen production predominantly by dendritic cells and resulted in the generation of CD8(+) cytotoxic T lymphocytes as well as in the development of a type 1 immune response. OBJECTIVE: We compared the in vivo efficiency of biolistic transfection with pFascin and plasmids containing the cytomegalovirus promoter (pCMV) in a mouse model of type I allergy. METHODS: BALB/c mice were sensitized with the model allergen beta-galactosidase to induce a distinctive type 2 immune response. The effect of prophylactic as well as therapeutic biolistic transfection with beta-galactosidase-encoding plasmids on the development of antibody titers was followed, and anaphylactic potential of sera was determined. Spleen cells were stimulated in vitro to analyze cytokine production and induction of CD8(+) effector T cells. RESULTS: Protective allergen gene transfer with pFascin efficiently prevented specific IgE production accompanied by immune deviation toward a T(H)1-polarized immune response as well as by the induction of IFN-gamma-producing CD8(+) effector T cells, being comparable to vaccination with pCMV. In a therapeutical setting, biolistic DNA vaccination with pFascin or pCMV transiently protected allergen-sensitized mice against the strong increase in specific IgE production caused by subsequent allergen challenge. CONCLUSION: We demonstrate for the first time that restricting transgene expression primarily to dendritic cells after DNA vaccination suffices to cause inhibition of IgE production prophylactically and therapeutically.


Subject(s)
Biolistics , Carrier Proteins/genetics , Dendritic Cells/immunology , Desensitization, Immunologic/methods , Immunoglobulin E/biosynthesis , Microfilament Proteins/genetics , Vaccination , Vaccines, DNA/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...