Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 9(91): eadh0152, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38181095

ABSTRACT

Immune tolerance is maintained in lymphoid organs (LOs). Despite the presence of complex immune cell networks in non-LOs, it is unknown whether self-tolerance is maintained in these tissues. We developed a technique to restrict genetic recombination to regulatory T cells (Tregs) only in skin. Selective depletion of skin Tregs resulted in T cell-mediated inflammation of hair follicles (HFs). Suppression did not rely on CTLA-4, but instead on high-affinity interleukin-2 (IL-2) receptor expression by skin Tregs, functioning exclusively in a cell-extrinsic manner. In a novel model of HF stem cell (HFSC)-driven autoimmunity, we reveal that skin Tregs immunologically protect the HFSC niche. Finally, we used spatial transcriptomics to identify aberrant IL-2 signaling at stromal-HF interfaces in a rare form of human alopecia characterized by HFSC destruction and alopecia areata. Collectively, these results reveal the fundamental biology of Tregs in skin uncoupled from the systemic pool and elucidate a mechanism of self-tolerance.


Subject(s)
Immune Privilege , T-Lymphocytes, Regulatory , Humans , Hair Follicle , Interleukin-2 , Stem Cell Niche
2.
J Immunother Cancer ; 9(10)2021 10.
Article in English | MEDLINE | ID: mdl-34675067

ABSTRACT

Viral antigens are among the strongest elicitors of immune responses. A significant proportion of the human population already carries pre-existing immunity against several childhood viruses, which could potentially be leveraged to fight cancer. We sought to provide proof of concept in mouse models that a pre-existing measles virus (MeV) immunity can be redirected to inhibit tumor growth by directly forcing expression of cognate antigens in the tumor. To this end, we designed DNA vaccines against known MeV cytotoxic and helper T epitopes, and administered these intradermally to mice that were subsequently challenged with syngeneic squamous cancer cells engineered to either express the cognate antigens or not. Alternatively, established wild-type tumors in vaccinated animals were treated intratumorally with in vitro transcribed mRNA encoding the cognate epitopes. Vaccination generated MeV cytotoxic T lymphocyte (CTL) immunity in mice as demonstrated by enhanced interferon gamma production, antigen-specific T cell proliferation, and CTL-mediated specific killing of antigen-pulsed target cells. When challenged with syngeneic tumor cells engineered to express the cognate antigens, 77% of MeV-vaccinated mice rejected the tumor versus 21% in control cohorts. Antitumor responses were largely dependent on the presence of CD8+ cells. Significant protection was observed even when only 25% of the tumor bulk expressed cognate antigens. We therefore tested the strategy therapeutically, allowing tumors to develop in vaccinated mice before intratumoral injection with Viromer nanoparticles complexed with mRNA encoding the cognate antigens. Treatment significantly enhanced overall survival compared with controls, including complete tumor regression in 25% of mice. Our results indicate that redirecting pre-existing viral immunity to fight cancer is a viable alternative that could meaningfully complement current cancer immune therapies such as personalized cancer vaccines and checkpoint inhibitor blockade.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/immunology , Immunologic Memory/immunology , Measles virus/immunology , Animals , Disease Models, Animal , Humans , Mice
3.
Eur J Immunol ; 51(1): 191-196, 2021 01.
Article in English | MEDLINE | ID: mdl-32648940

ABSTRACT

Interleukin-31 (IL-31) is a Th2 cell-derived cytokine that has been closely linked to pruritic skin inflammation. More recently, enhanced IL-31 serum levels have also been observed in patients with allergic rhinitis and allergic asthma. Therefore, the main aim of this study was to unravel the contribution of IL-31 to allergen-induced lung inflammation. We analyzed lung inflammation in response to the timothy grass (Phleum pratense) pollen allergen Phl p 5 in C57BL/6 wild-type (wt) mice, IL-31 transgenic (IL-31tg) mice, and IL-31 receptor alpha-deficient animals (IL-31RA-/- ). IL-31 and IL-31RA levels were monitored by qRT-PCR. Cellular infiltrate in bronchoalveolar lavage fluid (BALF) and lung tissue inflammation, mucus production as well as epithelial thickness were measured by flow cytometry and histomorphology. While allergen challenge induced IL-31RA expression in lung tissue of wt and IL-31tg mice, high IL-31 expression was exclusively observed in lung tissue of IL-31tg mice. Upon Phl p 5 challenge, IL-31tg mice showed reduced numbers of leukocytes and eosinophils in BALF and lung tissue as well as diminished mucin expression and less pronounced epithelial thickening compared to IL-31RA-/- or wt animals. These findings suggest that the IL-31/IL-31RA axis may regulate local, allergen-induced inflammation in the lungs.


Subject(s)
Allergens/adverse effects , Allergens/immunology , Interleukins/immunology , Plant Proteins/adverse effects , Plant Proteins/immunology , Pneumonia/immunology , Animals , Asthma/etiology , Asthma/immunology , Asthma/prevention & control , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Eosinophils/immunology , Female , Interleukins/genetics , Leukocytes/immunology , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phleum/adverse effects , Phleum/immunology , Pneumonia/etiology , Pneumonia/prevention & control , Pollen/adverse effects , Pollen/immunology , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology
4.
Dermatology ; 237(2): 296-302, 2021.
Article in English | MEDLINE | ID: mdl-32344413

ABSTRACT

BACKGROUND: Little information is available about the complexity and function of skin cells contributing to the high stability of tattoos. It has been shown that dermal macrophages play an important role in the storage and maintenance of pigment particles. By contrast, the impact of dermal fibroblasts, forming the connective tissue of the skin, on the stability of the tattoo is not known. METHOD: In this study, we compared the cell number and the particle load in dermal macrophages versus dermal fibroblasts, isolated from tail skin of tattooed mice. RESULTS: Microscopic analysis revealed that both cell populations contained the tattoo particles, although in largely different amounts. A small number of macrophages with high side scatter intensity contained a large quantity of pigment particles, whereas a high number of dermal fibroblasts harbored only a few pigment particles. Using the CD64dtr mouse model that allows for selective, diphtheria toxin-mediated depletion of macrophages, we have previously shown that macrophages hold the tattoo in place by capture-release and recapture cycles. In the tattooed skin of macrophage-depleted mice, the content of pigment particles in fibroblasts did not change; however, the total number of fibroblasts carrying particles increased. CONCLUSION: The present study demonstrates that dermal macrophages and fibroblasts contribute in different ways to the tattoo stability and further improves our knowledge on tattoo persistence.


Subject(s)
Coloring Agents , Dermis/cytology , Fibroblasts/physiology , Macrophages/physiology , Tattooing , Animals , Cell Count , Ink , Mice , Microscopy
5.
Sci Rep ; 10(1): 11164, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32636404

ABSTRACT

Human skin contains a population of memory T cells that supports tissue homeostasis and provides protective immunity. The study of human memory T cells is often restricted to in vitro studies and to human PBMC serving as primary cell source. Because the tissue environment impacts the phenotype and function of memory T cells, it is crucial to study these cells within their tissue. Here we utilized immunodeficient NOD-scid IL2rγnull (NSG) mice that carried in vivo-generated engineered human skin (ES). ES was generated from human keratinocytes and fibroblasts and was initially devoid of skin-resident immune cells. Upon adoptive transfer of human PBMC, this reductionist system allowed us to study human T cell recruitment from a circulating pool of T cells into non-inflamed human skin in vivo. Circulating human memory T cells preferentially infiltrated ES and showed diverse functional profiles of T cells found in fresh human skin. The chemokine and cytokine microenvironment of ES closely resembled that of non-inflamed human skin. Upon entering the ES T cells assumed a resident memory T cell-like phenotype in the absence of infection, and a proportion of these cutaneous T cells can be locally activated upon injection of monocyte derived dendritic cells (moDCs) that presented Candida albicans. Interestingly, we found that CD69+ memory T cells produced higher levels of effector cytokines in response to Candida albicans, compared to CD69- T cells. Overall, this model has broad utility in many areas of human skin immunology research, including the study of immune-mediated skin diseases.


Subject(s)
Immunologic Memory , Skin/immunology , T-Lymphocytes/immunology , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , Candida albicans/immunology , Female , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Middle Aged , Skin/cytology , Skin Transplantation , Tissue Engineering
7.
J Control Release ; 266: 87-99, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-28919557

ABSTRACT

Due to its unique immunological properties, the skin is an attractive target tissue for allergen-specific immunotherapy. In our current work, we combined a dendritic cell targeting approach with epicutaneous immunization using an ablative fractional laser to generate defined micropores in the upper layers of the skin. By coupling the major birch pollen allergen Bet v 1 to mannan from S. cerevisiae via mild periodate oxidation we generated hypoallergenic Bet-mannan neoglycoconjugates, which efficiently targeted CD14+ dendritic cells and Langerhans cells in human skin explants. Mannan conjugation resulted in sustained release from the skin and retention in secondary lymphoid organs, whereas unconjugated antigen showed fast renal clearance. In a mouse model, Bet-mannan neoglycoconjugates applied via laser-microporated skin synergistically elicited potent humoral and cellular immune responses, superior to intradermal injection. The induced antibody responses displayed IgE-blocking capacity, highlighting the therapeutic potential of the approach. Moreover, application via micropores, but not by intradermal injection, resulted in a mixed TH1/TH17-biased immune response. Our data clearly show that applying mannan-neoglycoconjugates to an organ rich in dendritic cells using laser-microporation is superior to intradermal injection. Due to their low IgE binding capacity and biodegradability, mannan neoglycoconjugates therefore represent an attractive formulation for allergen-specific epicutaneous immunotherapy.


Subject(s)
Allergens/administration & dosage , Antigens, Plant/administration & dosage , Dendritic Cells/immunology , Lasers , Mannans/administration & dosage , Skin/immunology , Vaccination/methods , Administration, Cutaneous , Animals , Complement Activation , Female , Humans , Immunoglobulin E/immunology , Mice, Inbred BALB C , Porosity , Th1 Cells/immunology , Th17 Cells/immunology
8.
J Immunol ; 199(5): 1626-1634, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28739880

ABSTRACT

The skin hosts a variety of dendritic cells (DCs), which act as professional APC to control cutaneous immunity. Langerhans cells (LCs) are the only DC subset in the healthy epidermis. However, due to the complexity of the skin DC network, their relative contribution to either immune activation or immune tolerance is still not entirely understood. To specifically study the function of LCs in vivo, without altering the DC subset composition in the skin, we have generated transgenic mouse models for tamoxifen-inducible de novo expression of Ags in LCs but no other langerin+ DCs. Therefore, this system allows for LC-restricted Ag presentation to T cells. Presentation of nonsecreted OVA (GFPOVA) by steady-state LCs resulted in transient activation of endogenous CTL in transgenic mice. However, when these mice were challenged with OVA by gene gun immunization in the contraction phase of the primary CTL response they did not respond with a recall of CTL memory but, instead, with robust Ag-specific CTL tolerance. We found regulatory T cells (Tregs) enriched in the skin of tolerized mice, and depletion of Tregs or adoptive experiments revealed that Tregs were critically involved in CTL tolerance. By contrast, when OVA was presented by activated LCs, a recallable CTL memory response developed in transgenic mice. Thus, neoantigen presentation by epidermal LCs results in either robust CTL tolerance or CTL memory, and this decision-making depends on the activation state of the presenting LCs.


Subject(s)
Immune Tolerance , Langerhans Cells/immunology , Skin/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation , Autoantigens/genetics , Autoantigens/immunology , Cells, Cultured , Gene Expression Regulation , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Ovalbumin/genetics , Ovalbumin/immunology , Tamoxifen/administration & dosage
9.
Anticancer Res ; 35(9): 4697-705, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26254359

ABSTRACT

BACKGROUND/AIM: Polycomb repressive complex 2 (PRC2), an epigenetic master regulator, contributes to progression and development of biliary tract cancer (BTC). The present study investigated the effects of the PRC2 inhibitor 3-deazaneplanocin A (DZNep) on BTC cell lines. MATERIALS AND METHODS: In vitro effects of DZNep treatment were analyzed for cell viability, gene expression and functional characteristics of cancer stem cell (CSC). RESULTS: DZNep treatment caused a cell line- and dose-dependent decrease in viability. In the EGI-1 cell line, a direct cytotoxic effect was accompanied by mRNA down-regulation of the PRC2 core components, cyclins as well as of CSC-related genes. Furthermore, DZNep affected putative CSCs by reduction of sphere formation and aldehyde dehydrogenase-1-positive cells. The stem cell characteristics of these subpopulations were verified by real-time polymerase chain reaction analysis. CONCLUSION: Taken together, our results show that DZNep might be a promising pharmacological agent for future therapies regarding BTC.


Subject(s)
Adenosine/analogs & derivatives , Biliary Tract Neoplasms/pathology , Neoplastic Stem Cells/pathology , Adenosine/pharmacology , Biliary Tract Neoplasms/genetics , Biomarkers, Tumor/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclins/genetics , Cyclins/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
PLoS One ; 10(6): e0128722, 2015.
Article in English | MEDLINE | ID: mdl-26030383

ABSTRACT

The skin accommodates multiple dendritic cell (DC) subsets with remarkable functional diversity. Immune reactions are initiated and modulated by the triggering of DC by pathogen-associated or endogenous danger signals. In contrast to these processes, the influence of intrinsic features of protein antigens on the strength and type of immune responses is much less understood. Therefore, we investigated the involvement of distinct DC subsets in immune reactions against two structurally different model antigens, E. coli beta-galactosidase (betaGal) and chicken ovalbumin (OVA) under otherwise identical conditions. After epicutaneous administration of the respective DNA vaccines with a gene gun, wild type mice induced robust immune responses against both antigens. However, ablation of langerin+ DC almost abolished IgG1 and cytotoxic T lymphocytes against betaGal but enhanced T cell and antibody responses against OVA. We identified epidermal Langerhans cells (LC) as the subset responsible for the suppression of anti-OVA reactions and found regulatory T cells critically involved in this process. In contrast, reactions against betaGal were not affected by the selective elimination of LC, indicating that this antigen required a different langerin+ DC subset. The opposing findings obtained with OVA and betaGal vaccines were not due to immune-modulating activities of either the plasmid DNA or the antigen gene products, nor did the differential cellular localization, size or dose of the two proteins account for the opposite effects. Thus, skin-borne protein antigens may be differentially handled by distinct DC subsets, and, in this way, intrinsic features of the antigen can participate in immune modulation.


Subject(s)
Dendritic Cells/immunology , Skin/immunology , Vaccines, DNA/immunology , Animals , Antigens, Surface/immunology , Biolistics/methods , Chickens , Escherichia coli/immunology , Immunoglobulin G/immunology , Langerhans Cells/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , T-Lymphocytes, Cytotoxic/immunology , beta-Galactosidase/immunology
11.
PLoS One ; 9(7): e102280, 2014.
Article in English | MEDLINE | ID: mdl-25025197

ABSTRACT

The type of immune response is critical for successful protection and typically determined by pathogen-associated danger molecules. In contrast, protein antigens are usually regarded as passive target structures. Here, we provide evidence that the structure of the antigen can profoundly influence the type of response that is elicited under else identical conditions. In mice, gene gun vaccines induce predominantly Th2-biased immune reactions against most antigens. One exception is E. coli beta-galactosidase (ßGal) that induces a balanced Th1/Th2 response. Because both, the delivered material (plasmid DNA-coated gold particles) as well as the procedure (biolistic delivery to the skin surface) is the same as for other antigens we hypothesized that Th1 induction could be a function of ßGal protein expressed in transfected cells. To test this we examined gene gun vaccines encoding structural or functional variants of the antigen. Employing a series of gene gun vaccines encoding individual structural domains of ßGal, we found that neither of them induced IgG2a antibodies. Even disruption of the homo-tetramer association of the native protein by deletion of a few N-terminal amino acids was sufficient to abrogate IgG2a production. However, enzymatically inactive ßGal with only one point mutation in the catalytic center retained the ability to induce Th1 reactions. Thus, structural but not functional integrity of the antigen must be retained for Th1 induction. ßGal is not a Th1 adjuvant in the classical sense because neither were ßGal-transgenic ROSA26 mice particularly Th1-biased nor did co-administration of a ßGal-encoding plasmid induce IgG2a against other antigens. Despite this, gene gun vaccines elicited Th1 reactions to antigens fused to the open reading frame of ßGal. We interpret these findings as evidence that different skin-borne antigens may be differentially handled by the immune system and that the three-dimensional structure of an antigen is an important determinant for this.


Subject(s)
Antigens, Bacterial/immunology , Escherichia coli Proteins/immunology , Th1 Cells/immunology , Vaccines, DNA/immunology , beta-Galactosidase/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Biolistics/methods , Cell Line , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Protein Interaction Domains and Motifs/immunology , Protein Multimerization/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines, DNA/administration & dosage , beta-Galactosidase/chemistry , beta-Galactosidase/genetics
12.
Int Arch Allergy Immunol ; 158(4): 326-34, 2012.
Article in English | MEDLINE | ID: mdl-22472723

ABSTRACT

BACKGROUND: Knowledge of allergen-specific T cell epitopes is a prerequisite not only for therapeutic approaches but also for elucidating immunological mechanisms of type I allergy. Ex vivo detection of allergen-specific T cells using class II tetramer technology has become an important tool for investigating immune responses in atopic and healthy individuals. METHODS: Using (3)H-thymidine incorporation assays, T cell epitopes specific for the major timothy grass pollen allergen Phl p 5.0101 were mapped in 11 allergic donors and two different mouse strains. Different protocols for expansion/restimulation of T cells from the blood of allergic donors and detection of allergen-specific T cells by Class II Ultimer staining were evaluated. RESULTS: We identified several new Phl p 5.0101 class II T cell epitopes in allergic patients and confirmed previously published ones. Additionally, we discovered the major T cell epitopes in BALB/c and C57BL/6 mice. Using a novel Class II Ultimer, we detected epitope-specific T cells expanded from the blood of an allergic donor. CONCLUSIONS: Epitope mapping of Phl p 5.0101 revealed an immunodominant epitope in BALB/c and C57BL/6 mice and an immunodominant region in humans (amino acids 259-282), which was recognized by 8 out of 11 allergic donors. Detection of Phl p 5-specific T cells was demonstrated using a Class II Ultimer specific for epitope 196-210. Successful detection of ultimer-positive T cells was strongly dependent on a resting phase after in vitro expansion.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Plant Proteins/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Epitope Mapping , Female , Humans , Immunodominant Epitopes/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Staining and Labeling
13.
J Immunol ; 188(11): 5319-26, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22539792

ABSTRACT

IL-31 is a T cell-derived cytokine that signals via a heterodimeric receptor composed of IL-31Rα and oncostatin M receptor ß. Although several studies have aimed to investigate IL-31-mediated effects, the biological functions of this cytokine are currently not well understood. IL-31 expression correlates with the expression of IL-4 and IL-13 and is associated with atopic dermatitis in humans, indicating that IL-31 is involved in Th2-mediated skin inflammation. Because dendritic cells are the main activators of Th cell responses, we posed the question of whether dendritic cells express the IL-31R complex and govern immune responses triggered by IL-31. In the current study, we report that primary human CD1c(+) as well as monocyte-derived dendritic cells significantly upregulate the IL-31Rα receptor chain upon stimulation with IFN-γ. EMSAs, chromatin immunoprecipitation assays, and small interfering RNA-based silencing assays revealed that STAT1 is the main transcription factor involved in IFN-γ-dependent IL-31Rα expression. Subsequent IL-31 stimulation resulted in a dose-dependent release of proinflammatory mediators, including TNF-α, IL-6, CXCL8, CCL2, CCL5, and CCL22. Because these cytokines are crucially involved in skin inflammation, we hypothesize that IL-31-specific activation of dendritic cells may be part of a positive feedback loop driving the progression of inflammatory skin diseases.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Inflammation Mediators/metabolism , Interferon-gamma/physiology , Receptors, Interleukin/biosynthesis , STAT1 Transcription Factor/physiology , Cells, Cultured , Dendritic Cells/pathology , Feedback , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/physiology , Receptors, Interleukin/genetics , Receptors, Interleukin/physiology , Skin Diseases/immunology , Skin Diseases/metabolism , Skin Diseases/pathology
14.
J Immunol ; 186(9): 5333-44, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21451110

ABSTRACT

Allergic inflammation is based on the cross-linking of mast cell and basophil-bound IgE Abs and requires at least two binding sites for IgE on allergens, which are difficult to characterize because they are often conformational in nature. We studied the IgE recognition of birch pollen allergen Bet v 1, a major allergen for >100 million allergic patients. Monoclonal and polyclonal Abs raised against Bet v 1-derived peptides were used to compete with allergic patients' IgE binding to Bet v 1 to search for sequences involved in IgE recognition. Strong inhibitions of patients' IgE binding to Bet v 1 (52-75%) were obtained with mAbs specific for two peptides comprising aa 29-58 (P2) and aa 73-103 (P6) of Bet v 1. As determined by surface plasmon resonance, mAb2 specific for P2 and mAb12 specific for P6 showed high affinity, but only polyclonal rabbit anti-P2 and anti-P6 Abs or a combination of mAbs inhibited allergen-induced basophil degranulation. Thus, P2 and P6 define a surface patch on the Bet v 1 allergen, which allows simultaneous binding of several different IgE Abs required for efficient basophil and mast cell activation. This finding explains the high allergenic activity of the Bet v 1 allergen. The approach of using peptide-specific Abs for the mapping of conformational IgE epitopes on allergens may be generally applicable. It may allow discriminating highly allergenic from less allergenic allergen molecules and facilitate the rational design of active and passive allergen-specific immunotherapy strategies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Antigens, Plant/immunology , Epitope Mapping/methods , Epitopes, B-Lymphocyte/immunology , Immunoglobulin E/immunology , Amino Acid Sequence , Animals , Antigens, Plant/chemistry , Basophils/immunology , Betula/immunology , Binding, Competitive , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitopes, B-Lymphocyte/chemistry , Humans , Hypersensitivity/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Pollen/immunology , Rabbits , Sequence Homology, Amino Acid
15.
J Immunol ; 186(3): 1377-83, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21187444

ABSTRACT

The C-type lectin langerin/CD207 was originally discovered as a specific marker for epidermal Langerhans cells (LC). Recently, additional and distinct subsets of langerin(+) dendritic cells (DC) have been identified in lymph nodes and peripheral tissues of mice. Although the role of LC for immune activation or modulation is now being discussed controversially, other langerin(+) DC appear crucial for protective immunity in a growing set of infection and vaccination models. In knock-in mice that express the human diphtheria toxin receptor under control of the langerin promoter, injection of diphtheria toxin ablates LC for several weeks whereas other langerin(+) DC subsets are replenished within just a few days. Thus, by careful timing of diphtheria toxin injections selective states of deficiency in either LC only or all langerin(+) cells can be established. Taking advantage of this system, we found that, unlike selective LC deficiency, ablation of all langerin(+) DC abrogated the activation of IFN-γ-producing and cytolytic CD8(+) T cells after gene gun vaccination. Moreover, we identified migratory langerin(+) dermal DC as the subset that directly activated CD8(+) T cells in lymph nodes. Langerin(+) DC were also critical for IgG1 but not IgG2a Ab induction, suggesting differential polarization of CD4(+) T helper cells by langerin(+) or langerin-negative DC, respectively. In contrast, protein vaccines administered with various adjuvants induced IgG1 independently of langerin(+) DC. Taken together, these findings reflect a highly specialized division of labor between different DC subsets both with respect to Ag encounter as well as downstream processes of immune activation.


Subject(s)
Antigens, Surface/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunoglobulin Heavy Chains/classification , Immunoglobulin Switch Region/genetics , Immunoglobulin gamma-Chains/biosynthesis , Lectins, C-Type/biosynthesis , Lymphocyte Activation/immunology , Mannose-Binding Lectins/biosynthesis , Skin/immunology , Vaccines, DNA/immunology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Surface/genetics , Biolistics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Death/genetics , Cell Death/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Dendritic Cells/metabolism , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Switch Region/immunology , Immunoglobulin gamma-Chains/classification , Immunoglobulin gamma-Chains/metabolism , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Mannose-Binding Lectins/deficiency , Mannose-Binding Lectins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Skin/cytology , Skin/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Vaccines, DNA/administration & dosage
16.
J Allergy Clin Immunol ; 124(5): 1070-7.e1-11, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19665781

ABSTRACT

BACKGROUND: The spread of type I allergic diseases has reached epidemic dimensions. The success of therapeutic intervention is limited, and hence prophylactic vaccination is now seriously considered. However, immunization of healthy individuals requires safety standards far beyond those applicable for therapeutic approaches. mRNAs encoding allergen molecules represent an attractive tool for preventive vaccination because of the inherent safety features of this vaccine type. OBJECTIVE: In the current study we investigated whether mRNA constructs would be capable of protecting against type I allergic reactions in a murine model using the grass pollen allergen Phl p 5 and 28 other major pollen, food, animal, mold, and latex allergens. METHODS: BALB/c mice were immunized intradermally either with conventional or replicase-based mRNA constructs. Subsequently, animals were sensitized by means of subcutaneous injection of allergen/alum, followed by airway provocation. IgG1/IgG2a/IgE titers were determined by using ELISAs. Allergen-specific functional IgE levels were assessed by using the basophil release assay. Measurement of cytokines in splenocyte cultures and bronchoalveolar lavage fluids were performed by using enzyme-linked immunosorbent spot assays/sandwich ELISAs. Eosinophil and CD8(+) counts in bronchoalveolar lavage specimens were determined by means of flow cytometry. Airway hyperreactivity was assessed with whole-body plethysmography and invasive resistance/dynamic compliance measurement. RESULTS: mRNA vaccination proved its antiallergic efficacy in terms of IgG subclass distribution, functional IgE suppression, reduction of IL-4 and IL-5 levels, induction of IFN-gamma-producing cells, and reduction of airway hyperreactivity and eosinophil counts in the lung. CONCLUSION: Immunization with mRNA induces T(H)1-biased immune responses similar to those elicited through DNA-based vaccination but additionally offers the advantage of a superior safety profile.


Subject(s)
Allergens/genetics , Hypersensitivity/prevention & control , RNA, Messenger/therapeutic use , Vaccination/methods , Vaccines/therapeutic use , Allergens/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/biosynthesis , Cytokines/immunology , Female , Immunoglobulin E/blood , Immunoglobulin G/blood , Injections, Intradermal , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/immunology , Vaccines/genetics , Vaccines/immunology
17.
J Immunol ; 182(10): 6298-306, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414783

ABSTRACT

Allergens and rhinovirus infections are among the most common elicitors of respiratory diseases. We report the construction of a recombinant combination vaccine for allergy and rhinovirus infections based on rhinovirus-derived VP1, the surface protein which is critically involved in infection of respiratory cells, and a nonallergenic peptide of the major grass pollen allergen Phl p 1. Recombinant hybrid molecules consisting of VP1 and a Phl p 1-derived peptide of 31 aa were expressed in Escherichia coli. The hybrid molecules did not react with IgE Abs from grass pollen allergic patients and lacked allergenic activity when exposed to basophils from allergic patients. Upon immunization of mice and rabbits, the hybrids did not sensitize against Phl p 1 but induced protective IgG Abs that cross-reacted with group 1 allergens from different grass species and blocked allergic patients' IgE reactivity to Phl p 1 as well as Phl p 1-induced basophil degranulation. Moreover, hybrid-induced IgG Abs inhibited rhinovirus infection of cultured human epithelial cells. The principle of fusing nonallergenic allergen-derived peptides onto viral carrier proteins may be used for the engineering of safe allergy vaccines which also protect against viral infections.


Subject(s)
Allergens/immunology , Common Cold/prevention & control , Hypersensitivity/prevention & control , Plant Proteins/immunology , Vaccines, Synthetic/immunology , Viral Proteins/immunology , Animals , Cross Reactions/immunology , Electrophoresis, Polyacrylamide Gel , Humans , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Rabbits , Recombinant Fusion Proteins/immunology , Rhinovirus , Vaccines, Combined/immunology
18.
J Immunol ; 180(11): 7451-60, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18490745

ABSTRACT

IgE-mediated sensitization to wheat flour belongs to the most frequent causes of occupational asthma. A cDNA library from wheat seeds was constructed and screened with serum IgE from baker's asthma patients. One IgE-reactive phage clone contained a full-length cDNA coding for an allergen with a molecular mass of 9.9 kDa and an isoelectric point of 6. According to sequence analysis it represents a member of the potato inhibitor I family, a group of serine proteinase inhibitors, and thus is the first allergen belonging to the group 6 pathogenesis-related proteins. The recombinant wheat seed proteinase inhibitor was expressed in Escherichia coli and purified to homogeneity. According to circular dichroism analysis, it represented a soluble and folded protein with high thermal stability containing mainly beta-sheets, random coils, and an alpha-helical element. The recombinant allergen showed allergenic activity in basophil histamine release assays and reacted specifically with IgE from 3 of 22 baker's asthma patients, but not with IgE from grass pollen allergic patients or patients suffering from food allergy to wheat. Allergen-specific Abs were raised to localize the allergen by immunogold electron microscopy in the starchy endosperm and the aleuron layer. The allergen is mainly expressed in mature wheat seeds and, despite an approximately 50% sequence identity, showed no relevant cross-reactivity with allergens from other plant-derived food sources such as maize, rice, beans, or potatoes. Recombinant wheat serine proteinase inhibitor, when used in combination with other specific allergens, may be useful for the diagnosis and therapy of IgE-mediated baker's asthma.


Subject(s)
Allergens , Asthma/immunology , Occupational Diseases/immunology , Serine Proteinase Inhibitors/immunology , Triticum/immunology , Wheat Hypersensitivity/immunology , Adult , Allergens/chemistry , Allergens/genetics , Allergens/immunology , Allergens/metabolism , Amino Acid Sequence , Base Sequence , Female , Gene Library , Humans , Immunoglobulin E , Male , Microscopy, Immunoelectron , Middle Aged , Molecular Sequence Data , Phylogeny , Seeds/chemistry , Sequence Alignment , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/metabolism , Triticum/chemistry
19.
J Immunol ; 179(2): 886-93, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617579

ABSTRACT

Gene gun immunization, i.e., bombardment of skin with DNA-coated particles, is an efficient method for the administration of DNA vaccines. Direct transfection of APC or cross-presentation of exogenous Ag acquired from transfected nonimmune cells enables MHC-I-restricted activation of CD8(+) T cells. Additionally, MHC-II-restricted presentation of exogenous Ag activates CD4(+) Th cells. Being the principal APC in the epidermis, Langerhans cells (LC) seem ideal candidates to accomplish these functions. However, the dependence on LC of gene gun-induced immune reactions has not yet been demonstrated directly. This was primarily hampered by difficulties to discriminate the contributions of LC from those of other dermal dendritic cells. To address this problem, we have used Langerin-diphtheria toxin receptor knockin mice that allow for selective inducible ablation of LC. LC deficiency, even over the entire duration of experiments, did not affect any of the gene gun-induced immune functions examined, including proliferation of CD4(+) and CD8(+) T cells, IFN-gamma secretion by spleen cells, Ab production, CTL activity, and development of protective antitumor immunity. Together, our data show that gene gun immunization is capable of inducing humoral and cell-mediated immune reactions independently of LC.


Subject(s)
Antibody Formation , Biolistics , Epidermis/immunology , Immunity, Cellular , Immunization/methods , Langerhans Cells/immunology , Animals , Antibody Formation/immunology , Cancer Vaccines/administration & dosage , Cell Proliferation , Epidermal Cells , Flow Cytometry , Heparin-binding EGF-like Growth Factor , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage
20.
Mol Immunol ; 44(8): 1879-87, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17070909

ABSTRACT

Gene gun immunization has been associated with the induction of a heterologous type of immune response characterized by a T(H)1-like immune reaction on the cellular level, i.e. generation of IFN-gamma secreting CD8(+) T-cells, yet a T(H)2 biased serology as indicated by high IgG1:IgG2a ratios and induction of IgE. Nevertheless, gene gun immunization using the model molecule beta-galactosidase has been argued to prevent IgE induction and to promote T(H)1 cells with respect to allergy DNA immunization. In our current study, we evaluated the potential of gene gun immunization to prevent type I allergic reactions comparing beta-galactosidase with two clinically relevant allergens, and further investigated the effect of gene gun immunization on relevant lung parameters. BALB/c mice were immunized with plasmids encoding the birch pollen allergen Bet v 1, the grass pollen allergen Phl p 5, or the model molecule beta-galactosidase, either by gene gun or intradermal injection followed by sensitization and intranasal provocation with the respective allergen. IgG1 and IgG2a antibody titers were determined by ELISA. IgE levels were evaluated in a rat basophil release assay. The severity of eosinophilia was determined in bronchoalveolar lavages, and the overall infiltrate was analyzed by histology on lung paraffin sections. Gene gun immunization induced a T(H)2-biased immune reaction, which did not prevent from production of IgE after subsequent sensitization. This T(H)2 effect was influenced by the nature of the antigen, with a more pronounced T(H)2-bias for the allergens Bet v 1 and Phl p 5 compared to beta-galactosidase. Gene gun immunization with all three antigens promoted eosinophil influx into the lung and did not alleviate lung pathology after intranasal provocation. In contrast to needle injection of plasmid DNA, which triggers a clearly T(H)1-biased and allergy-preventing immune response, gene gun application fails to induce anti-allergic reactions with all tested antigens and is therefore contraindicated for allergen-specific immunotherapy.


Subject(s)
Allergens/adverse effects , Biolistics , Desensitization, Immunologic/adverse effects , Hypersensitivity/complications , Immunization/adverse effects , Plant Proteins/adverse effects , Pulmonary Eosinophilia/chemically induced , Vaccines, DNA/adverse effects , Allergens/genetics , Allergens/immunology , Animals , Antigens, Plant , Basophils/immunology , Basophils/pathology , Bronchoalveolar Lavage Fluid , Desensitization, Immunologic/instrumentation , Desensitization, Immunologic/methods , Female , Hypersensitivity/immunology , Hypersensitivity/pathology , Hypersensitivity/prevention & control , Immunization/instrumentation , Immunization/methods , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Interferon-gamma/immunology , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Plant Proteins/genetics , Plant Proteins/immunology , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/pathology , Rats , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , beta-Galactosidase/genetics , beta-Galactosidase/immunology
SELECTION OF CITATIONS
SEARCH DETAIL