Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Allergy ; 78(6): 1605-1614, 2023 06.
Article En | MEDLINE | ID: mdl-36704937

BACKGROUND: Peanut allergy is a type-I hypersensitivity immune reaction mediated by the binding of peanut allergens to IgE-FcεRI complexes on mast cells and basophils and by their subsequent cellular degranulation. Of all major peanut allergens, Ara h 2 is considered the most anaphylactic. With few options but allergen avoidance, effective treatment of allergic patients is needed. Passive immunotherapy (herein called PIT) based on prophylactic administration of peanut-specific monoclonal antibodies (mAbs) may present a promising treatment option for this under-served disease. METHOD: Fully human recombinant anti-peanut IgG mAbs were tested in mice sensitized to peanut allergen extract. Allergic mice received intravenous immunotherapy with anti-peanut Ara h 2-specific IgG1 or IgG4 mAbs cocktails, and were then challenged by a systemic injection of high-dose peanut allergen extract. The protection from allergic anaphylaxis was measured by monitoring the core body temperature. RESULTS: PIT with peanut-specific mAbs was associated with a significant and dose-dependent reduction of anaphylactic reactions in peanut-sensitized mice challenged with peanut allergen extract. Complete protection was observed at doses approximately 0.3-0.6 mg mAbs. Mixtures of mAbs were more effective than single mAbs, and effective treatment could be obtained with mAbs of both IgG1 and IgG4 subclasses. The therapeutic effect of anti-Ara h 2 mAbs was based on allergen neutralization and independent of the Fcγ receptor and mast-cell inhibition. CONCLUSION: This is the first report that shows that human-derived anti-peanut mAbs can prevent allergic anaphylaxis in mice. The study demonstrates that neutralizing allergenic epitopes on Ara h 2 by mAbs may represent a promising treatment option in peanut-allergy.


Anaphylaxis , Hypersensitivity, Immediate , Peanut Hypersensitivity , Humans , Mice , Animals , Anaphylaxis/prevention & control , Antibodies, Monoclonal , Antigens, Plant , Peanut Hypersensitivity/prevention & control , Allergens , Recombinant Proteins , Immunoglobulin G , Arachis , Plant Extracts , 2S Albumins, Plant/chemistry
2.
Clin Exp Allergy ; 53(5): 550-560, 2023 05.
Article En | MEDLINE | ID: mdl-36629248

BACKGROUND: Peanut allergy accounts for the majority of food-induced hypersensitivity reactions and can lead to lethal anaphylaxis. Animal models can provide an insight into the immune mechanisms responsible for sensitization and allergic anaphylaxis. However, different mouse strains and sensitization protocols can influence the successful development of a peanut allergic mouse model. OBJECTIVE: We aimed at developing a systemic anaphylaxis model of peanut allergy that resembles human anaphylaxis. We compared the immunological and clinical responses in genetically different mouse strains. METHODS: Female BALB/c, C57BL/6, and C3H mice were intraperitoneally sensitized and later challenged with peanut proteins. Allergen-specific serology was done by ELISA, and anaphylaxis was evaluated by monitoring changes in body temperature upon systemic challenge. RESULTS: Sensitization to peanut was successful in C3H mice and triggered production of allergen-specific antibodies, cytokines and anaphylaxis. Allergic reactions were characterized by the release of allergic mediators and by changes in leukocyte populations in blood and in the peritoneal cavity. Among the identified major peanut allergens, Ara h 2 showed the strongest anaphylactic potential. Much lower or no trigger of peanut-specific antibodies was observed in BALB/c and C57BL/6 mice, which experienced no hypersensitivity reactions. CONCLUSIONS: Mouse strain matters for testing of peanut protein allergens. We identified C3H mice as a suitable strain for the development of a mouse model of peanut-allergic anaphylaxis. Pre-clinical, humoural and cellular responses resembled the responses observed in human patients. The described model can be useful for further studies on peanut allergy and for the development of new therapeutic strategies.


Anaphylaxis , Food Hypersensitivity , Peanut Hypersensitivity , Humans , Female , Mice , Animals , Arachis , Mice, Inbred C3H , Immunoglobulin E , Mice, Inbred C57BL , Allergens
4.
Front Immunol ; 13: 815609, 2022.
Article En | MEDLINE | ID: mdl-35173729

Conventional vaccines are very efficient in the prevention of bacterial infections caused by extracellular pathogens due to effective stimulation of pathogen-specific antibodies. In contrast, considering that intracellular surveillance by antibodies is not possible, they are typically less effective in preventing or treating infections caused by intracellular pathogens such as Mycobacterium tuberculosis. The objective of the current study was to use so-called photochemical internalization (PCI) to deliver a live bacterial vaccine to the cytosol of antigen-presenting cells (APCs) for the purpose of stimulating major histocompatibility complex (MHC) I-restricted CD8 T-cell responses. For this purpose, Mycobacterium bovis BCG (BCG) was combined with the photosensitiser tetraphenyl chlorine disulfonate (TPCS2a) and injected intradermally into mice. TPCS2a was then activated by illumination of the injection site with light of defined energy. Antigen-specific CD4 and CD8 T-cell responses were monitored in blood, spleen, and lymph nodes at different time points thereafter using flow cytometry, ELISA and ELISPOT. Finally, APCs were infected and PCI-treated in vitro for analysis of their activation of T cells in vitro or in vivo after autologous vaccination of mice. Combination of BCG with PCI induced stronger BCG-specific CD4 and CD8 T-cell responses than treatment with BCG only or with BCG and TPCS2a without light. The overall T-cell responses were multifunctional as characterized by the production of IFN-γ, TNF-α, IL-2 and IL-17. Importantly, PCI induced cross-presentation of BCG proteins for stimulation of antigen-specific CD8 T-cells that were particularly producing IFN-γ and TNF-α. PCI further facilitated antigen presentation by causing up-regulation of MHC and co-stimulatory proteins on the surface of APCs as well as their production of TNF-α and IL-1ß in vivo. Furthermore, PCI-based vaccination also caused local inflammation at the site of vaccination, showing strong infiltration of immune cells, which could contribute to the stimulation of antigen-specific immune responses. This study is the first to demonstrate that a live microbial vaccine can be combined with a photochemical compound and light for cross presentation of antigens to CD8 T cells. Moreover, the results revealed that PCI treatment strongly improved the immunogenicity of M. bovis BCG.


BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lung/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , BCG Vaccine/administration & dosage , Cross-Priming , Female , Inflammation/immunology , Injections, Intradermal , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mycobacterium bovis/immunology , Photosensitizing Agents/administration & dosage , Tumor Necrosis Factor-alpha/biosynthesis , Vaccination/methods
5.
ACS Chem Biol ; 16(5): 838-843, 2021 05 21.
Article En | MEDLINE | ID: mdl-33881303

Nanoparticle-based delivery systems have shown great promise for theranostics and bioimaging on the laboratory scale due to favorable pharmacokinetics and biodistribution. In this study, we examine the utility of a cage-forming variant of the protein lumazine synthase, which was previously designed and evolved to encapsulate biomacromolecular cargo. Linking antibody-binding domains to the exterior of the cage enabled binding of targeting immunoglobulins and cell-specific uptake of encapsulated cargo. Protein nanocages displaying antibody-binding domains appear to be less immunogenic than their unmodified counterparts, but they also recruit serum antibodies that can mask the efficacy of the targeting antibody. Our study highlights the strengths and limitations of a common targeting strategy for practical nanoparticle-based delivery applications.


Biocompatible Materials/chemistry , Multienzyme Complexes/chemistry , Nanocapsules/chemistry , Antibodies/chemistry , Antibodies/immunology , Cell Membrane Permeability , Drug Compounding , Drug Liberation , Humans , Immunoglobulins/chemistry , Immunoglobulins/immunology , Molecular Targeted Therapy , Protein Engineering , Surface Properties , Tissue Distribution
6.
J Control Release ; 332: 96-108, 2021 04 10.
Article En | MEDLINE | ID: mdl-33609623

Antigen cross-presentation to cytotoxic CD8+ T cells is crucial for the induction of anti-tumor and anti-viral immune responses. Recently, co-encapsulation of photosensitizers and antigens into microspheres and subsequent photochemical internalization (PCI) of antigens in antigen presenting cells has emerged as a promising new strategy for inducing antigen-specific CD8+ T cell responses in vitro and in vivo. However, the exact cellular mechanisms have hardly been investigated in vivo, i.e., which cell types take up antigen-loaded microspheres at the site of injection, or in which secondary lymphoid organ does T cell priming occur? We used spray-dried poly(lactic-co-glycolic acid) (PLGA) microspheres loaded with ovalbumin and the photosensitizer tetraphenyl chlorine disulfonate (TPCS2a) to investigate these processes in vivo. Intravital microscopy and flow cytometric analysis of the murine ear skin revealed that dendritic cells (DCs) take up PLGA microspheres in peripheral tissues. Illumination then caused photoactivation of TPCS2a and induced local tissue inflammation that enhanced CCR7-dependent migration of microsphere-containing DCs to tissue-draining lymph nodes (LNs), i.e., the site of CD8+ T cell priming. The results contribute to a better understanding of the functional mechanism of PCI-mediated vaccination and highlight the importance of an active transport of vaccine microspheres by antigen presenting cells to draining LNs.


Antigens , CD8-Positive T-Lymphocytes , Animals , Dendritic Cells , Lymph Nodes , Mice , Mice, Inbred C57BL , Ovalbumin , Receptors, CCR7
7.
Front Immunol ; 10: 1548, 2019.
Article En | MEDLINE | ID: mdl-31333674

Cytotoxic T lymphocytes (CTLs) are key players in fighting cancer, and their induction is a major focus in the design of therapeutic vaccines. Yet, therapeutic vaccine efficacy is limited, in part due to the suboptimal vaccine processing by antigen-presenting cells (APCs). Such processing typically takes place via the MHC class II pathway for CD4 T-cell activation and MHC class I pathway for activation of CD8 CTLs. We show that a combination of skin photochemical treatment and immunization, so-called photochemical internalization (PCI) facilitated CTL activation due to the photochemical adjuvant effect induced by photosensitizer, oxygen, and light. Mice were immunized intradermally with antigen and photosensitizer, followed by controlled light exposure. PCI-treated mice showed strong activation of CD8 T cells, with improved IFN-γ production and cytotoxicity, as compared to mice immunized without parallel PCI treatment. Surprisingly, the CD8 T-cell effector functions were not impaired in MHC class II- or CD4 T-cell-deficient mice. Moreover, PCI-based vaccination caused tumor regression independent of MHC class II or CD4 T cells presence in melanoma bearing mice. Together, the data demonstrate that PCI can act as a powerful adjuvant in cancer vaccines, even in hosts with impaired T-helper functions.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Photosensitivity Disorders/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Female , Immunization/methods , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Melanoma/immunology , Mice , Mice, Inbred C57BL , Photosensitizing Agents/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccination/methods
8.
J Immunol ; 195(1): 166-73, 2015 Jul 01.
Article En | MEDLINE | ID: mdl-26019274

The generation of CTLs is crucial in the immunological fight against cancer and many infectious diseases. To achieve this, vaccine Ags need to be targeted to the cytosol of dendritic cells, which can activate CD8 T cells via MHC class I (MHCI). Therefore, such targeting has become one of the major objectives of vaccine research. In this study, we aimed to bypass the unwanted and default MHC class II Ag presentation and trigger MHCI presentation by using a photosensitizer that, upon light activation, would facilitate cytosolic targeting of codelivered Ag. Poly(lactide-co-glycolide) microparticles ∼1 µm size were loaded with OVA and the photosensitizer tetraphenyl chlorine disulphonate (TPCS2a) and administered intradermally in mice, which were illuminated 1 d later for activation of the photosensitizer. Immunization in the presence of TPCS2a significantly increased activation of CD8 T cells compared with immunization without TPCS2a and as measured by CD8 T cell proliferation, production of proinflammatory IFN-γ, TNF-α, and IL-2, and prevention of tumor growth. Cytotoxicity was demonstrated by granzyme B production in vitro and by in vivo killing of CFSE-labeled targets. CD4-dependent Ab responses were abrogated in mice immunized with TPCS2a-containing particles, suggesting that photosensitization facilitated a shift from default MHC class II toward MHCI Ag presentation. Hence, vaccine particles with Ag and photosensitizers proved an effective vehicle or adjuvant for stimulation of CTLs, and they may find potential application in therapeutic cancer vaccination and in prophylactic and therapeutic vaccination against intracellular infections.


Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Dendritic Cells/drug effects , Melanoma, Experimental/prevention & control , Porphyrins/administration & dosage , Skin Neoplasms/prevention & control , T-Lymphocytes, Cytotoxic/drug effects , Animals , Antibodies, Neoplasm/biosynthesis , Antigens, Neoplasm/administration & dosage , Cytosol/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Drug Carriers/chemistry , Female , Granzymes/biosynthesis , Immunization , Injections, Intradermal , Interleukin-2/biosynthesis , Lactic Acid/chemistry , Light , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Neoplasm Transplantation , Ovalbumin/administration & dosage , Ovalbumin/immunology , Photosensitizing Agents/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Tumor Necrosis Factor-alpha/biosynthesis
9.
J Control Release ; 198: 10-7, 2015 Jan 28.
Article En | MEDLINE | ID: mdl-25482339

Cancer vaccines aim to induce CD8 T cells infiltrating the tumour. For protein-based vaccines, the main biological barrier to overcome is the default MHC class-II-pathway, with activation of CD4 T cells rather than CD8 T cells. The latter requires antigens to access the cytosol and MHC class I antigen presentation. We applied photosensitiser and light to trigger disruption of antigen-containing endosomes and thereby MHC class I cross-presentation of a model cancer vaccine. This "photochemical internalisation" resulted in activation, proliferation, and IFN-γ production of cytotoxic CD8 T cells, which suppressed tumour growth by infiltrating CD8 T cells and caspase-3-dependent apoptosis. The process was independent of MHC class II, MyD88, and TLR4 signalling, but dependent on trypsin- and caspase-like proteasome activity and partly also on chloroquine. This novel method of vaccination may find applications in cancer immunotherapy where the activation of CD8 T cells is important.


CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Melanoma/therapy , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Skin Neoplasms/therapy , Animals , Cell Proliferation , Cross-Priming , Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Interferon-gamma/immunology , Light , Melanoma/immunology , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Photosensitivity Disorders , Skin Neoplasms/immunology , Spleen/cytology
10.
J Control Release ; 174: 143-50, 2014 Jan 28.
Article En | MEDLINE | ID: mdl-24280261

The protection or treatment of several immunological disorders is dependent on the antigen-specific and cytotoxic CD8 T cells. However, vaccines aimed at stimulating CD8 T-cell responses are typically ineffective because vaccine antigens are primarily processed by the MHC class-II and not the MHC class-I pathway of antigen presentation: the latter requires cytosolic delivery of antigen. In order to facilitate targeting of antigen to cytosol, the antigen was combined with the photosensitiser TPCS2a (disulfonated tetraphenyl chlorin) and administered intradermally to mice. The photosensitiser was activated by illumination of the injection site. This photochemical internalization (PCI) strongly increased the stimulation of CD8 T-cell responses as measured by antigen-specific proliferation and secretion of pro-inflammatory cytokines. Fluorescence microscopy showed that delivery to cytosol was TPCS2a dependent and occurred by light-induced disruption of TPCS2a- and antigen-containing endosomes. PCI-based vaccination prevented growth of malignant B16 cells as compared with vaccination without PCI. In conclusion, PCI represents a potent tool for delivery of antigens to cytosol for stimulation of cytotoxic CD8 T-cell responses. This study demonstrated a first proof-of-principle for PCI-mediated immunisation with potential application in cancer immunotherapy.


Antigens/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Ovalbumin/pharmacology , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cell Line, Tumor , Cytosol/metabolism , DNA-Binding Proteins/genetics , Female , Genes, MHC Class I/genetics , Immunization , Injections, Intradermal , Light , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Photosensitizing Agents/radiation effects , Porphyrins/radiation effects
11.
Eur J Pharm Biopharm ; 85(1): 34-41, 2013 Sep.
Article En | MEDLINE | ID: mdl-23461859

Tumour chemotherapy with drugs is typically associated with severe systemic and local side effects for which reason immunotherapy represents a safer alternative. However, vaccination often fails to generate the required cytotoxic CD8 T-cell responses due to insufficient access of antigens to the cytosol and the MHC class I pathway of antigen presentation. One important issue of tumour research is therefore to develop strategies that allow cytosolic targeting or endosomal escape of tumour antigens. The objective of the current study was to test whether endocytosed antigen could be delivered to MHC class I by means of photochemical internalisation (PCI). Briefly, the antigen and the photosensitiser Amphinex were loaded in vitro onto bone-marrow-derived murine dendritic cells (DCs). After light activation, which is supposed to cause disruption of OVA- and Amphinex-containing endosomes, the DCs were cultured with OVA-specific CD8 T cells or used for immunisation of mice. PCI facilitated CD8 T-cell responses as measured by IFN-γ secretion in vitro and CD8 T-cell proliferation in vivo. In conclusion, the current proof-of-concept study is the first to describe PCI-mediated immunisation and the results revealed the feasibility of this novel technology in autologous vaccination for stimulation of CD8 T-cell responses.


Antigens/administration & dosage , Cytosol/metabolism , Genes, MHC Class I , Immunity, Cellular/drug effects , Immunization/methods , Photosensitizing Agents/pharmacology , T-Lymphocytes/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/radiation effects , Antigens/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/radiation effects , Cells, Cultured , Cytosol/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/radiation effects , Endocytosis/drug effects , Endocytosis/radiation effects , Endosomes/chemistry , Endosomes/metabolism , Endosomes/radiation effects , Feasibility Studies , Female , Immunity, Cellular/radiation effects , Interferon-gamma Release Tests , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Photosensitizing Agents/radiation effects , Specific Pathogen-Free Organisms , T-Lymphocytes/metabolism
12.
Exp Dermatol ; 22(2): 119-24, 2013 Feb.
Article En | MEDLINE | ID: mdl-23362870

Mycosis fungoides and its leukaemic counterpart Sézary syndrome are the most frequent cutaneous T-cell lymphomas (CTCL), and there is no cure for these diseases. We evaluated the effect of clinically approved antihistamines on the growth of CTCL cell lines. CTCL cell lines as well as blood lymphocytes from patients with Sézary syndrome were cultured with antihistamines, and the cell were analysed for proliferation, apoptosis and expression of programmed death molecules and transcription factors. The two antihistamines clemastine and desloratadine, currently used for symptom alleviation in allergy, induced potent reduction of the activities of the constitutively active transcription factors c-Myc, STAT3, STAT5a and STAT5b in mycosis fungoides and Sézary syndrome cell lines. This inhibition was followed by apoptosis and cell death, especially in the Sézary syndrome-derived cell line Hut78 that also showed increased expression of the programmed death-1 (PD-1) after clemastine treatment. In lymphocytes isolated from Sézary syndrome patients, the CD4-positive fraction underwent apoptosis after clemastine treatment, while CD4-negative lymphocytes were little affected. Because both c-Myc and STAT transcription factors are highly expressed in proliferating tumours, their inhibition by clemastine, desloratadine and other inhibitors could complement established chemotherapies not only for cutaneous T-cell lymphomas but perhaps also other cancers.


Apoptosis , Clemastine/pharmacology , Histamine Antagonists/pharmacology , Loratadine/analogs & derivatives , Lymphoma, T-Cell, Cutaneous/metabolism , Proto-Oncogene Proteins c-myc/metabolism , STAT3 Transcription Factor/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cytoplasm/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Loratadine/pharmacology
13.
Vaccine ; 31(7): 1057-64, 2013 Feb 04.
Article En | MEDLINE | ID: mdl-23273509

Vaccination with Mycobacterium bovis BCG provides limited protection against pulmonary tuberculosis and a risk of dissemination in immune-compromised vaccinees. For the development of new TB vaccines that stimulate strong T-cell responses a variety of strategies is being followed, especially recombinant BCG and attenuated M. tuberculosis. The objective of the current study was to test potential benefits of vaccination through direct lymph-node targeting of wildtype BCG; the recommended route of vaccination with BCG is intradermal. C57BL/6 mice were immunised with BCG by intradermal, subcutaneous or intralymphatic injections. Cellular immune responses and protection against M. tuberculosis were determined. Intralymphatic vaccination was 100-1000 times more effective in stimulating BCG-specific immune responses than intradermal or subcutaneous immunisation. Intralymphatic administration stimulated high frequencies of mycobacterium-specific lymphocytes with strong proliferating capacity and production of TNF-α, IL-2, IL-17 and, especially, IFN-γ secretion by. CD4 and CD8 T cells. Most importantly, intralymphatic vaccination with 2×10(3)CFU BCG induced sustained protection against M. tuberculosis in intratracheally challenged C57BL/6 mice, whereas subcutaneous vaccination with 2×10(5)CFU BCG conferred only a transient protection. Hence, direct administration of M. bovis BCG to lymph nodes demonstrates that efficient targeting to lymph nodes may help to overcome the efficacy problems of vaccination with BCG.


BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymph Nodes/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccination/methods , Animals , BCG Vaccine/administration & dosage , Cell Proliferation , Cytokines/metabolism , Disease Models, Animal , Female , Injections, Intradermal , Injections, Intralymphatic , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis, Pulmonary/immunology
14.
Blood ; 121(1): 85-94, 2013 Jan 03.
Article En | MEDLINE | ID: mdl-23144170

Immunoglobulin class switching from IgM to IgG in response to peptides is generally T cell-dependent and vaccination in T cell-deficient individuals is inefficient. We show that a vaccine consisting of a dense array of peptides on liposomes induced peptide-specific IgG responses totally independent of T-cell help. Independency was confirmed in mice lacking T cells and in mice deficient for MHC class II, CD40L, and CD28. The IgG titers were high, long-lived, and comparable with titers obtained in wild-type animals, and the antibody response was associated with germinal center formation, expression of activation-induced cytidine deaminase, and affinity maturation. The T cell-independent (TI) IgG response was strictly dependent on ligation of TLR4 receptors on B cells, and concomitant TLR4 and cognate B-cell receptor stimulation was required on a single-cell level. Surprisingly, the IgG class switch was mediated by TIR-domain-containing adapter inducing interferon-ß (TRIF), but not by MyD88. This study demonstrates that peptides can induce TI isotype switching when antigen and TLR ligand are assembled and appropriately presented directly to B lymphocytes. A TI vaccine could enable efficient prophylactic and therapeutic vaccination of patients with T-cell deficiencies and find application in diseases where induction of T-cell responses contraindicates vaccination, for example, in Alzheimer disease.


Adaptor Proteins, Vesicular Transport/physiology , Amyloid beta-Peptides/immunology , B-Lymphocytes/immunology , Immunoglobulin Class Switching/immunology , Peptide Fragments/immunology , Toll-Like Receptor 4/physiology , Vaccines, Subunit/immunology , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Adoptive Transfer , Amino Acid Sequence , Amyloid beta-Peptides/administration & dosage , Animals , Antigen Presentation , B-Lymphocytes/metabolism , CD28 Antigens/deficiency , CD28 Antigens/immunology , CD40 Ligand/deficiency , CD40 Ligand/immunology , Germinal Center/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Lipopolysaccharide Receptors/immunology , Liposomes , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Molecular Sequence Data , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Receptors, Antigen, B-Cell/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Vaccination , Vaccines, Subunit/administration & dosage
15.
J Allergy Clin Immunol ; 128(6): 1286-94, 2011 Dec.
Article En | MEDLINE | ID: mdl-21807405

BACKGROUND: Antihistamines are considered safe and used worldwide against allergy, pruritus, nausea, and cough and as sleeping aids. Nonetheless, a growing number of reports suggest that antihistamines also have immunoregulatory functions. OBJECTIVE: We examined the extent and by what potential mechanisms histamine-1-receptor (H1R) antagonists exert immune suppressive effects. METHODS: Immune suppression by antihistamines and immunosuppressants was tested in mice infected with Listeria monocytogenes. Potential modes of action were studied in vitro by using murine and human cells. We also tested whether injection of clemastine in healthy volunteers affected the activation of peripheral macrophages and monocytes. Finally, therapeutic application of clemastine-mediated immune suppression was tested in a murine model of sepsis. RESULTS: Clemastine and desloratadine strongly reduced innate responses to Listeria monocytogenes in mice as did dexamethasone. The immune suppression was MyD88 independent and characterized by inhibition of the mitogen-activated protein kinase-extracellular signal-regulated kinase signaling pathway, leading to overall impaired innate immunity with reduced TNF-α and IL-6 production. Surprisingly, the observed effects were H1R independent as demonstrated in H1R-deficient mice. Moreover, in a double-blind placebo-controlled clinical trial, 1 intravenous administration of clemastine reduced the TNF-α secretion potential of peripheral blood macrophages and monocytes. This inhibition could be exploited to treat sepsis in mice. CONCLUSIONS: The safety profile of antihistamines may need to be revisited. However, antihistamine-mediated immune suppression may also be exploited and find applications in the treatment of inflammatory diseases.


Clemastine/pharmacology , Cytokines/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Histamine H1 Antagonists/pharmacology , Immunity, Innate/drug effects , Animals , Cytokines/biosynthesis , Cytokines/immunology , Disease Susceptibility/immunology , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunoblotting , Immunohistochemistry , Immunosuppression Therapy/adverse effects , Listeriosis/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Real-Time Polymerase Chain Reaction
16.
Clin Vaccine Immunol ; 18(6): 907-13, 2011 Jun.
Article En | MEDLINE | ID: mdl-21471301

Pathogenic mycobacteria escape host innate immune responses by blocking phagosome-lysosome fusion. Avoiding lysosomal delivery may also be involved in the capacity of mycobacteria to evade major histocompatibility complex (MHC) class I- or II-dependent T-cell responses. In this study, we used a genetic mutant of Mycobacterium bovis BCG that is unable to escape lysosomal transfer and show that presentation of mycobacterial antigens is affected by the site of intracellular residence. Compared to infection with wild-type BCG, infection of murine bone marrow-derived dendritic cells with a mycobacterial mutant deficient in zinc metalloprotease 1 (Zmp1) resulted in increased presentation of MHC class II-restricted antigens, as assessed by activation of mycobacterial Ag85A-specific T-cell hybridomas. The zmp1 deletion mutant was more immunogenic in vivo, as measured by delayed-type hypersensitivity (DTH), antigen-specific lymphocyte proliferation, and the frequency of antigen-specific gamma interferon (IFN-γ)-producing lymphocytes of both CD4 and CD8 subsets. In conclusion, our results suggest that phagosome maturation and lysosomal delivery of BCG facilitate mycobacterial antigen presentation and enhance immunogenicity.


Antigen Presentation , Dendritic Cells/immunology , Lysosomes/microbiology , Metalloendopeptidases/metabolism , Mycobacterium bovis/immunology , Phagosomes/microbiology , Virulence Factors/metabolism , Animals , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Dendritic Cells/microbiology , Female , Gene Deletion , Humans , Interferon-gamma/metabolism , Metalloendopeptidases/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mycobacterium bovis/genetics , Virulence Factors/genetics
17.
Kidney Int ; 78(1): 38-47, 2010 Jul.
Article En | MEDLINE | ID: mdl-20393451

Renal proximal tubular epithelial cells, a target of infiltrating T cells during renal allograft rejection, may be protected from this injury by the cell surface protein CD274 (also termed PD-L1 for programmed death ligand 1). The co-inhibitory molecules PD-L1 (CD274) and PD-L2 (CD273) are ligands of PD-1 (programmed death 1; CD279). Here we determine the functional role of PD-1/PD-L pathways in human renal allograft rejection. Treatment of human primary tubular epithelial cells with interferon-beta and -gamma caused a dose-dependent and synergistic increase of PD-L1 and PD-L2 expression. Blockade of surface PD-L1, but not PD-L2, on interferon-treated tubular epithelial cells resulted in a significant increase in CD4+ T-cell proliferation and cytokine production by CD4+ and CD8+ T cells. The expression of PD-L1, PD-L2, and PD-1 mRNA and protein was upregulated in biopsies of patients with renal allograft rejection compared to the respective levels found in the pre-transplant biopsies. Induction of PD-L1 was significantly associated with acute vascular rejection. Our study suggests that the renal epithelial PD-1/PD-L1 pathway exerts an inhibitory effect of on alloreactive T-cell responses. The upregulation of PD-L1 on proximal tubular epithelial cells in patients with acute allograft rejection may reduce T-cell-mediated injury.


Antigens, CD/physiology , Kidney Tubules, Proximal/metabolism , T-Lymphocytes/immunology , B7-H1 Antigen , Cell Proliferation , Epithelial Cells/immunology , Epithelial Cells/metabolism , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism , Ligands , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Proteins/genetics , Proteins/immunology , Proteins/metabolism , T-Lymphocytes/metabolism , Up-Regulation
18.
Kidney Blood Press Res ; 30(6): 421-9, 2007.
Article En | MEDLINE | ID: mdl-17975322

BACKGROUND/AIMS: MHC molecules are upregulated on renal proximal tubular epithelial cells (TEC) under inflammatory conditions. This allows TEC to act as 'non-professional' antigen-presenting cells (APC). The aim of this study was to compare the costimulatory molecule expression pattern and the T cell activation capacity between renal TEC and professional APC, e.g. bone marrow-derived dendritic cells (BM-DC). METHODS: Flow cytometry analysis was used to study the costimulatory molecule surface expression on TEC or BM-DC. Ovalbumin-specific CD4 and CD8 T cell activation induced by TEC or BM-DC was compared, in terms of T cell proliferation, cytokine production and CTL activity. RESULTS: TEC did not constitutively express significant amounts of costimulatory molecules. Stimulation of TEC with IFN-beta or IFN-gamma, but not other tested cytokines, enhanced the expression of PD-L1, ICOS-L and CD40. Compared to BM-DC, TEC only induced suboptimal T cell activation. Blockade of PD-L1 on both APC strongly increased T cell activity. Furthermore, high PD-L1-expressing TEC were more resistant to the cytolysis by CTL. CONCLUSION: The low costimulatory molecule expression may explain the suboptimal T cell activation by TEC. The IFN-upregulated negative costimulatory molecule PD-L1 on TEC may play a protective role to limit tissue injury during renal parenchymal immune responses.


B7-1 Antigen/metabolism , CD40 Antigens/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Lymphocyte Activation/physiology , Membrane Glycoproteins/metabolism , Peptides/metabolism , Proteins/metabolism , T-Lymphocytes/physiology , Animals , B7-H1 Antigen , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Inducible T-Cell Co-Stimulator Ligand , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Kidney Tubules, Proximal/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/metabolism , T-Lymphocytes/immunology
19.
Nephron Exp Nephrol ; 107(1): e22-9, 2007.
Article En | MEDLINE | ID: mdl-17671397

BACKGROUND/AIM: TGF-beta expression is increased in immune-mediated and fibrotic renal diseases and modulates the tubulointerstitial T-cell response. We examined whether TGF-beta changes the expression of PD-L1 and CD40 in the renal proximal tubular epithelial cell (TEC), and whether the activation of CD8(+) cytotoxic T cells (CTLs) is influenced by TGF-beta treatment of TECs. METHODS: Murine TECs were treated with TGF-beta or IFN-gamma. The expression of PD-L1 and CD40 was examined with RT-PCR and flow cytometry. To investigate if TGF-beta treatment influenced the antigen presentation capacity of TECs, OT-1 CTLs were co-incubated with TGF-beta-treated, OVA(257-264) peptide-pulsed congeneic TECs. The cytotoxicity of OT-1 CTLs was estimated by their capacity to produce IFN-gamma and their cytolytic activity. RESULTS: TGF-beta treatment lead to a transition in morphology of renal TECs and downregulated the basal and the IFN-gamma-stimulated PD-L1 expression in TECs. Interestingly, TGF-beta treatment of TECs increased the constitutive and IFN-gamma-induced CD40 expression. In contrast to IFN-gamma which reduced the CTL activity, TGF-beta treatment of TECs elevated the OVA-specific CTL response. CONCLUSION: Our data show that TGF-beta changed the cellular phenotype and the expression of PD-L1 and CD40 on TECs and enhanced the activity of OVA peptide-specific CD8(+) T cells. TGF-beta may thereby play an important role in the progression of renal tubulointerstitial damage in CD8(+) T-cell-mediated renal diseases.


B7-1 Antigen/metabolism , CD40 Antigens/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Membrane Glycoproteins/metabolism , Ovalbumin/immunology , Peptides/metabolism , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta/pharmacology , Animals , B7-H1 Antigen , Cells, Cultured , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epitopes , Histocompatibility Antigens Class I/metabolism , Interferon-gamma/pharmacology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/immunology , Mice , Mice, Inbred Strains , Phenotype
20.
Nephrology (Carlton) ; 12(4): 357-63, 2007 Aug.
Article En | MEDLINE | ID: mdl-17635750

AIM: Autosomal dominant polycystic kidney disease (ADPKD) is characterized by an imbalance between tubular epithelial cell proliferation and apoptosis. We have previously shown that the mammalian target of rapamycin (mTOR) signalling pathway is aberrantly activated in the cystic kidneys of Han:SPRD rats with ADPKD. Because the Akt kinase is an upstream regulator of mTOR, we hypothesized that the activity of Akt could be enhanced in the kidneys of Han:SPRD rats. METHODS: Reverse transcription-polymerase chain reaction, western blot, enzyme-linked immunosorbent assay and immunohistochemistry were used to analyse Akt expression in rat polycystic kidneys. RESULTS: Wild-type (+/+) and heterozygous (Cy/+) Han:SPRD rats showed constitutive expression of Akt-1, -2 and -3 mRNA by reverse transcription-polymerase chain reaction analysis with no significant difference between Cy/+ and +/+ kidneys. Western blotting and enzyme-linked immunosorbent assay showed a significant increase in phosphorylated Akt in Cy/+ compared with +/+ kidneys. The pattern of immunoreactivity for phosphorylated Akt in kidney sections was the same in +/+ and in Cy/+ rats, with very low levels in interphase cells, but extremely bright signals in mitotic cells, beginning with the onset of the prophase. The in vivo incorporation of bromo-deoxyuridine revealed approximately a ninefold higher rate of proliferation in Cy/+ cyst epithelia compared with normal tubule epithelia in +/+ rats, while the expression of the cell cycle marker Ki67 revealed approximately a sixfold higher rate of proliferation. In summary, enhanced phosphorylation of Akt can be demonstrated in Cy/+ kidneys which correlates with a markedly elevated proliferation rate of epithelial cells in cysts. Mitotic but not resting cells display strong phosphorylation of Akt. CONCLUSION: Because Akt is a proximal target of mTOR, its inhibition with specific antagonists could be useful to prevent or halt cystogenesis in ADPKD.


Mitosis , Polycystic Kidney Diseases/genetics , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , Animals , Rats
...