1
|
Sudharson S, Kalic T, Eckl-Dorna J, Lengger N, Breiteneder H, Hafner C. Modulation of Bronchial Epithelial Barrier Integrity by Low Molecular Weight Components from Birch Pollen. Int J Mol Sci 2024; 25:7374. [PMID: 39000481 PMCID: PMC11242533 DOI: 10.3390/ijms25137374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Pollen, in addition to allergens, comprise low molecular weight components (LMC) smaller than 3 kDa. Emerging evidence indicates the relevance of LMC in allergic immune responses. However, the interaction of birch pollen (BP)-derived LMC and epithelial cells has not been extensively studied. We investigated epithelial barrier modifications induced by exposure to BP LMC, using the human bronchial epithelial cell line 16HBE14o-. Epithelial cell monolayers were apically exposed to the major BP allergen Bet v 1, aqueous BP extract or BP-derived LMC. Barrier integrity after the treatments was monitored by measuring transepithelial electrical resistance at regular intervals and by using the xCELLigence Real-Time Cell Analysis system. The polarized release of cytokines 24 h following treatment was measured using a multiplex immunoassay. Epithelial barrier integrity was significantly enhanced upon exposure to BP LMC. Moreover, BP LMC induced the repair of papain-mediated epithelial barrier damage. The apical release of CCL5 and TNF-α was significantly reduced after exposure to BP LMC, while the basolateral release of IL-6 significantly increased. In conclusion, the results of our study demonstrate that BP-derived LMC modify the physical and immunological properties of bronchial epithelial cells and thus regulate airway epithelial barrier responses.
Collapse
Affiliation(s)
- Srinidhi Sudharson
- Department of Dermatology, University Hospital Sankt Poelten, Karl Landsteiner University of Health Sciences, 3100 Sankt Poelten, Austria
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Tanja Kalic
- Department of Dermatology, University Hospital Sankt Poelten, Karl Landsteiner University of Health Sciences, 3100 Sankt Poelten, Austria
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, 1090 Vienna, Austria
| | - Nina Lengger
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Christine Hafner
- Department of Dermatology, University Hospital Sankt Poelten, Karl Landsteiner University of Health Sciences, 3100 Sankt Poelten, Austria
| |
Collapse
|
2
|
Zuberbier T, Stevanovic K, Ansotegui IJ, Anto JM, Bergmann KC, D'Amato G, Grüntuch-Ernst A, Haahtela T, Maurer M, Pietikäinen S, Christou D, Bousquet J. Green Roof Gardens - Selecting Allergy-Friendly Vegetation: A Global Allergy and Asthma Excellence Network (GA²LEN) Position Paper. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:347-354. [PMID: 37863318 DOI: 10.1016/j.jaip.2023.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/22/2023]
Abstract
Green roof gardens are important for planetary health by mitigating the effects of urbanization. Because of the nature of green roof gardens, only particular plants can be used. The allergologic impact of these plants remains ill-characterized and guidance on building allergy-friendly green roof gardens is missing. To address this gap, we investigated the plant spectrum of several German green roof companies and categorized plants based on their primary pollination mechanism. Except for grasses, most plants were insect-pollinated and of low allergenicity. In addition, we conducted a review on the allergologic impact of plants used for green roof gardens. Our aim was to provide landscape architects with guidance on how to develop allergy-friendly green roof gardens. We highlight the need for universally accepted standards for assessing the allergenicity of roof top plants. Also, we recommend the joint development, by green roof producers and allergists, of criteria for allergy-friendly roof gardens. Their implementation may help to reduce the risk of allergen sensitization and allergy exacerbation, such as by avoiding the use of wind-pollinated plants of proven allergenicity including grasses. Green infrastructure, such as green roofs, should benefit planetary health without increasing the prevalence and burden of allergies.
Collapse
Affiliation(s)
- Torsten Zuberbier
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany; Institute of Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Katarina Stevanovic
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany; Institute of Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ignacio J Ansotegui
- Department of Allergy and Immunology, Hospital Quironsalud Bizkaia, Bilbao, Spain
| | - Josep M Anto
- Barcelona Institute of Global Health, Barcelona, Spain; UGA (Management and Administration Unit) of Medicine and Life Sciences - MELIS, Universitat Pompeu Farbra, Barcelona, Spain
| | - Karl-Christian Bergmann
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany; Institute of Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gennaro D'Amato
- Division of Respiratory Diseases and Allergy, High Specialty Hospital A. Cardarelli, Naples, Italy; School of Specialization in Respiratory Disease, University of Naples, Naples, Italy
| | - Almut Grüntuch-Ernst
- IDAS Institute for Design and Architectural Strategies, Technische Universität Braunschweig, Germany
| | - Tari Haahtela
- Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Marcus Maurer
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany; Institute of Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Demetrios Christou
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany; Institute of Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jean Bousquet
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany; Institute of Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Allergic Rhinitis and its Impact on Asthma (ARIA), Montpellier, France
| |
Collapse
|
3
|
Tossavainen T, Martikainen MV, Loukola H, Roponen M. Common Pollen Modulate Immune Responses against Viral-Like Challenges in Airway Coculture Model. J Immunol Res 2023; 2023:6639092. [PMID: 37965270 PMCID: PMC10643028 DOI: 10.1155/2023/6639092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Recent research indicates that exposure to pollen increases the risk and severity of respiratory infections, while studies also suggest that it may possess a protective function. Our aim was to investigate how exposure to common pollen modifies airway cells' responses to viral- or bacterial-like challenges and vice versa. Cocultured A549 and THP-1 cells were exposed to three doses of four different pollens (Alnus glutinosa, Betula pendula, Phleum pratense, or Ambrosia artemisiifolia) and subsequently to Toll-like receptor (TLR) ligands mimicking bacterial and viral challenges (TLR3, TLR4, TLR7/8). The stimulation experiment was replicated in reverse order. Toxicological and immunological end points were analyzed. When cells were primed with pollen, especially with grass (P. pratense) or weed (A. artemisiifolia), the ability of cells to secrete cytokines in response to bacterial- and viral-like exposure was decreased. In contrast, cells primed with viral ligand TLR7/8 showed greater cytokine responses against pollen than cells exposed to ligands or pollen alone. Our results suggest that pollen exposure potentially weakens immune reactions to bacterial- or viral-like challenges by modulating cytokine production. They also indicate that TLR7/8-mediated viral challenges could elicit exaggerated immune responses against pollen. Both mechanisms could contribute to the acceleration and complication of infections during the pollen season.
Collapse
Affiliation(s)
- Tarleena Tossavainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maria-Viola Martikainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Hanna Loukola
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Marjut Roponen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
4
|
Caimmi D, Clark E, Fogel P, Heddebaut N, Aubier M, Demoly P. Efficacy of a Non-Pharmacological Intervention in Seasonal Allergic Rhinitis Symptoms. Int Arch Allergy Immunol 2023; 185:111-115. [PMID: 37926078 PMCID: PMC10836848 DOI: 10.1159/000533271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/21/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Patients suffering from allergic rhinitis seek for several therapeutic symptomatic options, including nonconventional treatments, to control their symptoms. OBJECTIVES Through the present proof-of-concept study, we prospectively investigated the potential role of Puressentiel® nasal protection spray (SNPA) in patients suffering from cypress pollen allergic rhinitis. METHODS In 15 adults, we performed two nasal provocation tests, with a cypress pollen extract, with a 15-day interval, with and without previous randomized administration of SNPA, and evaluated a nasal symptom score, the nasal inspiratory peak flow, and the concentration of inflammatory cytokines in the nasal lavage after the procedures. RESULTS Comparing results in patients challenged with and without the SNPA spray before the nasal challenge, we found a 57% mean decrease in symptoms, and a 62% average difference in inspiratory peak flow, after the use of the spray. CONCLUSIONS Puressentiel® SNPA is effective in reducing nasal symptoms, as assessed by nasal symptoms score and nasal inspiratory peak flow, and could be a valid natural non-pharmacological option in patients suffering from allergic rhinitis.
Collapse
Affiliation(s)
- Davide Caimmi
- Department Respiratory Medicine and Allergy, Allergy Unit, Hôpital Arnaud de Villeneuve, University Hospital of Montpellier, Montpellier, France
- Université de Montpellier, DESP, INSERM, Montpellier, France
| | - Evangéline Clark
- Department Respiratory Medicine and Allergy, Allergy Unit, Hôpital Arnaud de Villeneuve, University Hospital of Montpellier, Montpellier, France
- Université de Montpellier, DESP, INSERM, Montpellier, France
| | | | | | - Michel Aubier
- Faculty of Medicine, University Paris Cité, Paris, France
| | - Pascal Demoly
- Department Respiratory Medicine and Allergy, Allergy Unit, Hôpital Arnaud de Villeneuve, University Hospital of Montpellier, Montpellier, France
- Université de Montpellier, DESP, INSERM, Montpellier, France
| |
Collapse
|
5
|
Buhner S, Schäuffele S, Giesbertz P, Demir IE, Zeller F, Traidl-Hoffmann C, Schemann M, Gilles S. Allergen-free extracts from birch, ragweed, and hazel pollen activate human and guinea-pig submucous and spinal sensory neurons. Neurogastroenterol Motil 2023:e14559. [PMID: 36989179 DOI: 10.1111/nmo.14559] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 01/16/2023] [Accepted: 02/24/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Non-allergenic, low molecular weight components of pollen grains are suspected to trigger changes in gut functions, sometimes leading to inflammatory conditions. Based on extensive neuroimmune communication in the gut wall, we investigated the effects of aqueous pollen extracts (APE) on enteric and spinal sensory neurons. METHODS Using Ca2+ and fast potentiometric imaging, we recorded the responses of guinea-pig and human submucous and guinea-pig dorsal root ganglion (DRG) neurons to microejection of low (<3 kDa) and high (≥3 kDa) molecular weight APEs of birch, ragweed, and hazel. Histamine was determined pharmacologically and by mass spectrometry (LC-MS/MS). KEY RESULTS Birch APE<3kDa evoked strong [Ca+2 ]i signals in the vast majority of guinea-pig DRG neurons, and in guinea-pig and human enteric neurons. The effect of birch APE≥3kDa was much weaker. Fast neuroimaging in human enteric neurons revealed an instantaneous spike discharge after microejection of birch, ragweed, and hazel APE<3kDa [median (interquartile range) at 7.0 Hz (6.2/9.8), 5.7 Hz (4.4/7.1), and 8.4 Hz (4.3/12.5), respectively]. The percentage of responding neurons per ganglion were similar [birch 40.0% (33.3/100.0), ragweed 50.8% (34.4/85.6), and hazel 83.3% (57.1/100.0)]. A mixture of histamine receptor (H1-H3) blockers significantly reduced nerve activation evoked by birch and ragweed APEs<3kDa , but was ineffective on hazel. Histamine concentrations in ragweed, birch and hazel APE's < 3 kDa were 0.764, 0.047, and 0.013 μM, respectively. CONCLUSIONS Allergen-free APEs from birch, ragweed, and hazel evoked strong nerve activation. Altered nerve-immune signaling as a result of severe pollen exposure could be a pathophysiological feature of allergic and non-allergic gut inflammation.
Collapse
Affiliation(s)
- Sabine Buhner
- Chair of Human Biology, Technical University Munich, Freising, Germany
| | | | - Pieter Giesbertz
- Molecular Nutrition Unit, Technical University Munich, Freising, Germany
| | - Ihsan Ekin Demir
- University Hospital Rechts der Isar, Technical University Munich, Munich, Germany
| | - Florian Zeller
- Department of Surgery, Academic Hospital Freising, Freising, Germany
| | - Claudia Traidl-Hoffmann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Christine Kühne Center for Allergy Research and Education (CK-Care), Davos, Switzerland
| | - Michael Schemann
- Chair of Human Biology, Technical University Munich, Freising, Germany
| | - Stefanie Gilles
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
6
|
Martikainen MV, Tossavainen T, Hannukka N, Roponen M. Pollen, respiratory viruses, and climate change: Synergistic effects on human health. ENVIRONMENTAL RESEARCH 2023; 219:115149. [PMID: 36566960 DOI: 10.1016/j.envres.2022.115149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 06/17/2023]
Abstract
In recent years, evidence of the synergistic effects of pollen and viruses on respiratory health has begun to accumulate. Pollen exposure is a known risk factor for the incidence and severity of respiratory viral infections. However, recent evidence suggests that pollen exposure may also inhibit or weaken viral infections. A comprehensive summary has not been made and a consensus on the synergistic health effects has not been reached. It is highly possible that climate change will increase the significance of pollen exposure as a cause of respiratory problems and, at the same time, affect the risk of infectious disease outbreaks. It is important to accurately assess how these two factors affect human health separately and concurrently. In this review article, for the first time, the data from previous studies are combined and reviewed and potential research gaps concerning the synergistic effects of pollen and viral exposure are identified.
Collapse
Affiliation(s)
- Maria-Viola Martikainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Tarleena Tossavainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Noora Hannukka
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Marjut Roponen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
7
|
Ragweed Major Allergen Amb a 11 Recombinant Production and Clinical Implications. Biomolecules 2023; 13:biom13010182. [PMID: 36671567 PMCID: PMC9855870 DOI: 10.3390/biom13010182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/17/2023] Open
Abstract
Ragweed pollen is highly allergenic and elicits type I hypersensitivity reactions in the exposed populations. Amb a 11 is a recently discovered component of this pollen, and its biological role in allergy is still being researched. In our study, ragweed allergy patients were recruited prospectively over a three-year period; a comprehensive questionnaire was administered, and sera were collected and stored. The production of recombinant Amb a 11 was achieved in parallel with patients' recruitment. The gene coding for mature protein was inserted in E. coli and in Sf9 Spodoptera frugiperda cells. The recombinant allergens (designated eAmb a 11 and iAmb a 11) were tested for His-tag presence in Western blot. IgE reactivity was evaluated in 150 patients' sera for both recombinant allergen forms in ELISA, with 5 positive sera being tested further by hRBL (humanized rat basophilic leukemia) hexosaminidase release assay. Both allergen forms were proven to be IgE-reactive His-tagged proteins, with an extensive overlap of positive sera (92 toward the former recombinant allergen, 100 toward the latter) and an overall Amb a 11 sensitization prevalence estimated at 68.67%. The hRBL mediator release assay revealed a significant, slightly weaker effect of recombinant allergens when compared with nAmb a 1. Sensitization to this major allergen appears to be associated with more severe asthma symptoms (OR = 4.71, 95% CI = 1.81-12.21). In conclusion, recombinant Amb a 11 is a bona fide allergen, which is IgE-reactive and an inducer of hRBL degranulation. It is an important IgE-reactive component from ragweed pollen, with high IgE sensitization prevalence in the sample population and allergenicity of the recombinant allergen comparable to Amb a 1.
Collapse
|
8
|
Wang Z, Wang Q, Duan S, Zhang Y, Zhao L, Zhang S, Hao L, Li Y, Wang X, Wang C, Zhang N, Bachert C, Zhang L, Lan F. A diagnostic model for predicting type 2 nasal polyps using biomarkers in nasal secretion. Front Immunol 2022; 13:1054201. [PMID: 36618395 PMCID: PMC9811186 DOI: 10.3389/fimmu.2022.1054201] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Background Predicting type 2 chronic rhinosinusitis with nasal polyps (CRSwNP) may help for selection of appropriate surgical procedures or pharmacotherapies in advance. However, an accurate non-invasive method for diagnosis of type 2 CRSwNP is presently unavailable. Methods To optimize the technique for collecting nasal secretion (NasSec), 89 CRSwNP patients were tested using nasal packs made with four types of materials. Further, Th2low and Th2highCRSwNP defined by clustering analysis in another 142 CRSwNP patients using tissue biomarkers, in the meanwhile, inflammatory biomarkers were detected in NasSec of the same patients collected by the selected nasal pack. A diagnostic model was established by machine learning algorithms to predict Th2highCRSwNP using NasSecs biomarkers. Results Considering the area under receiver operating characteristic curve (AUC) for IL-5 in NasSec, nasal pack in polyvinyl alcohol (PVA) was superior to other materials for NasSec collection. When Th2low and Th2highCRSwNP clusters were defined, logistic regression and decision tree model for prediction of Th2highCRSwNP demonstrated high AUCs values of 0.92 and 0.90 respectively using biomarkers of NasSecs. Consequently, the pre-pruned decision tree model; based on the levels of IL-5 in NasSec (≤ 15.04 pg/mL), blood eosinophil count (≤ 0.475*109/L) and absence of comorbid asthma; was chosen to define Th2lowCRSwNP from Th2highCRSwNP for routine clinical use. Conclusions Taken together, a decision tree model based on a combination of NasSec biomarkers and clinical features can accurately define type 2 CRSwNP patients and therefore may be of benefit to patients in receiving appropriate therapies in daily clinical practice.
Collapse
Affiliation(s)
- Zaichuan Wang
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Qiqi Wang
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China
| | - Su Duan
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Yuling Zhang
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Limin Zhao
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Shujian Zhang
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Liusiqi Hao
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Yan Li
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China
| | - Xiangdong Wang
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Chenshuo Wang
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Nan Zhang
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium
| | - Luo Zhang
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China,Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China,*Correspondence: Luo Zhang, ; Feng Lan,
| | - Feng Lan
- Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing, China,*Correspondence: Luo Zhang, ; Feng Lan,
| |
Collapse
|
9
|
Guryanova SV, Finkina EI, Melnikova DN, Bogdanov IV, Bohle B, Ovchinnikova TV. How Do Pollen Allergens Sensitize? Front Mol Biosci 2022; 9:900533. [PMID: 35782860 PMCID: PMC9245541 DOI: 10.3389/fmolb.2022.900533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Plant pollen is one of the main sources of allergens causing allergic diseases such as allergic rhinitis and asthma. Several allergens in plant pollen are panallergens which are also present in other allergen sources. As a result, sensitized individuals may also experience food allergies. The mechanism of sensitization and development of allergic inflammation is a consequence of the interaction of allergens with a large number of molecular factors that often are acting in a complex with other compounds, for example low-molecular-mass ligands, which contribute to the induction a type 2-driven response of immune system. In this review, special attention is paid not only to properties of allergens but also to an important role of their interaction with lipids and other hydrophobic molecules in pollen sensitization. The reactions of epithelial cells lining the nasal and bronchial mucosa and of other immunocompetent cells will also be considered, in particular the mechanisms of the activation of B and T lymphocytes and the formation of allergen-specific antibody responses.
Collapse
Affiliation(s)
- Svetlana V. Guryanova
- Science-Educational Center, M. M. Shemyakin & Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, Moscow, Russia
- Medical Institute, Peoples’ Friendship University of Russia, The Ministry of Science and Higher Education of the Russian Federation, Moscow, Russia
| | - Ekaterina I. Finkina
- Science-Educational Center, M. M. Shemyakin & Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, Moscow, Russia
| | - Daria N. Melnikova
- Science-Educational Center, M. M. Shemyakin & Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, Moscow, Russia
| | - Ivan V. Bogdanov
- Science-Educational Center, M. M. Shemyakin & Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, Moscow, Russia
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Tatiana V. Ovchinnikova
- Science-Educational Center, M. M. Shemyakin & Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, Moscow, Russia
- Department of Biotechnology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- *Correspondence: Tatiana V. Ovchinnikova,
| |
Collapse
|
10
|
Hopkins GV, Cochrane S, Onion D, Fairclough LC. The Role of Lipids in Allergic Sensitization: A Systematic Review. Front Mol Biosci 2022; 9:832330. [PMID: 35495627 PMCID: PMC9047936 DOI: 10.3389/fmolb.2022.832330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Immunoglobulin E (IgE)-mediated allergies are increasing in prevalence, with IgE-mediated food allergies currently affecting up to 10% of children and 6% of adults worldwide. The mechanisms underpinning the first phase of IgE-mediated allergy, allergic sensitization, are still not clear. Recently, the potential involvement of lipids in allergic sensitization has been proposed, with reports that they can bind allergenic proteins and act on immune cells to skew to a T helper type 2 (Th2) response. Objectives: The objective of this systematic review is to determine if there is strong evidence for the role of lipids in allergic sensitization. Methods: Nineteen studies were reviewed, ten of which were relevant to lipids in allergic sensitization to food allergens, nine relevant to lipids in aeroallergen sensitization. Results: The results provide strong evidence for the role of lipids in allergies. Intrinsic lipids from allergen sources can interact with allergenic proteins to predominantly enhance but also inhibit allergic sensitization through various mechanisms. Proposed mechanisms included reducing the gastrointestinal degradation of allergenic proteins by altering protein structure, reducing dendritic cell (DC) uptake of allergenic proteins to reduce immune tolerance, regulating Th2 cytokines, activating invariant natural killer T (iNKT) cells through CD1d presentation, and directly acting upon toll-like receptors (TLRs), epithelial cells, keratinocytes, and DCs. Conclusion: The current literature suggests intrinsic lipids are key influencers of allergic sensitization. Further research utilising human relevant in vitro models and clinical studies are needed to give a reliable account of the role of lipids in allergic sensitization.
Collapse
Affiliation(s)
- Georgina V. Hopkins
- School of Life Sciences, The University of Nottingham, Nottingham, United Kingdom
| | - Stella Cochrane
- SEAC, Unilever, Colworth Science Park, Sharnbrook, United Kingdom
| | - David Onion
- School of Life Sciences, The University of Nottingham, Nottingham, United Kingdom
| | - Lucy C. Fairclough
- School of Life Sciences, The University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
11
|
Potocki L, Karbarz M, Adamczyk-Grochala J, Kasprzyk I, Pawlina-Tyszko K, Lewinska A, Wnuk M. Silver birch pollen-derived microRNAs promote NF-κB-mediated inflammation in human lung cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 800:149531. [PMID: 34392209 DOI: 10.1016/j.scitotenv.2021.149531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/17/2021] [Accepted: 08/04/2021] [Indexed: 06/13/2023]
Abstract
The pollen of Betula pendula Roth (silver birch) is considered to be the main cause of allergy-related rhinitis in Europe and its protein-based allergens such as Bet v 1 are well characterized. However, little is known about non-protein components of birch pollen, e.g., small RNAs and their proinflammatory activity. In the present study, next-generation sequencing (NGS) and bioinformatic approaches were used for silver birch pollen (SBP)-derived microRNA profiling and evaluation of microRNA target genes and pathways in human. Human lung cells, namely WI-38 fibroblasts and A549 alveolar epithelial cells were then stimulated with SBP microRNA in vitro and imaging cytometry-based analysis of the levels of proinflammatory cytokines, autophagy parameters and small RNA processing regulators was conducted. Bioinformatic analysis revealed that SBP microRNA may interfere with autophagy, inflammation and allergy pathways in human. SBP and SBP-derived microRNA induced NF-κB-mediated proinflammatory response in human lung cells as judged by increased levels of NF-κB p65, IL-8 and TNFα. NSUN2 and NSUN5 were involved in pollen-derived microRNA processing. Pollen-derived microRNA also modulated autophagic pathway by changes in the pools of LC3B and p62 that may affect autophagy-based adaptive responses during allergic lung inflammation. We postulate that SBP-derived microRNAs can be considered as novel proinflammatory environmental agents.
Collapse
Affiliation(s)
- Leszek Potocki
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Malgorzata Karbarz
- Department of Biology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Jagoda Adamczyk-Grochala
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Idalia Kasprzyk
- Department of Biology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Klaudia Pawlina-Tyszko
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland
| | - Anna Lewinska
- Department of Biotechnology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| | - Maciej Wnuk
- Department of Biology, Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland.
| |
Collapse
|
12
|
Abstract
The prevalence of allergic diseases is increasing rapidly and has already reached an epidemic level. Two major drivers of this development are climate change and globalization, which both induce an increase in allergens. Concomitant climate change fosters the spreading of the latter on a global scale. The increase in allergens not only aggravates the symptoms and the degree of suffering for patients who already are allergic, but also gives rise to new cases of allergies. The distribution of allergies in society follows a steep socioeconomic gradient worldwide. According to well-established theories of justice such a distribution of the allergy burden is unfair. This fact adds a major ethical dimension and challenge to the allergy epidemic. This chapter draws on the key points of policies for allergy prevention and treatment. It shows how related programs and measures can be conceptualized and prioritized according to the principles of distributional justice.
Collapse
Affiliation(s)
- Clemens Heuson
- Zentrum für Klimaresilienz, Universität Augsburg, Augsburg, Germany.
| |
Collapse
|
13
|
Rauer D, Gilles S, Wimmer M, Frank U, Mueller C, Musiol S, Vafadari B, Aglas L, Ferreira F, Schmitt‐Kopplin P, Durner J, Winkler JB, Ernst D, Behrendt H, Schmidt‐Weber CB, Traidl‐Hoffmann C, Alessandrini F. Ragweed plants grown under elevated CO 2 levels produce pollen which elicit stronger allergic lung inflammation. Allergy 2021; 76:1718-1730. [PMID: 33037672 DOI: 10.1111/all.14618] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Common ragweed has been spreading as a neophyte in Europe. Elevated CO2 levels, a hallmark of global climate change, have been shown to increase ragweed pollen production, but their effects on pollen allergenicity remain to be elucidated. METHODS Ragweed was grown in climate-controlled chambers under normal (380 ppm, control) or elevated (700 ppm, based on RCP4.5 scenario) CO2 levels. Aqueous pollen extracts (RWE) from control- or CO2 -pollen were administered in vivo in a mouse model for allergic disease (daily for 3-11 days, n = 5) and employed in human in vitro systems of nasal epithelial cells (HNECs), monocyte-derived dendritic cells (DCs), and HNEC-DC co-cultures. Additionally, adjuvant factors and metabolites in control- and CO2 -RWE were investigated using ELISA and untargeted metabolomics. RESULTS In vivo, CO2 -RWE induced stronger allergic lung inflammation compared to control-RWE, as indicated by lung inflammatory cell infiltrate and mediators, mucus hypersecretion, and serum total IgE. In vitro, HNECs stimulated with RWE increased indistinctively the production of pro-inflammatory cytokines (IL-8, IL-1β, and IL-6). In contrast, supernatants from CO2 -RWE-stimulated HNECs, compared to control-RWE-stimulated HNECS, significantly increased TNF and decreased IL-10 production in DCs. Comparable results were obtained by stimulating DCs directly with RWEs. The metabolome analysis revealed differential expression of secondary plant metabolites in control- vs CO2 -RWE. Mixes of these metabolites elicited similar responses in DCs as compared to respective RWEs. CONCLUSION Our results indicate that elevated ambient CO2 levels elicit a stronger RWE-induced allergic response in vivo and in vitro and that RWE increased allergenicity depends on the interplay of multiple metabolites.
Collapse
Affiliation(s)
- Denise Rauer
- Chair and Institute of Environmental Medicine UNIKA‐T, Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Stefanie Gilles
- Chair and Institute of Environmental Medicine UNIKA‐T, Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Maria Wimmer
- Center of Allergy & Environment (ZAUM) Technical University of Munich (TUM) and Helmholtz Zentrum München Munich Germany
- Members of the German Center of Lung Research (DZL) Munich Germany
| | - Ulrike Frank
- Institute of Biochemical Plant Pathology (BIOP) Helmholtz Zentrum München Neuherberg Germany
| | - Constanze Mueller
- BGC Research Unit Analytical BioGeoChemistry Helmholtz Zentrum München Neuherberg Germany
| | - Stephanie Musiol
- Center of Allergy & Environment (ZAUM) Technical University of Munich (TUM) and Helmholtz Zentrum München Munich Germany
- Members of the German Center of Lung Research (DZL) Munich Germany
| | - Behnam Vafadari
- Chair and Institute of Environmental Medicine UNIKA‐T, Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Lorenz Aglas
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Fatima Ferreira
- Department of Biosciences University of Salzburg Salzburg Austria
| | | | - Jörg Durner
- Institute of Biochemical Plant Pathology (BIOP) Helmholtz Zentrum München Neuherberg Germany
| | - Jana Barbro Winkler
- Research Unit Environmental Simulation Institute of Biochemical Plant Pathology Helmholtz Zentrum München Neuherberg Germany
| | - Dieter Ernst
- Institute of Biochemical Plant Pathology (BIOP) Helmholtz Zentrum München Neuherberg Germany
| | - Heidrun Behrendt
- Center of Allergy & Environment (ZAUM) Technical University of Munich (TUM) and Helmholtz Zentrum München Munich Germany
| | - Carsten B. Schmidt‐Weber
- Center of Allergy & Environment (ZAUM) Technical University of Munich (TUM) and Helmholtz Zentrum München Munich Germany
- Members of the German Center of Lung Research (DZL) Munich Germany
| | - Claudia Traidl‐Hoffmann
- Chair and Institute of Environmental Medicine UNIKA‐T, Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- Outpatient Clinic for Environmental Medicine University Clinic Augsburg Augsburg Germany
- Christine‐Kühne Center for Allergy Research and Education (CK‐Care) Davos Switzerland
| | - Francesca Alessandrini
- Center of Allergy & Environment (ZAUM) Technical University of Munich (TUM) and Helmholtz Zentrum München Munich Germany
- Members of the German Center of Lung Research (DZL) Munich Germany
| |
Collapse
|
14
|
Damialis A, Gilles S, Sofiev M, Sofieva V, Kolek F, Bayr D, Plaza MP, Leier-Wirtz V, Kaschuba S, Ziska LH, Bielory L, Makra L, Del Mar Trigo M, Traidl-Hoffmann C. Higher airborne pollen concentrations correlated with increased SARS-CoV-2 infection rates, as evidenced from 31 countries across the globe. Proc Natl Acad Sci U S A 2021; 118:e2019034118. [PMID: 33798095 PMCID: PMC7999946 DOI: 10.1073/pnas.2019034118] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pollen exposure weakens the immunity against certain seasonal respiratory viruses by diminishing the antiviral interferon response. Here we investigate whether the same applies to the pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is sensitive to antiviral interferons, if infection waves coincide with high airborne pollen concentrations. Our original hypothesis was that more airborne pollen would lead to increases in infection rates. To examine this, we performed a cross-sectional and longitudinal data analysis on SARS-CoV-2 infection, airborne pollen, and meteorological factors. Our dataset is the most comprehensive, largest possible worldwide from 130 stations, across 31 countries and five continents. To explicitly investigate the effects of social contact, we additionally considered population density of each study area, as well as lockdown effects, in all possible combinations: without any lockdown, with mixed lockdown-no lockdown regime, and under complete lockdown. We found that airborne pollen, sometimes in synergy with humidity and temperature, explained, on average, 44% of the infection rate variability. Infection rates increased after higher pollen concentrations most frequently during the four previous days. Without lockdown, an increase of pollen abundance by 100 pollen/m3 resulted in a 4% average increase of infection rates. Lockdown halved infection rates under similar pollen concentrations. As there can be no preventive measures against airborne pollen exposure, we suggest wide dissemination of pollen-virus coexposure dire effect information to encourage high-risk individuals to wear particle filter masks during high springtime pollen concentrations.
Collapse
Affiliation(s)
- Athanasios Damialis
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany;
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Stefanie Gilles
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Mikhail Sofiev
- Finnish Meteorological Institute, Helsinki FI-00101, Finland
| | | | - Franziska Kolek
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Daniela Bayr
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Maria P Plaza
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Vivien Leier-Wirtz
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Sigrid Kaschuba
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| | - Lewis H Ziska
- Mailman School of Public Health, Columbia University, New York, NY 10032
| | - Leonard Bielory
- Center for Environmental Prediction, Rutgers University, New Brunswick, NJ 08901
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ 08854
- Medicine, Allergy, Immunology and Ophthalmology Department, Hackensack Meridian School of Medicine, Nutley, NJ 07110
- New Jersey Center of Science, Technology and Mathematics, Kean University, Union, NJ 07083
| | - László Makra
- Institute of Economics and Rural Development, Faculty of Agriculture, University of Szeged, Szeged 6720, Hungary
| | - Maria Del Mar Trigo
- Department of Botany and Plant Physiology, University of Malaga, Malaga 29016, Spain
| | - Claudia Traidl-Hoffmann
- Chair of Environmental Medicine, Technical University of Munich, Augsburg 86156, Germany
- Institute of Environmental Medicine, Helmholtz Centre Munich, Augsburg 86156, Germany
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg 86156, Germany
| |
Collapse
|
15
|
Gökkaya M, Damialis A, Nussbaumer T, Beck I, Bounas-Pyrros N, Bezold S, Amisi MM, Kolek F, Todorova A, Chaker A, Aglas L, Ferreira F, Redegeld FA, Brunner JO, Neumann AU, Traidl-Hoffmann C, Gilles S. Defining biomarkers to predict symptoms in subjects with and without allergy under natural pollen exposure. J Allergy Clin Immunol 2020; 146:583-594.e6. [DOI: 10.1016/j.jaci.2020.02.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 01/31/2020] [Accepted: 02/24/2020] [Indexed: 01/11/2023]
|
16
|
Eguiluz‐Gracia I, Mathioudakis AG, Bartel S, Vijverberg SJH, Fuertes E, Comberiati P, Cai YS, Tomazic PV, Diamant Z, Vestbo J, Galan C, Hoffmann B. The need for clean air: The way air pollution and climate change affect allergic rhinitis and asthma. Allergy 2020; 75:2170-2184. [PMID: 31916265 DOI: 10.1111/all.14177] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/20/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
Air pollution and climate change have a significant impact on human health and well-being and contribute to the onset and aggravation of allergic rhinitis and asthma among other chronic respiratory diseases. In Westernized countries, households have experienced a process of increasing insulation and individuals tend to spend most of their time indoors. These sequelae implicate a high exposure to indoor allergens (house dust mites, pets, molds, etc), tobacco smoke, and other pollutants, which have an impact on respiratory health. Outdoor air pollution derived from traffic and other human activities not only has a direct negative effect on human health but also enhances the allergenicity of some plants and contributes to global warming. Climate change modifies the availability and distribution of plant- and fungal-derived allergens and increases the frequency of extreme climate events. This review summarizes the effects of indoor air pollution, outdoor air pollution, and subsequent climate change on asthma and allergic rhinitis in children and adults and addresses the policy adjustments and lifestyle changes required to mitigate their deleterious effects.
Collapse
Affiliation(s)
- Ibon Eguiluz‐Gracia
- Allergy Unit IBIMA‐Hospital Regional Universitario de Malaga‐UMA Malaga Spain
| | - Alexander G. Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine School of Biological Sciences The University of Manchester Manchester Academic Health Science Centre UK
- North West Lung Centre Wythenshawe Hospital Manchester University NHS Foundation Trust Southmoor Road Manchester UK
| | - Sabine Bartel
- Early Life Origins of Chronic Lung Disease, Research Center Borstel Leibniz Lung Center Member of the German Research Center for Lung Research (DZL) Borstel Germany
- Department of Pathology and Medical Biology University Medical Center Groningen GRIAC Research Institute University of Groningen Groningen The Netherlands
| | - Susanne J. H. Vijverberg
- Department of Respiratory Medicine Amsterdam UMC University of Amsterdam Amsterdam The Netherlands
| | - Elaine Fuertes
- National Heart and Lung Institute Imperial College London London UK
| | - Pasquale Comberiati
- Section of Paediatrics Department of Clinical and Experimental Medicine University of Pisa Pisa Italy
- Department of Clinical Immunology and Allergology Sechenov University Moscow Russia
| | - Yutong Samuel Cai
- Department of Epidemiology and Biostatistics MRC Centre for Environment and Health School of Public Health Imperial College London London UK
- The George Institute for Global Health University of Oxford Oxford UK
| | - Peter Valentin Tomazic
- Department of General ORL, Head and Neck Surgery Medical University of Graz Graz Austria
| | - Zuzana Diamant
- Department of Respiratory Medicine & Allergology Institute for Clinical Science Skane University Hospital Lund University Lund Sweden
- Department of Respiratory Medicine First Faculty of Medicine Charles University and Thomayer Hospital Prague Czech Republic
| | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine School of Biological Sciences The University of Manchester Manchester Academic Health Science Centre UK
- North West Lung Centre Wythenshawe Hospital Manchester University NHS Foundation Trust Southmoor Road Manchester UK
| | - Carmen Galan
- Department of Botany, Ecology and Plant Physiology International Campus of Excellence on Agrifood (ceiA3) University of Córdoba Córdoba Spain
| | - Barbara Hoffmann
- Institute for Occupational, Social and Environmental Medicine Medical Faculty University of Düsseldorf Düsseldorf Germany
| |
Collapse
|
17
|
Pointner L, Bethanis A, Thaler M, Traidl-Hoffmann C, Gilles S, Ferreira F, Aglas L. Initiating pollen sensitization - complex source, complex mechanisms. Clin Transl Allergy 2020; 10:36. [PMID: 32884636 PMCID: PMC7461309 DOI: 10.1186/s13601-020-00341-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/27/2020] [Accepted: 08/12/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanisms involved in the induction of allergic sensitization by pollen are not fully understood. Within the last few decades, findings from epidemiological and experimental studies support the notion that allergic sensitization is not only dependent on the genetics of the host and environmental factors, but also on intrinsic features of the allergenic source itself. In this review, we summarize the current concepts and newest advances in research focusing on the initial mechanisms inducing pollen sensitization. Pollen allergens are embedded in a complex and heterogeneous matrix composed of a myriad of bioactive molecules that are co-delivered during the allergic sensitization. Surprisingly, several purified allergens were shown to lack inherent sensitizing potential. Thus, growing evidence supports an essential role of pollen-derived components co-delivered with the allergens in the initiation of allergic sensitization. The pollen matrix, which is composed by intrinsic molecules (e.g. proteins, metabolites, lipids, carbohydrates) and extrinsic compounds (e.g. viruses, particles from air pollutants, pollen-linked microbiome), provide a specific context for the allergen and has been proposed as a determinant of Th2 polarization. In addition, the involvement of various pattern recognition receptors (PRRs), secreted alarmins, innate immune cells, and the dependency of DCs in driving pollen-induced Th2 inflammatory processes suggest that allergic sensitization to pollen most likely results from particular combinations of pollen-specific signals rather than from a common determinant of allergenicity. The exact identification and characterization of such pollen-derived Th2-polarizing molecules should provide mechanistic insights into Th2 polarization and pave the way for novel preventive and therapeutic strategies against pollen allergies.
Collapse
Affiliation(s)
- Lisa Pointner
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| | - Athanasios Bethanis
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| | - Michael Thaler
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| | - Claudia Traidl-Hoffmann
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
- Christine-Kühne-Center for Allergy Research and Education (CK-Care), Davos, Switzerland
| | - Stefanie Gilles
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Fatima Ferreira
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| | - Lorenz Aglas
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße. 34, 5020 Salzburg, Austria
| |
Collapse
|
18
|
Bergougnan C, Dittlein DC, Hümmer E, Riepl R, Eisenbart S, Böck D, Griesbaum L, Weigl A, Damialis A, Hartwig A, Neumann AU, Zenk J, Traidl-Hoffmann C, Gilles S. Physical and immunological barrier of human primary nasal epithelial cells from non-allergic and allergic donors. World Allergy Organ J 2020; 13:100109. [PMID: 32180893 PMCID: PMC7063333 DOI: 10.1016/j.waojou.2020.100109] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/04/2019] [Accepted: 02/12/2020] [Indexed: 11/16/2022] Open
Abstract
The epithelial cell-derived cytokine milieu has been discussed as a “master switch” in the development of allergic disease. To understand the role of innate immune response in nasal epithelial cells during allergic inflammation, we created and established a fast and minimally invasive method to isolate and culture human nasal epithelial cells from clinically and immunologically well characterized patients. Human nasal epithelial cells from non-atopic volunteers and from allergic rhinitis patients were compared in respect to their growth, barrier integrity, pattern recognition, receptor expression, and immune responses to allergens and an array of pathogen-associated molecular patterns and inflammasome activators. Cells from nasal scrapings were clearly identified as nasal epithelial cells by staining of pan-Cytokeratin, Cytokeratin-14 and Tubulin. Additionally, Mucin 5AC staining revealed the presence of goblet cells, while staining of tight-junction protein Claudin-1, Occludin and ZO-1 showed the ability of the cells to form a tight barrier. Cells of atopic donors grew slower than cells of non-atopic donors. All nasal epithelial cells expressed TLR1-6 and 9, yet the expression of TLR-9 was lower in cells from allergic rhinitis (AR) donors. Additionally, epithelial cells from AR donors responded with a different TLR expression pattern to stimulation with TLR ligands. TLR-3 was the most potent modulator of cytokine and chemokine secretion in all human nasal epithelial cells (HNECs). The secretion of IL-1β, CCL-5, IL-8, IL-18 and IL-33 was elevated in HNECs of AR donors as compared to cells of non-atopic donors. This was observed in the steady-state (IL-18, IL-33) as well as under stimulation with TLR ligands (IL-18, IL-33, CCL-5, IL-8), aqueous pollen extracts (IL-18, IL-33), or the inflammasome activator Nigericin (IL-1β). In conclusion, nasal epithelial cells of AR donors show altered physical barrier responses in steady-state and in response to allergen stimulation. Cells of AR donors show increased expression of pro-inflammatory and IL-1 family cytokines at baseline and under stimulation, which could contribute to a micromilieu which is favorable for Th2.
Collapse
Key Words
- ALI, Air liquid interphase
- APE, Aqueous pollen extract
- AR, Allergic rhinitis
- Allergic rhinitis
- HDM, House dust mite
- HNEC, Human nasal epithelial cell
- Inflammation
- LPS, Lipopolysaccharide from E. Coli K12 (TLR-4 ligand)
- MyD88, Myeloid differentiation primary response 88
- Nasal epithelium
- PAMP, Pathogen-associated molecular pattern
- PRR, Pattern recognition receptor
- Pattern recognition receptor
- Pollen
- PolyIC, Polyinosinic–polycytidylic acid (TLR-3 ligand)
- SAR, Seasonal allergic rhinitis
- SEM, Scanning electron microscopy
- TER, Transepithelial electrical resistance
- TLR, Toll-like receptor
- TRIF, TIR-domain-containing adapter-inducing interferon-β
Collapse
Affiliation(s)
- Carolin Bergougnan
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany.,Christine-Kühne-Center for Allergy Research and Education (CK-Care), Davos, Switzerland
| | - Daniela C Dittlein
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany
| | - Elke Hümmer
- Department of Otolaryngology, Augsburg University Medical School, Augsburg, Germany
| | - Rosalie Riepl
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany
| | - Selina Eisenbart
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany
| | - Dominik Böck
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany
| | - Lena Griesbaum
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany
| | - Anna Weigl
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany
| | - Athanasios Damialis
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany
| | | | - Avidan U Neumann
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany
| | - Johannes Zenk
- Department of Otolaryngology, Augsburg University Medical School, Augsburg, Germany
| | - Claudia Traidl-Hoffmann
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany.,Christine-Kühne-Center for Allergy Research and Education (CK-Care), Davos, Switzerland
| | - Stefanie Gilles
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and HelmholtzZentrum München, Augsburg, Germany
| |
Collapse
|
19
|
Gilles S, Blume C, Wimmer M, Damialis A, Meulenbroek L, Gökkaya M, Bergougnan C, Eisenbart S, Sundell N, Lindh M, Andersson L, Dahl Å, Chaker A, Kolek F, Wagner S, Neumann AU, Akdis CA, Garssen J, Westin J, Land B, Davies DE, Traidl‐Hoffmann C. Pollen exposure weakens innate defense against respiratory viruses. Allergy 2020; 75:576-587. [PMID: 31512243 DOI: 10.1111/all.14047] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 06/13/2019] [Accepted: 06/24/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Hundreds of plant species release their pollen into the air every year during early spring. During that period, pollen allergic as well as non-allergic patients frequently present to doctors with severe respiratory tract infections. Our objective was therefore to assess whether pollen may interfere with antiviral immunity. METHODS We combined data from real-life human exposure cohorts, a mouse model and human cell culture to test our hypothesis. RESULTS Pollen significantly diminished interferon-λ and pro-inflammatory chemokine responses of airway epithelia to rhinovirus and viral mimics and decreased nuclear translocation of interferon regulatory factors. In mice infected with respiratory syncytial virus, co-exposure to pollen caused attenuated antiviral gene expression and increased pulmonary viral titers. In non-allergic human volunteers, nasal symptoms were positively correlated with airborne birch pollen abundance, and nasal birch pollen challenge led to downregulation of type I and -III interferons in nasal mucosa. In a large patient cohort, numbers of rhinoviruspositive cases were correlated with airborne birch pollen concentrations. CONCLUSION The ability of pollen to suppress innate antiviral immunity, independent of allergy, suggests that high-risk population groups should avoid extensive outdoor activities when pollen and respiratory virus seasons coincide.
Collapse
Affiliation(s)
- Stefanie Gilles
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Cornelia Blume
- Faculty of Medicine Academic Unit of Clinical and Experimental Sciences University of Southampton Southampton UK
- Southampton NIHR Respiratory Biomedical Research Unit University Hospital Southampton Southampton UK
| | - Maria Wimmer
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- Division of Pharmacology Department of Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
| | - Athanasios Damialis
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Laura Meulenbroek
- Division of Pharmacology Department of Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
- Department of Immunology Nutricia Research Utrecht The Netherlands
| | - Mehmet Gökkaya
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Carolin Bergougnan
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Selina Eisenbart
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Nicklas Sundell
- Department of Infectious Diseases/Clinical Virology University of Gothenburg Gothenburg Sweden
| | - Magnus Lindh
- Department of Infectious Diseases/Clinical Virology University of Gothenburg Gothenburg Sweden
| | - Lars‐Magnus Andersson
- Department of Infectious Diseases/Clinical Virology University of Gothenburg Gothenburg Sweden
| | - Åslög Dahl
- Department of Biological and Environmental Sciences Faculty of Sciences University of Gothenburg Gothenburg Sweden
| | - Adam Chaker
- ENT Department Klinikum Rechts der Isar Technical University of Munich Munich Germany
| | - Franziska Kolek
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Sabrina Wagner
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Avidan U. Neumann
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University Zurich Davos Switzerland
- Christine‐Kühne‐Center for Allergy Research and Education (CK‐Care) Davos Switzerland
| | - Johan Garssen
- Division of Pharmacology Department of Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
- Department of Immunology Nutricia Research Utrecht The Netherlands
| | - Johan Westin
- Department of Infectious Diseases/Clinical Virology University of Gothenburg Gothenburg Sweden
| | - Belinda Land
- Department of Immunology Nutricia Research Utrecht The Netherlands
- Laboratory of Translational Immunology The Wilhelmina Children's Hospital University Medical Center Utrecht Utrecht The Netherlands
| | - Donna E. Davies
- Faculty of Medicine Academic Unit of Clinical and Experimental Sciences University of Southampton Southampton UK
- Southampton NIHR Respiratory Biomedical Research Unit University Hospital Southampton Southampton UK
| | - Claudia Traidl‐Hoffmann
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- Christine‐Kühne‐Center for Allergy Research and Education (CK‐Care) Davos Switzerland
| |
Collapse
|
20
|
Allergie als Volkskrankheit. ALLERGO JOURNAL 2019. [DOI: 10.1007/s15007-019-1866-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
21
|
Fish-derived low molecular weight components modify bronchial epithelial barrier properties and release of pro-inflammatory cytokines. Mol Immunol 2019; 112:140-150. [PMID: 31102986 PMCID: PMC6997027 DOI: 10.1016/j.molimm.2019.04.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 01/07/2023]
Abstract
The prevalence of fish allergy among fish-processing workers is higher than in the general population, possibly due to sensitization via inhalation and higher exposure. However, the response of the bronchial epithelium to fish allergens has never been explored. Parvalbumins (PVs) from bony fish are major sensitizers in fish allergy, while cartilaginous fish and their PVs are considered less allergenic. Increasing evidence demonstrates that components other than proteins from the allergen source, such as low molecular weight components smaller than 3 kDa (LMC) from pollen, may act as adjuvants during allergic sensitization. We investigated the response of bronchial epithelial cells to PVs and to LMC from Atlantic cod, a bony fish, and gummy shark, a cartilaginous fish. Polarized monolayers of the bronchial epithelial cell line 16HBE14o- were stimulated apically with fish PVs and/-or the corresponding fish LMC. Barrier integrity, transport of PVs across the monolayers and release of mediators were monitored. Intact PVs from both the bony and the cartilaginous fish were rapidly internalized by the cells and transported to the basolateral side of the monolayers. The PVs did not disrupt the epithelial barrier integrity nor did they modify the release of proinflammatory cytokines. In contrast, LMC from both fish species modified the physical and immunological properties of the epithelial barrier and the responses differed between bony and cartilaginous fish. While the barrier integrity was lowered by cod LMC 24 h after cell stimulation, it was increased by up to 2.3-fold by shark LMC. Furthermore, LMC from both fish species increased basolateral and apical release of IL-6 and IL-8, while CCL2 release was increased by cod but not by shark LMC. In summary, our study demonstrated the rapid transport of PVs across the epithelium which may result in their availability to antigen presenting cells required for allergic sensitization. Moreover, different cell responses to LMC derived from bony versus cartilaginous fish were observed, which may play a role in different allergenic potentials of these two fish classes.
Collapse
|
22
|
Damialis A, Häring F, Gökkaya M, Rauer D, Reiger M, Bezold S, Bounas-Pyrros N, Eyerich K, Todorova A, Hammel G, Gilles S, Traidl-Hoffmann C. Human exposure to airborne pollen and relationships with symptoms and immune responses: Indoors versus outdoors, circadian patterns and meteorological effects in alpine and urban environments. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 653:190-199. [PMID: 30408667 DOI: 10.1016/j.scitotenv.2018.10.366] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/27/2018] [Accepted: 10/27/2018] [Indexed: 06/08/2023]
Abstract
Pollen exposure is a major cause of respiratory allergies worldwide. However, it is unclear how everyday exposure is related to symptoms and how allergic patients may be affected spatially and temporally. Hence, we investigated the relationship of pollen, symptoms and immune responses under a controlled regime of 'high-low-moderate' pollen exposure in urban versus alpine environment. The research was conducted in 2016 in two locations in Germany: urban Augsburg (494 m) and Schneefernerhaus (UFS) on Zugspitze mountain (2656 m). Monitoring of airborne pollen took place using Hirst-type volumetric traps. On UFS, both indoor and outdoor samples were taken. Grass pollen allergic human volunteers were monitored daily during the peak of the grass pollen season, in Augsburg, on UFS, then again in Augsburg. Nasal biosamples were obtained throughout the study to investigate immune responses. All symptoms decreased significantly during the stay on UFS and remained low even after the return to Augsburg. The same was observed for nasal total IgE and IgM levels and for nasal type 2 cytokines and chemokines. Augsburg showed higher pollen concentrations than those on UFS. At all sites, pollen were present throughout each day, but were more abundant in Augsburg during morning. On UFS, outdoor pollen levels were up to 6-fold higher than those indoors. Nasal, ocular and pulmonary symptoms correlated with current and previous days' pollen concentrations and relative humidity. Stays in low-exposure environments during the peak pollen season can be an efficient means of reducing allergic symptoms and immune responses. However, in alpine environments, even occasional pollen exposure during short intervals may still trigger symptoms because of the additional environmental stress posed onto allergics. This highlights the need for the consideration of additional environmental factors, apart from symptom diaries and immune responses, so as to efficiently predict high-risk allergy periods.
Collapse
Affiliation(s)
- Athanasios Damialis
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany.
| | - Franziska Häring
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Mehmet Gökkaya
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Denise Rauer
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Matthias Reiger
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Sebastian Bezold
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Nikolaos Bounas-Pyrros
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Kilian Eyerich
- Department of Dermatology, Technical University of Munich, Munich, Germany
| | - Antonia Todorova
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Gertrud Hammel
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Stefanie Gilles
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany
| | - Claudia Traidl-Hoffmann
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany; Christine Kühne Center for Allergy Research and Education (CK Care), Davos, Switzerland; Outpatient Clinic for Environmental Medicine, Klinikum Augsburg, Augsburg, Germany
| |
Collapse
|
23
|
Mrkić I, Minić R, Popović D, Živković I, Gavrović-Jankulović M. Newly designed hemagglutinin-Der p 2 chimera is a potential candidate for allergen specific immunotherapy. Life Sci 2018; 213:158-165. [PMID: 30352241 DOI: 10.1016/j.lfs.2018.10.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/18/2018] [Accepted: 10/18/2018] [Indexed: 01/01/2023]
Abstract
Aim To investigate the immunomodulatory potential of a chimera composed of the receptor-binding domain of hemagglutinin 1 (H1s) from Influenza virus and Der p 2 (D2) allergen for allergen-specific immunotherapy of house-dust mite allergy (HDM). MAIN METHODS H1sD2 chimera and D2 allergen were produced by genetic engineering in E. coli. Recombinant antigens were extracted from inclusion bodies by urea, then refolded and purified by immobilized- metal affinity chromatography (IMAC). Purity was verified by 2D-PAGE and secondary structures were assessed by CD spectroscopy. IgE reactivity of H1sD2 and D2 was tested in western blot with sera from 8 persons with clinical history of HDM allergy. Immunogenicity of H1sD2 and D2 were analyzed in Balb/c mice. Cytokine profile was analyzed by ELISA after stimulation of mouse spleen cells with H1sD2 and D2. Leukocyte population abundance of cells isolated from spleen and lymph node was assessed by flow cytometry. KEY FINDINGS Purified recombinant proteins H1sD2 (42 kDa) and D2 (15 kDa) revealed well defined secondary structures, and preserved IgE reactive epitopes. Analysis of supernatants of mouse spleen cells after stimulation with H1sD2 and D2, revealed a qualitatively different cytokine profile from H1sD2 immunized mouse cells (increase in IL10). CD8+ cells were decreased in the lymph node of D2 immunized mice, whereas H1sD2 immunization led to an increase of CD8+ cells in both the lymph node and the spleen. SIGNIFICANCE H1sD2 chimera attenuates Der p 2-inherent Th2 response and directs the immune response toward Th1 and Treg phenotype.
Collapse
Affiliation(s)
- Ivan Mrkić
- Innovation Center, University of Belgrade - Faculty of Chemistry, Belgrade, Serbia
| | - Rajna Minić
- Institute of Virology, Vaccines and Sera, Torlak, Belgrade, Serbia
| | - Dragan Popović
- Department of Chemistry - IChTM, University of Belgrade, Belgrade, Serbia
| | - Irena Živković
- Institute of Virology, Vaccines and Sera, Torlak, Belgrade, Serbia
| | | |
Collapse
|
24
|
Zhu C, Farah J, Choël M, Gosselin S, Baroudi M, Petitprez D, Visez N. Uptake of ozone and modification of lipids in Betula Pendula pollen. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 242:880-886. [PMID: 30041161 DOI: 10.1016/j.envpol.2018.07.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 06/08/2023]
Abstract
Pollen allergy risk is modified by air pollutants, including ozone, but the chemical modifications induced on pollen grains are poorly understood. Pollen lipidic extract has been shown to act as an adjuvant to the allergenic reaction and therefore, the modification of lipids by air pollutants could have health implications. Birch pollen was exposed in vitro to ozone to explore the reactivity of O3 on its surface and on its lipidic fraction. Uptake coefficients of ozone were determined for ozone concentration of 117 ppb on the surface of native birch pollen (8.6 ± 0.8 × 10-6), defatted pollen (9.9 ± 0.9 × 10-6), and for crushed pollen grains (34±3 × 10-6). The mass of ozone uptaken was increased by a factor of four for crushed pollen compared to native pollen showing a higher susceptibility to ozone of cytoplasmic granules and broken pollen grains. A total mass of extractible lipids of 27 mg per gram of birch pollen was found and a fraction of these lipids was identified and quantified (fatty acids, alkanes, alkenes and aldehydes). The distribution of lipids was modified by ozone exposure of 115 and 1000 ppb for 16 h with the following reactivity: consumption of alkene, formation of aldehydes and formation of nonanoic acid and octadecanoic acid. The quantity of ozone trapped in the lipidic fraction during 15 min at 115 ppb is enough to contribute to the reactivity of one-third of the alkenes demonstrating that pollen could be susceptible to an atmospheric increase of ozone concentration even for a very short duration complicating the understanding of the link between pollen allergy and pollution.
Collapse
Affiliation(s)
- Chao Zhu
- Environment Research Institute, Shandong University, Jinan 250100, China; Univ. Lille, CNRS, UMR 8522 - PC2A - Physicochimie des Processus de Combustion et de l'Atmosphère, F-59000 Lille, France
| | - Jinane Farah
- Univ. Lille, CNRS, UMR 8522 - PC2A - Physicochimie des Processus de Combustion et de l'Atmosphère, F-59000 Lille, France; Lebanese University, Faculty of Public Health (FSP III), Water & Environment Science, Tripoli, Lebanon
| | - Marie Choël
- Univ. Lille, CNRS, UMR 8516 - LASIR - Laboratoire de Spectrochimie Infrarouge et Raman, F-59000 Lille, France
| | - Sylvie Gosselin
- Univ. Lille, CNRS, UMR 8522 - PC2A - Physicochimie des Processus de Combustion et de l'Atmosphère, F-59000 Lille, France
| | - Moomen Baroudi
- Lebanese University, Faculty of Public Health (FSP III), Water & Environment Science, Tripoli, Lebanon
| | - Denis Petitprez
- Univ. Lille, CNRS, UMR 8522 - PC2A - Physicochimie des Processus de Combustion et de l'Atmosphère, F-59000 Lille, France
| | - Nicolas Visez
- Univ. Lille, CNRS, UMR 8522 - PC2A - Physicochimie des Processus de Combustion et de l'Atmosphère, F-59000 Lille, France.
| |
Collapse
|
25
|
Bedeutung von Klima- und Umweltschutz für die Gesundheit mit besonderer Berücksichtigung von Schädigungen der Hautbarriere und allergischen Folgeerkrankungen. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2018; 61:684-696. [DOI: 10.1007/s00103-018-2742-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
26
|
Lehmann I. [Environmental pollutants as adjuvant factors of immune system derived diseases]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2018; 60:592-596. [PMID: 28466130 DOI: 10.1007/s00103-017-2545-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The main task of the immune system is to protect the body against invading pathogens. To be able to do so, immune cells must be able to recognize and combat exogenous challenges and at the same time tolerate body-borne structures. A complex regulatory network controls the sensitive balance between defense and tolerance. Perturbation of this network ultimately leads to the development of chronic inflammation, such as allergies, autoimmune reactions, and infections, because the immune system is no longer able to efficiently eliminate invading pathogens. Environmental pollutants can cause such perturbations by affecting the function of immune cells in such a way that they would react hypersensitively against allergens and the body's own structures, respectively, or that they would be no longer able to adequately combat pathogens. This indirect effect is also known as adjuvant effect. For pesticides, heavy metals, wood preservatives, or volatile organic compounds such adjuvant effects are well known. Examples of the mechanism by which environmental toxins contribute to chronic inflammatory diseases are manifold and will be discussed along asthma and allergies.While the immune system of healthy adults is typically well able to distinguish between foreign and endogenous substances even under adverse environmental conditions, that of children would react much more sensible upon comparable environmental challenges. To prevent priming for diseases by environmental cues during that highly sensitive period of early childhood children are to be particularly protected.
Collapse
Affiliation(s)
- Irina Lehmann
- Department Umweltimmunologie, Helmholtz-Zentrum für Umweltforschung, Permoserstraße 15, 04318, Leipzig, Deutschland.
| |
Collapse
|
27
|
Strasser L, Dang HH, Schwarz H, Asam C, Ferreira F, Horejs-Hoeck J, Huber CG. Unbiased Quantitative Proteomics Reveals a Crucial Role of the Allergen Context for the Activation of Human Dendritic Cells. Sci Rep 2017; 7:16638. [PMID: 29192156 PMCID: PMC5709417 DOI: 10.1038/s41598-017-16726-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 11/16/2017] [Indexed: 01/01/2023] Open
Abstract
Worldwide, more than 1 billion people suffer from allergic diseases. However, until now it is not fully understood how certain proteins can induce allergic immune responses, while others cannot. Studies suggest that allergenicity is a process not only determined by properties of the allergen itself but also by costimulatory factors, that are not classically associated with allergic reactions. To investigate the allergenicity of the major birch pollen allergen Bet v 1 and the impact of adjuvants associated with pollen, e.g. lipopolysaccharide (LPS), we performed quantitative proteome analysis to study the activation of monocyte-derived dendritic cells (moDCs). Thus, we treated cells with birch pollen extract (BPE), recombinant Bet v 1, and LPS followed by proteomic profiling via high-performance liquid chromatography and tandem mass spectrometry (HPLC-MS/MS) using isobaric labelling. Enrichment and pathway analysis revealed the influence of regulated proteins especially in cytokine signalling and dendritic cell activation. We found highly regulated, but differentially expressed proteins after treatment with BPE and LPS, whereas the cellular response to Bet v 1 was limited. Our findings lead to the conclusion that Bet v 1 needs a specific “allergen context” involving cofactors apart from LPS to induce an immune response in human moDCs.
Collapse
Affiliation(s)
- L Strasser
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - H-H Dang
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - H Schwarz
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - C Asam
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - F Ferreira
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - J Horejs-Hoeck
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - C G Huber
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria.
| |
Collapse
|
28
|
Roberts G, Boyle R, Crane J, Hogan SP, Saglani S, Wickman M, Woodfolk JA. Developments in the field of allergy in 2016 through the eyes of Clinical and Experimental Allergy. Clin Exp Allergy 2017; 47:1512-1525. [PMID: 29068551 DOI: 10.1111/cea.13049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this article, we described the development in the field of allergy as described by Clinical and Experimental Allergy in 2016. Experimental models of allergic disease, basic mechanisms, clinical mechanisms, allergens, asthma and rhinitis, and clinical allergy are all covered.
Collapse
Affiliation(s)
- G Roberts
- Clinical and Experimental Sciences and Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Isle of Wight, UK
| | - R Boyle
- Department of Paediatrics, Imperial College London, London, UK
| | - J Crane
- Department of Medicine, University of Otago Wellington, Wellington, New Zealand
| | - S P Hogan
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - S Saglani
- National Heart & Lung Institute, Imperial College London, London, UK
| | - M Wickman
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - J A Woodfolk
- Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
29
|
Reinmuth-Selzle K, Kampf CJ, Lucas K, Lang-Yona N, Fröhlich-Nowoisky J, Shiraiwa M, Lakey PSJ, Lai S, Liu F, Kunert AT, Ziegler K, Shen F, Sgarbanti R, Weber B, Bellinghausen I, Saloga J, Weller MG, Duschl A, Schuppan D, Pöschl U. Air Pollution and Climate Change Effects on Allergies in the Anthropocene: Abundance, Interaction, and Modification of Allergens and Adjuvants. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:4119-4141. [PMID: 28326768 PMCID: PMC5453620 DOI: 10.1021/acs.est.6b04908] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/07/2017] [Accepted: 03/22/2017] [Indexed: 05/13/2023]
Abstract
Air pollution and climate change are potential drivers for the increasing burden of allergic diseases. The molecular mechanisms by which air pollutants and climate parameters may influence allergic diseases, however, are complex and elusive. This article provides an overview of physical, chemical and biological interactions between air pollution, climate change, allergens, adjuvants and the immune system, addressing how these interactions may promote the development of allergies. We reviewed and synthesized key findings from atmospheric, climate, and biomedical research. The current state of knowledge, open questions, and future research perspectives are outlined and discussed. The Anthropocene, as the present era of globally pervasive anthropogenic influence on planet Earth and, thus, on the human environment, is characterized by a strong increase of carbon dioxide, ozone, nitrogen oxides, and combustion- or traffic-related particulate matter in the atmosphere. These environmental factors can enhance the abundance and induce chemical modifications of allergens, increase oxidative stress in the human body, and skew the immune system toward allergic reactions. In particular, air pollutants can act as adjuvants and alter the immunogenicity of allergenic proteins, while climate change affects the atmospheric abundance and human exposure to bioaerosols and aeroallergens. To fully understand and effectively mitigate the adverse effects of air pollution and climate change on allergic diseases, several challenges remain to be resolved. Among these are the identification and quantification of immunochemical reaction pathways involving allergens and adjuvants under relevant environmental and physiological conditions.
Collapse
Affiliation(s)
| | - Christopher J. Kampf
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
- Institute
of Inorganic and Analytical Chemistry, Johannes
Gutenberg University, Mainz, 55128, Germany
| | - Kurt Lucas
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Naama Lang-Yona
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | | | - Manabu Shiraiwa
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
- Department
of Chemistry, University of California, Irvine, California 92697-2025, United States
| | - Pascale S. J. Lakey
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Senchao Lai
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
- South
China University of Technology, School of
Environment and Energy, Guangzhou, 510006, China
| | - Fobang Liu
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Anna T. Kunert
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Kira Ziegler
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Fangxia Shen
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Rossella Sgarbanti
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Bettina Weber
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Iris Bellinghausen
- Department
of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, 55131, Germany
| | - Joachim Saloga
- Department
of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, 55131, Germany
| | - Michael G. Weller
- Division
1.5 Protein Analysis, Federal Institute
for Materials Research and Testing (BAM), Berlin, 12489, Germany
| | - Albert Duschl
- Department
of Molecular Biology, University of Salzburg, 5020 Salzburg, Austria
| | - Detlef Schuppan
- Institute
of Translational Immunology and Research Center for Immunotherapy,
Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, 55131 Germany
- Division
of Gastroenterology, Beth Israel Deaconess
Medical Center and Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Ulrich Pöschl
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| |
Collapse
|
30
|
Atopic dermatitis: new evidence on the role of allergic inflammation. Curr Opin Allergy Clin Immunol 2016; 16:458-64. [DOI: 10.1097/aci.0000000000000308] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|