1
|
Ma L, He Y, Xie H, Wang J, Chen J, Song S, Zhang L, Li L, Lai H, Liu Y, Chen H, Zhang X, Liu X, Zou Z, Zhang Q, Yan J, Tao A. Ferroptotic alveolar epithelial type II cells drive T H2 and T H17 mixed asthma triggered by birch pollen allergen Bet v 1. Cell Death Discov 2024; 10:96. [PMID: 38395974 PMCID: PMC10891108 DOI: 10.1038/s41420-024-01861-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Asthma is a common allergic disease characterized by airway hypersensitivity and airway remodeling. Ferroptosis is a regulated death marked by iron accumulation and lipid peroxidation. Several environmental pollutants and allergens have been shown to cause ferroptosis in epithelial cells, but the relationship between birch pollinosis and ferroptosis in asthma is poorly defined. Here, for the first time, we have identified ferroptosis of type II alveolar epithelial cells in mice with Bet v 1-induced asthma. Further analysis revealed that treatment with ferrostatin-1 reduced TH2/TH17-related inflammation and alleviated epithelial damage in mice with Bet v 1-induced asthma. In addition, ACSL4-knocked-down A549 cells are more resistant to Bet v 1-induced ferroptosis. Analysis of clinical samples verified higher serum MDA and 4-HNE concentrations compared to healthy individuals. We demonstrate that birch pollen allergen Bet v 1 induces ferroptosis underlaid TH2 and TH17 hybrid asthma. Lipid peroxidation levels can be considered as a biomarker of asthma severity, and treatment with a specific ferroptosis inhibitor could be a novel therapeutic strategy.
Collapse
Affiliation(s)
- Linyi Ma
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
- Department of Clinical Laboratory, General Hospital of the Yangtze River Shipping, Wuhan, 430005, China
| | - Ying He
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Huancheng Xie
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Jing Wang
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Jiaqian Chen
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Shijie Song
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Le Zhang
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Linmei Li
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - He Lai
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Yongping Liu
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Huifang Chen
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Xueyan Zhang
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Xueting Liu
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Zehong Zou
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Qingling Zhang
- Guangdong Provincial Key Laboratory of Allergy & Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jie Yan
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China.
| | - Ailin Tao
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China.
| |
Collapse
|
2
|
Akkoc T, O'Mahony L, Ferstl R, Akdis C, Akkoc T. Mouse Models of Asthma: Characteristics, Limitations and Future Perspectives on Clinical Translation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1376:119-133. [PMID: 34398449 DOI: 10.1007/5584_2021_654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Asthma is a complex and heterogeneous inflammatory airway disease primarily characterized by airway obstruction, which affects up to 15% of the population in Westernized countries with an increasing prevalence. Descriptive laboratory and clinical studies reveal that allergic asthma is due to an immunological inflammatory response and is significantly influenced by an individual's genetic background and environmental factors. Due to the limitations associated with human experiments and tissue isolation, direct mouse models of asthma provide important insights into the disease pathogenesis and in the discovery of novel therapeutics. A wide range of asthma models are currently available, and the correct model system for a given experimental question needs to be carefully chosen. Despite recent advances in the complexity of murine asthma models, for example humanized murine models and the use of clinically relevant allergens, the limitations of the murine system should always be acknowledged, and it remains to be seen if any single murine model can accurately replicate all the clinical features associated with human asthmatic disease.
Collapse
Affiliation(s)
- Tolga Akkoc
- Genetic Engineering and Biotechnology Institute, Tubitak Marmara Research Center, Kocaeli, Turkey.
| | - Liam O'Mahony
- Department of Medicine and Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ruth Ferstl
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Cezmi Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Davos, Switzerland
| | - Tunc Akkoc
- Department of Pediatric Allergy-Immunology, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
3
|
Sarate PJ, Srutkova D, Geissler N, Schwarzer M, Schabussova I, Inic-Kanada A, Kozakova H, Wiedermann U. Pre- and Neonatal Imprinting on Immunological Homeostasis and Epithelial Barrier Integrity by Escherichia coli Nissle 1917 Prevents Allergic Poly-Sensitization in Mice. Front Immunol 2021; 11:612775. [PMID: 33679699 PMCID: PMC7927790 DOI: 10.3389/fimmu.2020.612775] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/31/2020] [Indexed: 12/29/2022] Open
Abstract
A steady rise in the number of poly-sensitized patients has increased the demand for effective prophylactic strategies against multi-sensitivities. Probiotic bacteria have been successfully used in clinics and experimental models to prevent allergic mono-sensitization. In the present study, we have investigated whether probiotic bacteria could prevent poly-sensitization by imprinting on the immune system early in life. We used two recombinant variants of probiotic Escherichia coli Nissle 1917 (EcN): i) EcN expressing birch and grass pollen, poly-allergen chimera construct (EcN-Chim), and ii) an “empty” EcN without allergen expression (EcN-Ctrl). Conventional mice (CV) were treated with either EcN-Chim or EcN-Ctrl in the last week of the gestation and lactation period. Gnotobiotic mice received one oral dose of either EcN-Chim or EcN-Ctrl before mating. The offspring from both models underwent systemic allergic poly-sensitization and intranasal challenge with recombinant birch and grass pollen allergens (rBet v 1, rPhl p 1, and rPhl p 5). In the CV setting, the colonization of offspring via treatment of mothers reduced allergic airway inflammation (AAI) in offspring compared to poly-sensitized controls. Similarly, in a gnotobiotic model, AAI was reduced in EcN-Chim and EcN-Ctrl mono-colonized offspring. However, allergy prevention was more pronounced in the EcN-Ctrl mono-colonized offspring as compared to EcN-Chim. Mono-colonization with EcN-Ctrl was associated with a shift toward mixed Th1/Treg immune responses, increased expression of TLR2 and TLR4 in the lung, and maintained levels of zonulin-1 in lung epithelial cells as compared to GF poly-sensitized and EcN-Chim mono-colonized mice. This study is the first one to establish the model of allergic poly-sensitization in gnotobiotic mice. Using two different settings, gnotobiotic and conventional mice, we demonstrated that an early life intervention with the EcN without expressing an allergen is a powerful strategy to prevent poly-sensitization later in life.
Collapse
Affiliation(s)
- Priya J Sarate
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dagmar Srutkova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Nora Geissler
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Aleksandra Inic-Kanada
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Hana Kozakova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
E. coli Nissle 1917 is a safe mucosal delivery vector for a birch-grass pollen chimera to prevent allergic poly-sensitization. Mucosal Immunol 2019; 12:132-144. [PMID: 30242254 DOI: 10.1038/s41385-018-0084-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/27/2018] [Accepted: 08/25/2018] [Indexed: 02/04/2023]
Abstract
Allergic poly-sensitization affects a large number of allergic patients and poses a great challenge for their treatment. In this study we evaluated the effects of the probiotic Escherichia coli Nissle 1917 (EcN) expressing a birch and grass pollen allergen chimera 'Bet v 1, Phl p 1 and Phl p 5' (EcN-Chim) on allergy prevention after oral or intranasal application in poly-sensitized mice. In contrast to oral application, intranasal pretreatment with EcN-Chim prior to poly-sensitization led to a significant reduction of lung inflammation (eosinophils, IL-5, and IL-13 in bronchoalveolar lavage) along with suppressed levels of allergen-specific serum IgE. The suppression was associated with increased levels of allergen-specific IgA in lungs and serum IgG2a along with increased Foxp3, TGF-β, and IL-10 mRNA in bronchial lymph nodes. In vitro EcN induced high levels of IL-10 and IL-6 in both lung and intestinal epithelial cells. Importantly, using in vivo imaging techniques we demonstrated that intranasally applied EcN do not permanently colonize nose, lung, and gut and this strain might therefore be a safe delivery vector against allergy in humans. In conclusion, our data show that intranasal application of recombinant EcN expressing a multiallergen chimera presents a novel and promising treatment strategy for prevention of allergic poly-sensitization.
Collapse
|
5
|
Wagner A, Schabussova I, Drinic M, Akgün J, Loupal G, Kundi M, Joachim A, Wiedermann U. Oocyst-Derived Extract of Toxoplasma Gondii Serves as Potent Immunomodulator in a Mouse Model of Birch Pollen Allergy. PLoS One 2016; 11:e0155081. [PMID: 27149118 PMCID: PMC4857930 DOI: 10.1371/journal.pone.0155081] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/24/2016] [Indexed: 12/22/2022] Open
Abstract
Introduction Previously, we have shown that oral infection with Toxoplasma gondii oocysts prevented type I allergy in mice. Here we investigated whether the application of a T. gondii oocyst lysate antigen (OLA) could also reduce allergy development. BALB/c mice were immunised twice with OLA followed by sensitisation with the major birch pollen (BP) allergen Bet v 1 and an aerosol challenge with BP extract. Methods First, we tested OLA in vitro. Stimulation of splenocytes and bone marrow-derived dendritic cells (BMDC) with OLA led to the production of pro-inflammatory and regulatory cytokines such as IL-6, IFN-γ and IL-10. Moreover, BMDC exposed to OLA upregulated the maturation markers CD40, CD80, CD86, and MHCII. Furthermore, OLA was recognised by TLR2-transfected human embryonic kidney cells. Results Immunisation of mice with OLA induced high levels of Toxoplasma-specific IgG antibodies in sera along with increased production of IFN-γ and IL-10 in Toxoplasma-antigen restimulated splenocytes. OLA reduced allergic airway inflammation as manifested by significant reduction of eosinophils in bronchoalveolar fluids, decreased cellular infiltrates and mucus production in the lungs. Accordingly, Bet v 1-specific IgE was decreased in OLA-pretreated mice. The reduced allergic immune responses were accompanied by increased numbers of CD4+CD25highFoxp3+ regulatory T cells in spleens as well as by increased numbers of granulocytic myeloid-derived suppressor cells in lungs when compared to sensitised controls suggesting that these two cell populations might be involved in the suppression of the allergic immune responses. Conclusion Our data demonstrate that pretreatment with the oocyst extract can exert anti-allergic effects comparable to T. gondii infection. Thus, the immunomodulatory properties of the parasite extract indicate that this extract and in the future defined molecules thereof might serve as immunomodulatory adjuvants in allergy treatment and prophylaxis.
Collapse
Affiliation(s)
- Angelika Wagner
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Mirjana Drinic
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Johnnie Akgün
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerhard Loupal
- Institute of Pathology and Forensic Veterinary Medicine, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Michael Kundi
- Institute of Environmental Health, Center for Public Health, Medical University of Vienna, Vienna, Austria
| | - Anja Joachim
- Department of Pathobiology, Institute of Parasitology, University of Veterinary Medicine, Vienna, Austria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
6
|
Folded or Not? Tracking Bet v 1 Conformation in Recombinant Allergen Preparations. PLoS One 2015; 10:e0132956. [PMID: 26186356 PMCID: PMC4506129 DOI: 10.1371/journal.pone.0132956] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/20/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Recombinant Bet v 1a (rBet v 1a) has been used in allergy research for more than three decades, including clinical application of so-called hypoallergens. Quantitative IgE binding to rBet v 1a depends on its native protein conformation, which might be compromised upon heterologous expression, purification, or mutational engineering of rBet v 1a. OBJECTIVE To correlate experimental/theoretical comparisons of IgE binding of defined molar ratios of folded/misfolded recombinant Bet v 1a variants and to determine accuracy and precision of immuno- and physicochemical assays routinely used to assess the quality of recombinant allergen preparations. METHODS rBet v 1a and its misfolded variant rBet v 1aS112P/R145P were heterologously expressed and purified from Escherichia coli. Structural integrities and oligomerisation of the recombinant allergens were evaluated by 1H-nuclear magnetic resonance (1H-NMR), circular dichroism (CD) spectroscopy, and dynamic light scattering (DLS). IgE binding of defined combinations of rBet v 1a and rBet v 1aS112P/R145P was assessed using immunoblotting (IB), enzyme-linked immunosorbent assay (ELISA) and mediator release (MR) of humanized rat basophilic leukemia cells sensitized with serum IgE of subjects allergic to birch pollen. Experimental and theoretically expected results of the analyses were compared. RESULTS 1H-NMR spectra of rBet v 1a and rBet v 1aS112P/R145P demonstrate a native and highly disordered protein conformations, respectively. The CD spectra suggested typical alpha-helical and beta-sheet secondary structure content of rBet v 1a and random coil for rBet v 1aS112P/R145P. The hydrodynamic radii (RH) of 2.49 ± 0.39 nm (rBet v 1a) and 3.1 ± 0.56 nm (rBet v 1aS112P/R145P) showed monomeric dispersion of both allergens in solution. Serum IgE of birch pollen allergic subjects bound to 0.1% rBet v 1a in the presence of 99.9% of non-IgE binding rBet v 1aS112P/R145P. Immunoblot analysis overestimated, whereas ELISA and mediator release assay underestimated the actual quantity of IgE-reactive rBet v 1a in mixtures of rBet v 1a/rBet v 1aS112P/R145P with a molar ratio of rBet v 1a ≤ 10%. CONCLUSION Valid conclusions on quantitative IgE binding of recombinant Bet v 1a preparations depend on the accuracy and precision of physico- and immunochemical assays with which natively folded allergen is detected.
Collapse
|
7
|
Prime-boost vaccination with toxoplasma lysate antigen, but not with a mixture of recombinant protein antigens, leads to reduction of brain cyst formation in BALB/c mice. PLoS One 2015; 10:e0126334. [PMID: 26010355 PMCID: PMC4444244 DOI: 10.1371/journal.pone.0126334] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 03/24/2015] [Indexed: 01/01/2023] Open
Abstract
Introduction Infection with the ubiquitous parasite Toxoplasma gondii is a threat for immunocompromised patients and pregnant women and effective immune-prophylaxis is still lacking. Methods Here we tested a mixture of recombinant T. gondii antigens expressed in different developmental stages, i.e., SAG1, MAG1 and GRA7 (SMG), and a lysate derived from T. gondii tachyzoites (TLA) for prophylactic vaccination against cyst formation. Both vaccine formulations were applied systemically followed by an oral TLA-booster in BALB/c mice. Results Systemic priming with SMG and oral TLA-booster did not show significant induction of protective immune responses. In contrast, systemic priming and oral booster with TLA induced higher levels of Toxoplasma-specific IgG, IgG1 and IgG2a in sera as well as high levels of Toxoplasma-specific IgG1 in small intestines. Furthermore, high levels of Toxoplasma-specific Th1-, Th17- and Th2-associated cytokines were only detected in restimulated splenocytes of TLA-vaccinated mice. Importantly, in mice orally infected with T. gondii oocysts, only TLA-vaccination and booster reduced brain cysts. Furthermore, sera from these mice reduced tachyzoites invasion of Vero cells in vitro, indicating that antibodies may play a critical role for protection against Toxoplasma infection. Additionally, supernatants from splenocyte cultures of TLA-vaccinated mice containing high levels of IFN-γ lead to substantial production of nitric oxide (NO) after incubation with macrophages in vitro. Since NO is involved in the control of parasite growth, the high levels of IFN-γ induced by vaccination with TLA may contribute to the protection against T. gondii. Conclusion In conclusion, our data indicate that prime-boost approach with TLA, but not with the mixture of recombinant antigens SMG, induces effective humoral and cellular Toxoplasma-specific responses and leads to significant reduction of cerebral cysts, thereby presenting a viable strategy for further vaccine development against T. gondii infection.
Collapse
|
8
|
Akgün J, Schabussova I, Schwarzer M, Kozakova H, Kundi M, Wiedermann U. The Role of Alveolar Epithelial Type II-Like Cells in Uptake of Structurally Different Antigens and in Polarisation of Local Immune Responses. PLoS One 2015; 10:e0124777. [PMID: 25894334 PMCID: PMC4404363 DOI: 10.1371/journal.pone.0124777] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/05/2015] [Indexed: 01/17/2023] Open
Abstract
Background Our previous studies on intranasal tolerance induction demonstrated reduction of allergic responses with different allergen constructs. The underlying mechanisms varied depending on their conformation or size. Objective The aim of the present study was to compare the uptake of two structurally different allergen molecules within the respiratory tract following intranasal application. Methods The three-dimensional Bet v 1 (Bv1-Protein) and the T cell epitope peptide of Bet v 1 (Bv1-Peptide) were labelled with 5,6-Carboxyfluorescein (FAM) and their uptake was investigated in lung cells and cells of the nasal associated lymphoid tissue from naive and sensitised BALB/c mice. Phenotypic characterisation of FAM+ lung cells after antigen incubation in vitro and after intranasal application was performed by flow cytometry. Impact of Bv1-Protein and Bv1-Peptide on cytokine profiles and gene expression in vivo or in an alveolar epithelial type II (ATII) cell line were assessed in mono- and co-cultures with monocytes using ELISA and quantitative real-time PCR. Results Both antigens were taken up preferably by ATII-like cells (ATII-LCs) in naive mice, and by macrophages in sensitised mice. After intranasal application, Bv1-Peptide was taken up faster and more efficiently than Bv1-Protein. In vivo and in vitro experiments revealed that Bv1-Protein induced the transcription of thymic stromal lymphopoietin mRNA while Bv1-Peptide induced the transcription of IL-10 and MCP1 mRNA in ATII-LCs. Conclusion and Clinical Relevance Both tested antigens were taken up by ATII-LCs under steady state conditions and induced different polarisation of the immune responses. These data may have an important impact for the generation of novel and more effective prophylactic or therapeutic tools targeting the respiratory mucosa.
Collapse
Affiliation(s)
- Johnnie Akgün
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology, v.v.i., Academy of Sciences of the Czech Republic, Novy Hradek, Czech Republic
| | - Hana Kozakova
- Laboratory of Gnotobiology, Institute of Microbiology, v.v.i., Academy of Sciences of the Czech Republic, Novy Hradek, Czech Republic
| | - Michael Kundi
- Institute of Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Göteborg, Göteborg, Sweden
| |
Collapse
|
9
|
Ai C, Zhang Q, Ren C, Wang G, Liu X, Tian F, Zhao J, Zhang H, Chen YQ, Chen W. Protective effect of Streptococcus thermophilus CCFM218 against house dust mite allergy in a mouse model. Food Control 2015. [DOI: 10.1016/j.foodcont.2014.08.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
10
|
Xiao X, Zeng X, Zhang X, Ma L, Liu X, Yu H, Mei L, Liu Z. Effects of Caryota mitis profilin-loaded PLGA nanoparticles in a murine model of allergic asthma. Int J Nanomedicine 2013; 8:4553-62. [PMID: 24376349 PMCID: PMC3843607 DOI: 10.2147/ijn.s51633] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Pollen allergy is the most common allergic disease. However, tropical pollens, such as those of Palmae, have seldom been investigated compared with the specific immunotherapy studies done on hyperallergenic birch, olive, and ragweed pollens. Although poly(lactic-co-glycolic acid) (PLGA) has been extensively applied as a biodegradable polymer in medical devices, it has rarely been utilized as a vaccine adjuvant to prevent and treat allergic disease. In this study, we investigated the immunotherapeutic effects of recombinant Caryota mitis profilin (rCmP)-loaded PLGA nanoparticles and the underlying mechanisms involved. Methods A mouse model of allergenic asthma was established for specific immunotherapy using rCmP-loaded PLGA nanoparticles as the adjuvant. The model was evaluated by determining airway hyperresponsiveness and levels of serum-specific antibodies (IgE, IgG, and IgG2a) and cytokines, and observing histologic sections of lung tissue. Results The rCmP-loaded PLGA nanoparticles effectively inhibited generation of specific IgE and secretion of the Th2 cytokine interleukin-4, facilitated generation of specific IgG2a and secretion of the Th1 cytokine interferon-gamma, converted the Th2 response to Th1, and evidently alleviated allergic symptoms. Conclusion PLGA functions more appropriately as a specific immunotherapy adjuvant for allergen vaccines than does conventional Al(OH)3 due to its superior efficacy, longer potency, and markedly fewer side effects. The rCmP-loaded PLGA nanoparticles developed herein offer a promising avenue for specific immunotherapy in allergic asthma.
Collapse
Affiliation(s)
- Xiaojun Xiao
- Institute of allergy and Immunology, School of Medicine, Shenzhen University, Shenzhen, People's Republic of China
| | - Xiaowei Zeng
- Division of life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, People's Republic of China
| | - Xinxin Zhang
- Faculty of Basic Medical Science, Nanchang University, Nanchang, People's Republic of China
| | - Li Ma
- Faculty of Basic Medical Science, Nanchang University, Nanchang, People's Republic of China
| | - Xiaoyu Liu
- Institute of allergy and Immunology, School of Medicine, Shenzhen University, Shenzhen, People's Republic of China
| | - Haiqiong Yu
- Institute of allergy and Immunology, School of Medicine, Shenzhen University, Shenzhen, People's Republic of China
| | - Lin Mei
- Division of life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen, People's Republic of China
| | - Zhigang Liu
- Institute of allergy and Immunology, School of Medicine, Shenzhen University, Shenzhen, People's Republic of China
| |
Collapse
|
11
|
Tourdot S, Airouche S, Berjont N, Moussu H, Betbeder D, Nony E, Bordas-Le Floch V, Baron-Bodo V, Mascarell L, Moingeon P. Efficacy of sublingual vectorized recombinant Bet v 1a in a mouse model of birch pollen allergic asthma. Vaccine 2013; 31:2628-37. [PMID: 23583462 DOI: 10.1016/j.vaccine.2013.03.041] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 02/04/2013] [Accepted: 03/26/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Second generation sublingual allergy vaccines based upon recombinant allergens combined with vector systems are being developed as an alternative to conventional allergen extracts. Herein, we evaluated the efficacy of a recombinant form of the major allergen Bet v 1a (rBet v 1a) formulated as a mucoadhesive particle in a preclinical model of birch pollen (BP) respiratory allergy. MATERIALS AND METHODS BALB/c mice were sensitized to BP extracts by intraperitoneal injections followed by aerosol exposures. Sensitized mice underwent sublingual immunotherapy (SLIT) twice a week for eight weeks with either a BP extract or rBet v 1a formulated in amylopectin-based microparticles (MPA). SLIT efficacy was assessed using whole body plethysmography, lung histology and cell counts in broncho-alveolar lavages (BAL) as read outs. BP and/or rBet v 1a-specific T cell and antibody responses were monitored in lung and serum, respectively. IgA levels were measured in saliva. RESULTS Mice sensitized to BP exhibit chronic airway hyperresponsiveness (AHR), lung inflammation (documented by compliance and resistance measurements), eosinophil infiltrates in BAL, as well as Bet v 1-specific Th2 biased responses. Both SLIT with soluble rBet v 1a (50μg/dose) and BP extract (equivalent to 50μg rBet v 1 per dose) lead to a significant reduction in AHR, lung eosinophilia and Th2 responses. A sub-optimal dose of 5μg of rBet v 1a displays a similar level of efficacy with a significant decrease of Th2 responses when formulated with MPA microparticles. In addition, allergen vectorization with mucoadhesive particles allows a faster reduction in AHR in sensitized animals. CONCLUSION We demonstrate in a murine model of chronic BP respiratory allergy the efficacy of SLIT with vectorized rBet v 1a. Thus, combining recombinant allergens with mucoadhesive vector systems paves the ground for improved second generation sublingual allergy vaccines.
Collapse
Affiliation(s)
- Sophie Tourdot
- Research and Development, Stallergenes SA, Antony, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Schabussova I, Hufnagl K, Tang MLK, Hoflehner E, Wagner A, Loupal G, Nutten S, Zuercher A, Mercenier A, Wiedermann U. Perinatal maternal administration of Lactobacillus paracasei NCC 2461 prevents allergic inflammation in a mouse model of birch pollen allergy. PLoS One 2012; 7:e40271. [PMID: 22792257 PMCID: PMC3391241 DOI: 10.1371/journal.pone.0040271] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 06/04/2012] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The hygiene hypothesis implies that microbial agents including probiotic bacteria may modulate foetal/neonatal immune programming and hence offer effective strategies for primary allergy prevention; however their mechanisms of action are poorly understood. We investigated whether oral administration of Lactobacillus paracasei NCC 2461 to mothers during gestation/lactation can protect against airway inflammation in offspring in a mouse model of birch pollen allergy, and examined the immune mechanisms involved. METHODS BALB/c mice were treated daily with L. paracasei in drinking water or drinking water alone in the last week of gestation and during lactation. Their offspring were sensitized with recombinant Bet v 1, followed by aerosol challenge with birch pollen extract. RESULTS Maternal exposure to L. paracasei prevented the development of airway inflammation in offspring, as demonstrated by attenuation of eosinophil influx in the lungs; reduction of IL-5 levels in bronchoalveolar lavage, and in lung and mediastinal lymph node cell cultures; and reduced peribronchial inflammatory infiltrate and mucus hypersecretion. While allergen-specific IgE and IgG antibody levels remained unchanged by the treatment, IL-4 and IL-5 production in spleen cell cultures were significantly reduced upon allergen stimulation in offspring of L. paracasei treated mice. Offspring of L. paracasei supplemented mothers had significantly reduced Bet v 1-specific as well as Concanavalin A-induced responses in spleen and mesenteric lymph node cell cultures, suggesting the modulation of both antigen-specific and mitogen-induced immune responses in offspring. These effects were associated with increased Foxp3 mRNA expression in the lungs and increased TGF-beta in serum. CONCLUSION Our data show that in a mouse model of birch pollen allergy, perinatal administration of L. paracasei NCC 2461 to pregnant/lactating mothers protects against the development of airway inflammation in offspring by activating regulatory pathways, likely through TLR2/4 signalling.
Collapse
Affiliation(s)
- Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Karin Hufnagl
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Mimi L. K. Tang
- Department of Allergy and Immunology, Royal Children’s Hospital, The University of Melbourne, Melbourne, Australia
- Department of Allergy and Immune Disorders, Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, Royal Children’s Hospital, The University of Melbourne, Melbourne, Australia
| | - Elisabeth Hoflehner
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Angelika Wagner
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerhard Loupal
- Department of Pathobiology, Institute of Pathology and Forensic Veterinary Medicine, The University of Veterinary Medicine Vienna, Vienna, Austria
| | - Sophie Nutten
- Nutrition and Health Department, Nestlé Research Center, Lausanne, Switzerland
| | | | - Annick Mercenier
- Nutrition and Health Department, Nestlé Research Center, Lausanne, Switzerland
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Hoflehner E, Hufnagl K, Schabussova I, Jasinska J, Hoffmann-Sommergruber K, Bohle B, Maizels RM, Wiedermann U. Prevention of birch pollen-related food allergy by mucosal treatment with multi-allergen-chimers in mice. PLoS One 2012; 7:e39409. [PMID: 22768077 PMCID: PMC3387141 DOI: 10.1371/journal.pone.0039409] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 05/24/2012] [Indexed: 12/24/2022] Open
Abstract
Background Among birch pollen allergic patients up to 70% develop allergic reactions to Bet v 1-homologue food allergens such as Api g 1 (celery) or Dau c 1 (carrot), termed as birch pollen-related food allergy. In most cases, specific immunotherapy with birch pollen extracts does not reduce allergic symptoms to the homologue food allergens. We therefore genetically engineered a multi-allergen chimer and tested if mucosal treatment with this construct could represent a novel approach for prevention of birch pollen-related food allergy. Methodology BALB/c mice were poly-sensitized with a mixture of Bet v 1, Api g 1 and Dau c 1 followed by a sublingual challenge with carrot, celery and birch pollen extracts. For prevention of allergy sensitization an allergen chimer composed of immunodominant T cell epitopes of Api g 1 and Dau c 1 linked to the whole Bet v 1 allergen, was intranasally applied prior to sensitization. Results Intranasal pretreatment with the allergen chimer led to significantly decreased antigen-specific IgE-dependent β-hexosaminidase release, but enhanced allergen-specific IgG2a and IgA antibodies. Accordingly, IL-4 levels in spleen cell cultures and IL-5 levels in restimulated spleen and cervical lymph node cell cultures were markedly reduced, while IFN-γ levels were increased. Immunomodulation was associated with increased IL-10, TGF-β and Foxp3 mRNA levels in NALT and Foxp3 in oral mucosal tissues. Treatment with anti-TGF-β, anti-IL10R or anti-CD25 antibodies abrogated the suppression of allergic responses induced by the chimer. Conclusion Our results indicate that mucosal application of the allergen chimer led to decreased Th2 immune responses against Bet v 1 and its homologue food allergens Api g 1 and Dau c 1 by regulatory and Th1-biased immune responses. These data suggest that mucosal treatment with a multi-allergen vaccine could be a promising treatment strategy to prevent birch pollen-related food allergy.
Collapse
MESH Headings
- Administration, Intranasal
- Allergens/immunology
- Allergens/pharmacology
- Animals
- Antibodies, Blocking/pharmacology
- Antigens, Plant/immunology
- Basophils/drug effects
- Basophils/immunology
- Basophils/physiology
- Betula/immunology
- Cell Degranulation/drug effects
- Cytokines/biosynthesis
- Epitopes/immunology
- Female
- Food Hypersensitivity/immunology
- Food Hypersensitivity/pathology
- Food Hypersensitivity/prevention & control
- Gene Expression Regulation/drug effects
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Immunity, Humoral/drug effects
- Immunity, Humoral/immunology
- Immunization
- Immunoglobulin E/immunology
- Mice
- Mice, Inbred BALB C
- Nasal Mucosa/drug effects
- Nasal Mucosa/immunology
- Pollen/immunology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Recombinant Proteins/immunology
- Recombinant Proteins/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Elisabeth Hoflehner
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Karin Hufnagl
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Joanna Jasinska
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Karin Hoffmann-Sommergruber
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Rick M. Maizels
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
14
|
Broos S, Lundberg K, Akagi T, Kadowaki K, Akashi M, Greiff L, Borrebaeck CAK, Lindstedt M. Immunomodulatory nanoparticles as adjuvants and allergen-delivery system to human dendritic cells: Implications for specific immunotherapy. Vaccine 2010; 28:5075-85. [PMID: 20478343 DOI: 10.1016/j.vaccine.2010.05.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 04/27/2010] [Accepted: 05/03/2010] [Indexed: 01/29/2023]
Abstract
Novel adjuvants and antigen-delivery systems with immunomodulatory properties that shift the allergenic Th2 response towards a Th1 or regulatory T cell response are desired for allergen-specific immunotherapy. This study demonstrates that 200-nm sized biodegradable poly(gamma-glutamic acid) (gamma-PGA) nanoparticles (NPs) are activators of human monocyte-derived dendritic cells (MoDCs). Gamma-PGA NPs are efficiently internalized by immature MoDCs and strongly stimulate production of chemokines and inflammatory cytokines as well as up-regulation of co-stimulatory molecules and immunomodulatory mediators involved in efficient T cell priming. Furthermore, MoDCs from allergic subjects stimulated in vitro with a mixture of gamma-PGA NPs and extract of grass pollen allergen Phleum pratense (Phl p) augment allergen-specific IL-10 production and proliferation of autologous CD4(+) memory T cells. Thus, gamma-PGA NPs are promising as sophisticated adjuvants and allergen-delivery systems in allergen-specific immunotherapy.
Collapse
Affiliation(s)
- Sissela Broos
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Wagner A, Förster-Waldl E, Garner-Spitzer E, Schabussova I, Kundi M, Pollak A, Scheiner O, Joachim A, Wiedermann U. Immunoregulation by Toxoplasma gondii infection prevents allergic immune responses in mice. Int J Parasitol 2009; 39:465-72. [DOI: 10.1016/j.ijpara.2008.09.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 07/24/2008] [Accepted: 09/09/2008] [Indexed: 01/18/2023]
|
16
|
Repa A, Kozakova H, Hudcovic T, Stepankova R, Hrncir T, Tlaskalova-Hogenova H, Pollak A, Wiedermann U. Susceptibility to nasal and oral tolerance induction to the major birch pollen allergen Bet v 1 is not dependent on the presence of the microflora. Immunol Lett 2007; 117:50-6. [PMID: 18241932 DOI: 10.1016/j.imlet.2007.11.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Revised: 11/27/2007] [Accepted: 11/28/2007] [Indexed: 11/16/2022]
Abstract
The indigenous microflora plays an integrative role in the maintenance of immunological homeostasis. Several studies reported that immunological tolerance is dependent on microbial colonization of the gut. In the present study, we investigated whether the absence of the microflora influences the sensitization process to an allergen as well as the ability to develop mucosal tolerance in a mouse model of birch pollen allergy. Germ-free or conventional BALB/c mice were intranasally or intragastrically pre-treated with the major birch pollen allergen Bet v 1 prior to sensitization with this allergen. Both germ-free and conventional mice displayed comparable Th2 biased immune responses after allergic sensitization. Oral as well as intranasal tolerization led to suppression of allergen-specific serum antibodies (IgG1, IgE, IgA) as well as cytokine production by splenocytes (IL-5, IFN-gamma) in both germ-free and conventional animals. Peyer's patches of germ-free animals were approximately 20 times smaller than in conventional animals, but the relative distribution of lymphocyte subpopulations was equal. We conclude that the absence of the microflora does not influence the ability to mount Th2 responses nor to establish tolerance towards the aeroallergen Bet v 1. Our findings may challenge the view that the commensal microflora is a key factor for breakdown of physiological tolerance and allergy development.
Collapse
Affiliation(s)
- Andreas Repa
- Department of Specific Prophylaxis and Tropical Medicine, Center for Physiology, Pathophysiology & Immunology, Medical University of Vienna, Kinderspitalgasse 15, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Brimnes J, Kildsgaard J, Jacobi H, Lund K. Sublingual immunotherapy reduces allergic symptoms in a mouse model of rhinitis. Clin Exp Allergy 2007; 37:488-97. [PMID: 17430344 DOI: 10.1111/j.1365-2222.2006.02624.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Sublingual immunotherapy (SLIT) is a clinically effective treatment in both pollen and house dust mite-induced rhinitis and asthma. However, the mechanisms by which this is accomplished are not clear. OBJECTIVE The objective of the current study was to establish a mouse model of rhinitis in order to study the effect and mechanisms of SLIT. METHODS Mice were sensitized by intraperitoneal injections of alum-adsorbed Phleum pratense extract. Sensitized mice were SLIT-treated and subsequently challenged intranasally and analysed for clinical symptoms, antibody levels, eosinophilia and T cell response. RESULTS Intranasal challenge of sensitized mice led to the development of rhinitis characterized by significantly increased sneezing and influx of eosinophils into the nose. Levels of specific IgE were fivefold increased in nasopharyngeal lavage (NAL) fluid and more than doubled in serum. Furthermore, a T-helper type 2 (Th2) like T cell response was observed in local draining lymph nodes. SLIT treatment of sensitized mice reduced sneezing, eosinophilia and IgE levels in the NAL by more than 50%. Moreover, serum levels of IgE and IgG1 as well as T cell response in the draining lymph nodes were also significantly reduced. Treatment for a shorter time or with a lower dose only led to minor reductions of the clinical and immunological parameters, indicating that the effect of SLIT is time and dose dependent. CONCLUSION In the present study, we have established a mouse model displaying the hallmarks of allergic rhinitis using a clinically relevant allergen. Using this model, we have demonstrated that SLIT treatment is able to reduce allergic symptoms in a time- and dose-dependent manner.
Collapse
Affiliation(s)
- J Brimnes
- Department of Experimental Immunology, ALK-Abelló A/S, Hoersholm, Denmark.
| | | | | | | |
Collapse
|
18
|
|
19
|
Wagner S, Jasinska J, Breiteneder H, Kundi M, Pehamberger H, Scheiner O, Zielinski CC, Wiedermann U. Delayed tumor onset and reduced tumor growth progression after immunization with a Her-2/neu multi-peptide vaccine and IL-12 in c-neu transgenic mice. Breast Cancer Res Treat 2007; 106:29-38. [PMID: 17203384 DOI: 10.1007/s10549-006-9469-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Accepted: 11/25/2006] [Indexed: 11/28/2022]
Abstract
Passive immunotherapy with monoclonal antibodies is a routinely performed but cost intensive treatment against certain cancers. Induction of humoral anti-tumor responses by active peptide immunization has therefore become a favorable treatment concept. We have recently identified three peptides representing B-cell epitopes of the extracellular domain of Her-2/neu each of them inducing Her-2/neu specific immune responses with anti-tumor activity in vitro. The present study was performed to evaluate the in vivo protective capacity of a combined vaccination with these three peptides in FVB/N transgenic mice spontaneously developing c-neu overexpressing breast cancers. The three Her-2/neu peptides coupled to tetanus toxoid were administered with or without addition of recombinant IL-12. At the time all untreated mice had developed tumors about 40% of peptide-immunized mice and nearly 60% of mice immunized with the peptide vaccine co-applied with IL-12 remained tumor free. Moreover, co-administration of IL-12 had a significant impact on the retardation of tumor progression. The enhanced anti-tumor efficacy of the vaccine by IL-12 was associated with a Th1 biased immune response as demonstrated by an increased IFN-gamma production in vitro and elevated Her-2-specific IgG levels. Our findings clearly demonstrate that this multi-peptide vaccine is effective in tumor prevention and support its use against minimal disease, drug-resistant tumors or even for prophylaxis against cancers overexpressing Her-2/neu.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/therapeutic use
- Animals
- Antibodies/blood
- Antibodies/metabolism
- Antibody Formation
- Antibody Specificity
- Cancer Vaccines/pharmacology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Epitopes, B-Lymphocyte/immunology
- Female
- Glycoproteins/genetics
- Glycoproteins/immunology
- Glycoproteins/metabolism
- Humans
- Immunoglobulin G/blood
- Interferon-gamma/blood
- Interleukin-12/pharmacology
- Interleukin-12/therapeutic use
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Rats
- Receptor, ErbB-2/immunology
- Recombinant Proteins/pharmacology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Time Factors
- Vaccines, Subunit/pharmacology
- Vaccines, Subunit/therapeutic use
Collapse
Affiliation(s)
- Stefan Wagner
- Department of Pathophysiology, Center for Physiology and Pathophysiology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Cortes-Perez NG, Ah-Leung S, Bermúdez-Humarán LG, Corthier G, Wal JM, Langella P, Adel-Patient K. Intranasal coadministration of live lactococci producing interleukin-12 and a major cow's milk allergen inhibits allergic reaction in mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2007; 14:226-33. [PMID: 17202306 PMCID: PMC1828845 DOI: 10.1128/cvi.00299-06] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The Th1/Th2 balance deregulation toward a Th2 immune response plays a central role in allergy. We previously demonstrated that administration of recombinant Lactococcus lactis strains expressing bovine beta-lactoglobulin (BLG), a major cow's milk allergen, partially prevents mice from sensitization. In the present study, we aimed to improve this preventive effect by coadministration of L. lactis BLG and a second recombinant L. lactis strain producing biologically active interleukin-12 (IL-12). This L. lactis strain producing IL-12 was previously used to enhance the Th1 immune response in a tumoral murine model (L. G. Bermúdez-Humarán et al., J. Immunol. 175:7297-7302, 2005). A comparison of the administration of either BLG alone or BLG in the presence of IL-12 was conducted. A BLG-specific primary Th1 immune response was observed only after intranasal coadministration of both L. lactis BLG and IL-12-producing L. lactis, as demonstrated by the induction of serum-specific immunoglobulin G2a (IgG2a) concomitant with gamma interferon secretion by splenocytes, confirming the adjuvanticity of IL-12-producing L. lactis. Immunized mice were further sensitized by intraperitoneal administration of purified BLG, and the allergic reaction was elicited by intranasal challenge with purified BLG. Mice pretreated with BLG in either the presence or the absence of IL-12 were rendered completely tolerant to further allergic sensitization and elicitation. Pretreatment with either L. lactis BLG or L. lactis BLG and IL-12-producing L. lactis induces specific anti-BLG IgG2a production in serum and bronchoalveolar lavage (BAL) fluid. Although specific serum IgE was not affected by these pretreatments, the levels of eosinophilia and IL-5 secretion in BAL fluid were significantly reduced after BLG challenge in the groups pretreated with L. lactis BLG and L. lactis BLG-IL-12-producing L. lactis, demonstrating a decreased allergic reaction. Our data demonstrate for the first time (i) the induction of a protective Th1 response by the association of L. lactis BLG and IL-12-producing L. lactis which inhibits the elicitation of the allergic reaction to BLG in mice and (ii) the efficiency of intranasal administration of BLG for the induction of tolerance.
Collapse
Affiliation(s)
- Naima G Cortes-Perez
- Laboratoire INRA d'Immuno-Allergie Alimentaire, DSV/SPI-Bâtiment 136, CEA de Saclay, 91191 Gif-sur-Yvette cedex, France
| | | | | | | | | | | | | |
Collapse
|
21
|
Wild C, Wallner M, Hufnagl K, Fuchs H, Hoffmann-Sommergruber K, Breiteneder H, Scheiner O, Ferreira F, Wiedermann U. A recombinant allergen chimer as novel mucosal vaccine candidate for prevention of multi-sensitivities. Allergy 2007; 62:33-41. [PMID: 17156339 DOI: 10.1111/j.1398-9995.2006.01245.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND As conventional immunotherapy is less efficacious in patients with allergic multi-sensitivities compared with mono-sensitized subjects, new intervention strategies are needed. Therefore, an allergen chimer was genetically engineered for treatment of multi-sensitization with birch and grass pollen on the basis of mucosal tolerance induction. METHODS The major birch pollen allergen Bet v 1 served as a scaffold for N- and C-terminal linkage of the immunodominant peptides of the grass pollen allergens Phl p 1 and Phl p 5 and this new construct was cloned and expressed in Escherichia coli. After purification, physicochemical and immunological characterization the chimer was used for intranasal tolerance induction prior to poly-sensitization with Bet v 1, Phl p 1 and Phl p 5. RESULTS The immunological characterization revealed that the conformation of Bet v 1 within the chimer was comparable to that of natural as well as recombinant Bet v 1. The chimer was immunogenic in mice for T and B cell responses to the three allergens. Intranasal application of the chimer prior to poly-sensitization significantly suppressed humoral and cellular allergen-specific Th2 responses and prevented development of airway inflammation upon allergen challenge. Moreover, local allergen-specific IgA antibodies were induced by the chimer. The mechanisms of poly-tolerance induction seemed to be mediated by regulatory cytokines, since TGF-beta and IL-10 mRNA in splenocytes were upregulated and tolerance was transferable with these cells. CONCLUSION The data indicate that such allergen chimers harboring several unrelated allergens or allergen peptides could serve as mucosal polyvalent vaccines for prevention of multi-sensitivities.
Collapse
Affiliation(s)
- C Wild
- Department of Specific Prophylaxis and Tropical Medicine, Center for Physiology and Pathophysiology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Tournoy KG, Hove C, Grooten J, Moerloose K, Brusselle GG, Joos GF. Animal models of allergen-induced tolerance in asthma: are T-regulatory-1 cells (Tr-1) the solution for T-helper-2 cells (Th-2) in asthma? Clin Exp Allergy 2006; 36:8-20. [PMID: 16393260 DOI: 10.1111/j.1365-2222.2005.02385.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Non-specific anti-inflammatory medication is actually the treatment of choice for controlling the T-helper type 2 (Th-2) cell-driven airway inflammation in asthma. The induction of counterbalancing Th-1 cell clones, long considered a promising approach for immunotherapy, has failed to fulfil its promise because of potentially detrimental side-effects. This is therefore probably not a valid option for the treatment of asthma. With the increasing awareness that active immune mechanisms exist to control inflammatory responses, interest rises to investigate whether these can be exploited to control allergen-induced airway disease. The induction of antigen-specific T cells with suppressive characteristics (regulatory T cells) is therefore a potentially interesting approach. These regulatory T cells mediate tolerance in healthy, non-atopic individuals and have the potential of becoming an effective means of preventing allergen-induced airway inflammation and possibly of suppressing ongoing allergic immune responses. Here we review the available knowledge about allergen-induced suppressive immunity obtained from animal models taking into account the different developmental stages of allergic airway disease.
Collapse
Affiliation(s)
- K G Tournoy
- Department of Respiratory Diseases, Ghent University Hospital, Ghent University, Flanders Interuniversity Institute for Biotechnology, Ghent, Belgium.
| | | | | | | | | | | |
Collapse
|
23
|
Palomares O, Batanero E, Cañamero M, Villalba M, Rodríguez R. Prophylactic Intranasal Treatment with Fragments of 1,3-β-Glucanase Olive Pollen Allergen Prevents Airway Inflammation in a Murine Model of Type I Allergy. Int Arch Allergy Immunol 2006; 139:175-80. [PMID: 16439855 DOI: 10.1159/000091162] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Accepted: 10/19/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Olive pollen is an important cause of allergy in Mediterranean countries. More than 50% of olive-pollen-allergic patients are sensitized against the 1,3-beta-glucanase Ole e 9. To date, prophylactic and therapeutic treatments using purified recombinant allergens have not been studied in animal models of olive pollen allergy. METHODS BALB/c mice were immunized against Ole e 9 combining intraperitoneal injections of the allergen in Al(OH)3 with airway allergen challenges. A prophylactic treatment was performed by intranasal administration of a mixture of the recombinant fragments of the allergen prior to Ole e 9 sensitization. Serum levels of specific IgE, IgG1, IgG2a and IgG2b were measured by ELISA, and total IgE levels by sandwich ELISA. Bronchoalveolar lavage and lungs from mice were collected to study airway inflammation by light microscopy. RESULTS BALB/c mice immunized against Ole e 9 developed a predominantly Th2-like immune response with allergen-specific immunoglobulin induction and airway inflammation accompanied by the infiltration of eosinophils, lymphocytes, and neutrophils in the lung. Prophylactic treatment by intranasal application of the recombinant fragments of Ole e 9 avoids airway inflammation induced by sensitization with this allergen although the levels of Ole e 9-specific antibodies remain unchanged. CONCLUSIONS Prophylactic intranasal treatment with recombinant fragments of Ole e 9 prevents airway inflammation triggered by immunization to this allergen in a murine model of type I allergy.
Collapse
MESH Headings
- Administration, Intranasal
- Allergens/administration & dosage
- Allergens/immunology
- Animals
- Antigens, Plant
- Bronchoalveolar Lavage Fluid/cytology
- Disease Models, Animal
- Female
- Immunization/methods
- Immunoglobulin E/blood
- Immunoglobulin G/blood
- Immunoglobulin Isotypes/blood
- Lung/pathology
- Mice
- Mice, Inbred BALB C
- Olea/immunology
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Plant Proteins/administration & dosage
- Plant Proteins/immunology
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/immunology
- Rhinitis, Allergic, Seasonal/immunology
- Rhinitis, Allergic, Seasonal/pathology
- Rhinitis, Allergic, Seasonal/prevention & control
- beta-Glucosidase/administration & dosage
- beta-Glucosidase/immunology
Collapse
Affiliation(s)
- O Palomares
- Departamento de Bioquímica y Biología Molecular, Facultad de Química, Universidad Complutense de Madrid, and Departamento de Anatomía Patológica, Hospital Ruber Internacional, Spain
| | | | | | | | | |
Collapse
|
24
|
Winkler B, Hufnagl K, Spittler A, Ploder M, Kállay E, Vrtala S, Valenta R, Kundi M, Renz H, Wiedermann U. The role of Foxp3+ T cells in long-term efficacy of prophylactic and therapeutic mucosal tolerance induction in mice. Allergy 2006; 61:173-80. [PMID: 16409192 DOI: 10.1111/j.1398-9995.2006.01014.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Mucosal tolerance induction is suggested as treatment strategy for allergic diseases. Using a murine model of birch pollen (BP) allergy we investigated the long-term efficacy and the underlying mechanisms of mucosal tolerance induction with two structurally different molecules in a prophylactic and in a therapeutic set-up. METHODS The three-dimensional major BP allergen Bet v 1 or a nonconformational hypoallergenic fragment thereof was intranasally applied before (prophylaxis) or after sensitization (therapy). RESULTS In the prophylactic application both the Bet v 1 allergen and the fragment prevented allergic sensitization, and this effect lasted for 1 year. In the therapeutic approach established allergic immune responses were also suppressed after treatment with either of the molecules. However, a long-lasting curative effect (6 months) was only achieved with the Bet v 1 allergen but not with the Bet v 1 fragment. Real-time reverse transcriptase polymerase chain reaction (RT-PCR) analysis of splenocytes revealed that tolerance induction with the Bet v 1 allergen was associated with enhanced expression of transforming growth factor (TGF)-beta, interleukin (IL)-10, and Foxp3 mRNA in CD4+ T cells, whereas treatment with the fragment led to the induction of either Foxp3 (prophylaxis) or IL-10 (therapy) alone. CONCLUSION From these data we concluded (i) that the mechanisms underlying peripheral tolerance are linked to the conformation of the antigen, (ii) that mucosal tolerance is mediated by separate regulatory cell subsets, and (iii) that the long-term efficacy of immunosuppression is associated with the presence of Foxp3+ T cells.
Collapse
Affiliation(s)
- B Winkler
- Department of Specific Prophylaxis and Tropical Medicine, Center for Physiology and Pathophysiology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Liu YH, Tsai JJ. Production of salivary immunoglobulin A and suppression of Dermatophagoides pteronyssinus-induced airway inflammation by local nasal immunotherapy. Int Arch Allergy Immunol 2005; 138:161-8. [PMID: 16192741 DOI: 10.1159/000088438] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Accepted: 04/29/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Local nasal immunotherapy (LNIT) is an effective immunotherapeutic modality, especially when targeting a single immunodominant peptide from an allergen. However, the working mechanisms of LNIT are poorly understood. We hypothesized that LNIT with a mixture of group 2 allergens of Dermatophagoides pteronyssinus (Der p 2) protein and fungal immunomodulatory peptide (FIP) would generate suppression of Der p-induced airway inflammation through immunoglobulin (Ig) A secretion in the airways. METHOD Mice were sensitized with recombinant Der p 2 (rDer p 2) and Der p followed by LNIT with rDer p 2 in conjunction with FIP. After intratracheal challenge with rDer p 2 and Der p, the airway inflammation was determined by analyzing the cell subpopulation and cytokine concentration in the bronchoalveolar lavage fluid. The allergen-specific IgE, IgG2a and IgG in the sera and IgA in the saliva were measured by ELISA. RESULTS LNIT with rDer p 2 in conjunction with FIP could downregulate the lymphocyte infiltration in both rDer p 2- and Der p-induced airway inflammation. Both total and specific IgA in the saliva were increased after LNIT. Serum levels of IL-4, IL-10 and specific IgE were reduced and the specific IgG2a and IgG increased after LNIT. After LNIT, there was a reduction of airway hypersensitivity at 30 min after allergen challenge in the rDer p 2-and Der p-sensitized mice, with a significant decrease only in rDer p 2-sensitized mice. CONCLUSION LNIT with rDer p 2 in conjunction with FIP was not only able to suppress rDer p 2-induced airway inflammation but also generate suppression of Der p-induced airway inflammation. The simultaneous reduction of IL-4 and IL-10 indicated that IL-10-producing cells were not activated by LNIT. The increment of IgA in the airway might play a role in the prevention of airway inflammation.
Collapse
Affiliation(s)
- Yi-Hsia Liu
- Section of Allergy and Immunology, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| | | |
Collapse
|
26
|
McCusker CT. Use of mouse models of allergic rhinitis to study the upper and lower airway link. Curr Opin Allergy Clin Immunol 2004; 4:11-6. [PMID: 15090913 DOI: 10.1097/00130832-200402000-00004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Allergic rhinitis and asthma are examples of a continuum of airway diseases with diverse clinical manifestations. This review examines the most recent work in mouse models studying upper and lower airway links and interactions. RECENT FINDINGS The concept of united airways has been supported by investigative and epidemiological studies. Studies using mouse models of asthma and models of allergic rhinitis have demonstrated that analogous pathways lead to inflammation and airway hyperresponsiveness. Th2-type T cells and IL-13 play important immunopathologic roles. Recent studies have examined upper airway mucosal immune responses and development of both allergic and tolerant phenotypes. In a model of allergic airways disease, there is evidence of lower airway inflammation and airways hyperresponsiveness following application of allergen only to the nares, suggesting local stimulation can activate distal allergic responses. Immunomodulatory properties of the airway mucosa have also been explored. Allergen-specific tolerance can be induced by appropriate stimulation of airway mucosa and is associated with activation of IL-10-producing T cells. This effect is mediated by antigen presenting cells, especially dendritic cells. SUMMARY Immune stimulation of the airway mucosa, both in the upper and lower airways, results in active T-cell-mediated immune responses leading toward tolerance or asthma and allergic rhinitis. Regulation of these T-cell responses is currently under investigation. It is clear from these studies that antigenic stimulation of any part of the respiratory mucosa can have ripple effects along the entire airway and supports the concept of united airways.
Collapse
Affiliation(s)
- Christine T McCusker
- Division of Allergy and Immunology, Montreal Children's Hospital and Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
27
|
Herz U, Renz H, Wiedermann U. Animal models of type I allergy using recombinant allergens. Methods 2004; 32:271-80. [PMID: 14962762 DOI: 10.1016/j.ymeth.2003.08.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2003] [Indexed: 11/17/2022] Open
Abstract
Various animal models including guinea pigs, monkeys, dogs, rats, and mice have been established in an attempt to provide insights into the complex immunological and pathophysiological mechanisms of human type I allergic diseases. The detailed knowledge of the murine genome, the various components of the murine immune system, and the generation of engineered mice has made the murine system the most attractive among all animal models. The availability of multitude technologies and reagents to characterize and manipulate immunological pathways and mediators adds to the outstanding opportunities to assess the pathology of allergic diseases and to develop novel therapeutic strategies in mice. Numerous sensitization protocols with food and aero-allergens are used to establish an allergic/asthma-like phenotype in mice. Requirements for an appropriate murine model include a close resemblance to the pathology of the disease in humans, the objective measurement of the physiologic parameters, as well as reliability and reproducibility of the experimental data. With respect to reproducible experimental conditions, it has been recognized that extract preparations from natural allergen sources can vary in their allergen-content and -composition. This might influence the degree of sensitization or the outcome of treatment strategies in dependence of the applied extract preparation. The use of recombinant allergens in experimental in vivo and in vitro systems can overcome these problems. Another aspect, that has become obvious from the experimental studies, is that allergens can differ in their immunogenicity as well as in their capacity to act as tolerogens. Therefore, it seems important that the efficacy of the different allergen-molecules to act as therapeutic agents is individually examined. In this review, examples of animal models are described, in which recombinant allergens have been used for sensitization and/or treatment of allergic responses and how they have been used to enhance our understanding of the pathology of allergic diseases.
Collapse
Affiliation(s)
- Udo Herz
- Department of Clinical Chemistry and Molecular Diagnostics, Hospital of the Philipps-University, Marburg, Germany
| | | | | |
Collapse
|
28
|
Repa A, Wild C, Hufnagl K, Winkler B, Bohle B, Pollak A, Wiedermann U. Influence of the route of sensitization on local and systemic immune responses in a murine model of type I allergy. Clin Exp Immunol 2004; 137:12-8. [PMID: 15196238 PMCID: PMC1809092 DOI: 10.1111/j.1365-2249.2004.02519.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The pathophysiological and immunological characteristics of allergic immune responses are controlled by a variety of factors. We have studied the extent to which the route of sensitization influences allergen-specific IgE synthesis and local airway inflammation using a mouse model of allergic sensitization to the major birch pollen allergen Bet v 1. Sensitization of BALB/c mice with recombinant (r)Bet v 1 was performed using intraperitoneal (i.p.), subcutaneous (s.c.) or aerosol (a.s.) sensitization protocols. Mice were analysed for allergen-specific serum antibodies by ELISA and IgE-dependent basophil degranulation. Proliferative responses and cytokine production of splenocytes were measured upon Bet v 1 stimulation in vitro. Bronchoalveolar lavages were performed after airway challenge with aerosolized birch pollen extract for assessment of eosinophilic airway inflammation and local cytokine production in vivo. Highest allergen specific IgE levels and IgE-dependent basophil degranulation were achieved using the SC route. High IL-5 production by spleen and lung cells was associated with pronounced eosinophilia in bronchoalveolar lavages. After i.p. sensitization, despite giving the highest IgG levels, only low IgE levels, basophil degranulation and IL-5 production were seen. On the other hand, a.s. sensitization, resulting in the lowest systemic IgE and IL-5 levels, led to a comparably strong airway inflammation as the s.c. route. Our finding that the route of sensitization can result in a dissociation of local and systemic immune responses may contribute to a better understanding of the pathogenesis of allergic diseases and help to develop new treatment strategies.
Collapse
Affiliation(s)
- A Repa
- Department of Pathophysiology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
29
|
Wagner B, Hufnagl K, Radauer C, Wagner S, Baier K, Scheiner O, Wiedermann U, Breiteneder H. Expression of the B subunit of the heat-labile enterotoxin of Escherichia coli in tobacco mosaic virus-infected Nicotiana benthamiana plants and its characterization as mucosal immunogen and adjuvant. J Immunol Methods 2004; 287:203-15. [PMID: 15099768 DOI: 10.1016/j.jim.2004.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2003] [Revised: 01/26/2004] [Accepted: 02/02/2004] [Indexed: 11/20/2022]
Abstract
We have produced biologically active recombinant (r) LTB, the nontoxic B subunit of heat-labile toxin (LT) of Escherichia coli in tobacco mosaic virus (TMV)-infected Nicotiana benthamiana plants. We amplified the LTB encoding sequence with its leader and introduced a hexahistidyl tag and an endoplasmic reticulum retention signal. The resulting product was ligated into a TMV-based plant viral expression vector that was used for the generation of recombinant viral RNA. Eighty-nine percent of N. benthamiana plants inoculated with the recombinant viral RNA were systemically infected as determined by anti-TMV enzyme-linked immunosorbent assay (ELISA) experiments. The rLTB monomer was identified by LT-specific as well as by histidyl-tag-specific immunoblots. rLTB from plant extracts of TMV-infected N. benthamiana leaves was purified to give 75 microg rLTB pentamers per gram fresh plant material and was capable of binding G(M)1 ganglioside. The immunogenicity of the plant-produced rLTB was tested in mice and showed that intranasal application of rLTB (15 microg per mouse) induced LTB-specific IgG1 antibodies. To prove its adjuvanticity, rLTB was intranasally co-administered with the Hevea latex allergen Hev b 3, leading to allergen-specific IgG1 and IgG2a antibody production. The fact that intranasal application of rLTB and Hev b 3 prior to systemic challenge with the allergen enhanced the Th2 responses at the humoral and cellular level indicated that rLTB promoted immune responses that were naturally induced by the antigen/allergen. In conclusion, these results indicate that the plant viral expression system is suitable for the rapid large-scale production of biologically active LTB with strong mucosal adjuvant capacity.
Collapse
Affiliation(s)
- Birgit Wagner
- Department of Pathophysiology, Medical University of Vienna, General Hospital Vienna EBO 3.Q, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Winkler B, Bolwig C, Seppälä U, Spangfort MD, Ebner C, Wiedermann U. Allergen-specific immunosuppression by mucosal treatment with recombinant Ves v 5, a major allergen of Vespula vulgaris venom, in a murine model of wasp venom allergy. Immunology 2003; 110:376-85. [PMID: 14632666 PMCID: PMC1783061 DOI: 10.1046/j.1365-2567.2003.01751.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Up to 5% of the population suffer from systemic, 19% from local allergic hypersensitivity reactions to stinging insects. Even though specific immunotherapy is very effective in treating allergy to insect venom, new concepts of treatment strategies with only the disease eliciting allergen in recombinant form, along with antigen application via a less invasive route might be suggested for enhanced treatment efficacy and compliance. In the present study we aimed (i) to establish a mouse model of wasp venom allergy, mimicking the natural mode of sensitization, and (ii) to develop a prophylactic treatment strategy based on mucosal tolerance induction, using one major wasp venom allergen in recombinant form, i.e. recombinant (r)Ves v 5. Immunization with wasp venom--with or without the use of the adjuvant aluminium hydroxide--led to comparable T helper 2-like immune responses in vivo and in vitro. Intranasal administration of rVes v 5 prior to sensitization with wasp venom resulted in a significant reduction of wasp venom-specific antibody levels (immunoglobulin E (IgE)/IgG2a), type I hypersensitivity reactions in vivo and cytokine production in vitro. Pretreatment with the whole venom was less effective and caused toxic side reactions in higher concentrations, suggesting a favourable use of the recombinant venom allergen for mucosal application. Increased mRNA levels of transforming growth factor-beta and interleukin-10, along with adoptive cell transfer experiments indicated that the immunosuppression after intranasal rVes v 5-application has been mediated by regulatory mechanisms. This is further supported by the fact that the immunosuppression to rVes v 5 was associated with a bystander suppression to the unrelated aero-allergen Bet v 1. In conclusion, we demonstrated that the intranasal application of recombinant Ves v 5 prevented subsequent allergic sensitization to all components of the whole wasp venom. As allergy to insect venom develops in dependence of the frequency of insect stings, a prophylactic treatment based on mucosal tolerance induction with recombinant allergens might be of interest for people at high risk to frequent exposure to the stinging insects.
Collapse
Affiliation(s)
- Birgit Winkler
- Department of Pathophysiology, University of Vienna, Austria
| | | | | | | | | | | |
Collapse
|
31
|
Westritschnig K, Valenta R. Can we genetically engineer safer and more effective immunotherapy reagents? Curr Opin Allergy Clin Immunol 2003; 3:495-500. [PMID: 14612675 DOI: 10.1097/00130832-200312000-00012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Progress in allergen-specific immunotherapy, the only causative form of allergy treatment, was limited by the lack of defined allergen molecules for vaccine formulation. Today the genetic informations for the most common allergens have been obtained. Here we review recombinant allergen-based technologies for the improvement of diagnosis and therapy of allergy. RECENT FINDINGS Numerous strategies, including the genetic engineering of allergens for reduction of allergenic activity, have been developed to improve allergen-specific immunotherapy. Genetically modified allergen derivatives with reduced allergenic activity, preserved T cell epitope repertoire and retained immunogenicity have been characterized in vitro and in vivo. SUMMARY Based on the review of the recently published data we argue that it is possible to genetically engineer safer and more effective immunotherapy reagents.
Collapse
Affiliation(s)
- Kerstin Westritschnig
- Department of Pathophysiology, Vienna General Hospital, University of Vienna, Austria
| | | |
Collapse
|
32
|
Repa A, Grangette C, Daniel C, Hochreiter R, Hoffmann-Sommergruber K, Thalhamer J, Kraft D, Breiteneder H, Mercenier A, Wiedermann U. Mucosal co-application of lactic acid bacteria and allergen induces counter-regulatory immune responses in a murine model of birch pollen allergy. Vaccine 2003; 22:87-95. [PMID: 14604575 DOI: 10.1016/s0264-410x(03)00528-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Recent epidemiological studies and clinical trials suggest a possible role of certain lactic acid bacteria (LAB) strains in the prevention of allergic diseases. In this study, we aimed at evaluating the immunomodulatory potential of two LAB strains, Lactococcus lactis and Lactobacillus plantarum, for prophylaxis and therapy of allergic immune responses. Both LAB strains-induced high levels of IL-12 and IFN-gamma in naive murine spleen cell cultures. Intranasal co-application with recombinant Bet v 1, the major birch pollen allergen, prior or after allergic sensitization, led to increased levels of allergen-specific IgG2a antibodies and in vitro IFN-gamma production, indicating a shift towards Th1 responses. Successful immunomodulation by the mucosal pre-treatment was further demonstrated by suppression of allergen-induced basophil degranulation. We conclude that these LAB strains in combination with an allergen could be promising candidates for mucosal vaccination against type I allergy.
Collapse
Affiliation(s)
- Andreas Repa
- Department of Pathophysiology, Medical School AKH EBO3Q, University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mine Y, Rupa P. Genetic attachment of undecane peptides to ovomucoid third domain can suppress the production of specific IgG and IgE antibodies. Biochem Biophys Res Commun 2003; 311:223-8. [PMID: 14575717 DOI: 10.1016/j.bbrc.2003.09.201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
An undecane peptide (Gly-Ser-Pro-Gly-Ile-Pro-Gly-Ser-Thr-Gly-Met) was genetically attached to the N-terminus of ovomucoid third domain (DIII) to investigate structural characteristics of linear IgE and IgG (B cell) epitopes in DIII with respect to modulation of the immune response towards antigenicity and allergenicity. Balb/c mice were sensitized with native DIII, wild type recombinant DIII, and recombinant modified DIII containing the extra amino acid stretch. The immune responses to the antigens were compared using enzyme-linked immunosorbent assay. Interestingly, specific IgE and IgG levels were suppressed when the modified DIII was used as antigen. This was further confirmed by synthesizing immunodominant IgE and IgG epitopes of DIII on cellulose acetate membrane (SPOTs) and probing them with antibodies raised against DIII antigens. Anti-recombinant wild type DIII anti-serum showed strong binding activities to immunodominant IgE and IgG epitopes, while anti-modified DIII serum did not show any significant binding to the IgE and IgG epitopes. Thus, it is clearly demonstrated that the amino acid stretch in DIII is masking the immune reactive epitope. Genetical attachment of peptides into DIII was found to be effective in reducing the production of specific IgE and IgG antibodies in mice.
Collapse
Affiliation(s)
- Yoshinori Mine
- Department of Food Science, University of Guelph, Ont., Canada.
| | | |
Collapse
|
34
|
Pinto EF, de Mello Cortezia M, Rossi-Bergmann B. Interferon-gamma-inducing oral vaccination with Leishmania amazonensis antigens protects BALB/c and C57BL/6 mice against cutaneous leishmaniasis. Vaccine 2003; 21:3534-41. [PMID: 12922080 DOI: 10.1016/s0264-410x(03)00427-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The induction of oral tolerance against disease-inducing antigens has emerged as a feasible strategy to prevent immunopathologies. In this study, we investigated the effect of oral immunization with whole antigens of Leishmania amazonensis promastigotes (LaAg) on murine cutaneous leishmaniasis. We found that two oral doses with 100 microg LaAg rendered BALB/c and C57BL/6 mice more resistant against subsequent infection with L. amazonensis. The oral vaccine also partially protected BALB/c mice against Leishmania major infection. Unlike the oral route, hepatic immunization was without effect, indicating a requirement for antigen passage through the gut mucosa. Oral LaAg significantly impaired the capacity of infected BALB/c mice to mount a disease-associated hypersensitivity response, compatible with peripheral tolerization. Both IFN-gamma and IL-10, but not IL-4 were greatly elevated in the mesenteric lymph nodes whereas only IFN-gamma was increased in the peripheral lymph nodes, compatible with a TH1 cytokine response. Gamma delta TCR+ T cells may be an important component in antigenic sensitization of the gut mucosa since their depletion during oral immunization reverted protection. These results demonstrate for the first time the feasibility of using the oral route of immunization to induce protection against cutaneous leishmaniasis using a crude parasite antigen.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antigens, Protozoan/immunology
- Cytokines/biosynthesis
- Drug Hypersensitivity/immunology
- Injections
- Injections, Subcutaneous
- Interferon Inducers/pharmacology
- Interferon-gamma/biosynthesis
- Leishmania mexicana/immunology
- Leishmaniasis, Cutaneous/immunology
- Leishmaniasis, Cutaneous/pathology
- Leishmaniasis, Cutaneous/prevention & control
- Liver/physiology
- Lymph Nodes/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Protozoan Vaccines/administration & dosage
- Protozoan Vaccines/adverse effects
- Protozoan Vaccines/immunology
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Skin/pathology
- T-Lymphocytes/immunology
- Vaccination
Collapse
Affiliation(s)
- Eduardo Fonseca Pinto
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21.949-900 Rio de Janeiro, Brazil
| | | | | |
Collapse
|
35
|
Hufnagl K, Wagner B, Winkler B, Baier K, Hochreiter R, Thalhamer J, Kraft D, Scheiner O, Breiteneder H, Wiedermann U. Induction of mucosal tolerance with recombinant Hev b 1 and recombinant Hev b 3 for prevention of latex allergy in BALB/c mice. Clin Exp Immunol 2003; 133:170-6. [PMID: 12869021 PMCID: PMC1808768 DOI: 10.1046/j.1365-2249.2003.02210.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The prevalence of type I allergy to Hevea brasiliensis latex is particularly high among individuals with frequent exposure to latex products, such as health-care workers (HCW) and patients with spina bifida (SB). Treatment of latex allergy seems problematic as preventive measures, such as allergen avoidance, are not always possible and conventional immunotherapy with standardized latex extracts is not performed routinely. Thus, the aim of the present study was to establish a mouse model of latex allergy using two major latex allergens for HCWs and SB patients, Hev b 1 and Hev b 3, for sensitization. Prophylactic measures on the basis of mucosal tolerance induction with the recombinant allergens were tested in this model. Female BALB/c mice immunized intraperitoneally with recombinant (r)Hev b 1 or rHev b 3 displayed strong immune responses in vivo and in vitro. Intranasal treatment with rHev b 1 and rHev b 3 prior to sensitization led to reduced allergen-specific IgG1/IgE levels and significantly suppressed allergen-induced basophil degranulation. Moreover, lymphocyte proliferation and cytokine production (IL-4, IL-5, IFN-gamma) in vitro were significantly suppressed after pretreatment with both allergens. Suppressive cytokines, such as interleukin (IL)-10 and transforming growth factor (TGF)-beta, remained unchanged after the intranasal pretreatment, indicating mechanism of anergy rather than active immunosuppression. Taken together, these results suggest that mucosal tolerance induction with recombinant allergens could present a promising prevention strategy against latex allergy.
Collapse
Affiliation(s)
- K Hufnagl
- Department of Pathophysiology, University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Takabayashi K, Libet L, Chisholm D, Zubeldia J, Horner AA. Intranasal immunotherapy is more effective than intradermal immunotherapy for the induction of airway allergen tolerance in Th2-sensitized mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:3898-905. [PMID: 12646659 DOI: 10.4049/jimmunol.170.7.3898] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunotherapy (IT) by injection more readily induces clinical tolerance to stinging insects than to respiratory allergens. However, while systemic immunization induces adaptive responses systemically, the induction of mucosal immunity generally requires local Ag exposure. Taken together, these observations suggest that the poor success rate of systemic IT for asthma could be a consequence of inadequate immune modulation in the airways. In support of this position, investigations presented in this report demonstrate that allergen IT more effectively induces airway allergen tolerance in Th2-sensitized mice, when delivered by the intranasal (i.n.) vs the intradermal (i.d.) route. Moreover, compared with native allergen, allergen immunostimulatory sequence oligodeoxynucleotide conjugate proved to be a more effective i.n. IT reagent for protecting allergic mice from airway hypersensitivity responses. Furthermore, for both native allergen and allergen immunostimulatory sequence oligodeoxynucleotide conjugate, i.n. and i.d. IT delivery were similarly effective in modulating systemic immune profiles in Th2-sensitized mice, while only i.n. IT had significant immunomodulatory activity on B and T cell responses in the airways. The present investigations may be the first to suggest that i.n. IT is more effective than i.d. IT for the treatment of asthma. Furthermore, our results suggest that modulating airway rather than systemic immunity may be the more important therapeutic target for the induction of clinical tolerance to respiratory allergens.
Collapse
Affiliation(s)
- Kenji Takabayashi
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093-0663, USA
| | | | | | | | | |
Collapse
|
37
|
|