1
|
Tisch LJ, Bartone RD, Antoniak S, Bonner JC. Protease-activated receptor-2 (PAR2) mutation attenuates airway fibrosis in mice during the exacerbation of house dust mite‑induced allergic lung disease by multi‑walled carbon nanotubes. Respir Res 2025; 26:90. [PMID: 40057754 PMCID: PMC11889904 DOI: 10.1186/s12931-025-03168-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Pulmonary exposure to multi-walled carbon nanotubes (MWCNTs) induces potent pro-inflammatory and pro-fibrotic responses in mouse models of allergic lung disease. We recently reported that MWCNTs exacerbated components of house dust mite (HDM)-induced allergic lung disease, including eosinophilic inflammation, mucous cell metaplasia and airway fibrosis. Protease-activated receptor 2 (PAR2) plays a significant role in the development of various respiratory diseases, including asthma and pulmonary fibrosis. However, studies investigating the function of PAR2 in allergic lung disease have produced variable results. To further define the role of PAR2 in pulmonary pathology, we investigated the effects of MWCNTs on HDM-induced allergic lung disease in PAR2-mutant mice. METHODS The PAR2-mutant mice used were previously generated by replacing a 1.8-kb region of the PAR2 coding sequence with a neomycin resistance gene, which did not entirely delete the gene. Wild-type (WT) male C57BL/6J mice and PAR2-mutant male mice were exposed to a vehicle solution, MWCNTs, HDM extract, or both via oropharyngeal aspiration six times over 3 weeks. Bronchoalveolar lavage fluid (BALF) was collected to measure changes in inflammatory cells, total protein, and lactate dehydrogenase (LDH). Lung protein and mRNA were assayed for pro-inflammatory and profibrotic mediators, and formalin-fixed lung sections were evaluated for histopathology. RESULTS In WT and PAR2-mutant mice, co-exposure to MWCNTs and HDM extract significantly increased eosinophilic lung inflammation, mucous cell metaplasia, increased BALF cellularity, BALF total protein, and LDH levels. These results were not significantly different between genotypes. Additionally, MWCNTs and HDM extract co-exposure significantly increased airway fibrosis in WT and PAR2-mutant mice, characterized by increased airway collagen deposition and Col1a1 mRNA expression. Quantitative morphometry revealed a significant decrease in airway fibrosis in PAR2-mutant mice compared to WT mice, accompanied by reduced Col1a1 mRNA as detected by PCR. Despite this reduction, the pro-fibrotic mediator arginase 1 (Arg-1) protein and mRNA levels were significantly upregulated in PAR2-mutant mice. CONCLUSION Our study demonstrates that PAR2 mediates airway fibrosis but does not influence eosinophilic lung inflammation or mucous cell metaplasia caused by co-exposure to MWCNTs and HDM allergen.
Collapse
Affiliation(s)
- Logan J Tisch
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Ryan D Bartone
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Silvio Antoniak
- UNC Blood Research Center, Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James C Bonner
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
2
|
Xu K, Wang L, Lin M, He G. Update on protease-activated receptor 2 in inflammatory and autoimmune dermatological diseases. Front Immunol 2024; 15:1449126. [PMID: 39364397 PMCID: PMC11446762 DOI: 10.3389/fimmu.2024.1449126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024] Open
Abstract
Protease-activated receptor 2 (PAR2) is a cell-surface receptor expressed in various cell types, including keratinocytes, neurons, immune and inflammatory cells. Activation of PAR2, whether via its canonical or biased pathways, triggers a series of signaling cascades that mediate numerous functions. This review aims to highlight the emerging roles and interactions of PAR2 in different skin cells. It specifically summarizes the latest insights into the roles of PAR2 in skin conditions such as atopic dermatitis (AD), psoriasis, vitiligo and melasma. It also considers these roles from the perspective of the cutaneous microenvironment in relation to other inflammatory and autoimmune dermatological disorders. Additionally, the review explores PAR2's involvement in associated comorbidities from both cutaneous and extracutaneous diseases. Therefore, PAR2 may serve as a key target for interactions among various cells within the local skin environment.
Collapse
Affiliation(s)
- Kejia Xu
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Mao Lin
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Gu He
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Cao TBT, Luu Quoc Q, Jang JH, Yang EM, Ryu MS, Choi Y, Park HS. Serum Galectin-10: A biomarker for persistent airflow limitation in adult asthmatics. World Allergy Organ J 2024; 17:100955. [PMID: 39252790 PMCID: PMC11382115 DOI: 10.1016/j.waojou.2024.100955] [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: 02/13/2024] [Revised: 06/25/2024] [Accepted: 08/01/2024] [Indexed: 09/11/2024] Open
Abstract
Background Inhaled corticosteroids (ICS) are primary anti-inflammatory medications to control eosinophilic airway inflammation, and prevent asthma exacerbation. However, persistent airflow limitation (PAL) presents in some asthmatics even on ICS treatment, leading to lung function decline. Thus, we evaluated clinical associations of serum galectin-10 (Gal10) and galectin-3 (Gal3) levels in adult asthmatics who had maintained anti-asthma medication. Methods Sixty-seven asthmatics and 78 healthy controls (HCs) were recruited. Serum Gal10 and Gal3 levels were measured by enzyme-linked immunosorbent assay, and their clinical relevance with inflammatory and lung function parameters was evaluated. Spirometry was performed to assess PAL and small airway dysfunction (SAD). Airway epithelial cells were cocultured with eosinophils/neutrophils, and were exposed to house dust mites to assess the production of Gal10 and Gal3. Results Serum Gal10 (not Gal3) levels were significantly higher in asthmatics than in HCs (P < 0.001), in asthmatics with PAL than in those without PAL (P = 0.005), and in those with SAD than in those without SAD (P = 0.004). The Gal10-high group had significantly higher levels of peripheral CD66+ neutrophil counts, serum periostin and Gal3, and lower values of FEV1% and MMEF% than the Gal10-low group (P < 0.050 for all). The production of Gal10 and Gal3 was increased in eosinophilic airway model, while Gal10 (not Gal3) levels were increased in neutrophilic airway model as well as house dust mite stimulation. Conclusion Our findings suggest that serum Gal10 level may be a potential biomarker for PAL in adult asthmatics.
Collapse
Affiliation(s)
- Thi Bich Tra Cao
- Department of Allergy and Clinical Immunology, Ajou University Medical Center, Suwon, South Korea
| | - Quang Luu Quoc
- Department of Oral & Maxillofacial Surgery, Loma Linda University, School of Dentistry, CA, USA
| | - Jae-Hyuk Jang
- Department of Allergy and Clinical Immunology, Ajou University Medical Center, Suwon, South Korea
| | - Eun-Mi Yang
- Department of Allergy and Clinical Immunology, Ajou University Medical Center, Suwon, South Korea
| | - Min Sook Ryu
- Department of Allergy and Clinical Immunology, Ajou University Medical Center, Suwon, South Korea
| | - Youngwoo Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, South Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University Medical Center, Suwon, South Korea
| |
Collapse
|
4
|
Baglivo I, Quaranta VN, Dragonieri S, Colantuono S, Menzella F, Selvaggio D, Carpagnano GE, Caruso C. The New Paradigm: The Role of Proteins and Triggers in the Evolution of Allergic Asthma. Int J Mol Sci 2024; 25:5747. [PMID: 38891935 PMCID: PMC11171572 DOI: 10.3390/ijms25115747] [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: 04/18/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Epithelial barrier damage plays a central role in the development and maintenance of allergic inflammation. Rises in the epithelial barrier permeability of airways alter tissue homeostasis and allow the penetration of allergens and other external agents. Different factors contribute to barrier impairment, such as eosinophilic infiltration and allergen protease action-eosinophilic cationic proteins' effects and allergens' proteolytic activity both contribute significantly to epithelial damage. In the airways, allergen proteases degrade the epithelial junctional proteins, allowing allergen penetration and its uptake by dendritic cells. This increase in allergen-immune system interaction induces the release of alarmins and the activation of type 2 inflammatory pathways, causing or worsening the main symptoms at the skin, bowel, and respiratory levels. We aim to highlight the molecular mechanisms underlying allergenic protease-induced epithelial barrier damage and the role of immune response in allergic asthma onset, maintenance, and progression. Moreover, we will explore potential clinical and radiological biomarkers of airway remodeling in allergic asthma patients.
Collapse
Affiliation(s)
- Ilaria Baglivo
- Centro Malattie Apparato Digerente (CEMAD) Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Vitaliano Nicola Quaranta
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Silvano Dragonieri
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Stefania Colantuono
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Francesco Menzella
- Pulmonology Unit, S. Valentino Hospital-AULSS2 Marca Trevigiana, 31100 Treviso, Italy
| | - David Selvaggio
- UOS di Malattie dell’Apparato Respiratorio Ospedale Cristo Re, 00167 Roma, Italy
| | - Giovanna Elisiana Carpagnano
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Cristiano Caruso
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
5
|
Ouyang X, Reihill JA, Douglas LEJ, Martin SL. Airborne indoor allergen serine proteases and their contribution to sensitisation and activation of innate immunity in allergic airway disease. Eur Respir Rev 2024; 33:230126. [PMID: 38657996 PMCID: PMC11040391 DOI: 10.1183/16000617.0126-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 02/28/2024] [Indexed: 04/26/2024] Open
Abstract
Common airborne allergens (pollen, animal dander and those from fungi and insects) are the main triggers of type I allergic disorder in the respiratory system and are associated with allergic rhinitis, allergic asthma, as well as immunoglobulin E (IgE)-mediated allergic bronchopulmonary aspergillosis. These allergens promote IgE crosslinking, vasodilation, infiltration of inflammatory cells, mucosal barrier dysfunction, extracellular matrix deposition and smooth muscle spasm, which collectively cause remodelling of the airways. Fungus and insect (house dust mite and cockroaches) indoor allergens are particularly rich in proteases. Indeed, more than 40 different types of aeroallergen proteases, which have both IgE-neutralising and tissue-destructive activities, have been documented in the Allergen Nomenclature database. Of all the inhaled protease allergens, 85% are classed as serine protease activities and include trypsin-like, chymotrypsin-like and collagenolytic serine proteases. In this article, we review and compare the allergenicity and proteolytic effect of allergen serine proteases as listed in the Allergen Nomenclature and MEROPS databases and highlight their contribution to allergic sensitisation, disruption of the epithelial barrier and activation of innate immunity in allergic airways disease. The utility of small-molecule inhibitors of allergen serine proteases as a potential treatment strategy for allergic airways disease will also be discussed.
Collapse
Affiliation(s)
- Xuan Ouyang
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | | | | | | |
Collapse
|
6
|
Brister DL, Omer H, Whetstone CE, Ranjbar M, Gauvreau GM. Multifactorial Causes and Consequences of TLSP Production, Function, and Release in the Asthmatic Airway. Biomolecules 2024; 14:401. [PMID: 38672419 PMCID: PMC11048646 DOI: 10.3390/biom14040401] [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: 02/29/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Disruption of the airway epithelium triggers a defensive immune response that begins with the production and release of alarmin cytokines. These epithelial-derived alarmin cytokines, including thymic stromal lymphopoietin (TSLP), are produced in response to aeroallergens, viruses, and toxic inhalants. An alarmin response disproportionate to the inhaled trigger can exacerbate airway diseases such as asthma. Allergens inhaled into previously sensitized airways are known to drive a T2 inflammatory response through the polarization of T cells by dendritic cells mediated by TSLP. Harmful compounds found within air pollution, microbes, and viruses are also triggers causing airway epithelial cell release of TSLP in asthmatic airways. The release of TSLP leads to the development of inflammation which, when unchecked, can result in asthma exacerbations. Genetic and inheritable factors can contribute to the variable expression of TSLP and the risk and severity of asthma. This paper will review the various triggers and consequences of TSLP release in asthmatic airways.
Collapse
Affiliation(s)
| | | | | | | | - Gail M. Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (D.L.B.); (H.O.); (C.E.W.); (M.R.)
| |
Collapse
|
7
|
Soh WT, Zhang J, Hollenberg MD, Vliagoftis H, Rothenberg ME, Sokol CL, Robinson C, Jacquet A. Protease allergens as initiators-regulators of allergic inflammation. Allergy 2023; 78:1148-1168. [PMID: 36794967 PMCID: PMC10159943 DOI: 10.1111/all.15678] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/05/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Tremendous progress in the last few years has been made to explain how seemingly harmless environmental proteins from different origins can induce potent Th2-biased inflammatory responses. Convergent findings have shown the key roles of allergens displaying proteolytic activity in the initiation and progression of the allergic response. Through their propensity to activate IgE-independent inflammatory pathways, certain allergenic proteases are now considered as initiators for sensitization to themselves and to non-protease allergens. The protease allergens degrade junctional proteins of keratinocytes or airway epithelium to facilitate allergen delivery across the epithelial barrier and their subsequent uptake by antigen-presenting cells. Epithelial injuries mediated by these proteases together with their sensing by protease-activated receptors (PARs) elicit potent inflammatory responses resulting in the release of pro-Th2 cytokines (IL-6, IL-25, IL-1β, TSLP) and danger-associated molecular patterns (DAMPs; IL-33, ATP, uric acid). Recently, protease allergens were shown to cleave the protease sensor domain of IL-33 to produce a super-active form of the alarmin. At the same time, proteolytic cleavage of fibrinogen can trigger TLR4 signaling, and cleavage of various cell surface receptors further shape the Th2 polarization. Remarkably, the sensing of protease allergens by nociceptive neurons can represent a primary step in the development of the allergic response. The goal of this review is to highlight the multiple innate immune mechanisms triggered by protease allergens that converge to initiate the allergic response.
Collapse
Affiliation(s)
- Wai Tuck Soh
- Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jihui Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Morley D. Hollenberg
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Harissios Vliagoftis
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, and Alberta Respiratory Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Caroline L. Sokol
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clive Robinson
- Institute for Infection and Immunity, St George’s University of London, London, UK
| | - Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
8
|
Kopp EB, Agaronyan K, Licona-Limón I, Nish SA, Medzhitov R. Modes of type 2 immune response initiation. Immunity 2023; 56:687-694. [PMID: 37044059 DOI: 10.1016/j.immuni.2023.03.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023]
Abstract
Type 2 immunity defends against macro-parasites and can cause allergic diseases. Our understanding of the mechanisms governing the initiation of type 2 immunity is limited, whereas we know more about type 1 immune responses. Type 2 immunity can be triggered by a wide array of inducers that do not share common features and via diverse pathways and mechanisms. To address the complexity of the type 2 initiation pathways, we suggest a framework that conceptualizes different modes of induction of type 2 immunity. We discuss categories of type 2 inducers and their immunogenicity, types of tissue perturbations that are caused by these inducers, sensing strategies for the initiation of Th2 immune responses, and categorization of the signals that are produced in response to type 2 challenges. We describe tissue-specific examples of functional disruption that could lead to type 2 inflammation and propose that different sensing strategies that operate at the tissue level converge on the initiation of type 2 immune responses.
Collapse
Affiliation(s)
- Elizabeth B Kopp
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Karen Agaronyan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, New Haven, CT 06510, USA
| | - Ileana Licona-Limón
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Simone A Nish
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, New Haven, CT 06510, USA; Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
9
|
Miller MH, Swaby LG, Vailoces VS, LaFratta M, Zhang Y, Zhu X, Hitchcock DJ, Jewett TJ, Zhang B, Tigno-Aranjuez JT. LMAN1 is a receptor for house dust mite allergens. Cell Rep 2023; 42:112208. [PMID: 36870056 PMCID: PMC10105285 DOI: 10.1016/j.celrep.2023.112208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/01/2022] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Development of therapies with the potential to change the allergic asthmatic disease course will require the discovery of targets that play a central role during the initiation of an allergic response, such as those involved in the process of allergen recognition. We use a receptor glycocapture technique to screen for house dust mite (HDM) receptors and identify LMAN1 as a candidate. We verify the ability of LMAN1 to directly bind HDM allergens and demonstrate that LMAN1 is expressed on the surface of dendritic cells (DCs) and airway epithelial cells (AECs) in vivo. Overexpression of LMAN1 downregulates NF-κB signaling in response to inflammatory cytokines or HDM. HDM promotes binding of LMAN1 to the FcRγ and recruitment of SHP1. Last, peripheral DCs of asthmatic individuals show a significant reduction in the expression of LMAN1 compared with healthy controls. These findings have potential implications for the development of therapeutic interventions for atopic disease.
Collapse
Affiliation(s)
- Madelyn H Miller
- Biotechnology and Immunology Research, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Lindsay G Swaby
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Vanessa S Vailoces
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Maggie LaFratta
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Yuan Zhang
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Xiang Zhu
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Dorilyn J Hitchcock
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Travis J Jewett
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Bin Zhang
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Justine T Tigno-Aranjuez
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.
| |
Collapse
|
10
|
Jacquet A. The HDM allergen orchestra and its cysteine protease maestro: Stimulators of kaleidoscopic innate immune responses. Mol Immunol 2023; 156:48-60. [PMID: 36889186 DOI: 10.1016/j.molimm.2023.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/29/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023]
Abstract
House dust mite (HDM) encloses an explosive cocktail of allergenic proteins sensitizing hundreds of millions of people worldwide. To date, the innate cellular and molecular mechanism(s) orchestrating the HDM-induced allergic inflammation remains partially deciphered. Understanding the kaleidoscope of HDM-induced innate immune responses is hampered by (1) the large complexity of the HDM allergome with very diverse functional bioreactivities, (2) the perpetual presence of microbial compounds (at least LPS, β-glucan, chitin) promoting as well pro-Th2 innate signaling pathways and (3) multiple cross-talks involving structural, neuronal and immune cells. The present review provides an update on the innate immune properties, identified so far, of multiple HDM allergen groups. Experimental evidence highlights the importance of HDM allergens displaying protease or lipid-binding activities on the initiation of the allergic responses. Specifically, group 1 HDM cysteine proteases are considered as the key initiators of the allergic response through their capacities to impair the epithelial barrier integrity, to stimulate the release of pro-Th2 danger-associated molecular patterns (DAMPs) in epithelial cells, to produce super-active forms of IL-33 alarmin and to mature thrombin leading to Toll-like receptor 4 (TLR4) activation. Remarkably, the recently evidenced primary sensing of cysteine protease allergens by nociceptive neurons confirms the critical role of this HDM allergen group in the early events leading to Th2 differentiation.
Collapse
Affiliation(s)
- Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
11
|
Dramburg S, Hilger C, Santos AF, de Las Vecillas L, Aalberse RC, Acevedo N, Aglas L, Altmann F, Arruda KL, Asero R, Ballmer-Weber B, Barber D, Beyer K, Biedermann T, Bilo MB, Blank S, Bosshard PP, Breiteneder H, Brough HA, Bublin M, Campbell D, Caraballo L, Caubet JC, Celi G, Chapman MD, Chruszcz M, Custovic A, Czolk R, Davies J, Douladiris N, Eberlein B, Ebisawa M, Ehlers A, Eigenmann P, Gadermaier G, Giovannini M, Gomez F, Grohman R, Guillet C, Hafner C, Hamilton RG, Hauser M, Hawranek T, Hoffmann HJ, Holzhauser T, Iizuka T, Jacquet A, Jakob T, Janssen-Weets B, Jappe U, Jutel M, Kalic T, Kamath S, Kespohl S, Kleine-Tebbe J, Knol E, Knulst A, Konradsen JR, Korošec P, Kuehn A, Lack G, Le TM, Lopata A, Luengo O, Mäkelä M, Marra AM, Mills C, Morisset M, Muraro A, Nowak-Wegrzyn A, Nugraha R, Ollert M, Palosuo K, Pastorello EA, Patil SU, Platts-Mills T, Pomés A, Poncet P, Potapova E, Poulsen LK, Radauer C, Radulovic S, Raulf M, Rougé P, Sastre J, Sato S, Scala E, Schmid JM, Schmid-Grendelmeier P, Schrama D, Sénéchal H, Traidl-Hoffmann C, Valverde-Monge M, van Hage M, van Ree R, Verhoeckx K, Vieths S, Wickman M, Zakzuk J, Matricardi PM, et alDramburg S, Hilger C, Santos AF, de Las Vecillas L, Aalberse RC, Acevedo N, Aglas L, Altmann F, Arruda KL, Asero R, Ballmer-Weber B, Barber D, Beyer K, Biedermann T, Bilo MB, Blank S, Bosshard PP, Breiteneder H, Brough HA, Bublin M, Campbell D, Caraballo L, Caubet JC, Celi G, Chapman MD, Chruszcz M, Custovic A, Czolk R, Davies J, Douladiris N, Eberlein B, Ebisawa M, Ehlers A, Eigenmann P, Gadermaier G, Giovannini M, Gomez F, Grohman R, Guillet C, Hafner C, Hamilton RG, Hauser M, Hawranek T, Hoffmann HJ, Holzhauser T, Iizuka T, Jacquet A, Jakob T, Janssen-Weets B, Jappe U, Jutel M, Kalic T, Kamath S, Kespohl S, Kleine-Tebbe J, Knol E, Knulst A, Konradsen JR, Korošec P, Kuehn A, Lack G, Le TM, Lopata A, Luengo O, Mäkelä M, Marra AM, Mills C, Morisset M, Muraro A, Nowak-Wegrzyn A, Nugraha R, Ollert M, Palosuo K, Pastorello EA, Patil SU, Platts-Mills T, Pomés A, Poncet P, Potapova E, Poulsen LK, Radauer C, Radulovic S, Raulf M, Rougé P, Sastre J, Sato S, Scala E, Schmid JM, Schmid-Grendelmeier P, Schrama D, Sénéchal H, Traidl-Hoffmann C, Valverde-Monge M, van Hage M, van Ree R, Verhoeckx K, Vieths S, Wickman M, Zakzuk J, Matricardi PM, Hoffmann-Sommergruber K. EAACI Molecular Allergology User's Guide 2.0. Pediatr Allergy Immunol 2023; 34 Suppl 28:e13854. [PMID: 37186333 DOI: 10.1111/pai.13854] [Show More Authors] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 05/17/2023]
Abstract
Since the discovery of immunoglobulin E (IgE) as a mediator of allergic diseases in 1967, our knowledge about the immunological mechanisms of IgE-mediated allergies has remarkably increased. In addition to understanding the immune response and clinical symptoms, allergy diagnosis and management depend strongly on the precise identification of the elicitors of the IgE-mediated allergic reaction. In the past four decades, innovations in bioscience and technology have facilitated the identification and production of well-defined, highly pure molecules for component-resolved diagnosis (CRD), allowing a personalized diagnosis and management of the allergic disease for individual patients. The first edition of the "EAACI Molecular Allergology User's Guide" (MAUG) in 2016 rapidly became a key reference for clinicians, scientists, and interested readers with a background in allergology, immunology, biology, and medicine. Nevertheless, the field of molecular allergology is moving fast, and after 6 years, a new EAACI Taskforce was established to provide an updated document. The Molecular Allergology User's Guide 2.0 summarizes state-of-the-art information on allergen molecules, their clinical relevance, and their application in diagnostic algorithms for clinical practice. It is designed for both, clinicians and scientists, guiding health care professionals through the overwhelming list of different allergen molecules available for testing. Further, it provides diagnostic algorithms on the clinical relevance of allergenic molecules and gives an overview of their biology, the basic mechanisms of test formats, and the application of tests to measure allergen exposure.
Collapse
Affiliation(s)
- Stephanie Dramburg
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christiane Hilger
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
- Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital, London, United Kingdom
| | | | - Rob C Aalberse
- Sanquin Research, Dept Immunopathology, University of Amsterdam, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia, Colombia
| | - Lorenz Aglas
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Salzburg, Austria
| | - Friedrich Altmann
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Karla L Arruda
- Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Sao Paulo, Brasil, Brazil
| | - Riccardo Asero
- Ambulatorio di Allergologia, Clinica San Carlo, Paderno Dugnano, Italy
| | - Barbara Ballmer-Weber
- Klinik für Dermatologie und Allergologie, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Domingo Barber
- Institute of Applied Molecular Medicine Nemesio Diez (IMMAND), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
- RETIC ARADyAL and RICORS Enfermedades Inflamatorias (REI), Madrid, Spain
| | - Kirsten Beyer
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University Munich, Munich, Germany
| | - Maria Beatrice Bilo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
- Allergy Unit Department of Internal Medicine, University Hospital Ospedali Riuniti di Ancona, Torrette, Italy
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
| | - Philipp P Bosshard
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Helen A Brough
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Merima Bublin
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Dianne Campbell
- Department of Allergy and Immunology, Children's Hospital at Westmead, Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
- Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia, Colombia
| | - Jean Christoph Caubet
- Pediatric Allergy Unit, Department of Child and Adolescent, University Hospitals of Geneva, Geneva, Switzerland
| | - Giorgio Celi
- Centro DH Allergologia e Immunologia Clinica ASST- MANTOVA (MN), Mantova, Italy
| | | | - Maksymilian Chruszcz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Adnan Custovic
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Rebecca Czolk
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Janet Davies
- Queensland University of Technology, Centre for Immunology and Infection Control, School of Biomedical Sciences, Herston, Queensland, Australia
- Metro North Hospital and Health Service, Emergency Operations Centre, Herston, Queensland, Australia
| | - Nikolaos Douladiris
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Bernadette Eberlein
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University Munich, Munich, Germany
| | - Motohiro Ebisawa
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Kanagawa, Japan
| | - Anna Ehlers
- Chemical Biology and Drug Discovery, Utrecht University, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Philippe Eigenmann
- Pediatric Allergy Unit, Department of Child and Adolescent, University Hospitals of Geneva, Geneva, Switzerland
| | - Gabriele Gadermaier
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Salzburg, Austria
| | - Mattia Giovannini
- Allergy Unit, Department of Pediatrics, Meyer Children's University Hospital, Florence, Italy
| | - Francisca Gomez
- Allergy Unit IBIMA-Hospital Regional Universitario de Malaga, Malaga, Spain
- Spanish Network for Allergy research RETIC ARADyAL, Malaga, Spain
| | - Rebecca Grohman
- NYU Langone Health, Department of Internal Medicine, New York, New York, USA
| | - Carole Guillet
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Christine Hafner
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Robert G Hamilton
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Hauser
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Salzburg, Austria
| | - Thomas Hawranek
- Department of Dermatology and Allergology, Paracelsus Private Medical University, Salzburg, Austria
| | - Hans Jürgen Hoffmann
- Institute for Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | | | - Tomona Iizuka
- Laboratory of Protein Science, Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thilo Jakob
- Department of Dermatology and Allergology, University Medical Center, Justus Liebig University Gießen, Gießen, Germany
| | - Bente Janssen-Weets
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Uta Jappe
- Division of Clinical and Molecular Allergology, Priority Research Area Asthma and Allergy, Research Center Borstel, Borstel, Germany
- Leibniz Lung Center, Airway Research Center North (ARCN), Member of the German Center for Lung Research, Germany
- Interdisciplinary Allergy Outpatient Clinic, Dept. of Pneumology, University of Lübeck, Lübeck, Germany
| | - Marek Jutel
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland
| | - Tanja Kalic
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, St. Poelten, Austria
| | - Sandip Kamath
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
- Molecular Allergy Research Laboratory, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
| | - Sabine Kespohl
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr- Universität Bochum, Bochum, Germany
| | - Jörg Kleine-Tebbe
- Allergy & Asthma Center Westend, Outpatient Clinic and Clinical Research Center, Berlin, Germany
| | - Edward Knol
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - André Knulst
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jon R Konradsen
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Korošec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Annette Kuehn
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Gideon Lack
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
- Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Thuy-My Le
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Andreas Lopata
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
- Molecular Allergy Research Laboratory, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
| | - Olga Luengo
- RETIC ARADyAL and RICORS Enfermedades Inflamatorias (REI), Madrid, Spain
- Allergy Section, Internal Medicine Department, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mika Mäkelä
- Division of Allergy, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Pediatric Department, Skin and Allergy Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | | | - Clare Mills
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | | | - Antonella Muraro
- Food Allergy Referral Centre, Department of Woman and Child Health, Padua University Hospital, Padua, Italy
| | - Anna Nowak-Wegrzyn
- Division of Pediatric Allergy and Immunology, NYU Grossman School of Medicine, Hassenfeld Children's Hospital, New York, New York, USA
- Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Roni Nugraha
- Molecular Allergy Research Laboratory, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
- Department of Aquatic Product Technology, Faculty of Fisheries and Marine Science, IPB University, Bogor, Indonesia
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Kati Palosuo
- Department of Allergology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | | | - Sarita Ulhas Patil
- Division of Rheumatology, Allergy and Immunology, Departments of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Division of Allergy and Immunology, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Thomas Platts-Mills
- Division of Allergy and Clinical Immunology, University of Virginia, Charlottesville, Virginia, USA
| | | | - Pascal Poncet
- Institut Pasteur, Immunology Department, Paris, France
- Allergy & Environment Research Team Armand Trousseau Children Hospital, APHP, Paris, France
| | - Ekaterina Potapova
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Lars K Poulsen
- Allergy Clinic, Department of Dermatology and Allergy, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
| | - Christian Radauer
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Suzana Radulovic
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
- Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Monika Raulf
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr- Universität Bochum, Bochum, Germany
| | - Pierre Rougé
- UMR 152 PharmaDev, IRD, Université Paul Sabatier, Faculté de Pharmacie, Toulouse, France
| | - Joaquin Sastre
- Allergy Service, Fundación Jiménez Díaz; CIBER de Enfermedades Respiratorias (CIBERES); Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
| | - Sakura Sato
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Enrico Scala
- Clinical and Laboratory Molecular Allergy Unit - IDI- IRCCS, Fondazione L M Monti Rome, Rome, Italy
| | - Johannes M Schmid
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Schmid-Grendelmeier
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Christine Kühne Center for Allergy Research and Education CK-CARE, Davos, Switzerland
| | - Denise Schrama
- Centre of Marine Sciences (CCMAR), Universidade do Algarve, Faro, Portugal
| | - Hélène Sénéchal
- Allergy & Environment Research Team Armand Trousseau Children Hospital, APHP, Paris, France
| | - Claudia Traidl-Hoffmann
- Christine Kühne Center for Allergy Research and Education CK-CARE, Davos, Switzerland
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Marcela Valverde-Monge
- Allergy Service, Fundación Jiménez Díaz; CIBER de Enfermedades Respiratorias (CIBERES); Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
| | - Marianne van Hage
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ronald van Ree
- Department of Experimental Immunology and Department of Otorhinolaryngology, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Kitty Verhoeckx
- Department of Immunology and Dermatology/ Allergology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Stefan Vieths
- Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Magnus Wickman
- Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Josefina Zakzuk
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia, Colombia
| | - Paolo M Matricardi
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
12
|
Gandhi VD, Shrestha Palikhe N, Vliagoftis H. Protease-activated receptor-2: Role in asthma pathogenesis and utility as a biomarker of disease severity. Front Med (Lausanne) 2022; 9:954990. [PMID: 35966869 PMCID: PMC9372307 DOI: 10.3389/fmed.2022.954990] [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: 05/27/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
PAR2, a receptor activated by serine proteases, has primarily pro-inflammatory roles in the airways and may play a role in asthma pathogenesis. PAR2 exerts its effects in the lungs through activation of a variety of airway cells, but also activation of circulating immune cells. There is evidence that PAR2 expression increases in asthma and other inflammatory diseases, although the regulation of PAR2 expression is not fully understood. Here we review the available literature on the potential role of PAR2 in asthma pathogenesis and propose a model of PAR2-mediated development of allergic sensitization. We also propose, based on our previous work, that PAR2 expression on peripheral blood monocyte subsets has the potential to serve as a biomarker of asthma severity and/or control.
Collapse
Affiliation(s)
- Vivek Dipak Gandhi
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Alberta Respiratory Centre, University of Alberta, Edmonton, AB, Canada
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Nami Shrestha Palikhe
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Alberta Respiratory Centre, University of Alberta, Edmonton, AB, Canada
| | - Harissios Vliagoftis
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Alberta Respiratory Centre, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Harissios Vliagoftis,
| |
Collapse
|
13
|
Whetstone CE, Ranjbar M, Omer H, Cusack RP, Gauvreau GM. The Role of Airway Epithelial Cell Alarmins in Asthma. Cells 2022; 11:1105. [PMID: 35406669 PMCID: PMC8997824 DOI: 10.3390/cells11071105] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/01/2023] Open
Abstract
The airway epithelium is the first line of defense for the lungs, detecting inhaled environmental threats through pattern recognition receptors expressed transmembrane or intracellularly. Activation of pattern recognition receptors triggers the release of alarmin cytokines IL-25, IL-33, and TSLP. These alarmins are important mediators of inflammation, with receptors widely expressed in structural cells as well as innate and adaptive immune cells. Many of the key effector cells in the allergic cascade also produce alarmins, thereby contributing to the airways disease by driving downstream type 2 inflammatory processes. Randomized controlled clinical trials have demonstrated benefit when blockade of TSLP and IL-33 were added to standard of care medications, suggesting these are important new targets for treatment of asthma. With genome-wide association studies demonstrating associations between single-nucleotide polymorphisms of the TSLP and IL-33 gene and risk of asthma, it will be important to understand which subsets of asthma patients will benefit most from anti-alarmin therapy.
Collapse
Affiliation(s)
| | | | | | | | - Gail M. Gauvreau
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (C.E.W.); (M.R.); (H.O.); (R.P.C.)
| |
Collapse
|
14
|
Karaguzel D, Sarac BE, Akel Bilgic H, Summak GY, Unal MA, Kalayci O, Karaaslan C. House dust mite-derived allergens effect on matrix metalloproteases in airway epithelial cells. Exp Lung Res 2021; 47:436-450. [PMID: 34739337 DOI: 10.1080/01902148.2021.1998734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aim of the Study: Many allergens have protease activities. Although the immunomodulatory effects of these antigens are well known, the effects attributed to their protease activities are not thoroughly investigated. We set out to determine the effects of house dust mite (HDM) allergens with varying protease activities on bronchial epithelial cell functions. Materials and methods: BEAS-2B cells were maintained in ALI-culture and stimulated with Der p1 (cysteine protease), Der p6 (serine protease), and Der p2 (non-protease) with and without specific protease inhibitors or heat denaturation. Cell viability and epithelial permeability were measured with MTT and paracellular flux assay, respectively. The effect of heat denaturation on allergen structure was examined using in silico models. Matrix metalloproteinases (MMPs) were investigated at the transcription (qPCR), protein (ELISA), and functional (zymography) levels. Results: Epithelial permeability increased only after Der p6 but not after Der p1 or Der p2 stimulation. Der p2 increased both MMP-2 and MMP-9 expression, while Der p1 increased only MMP-9 expression. The heat-denatured form of Der p1 unexpectedly increased MMP-9 gene expression, which, through the use of in silico models, was attributed to its ability to change receptor connections by the formation of new electrostatic and hydrogen bonds. IL-8 and GM-CSF production were increased after Der p1 and Der p2 but decreased after Der p6 stimulation. IL-6 decreased after Der p1 but increased following stimulation with Der p6 and heat-denatured Der p2. Conclusion: Allergens in house dust mites are capable of inducing various changes in the epithelial cell functions by virtue of their protease activities.
Collapse
Affiliation(s)
- Dilara Karaguzel
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Turkey
| | - Basak Ezgi Sarac
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Turkey
| | - Hayriye Akel Bilgic
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Turkey
| | - Gokce Yagmur Summak
- Department of Physics Engineering, Faculty of Engineering, Ankara University, Ankara, Turkey
| | - Mehmet Altay Unal
- Department of Physics Engineering, Faculty of Engineering, Ankara University, Ankara, Turkey
| | - Omer Kalayci
- Pediatric Allergy and Asthma Unit, Hacettepe University School of Medicine, Ankara, Turkey
| | - Cagatay Karaaslan
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Turkey
| |
Collapse
|
15
|
Erban T, Klimov PB, Harant K, Talacko P, Nesvorna M, Hubert J. Label-free proteomic analysis reveals differentially expressed Wolbachia proteins in Tyrophagus putrescentiae: Mite allergens and markers reflecting population-related proteome differences. J Proteomics 2021; 249:104356. [PMID: 34438106 DOI: 10.1016/j.jprot.2021.104356] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 12/21/2022]
Abstract
Tyrophagus putrescentiae is an astigmatid mite of great economic, medical and veterinary importance. The microbiome, especially intracellular bacteria, may affect allergy/allergen expression. We targeted Wolbachia proteins, allergen comparisons and markers in Wolbachia-mite interactions in three mite populations. A decoy database was constructed by proteogenomics using the T. putrescentiae draft genome, Wolbachia transcriptome assembly and current T. putrescentiae-related sequences in GenBank. Among thousands of mite-derived proteins, 18 Wolbachia proteins were reliably identified. We suggest that peroxiredoxin, bacterioferritin, ankyrin repeat domain-containing protein and DegQ family serine endoprotease indicate a higher-level bacterium-bacterium-host interaction. We produced evidence that the host-Wolbachia interaction is modulated through pattern recognition receptors (PRRs), mannose-binding lectins/mannose receptors, the cholinergic anti-inflammatory pathway with TNF-α, and others. We observed Tyr p 3 suppression in mites with Wolbachia, linking trypsin to PRR modulation. Nine out of the 12 current WHO/IUIS official allergens were reliably identified, but the remaining three allergens, Tyr p 1, 8 and 35, were detected as only trace hits. This study provides numerous markers for further Wolbachia-host interaction research. For accuracy, mite allergens should be considered according to abundance in species, but mite populations/strains, as well as their microbiome structure, may be key factors. SIGNIFICANCE: The astigmatid mites occurring in homes are significant producers of allergens that are highly dangerous to humans and domesticated animals. Mites are tightly associated with microorganisms that affect their biology and consequently allergy signatures. Mite populations were found to be infected with certain intracellular bacteria, but some populations lacked an intracellular bacterium. Our previous research showed that some populations of Tyrophagus putrescentiae are infected with Wolbachia, but some populations host additional bacteria of interest. Thus, there are not only interactions between the mites and Wolbachia but also likely an additional level of interaction that can be found in the interaction between different bacteria in the mites. These "higher-level" signatures and consequences that bacteria affect, including allergen production, are not understood in mites. In this study, we identified Wolbachia-specific proteins in mites for the first time. This study provides Wolbachia- and mite-derived markers that can be clues for describing "higher-level" mite-bacterium-bacterium interactions. Indeed, the microbiome contribution to allergies can potentially be derived directly from bacterial proteins, especially if they are abundant.
Collapse
Affiliation(s)
- Tomas Erban
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne CZ-16106, Czechia.
| | - Pavel B Klimov
- School of Natural Sciences, Bangor University, Bangor LL57 2 UW, UK; Institute of Biology, University of Tyumen, Pirogova 3, 625043 Tyumen, Russia
| | - Karel Harant
- Proteomics Core Facility, Faculty of Science, Charles University, BIOCEV, Prumyslova 595, Vestec CZ-25242, Czechia; Institute for Environmental Studies, Faculty of Science, Charles University, Benatska 2, Prague 2 CZ-128 01, Czechia
| | - Pavel Talacko
- Proteomics Core Facility, Faculty of Science, Charles University, BIOCEV, Prumyslova 595, Vestec CZ-25242, Czechia
| | - Marta Nesvorna
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne CZ-16106, Czechia
| | - Jan Hubert
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne CZ-16106, Czechia
| |
Collapse
|
16
|
Wang YJ, Yu SJ, Tsai JJ, Yu CH, Liao EC. Antagonism of Protease Activated Receptor-2 by GB88 Reduces Inflammation Triggered by Protease Allergen Tyr-p3. Front Immunol 2021; 12:557433. [PMID: 34566947 PMCID: PMC8456102 DOI: 10.3389/fimmu.2021.557433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 08/24/2021] [Indexed: 11/29/2022] Open
Abstract
The occurrence of allergic diseases induced by aeroallergens has increased in the past decades. Among inhalant allergens, mites remain the important causal agent of allergic diseases. Storage mites- Tyrophagus putrescentiae are found in stored products or domestic environments. Major allergen Tyr-p3 plays a significant role in triggering IgE-mediated hypersensitivity. However, its effects on pulmonary inflammation, internalization, and activation in human epithelium remain elusive. Protease-activated receptors (PARs) are activated upon cleavage by proteases. A549 cells were used as an epithelial model to examine the PAR activation by Tyr-p3 and therapeutic potential of PAR-2 antagonist (GB88) in allergic responses. Enzymatic properties and allergen localization of Tyr-p3 were performed. The release of inflammatory mediators, phosphorylation of mitogen-activated protein kinase (MAPK), and cell junction disruptions were evaluated after Tyr-p3 challenge. Enzymatic properties determined by substrate digestion and protease inhibitors indicated that Tyr-p3 processes a trypsin-like serine protease activity. The PAR-2 mRNA levels were significantly increased by nTyr-p3 but inhibited by protease inhibitors or GB88. Protease allergen of nTyr-p3 significantly increased the levels of pro-inflammatory cytokines (IL-6 and TNF-α), chemokine (IL-8), and IL-1β in epithelial cells. nTyr-p3 markedly increased phosphorylation of extracellular signal-regulated kinase (ERK)1/2 and MAP kinase. When cells were pretreated with GB88 then added nTyr-p3, the phosphorylated ERK1/2 did not inhibit by GB88. GB88 increased ERK1/2 phosphorylation in human epithelium cells. GB88 is able to block PAR-2-mediated calcium signaling which inhibits the nTyr-p3-induced Ca2+ release. Among the pharmacologic inhibitors, the most effective inhibitor of the nTyr-p3 in the induction of IL-8 or IL-1β levels was GB88 followed by SBTI, MAPK/ERK, ERK, and p38 inhibitors. Levels of inflammatory mediators, including GM-CSF, VEGF, COX-2, TSLP, and IL-33 were reduced by treatment of GB88 or SBTI. Further, GB88 treatment down-regulated the nTyr-p3-induced PAR-2 expression in allergic patients with asthma or rhinitis. Tight junction and adherens junction were disrupted in epithelial cells by nTyr-p3 exposure; however, this effect was avoided by GB88. Immunostaining with frozen sections of the mite body showed the presence of Tyr-p3 throughout the intestinal digestive system, especially in the hindgut around the excretion site. In conclusion, our findings suggest that Tyr-p3 from domestic mites leads to disruption of the airway epithelial barrier after inhalation. Proteolytic activity of Tyr-p3 causes the PAR-2 mRNA expression, thus leading to the release of numerous inflammatory mediators. Antagonism of PAR2 activity suggests GB88 as the therapeutic potential for anti-inflammation medicine, especially in allergy development triggered by protease allergens.
Collapse
Affiliation(s)
- Yun-Ju Wang
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Sheng-Jie Yu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Jaw-Ji Tsai
- Division of Allergy, Immunology & Rheumatology, Department of Internal Medicine, Asia University Hospital, Taichung, Taiwan.,Division of Allergy, Immunology & Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ching-Hsiang Yu
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - En-Chih Liao
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| |
Collapse
|
17
|
Caraballo L, Valenta R, Acevedo N, Zakzuk J. Are the Terms Major and Minor Allergens Useful for Precision Allergology? Front Immunol 2021; 12:651500. [PMID: 33763086 PMCID: PMC7982392 DOI: 10.3389/fimmu.2021.651500] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Division of Immunopathology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia.,Karl Landsteiner University of Health Sciences, Krems, Austria.,NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| | - Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Josefina Zakzuk
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| |
Collapse
|
18
|
Abu Khweek A, Kim E, Joldrichsen MR, Amer AO, Boyaka PN. Insights Into Mucosal Innate Immune Responses in House Dust Mite-Mediated Allergic Asthma. Front Immunol 2020; 11:534501. [PMID: 33424827 PMCID: PMC7793902 DOI: 10.3389/fimmu.2020.534501] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 09/01/2020] [Indexed: 01/09/2023] Open
Abstract
The prevalence of asthma has been rising steadily for several decades, and continues to be a major public health and global economic burden due to both direct and indirect costs. Asthma is defined as chronic heterogeneous inflammatory diseases characterized by airway obstruction, mucus production and bronchospasm. Different endotypes of asthma are being recognized based on the distinct pathophysiology, genetic predisposition, age, prognosis, and response to remedies. Mucosal innate response to environmental triggers such as pollen, cigarette smoke, fragrances, viral infection, and house dust mite (HDM) are now recognized to play an important role in allergic asthma. HDM are the most pervasive allergens that co-habitat with us, as they are ubiquitous in-house dusts, mattress and bedsheets, and feed on a diet of exfoliated human skin flakes. Dermatophagoides pteronyssinus, is one among several HDM identified up to date. During the last decade, extensive studies have been fundamental in elucidating the interactions between HDM allergens, the host immune systems and airways. Moreover, the paradigm in the field of HDM-mediated allergy has been shifted away from being solely a Th2-geared to a complex response orchestrated via extensive crosstalk between the epithelium, professional antigen presenting cells (APCs) and components of the adaptive immunity. In fact, HDM have several lessons to teach us about their allergenicity, the complex interactions that stimulate innate immunity in initiating and perpetuating the lung inflammation. Herein, we review main allergens of Dermatophagoides pteronyssinus and their interactions with immunological sentinels that promote allergic sensitization and activation of innate immunity, which is critical for the development of the Th2 biased adaptive immunity to HDM allergens and development of allergic asthma.
Collapse
Affiliation(s)
- Arwa Abu Khweek
- Department of Biology and Biochemistry, Birzeit University, Birzeit, Palestine.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Marisa R Joldrichsen
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Amal O Amer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,The Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Prosper N Boyaka
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States.,The Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
19
|
Aspergillus fumigatus Protease Alkaline Protease 1 (Alp1): A New Therapeutic Target for Fungal Asthma. J Fungi (Basel) 2020; 6:jof6020088. [PMID: 32560087 PMCID: PMC7345148 DOI: 10.3390/jof6020088] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 12/26/2022] Open
Abstract
We review three recent findings that have fundamentally altered our understanding of causative mechanisms underlying fungal-related asthma. These mechanisms may be partially independent of host inflammatory processes but are strongly dependent upon the actions of Alp1 on lung structural cells. They entail (i) bronchial epithelial sensing of Alp1; (ii) Alp1-induced airway smooth muscle (ASM) contraction; (iii) Alp1-induced airflow obstruction. Collectively, these mechanisms point to Alp1 as a new target for intervention in fungal asthma.
Collapse
|
20
|
Gaspar R, de Matos MR, Cortes L, Nunes-Correia I, Todo-Bom A, Pires E, Veríssimo P. Pollen Proteases Play Multiple Roles in Allergic Disorders. Int J Mol Sci 2020; 21:ijms21103578. [PMID: 32438574 PMCID: PMC7278992 DOI: 10.3390/ijms21103578] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/14/2020] [Accepted: 05/16/2020] [Indexed: 12/15/2022] Open
Abstract
Allergic diseases are a major health concern worldwide. Pollens are important triggers for allergic rhinitis, conjunctivitis and asthma. Proteases released upon pollen grain hydration appear to play a major role in the typical immunological and inflammatory responses that occur in patients with allergic disorders. In this study, we aimed to identify specific proteolytic activity in a set of pollens with diverse allergenic potential. Diffusates from Chenopodium album, Plantago lanceolata and Eucalyptus globulus were added to a confluent monolayer of Calu-3 cells grown in an air-liquid interface system. We identified serine proteases and metalloproteinases in all pollen diffusates investigated. Proteases found in these pollen diffusates were shown to compromise the integrity of the lung epithelial barrier by disrupting transmembrane adhesion proteins E-cadherin, claudin-1 and Occludin, as well as, the cytosolic complex zonula occludens-1 (ZO-1) resulting in a time-dependent increase in transepithelial permeability. Tight junction disruption and increased transepithelial permeability facilitates allergen exposure to epithelial sub-layers contributing to the sensitization to a wide range of allergens. These pollen extracts also induced an increase in the release of interleukin 6 (IL-6) and interleukin 8 (IL-8) cytokines measured by flow cytometry possibly as a result of the activation of protease-activated receptors 2 (PAR-2).
Collapse
Affiliation(s)
- Ricardo Gaspar
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (R.G.); (M.R.d.M.); (L.C.); (I.N.-C.); (E.P.)
| | - Mafalda Ramos de Matos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (R.G.); (M.R.d.M.); (L.C.); (I.N.-C.); (E.P.)
| | - Luísa Cortes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (R.G.); (M.R.d.M.); (L.C.); (I.N.-C.); (E.P.)
- Microscopy Unit Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Isabel Nunes-Correia
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (R.G.); (M.R.d.M.); (L.C.); (I.N.-C.); (E.P.)
| | - Ana Todo-Bom
- Immunoallergology Service, Coimbra University Hospital, 3004-504 Coimbra, Portugal;
- Faculty of Medicine, University of Coimbra, 3000-504 Coimbra, Portugal
| | - Euclides Pires
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (R.G.); (M.R.d.M.); (L.C.); (I.N.-C.); (E.P.)
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Paula Veríssimo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (R.G.); (M.R.d.M.); (L.C.); (I.N.-C.); (E.P.)
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
21
|
Azmeh R, Greydanus DE, Agana MG, Dickson CA, Patel DR, Ischander MM, Lloyd RD. Update in Pediatric Asthma: Selected Issues. Dis Mon 2020; 66:100886. [PMID: 31570159 DOI: 10.1016/j.disamonth.2019.100886] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Asthma is a complex condition that affects 14% of the world's children and the approach to management includes both pharmacologic as well as non-pharmacologic strategies including attention to complex socioeconomic status phenomena. After an historical consideration of asthma, allergic and immunologic aspects of asthma in children and adolescents are presented. Concepts of socioeconomic aspects of asthma are considered along with environmental features and complications of asthma disparities. Also reviewed are links of asthma with mental health disorders, sleep disturbances and other comorbidities. A stepwise approach to asthma management is discussed that includes pharmacologic and non-pharmacologic strategies in the pediatric population. The role of immunotherapy and use of various immunomodulators are considered as well.
Collapse
Affiliation(s)
- Roua Azmeh
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Donald E Greydanus
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States.
| | - Marisha G Agana
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Cheryl A Dickson
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States; Health Equity and Community Affairs, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, Michigan, United States
| | - Dilip R Patel
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Mariam M Ischander
- Department of Pediatric and Adolescent Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Robert D Lloyd
- Pacific Northwest University of Health Sciences College of Osteopathic Medicine, Yakima, Washington, United States
| |
Collapse
|
22
|
Goettig P, Brandstetter H, Magdolen V. Surface loops of trypsin-like serine proteases as determinants of function. Biochimie 2019; 166:52-76. [PMID: 31505212 PMCID: PMC7615277 DOI: 10.1016/j.biochi.2019.09.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023]
Abstract
Trypsin and chymotrypsin-like serine proteases from family S1 (clan PA) constitute the largest protease group in humans and more generally in vertebrates. The prototypes chymotrypsin, trypsin and elastase represent simple digestive proteases in the gut, where they cleave nearly any protein. Multidomain trypsin-like proteases are key players in the tightly controlled blood coagulation and complement systems, as well as related proteases that are secreted from diverse immune cells. Some serine proteases are expressed in nearly all tissues and fluids of the human body, such as the human kallikreins and kallikrein-related peptidases with specialization for often unique substrates and accurate timing of activity. HtrA and membrane-anchored serine proteases fulfill important physiological tasks with emerging roles in cancer. The high diversity of all family members, which share the tandem β-barrel architecture of the chymotrypsin-fold in the catalytic domain, is conferred by the large differences of eight surface loops, surrounding the active site. The length of these loops alters with insertions and deletions, resulting in remarkably different three-dimensional arrangements. In addition, metal binding sites for Na+, Ca2+ and Zn2+ serve as regulatory elements, as do N-glycosylation sites. Depending on the individual tasks of the protease, the surface loops determine substrate specificity, control the turnover and allow regulation of activation, activity and degradation by other proteins, which are often serine proteases themselves. Most intriguingly, in some serine proteases, the surface loops interact as allosteric network, partially tuned by protein co-factors. Knowledge of these subtle and complicated molecular motions may allow nowadays for new and specific pharmaceutical or medical approaches.
Collapse
Affiliation(s)
- Peter Goettig
- Division of Structural Biology, Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria.
| | - Hans Brandstetter
- Division of Structural Biology, Department of Biosciences, University of Salzburg, Billrothstrasse 11, 5020, Salzburg, Austria
| | - Viktor Magdolen
- Clinical Research Unit, Department of Obstetrics and Gynecology, School of Medicine, Technical University of Munich, Ismaninger Strasse 22, 81675, München, Germany
| |
Collapse
|
23
|
Daines M, Zhu L, Pereira R, Zhou X, Bondy C, Pryor BM, Zhou J, Chen Y. Alternaria induces airway epithelial cytokine expression independent of protease-activated receptor. Respirology 2019; 25:502-510. [PMID: 31430011 DOI: 10.1111/resp.13675] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 04/24/2019] [Accepted: 07/01/2019] [Indexed: 01/24/2023]
Abstract
BACKGROUND AND OBJECTIVE A novel fungal allergen, Alternaria (Alt), has been previously shown to associate with the pathogenesis of allergic rhinitis and bronchial asthma, particularly in arid and semi-arid regions. Airway epithelial cells are among the first to encounter Alt, and epithelial cytokine production and subsequent airway inflammation are early events in the response to Alt exposure. However, the underlying mechanism is unclear. As protease-activated receptor 2 (PAR2) has been implicated in most of the Alt-induced biological events, we investigated the regulation of airway inflammation and epithelial cytokine expression by PAR2. METHODS Wild-type (WT) and Par2 knockout (Par2-KO) mice were used to evaluate the in vivo role of PAR2. Primary human and mouse airway epithelial cells were used to examine the mechanistic basis of epithelial cytokine regulation in vitro. RESULTS Surprisingly, Par2 deficiency had no negative impact on the change of lung function, inflammation and cytokine production in the mouse model of Alt-induced asthma. Alt-induced cytokine production in murine airway epithelial cells from Par2-KO mice was not significantly different from the WT cells. Consistently, PAR2 knockdown in human cells also had no effect on cytokine expression. In contrast, the cytokine expressions induced by synthetic PAR2 agonist or other asthma-related allergens (e.g. cockroach extracts) were indeed mediated via a PAR2-dependent mechanism. Finally, we found that EGFR pathway was responsible for Alt-induced epithelial cytokine expression. CONCLUSION The activation of EGFR, but not PAR2, was likely to drive the airway inflammation and epithelial cytokine production induced by Alt.
Collapse
Affiliation(s)
- Michael Daines
- Department of Internal Medicine, School of Medicine, University of Arizona, Tucson, AZ, USA.,Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
| | - Lingxiang Zhu
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Rhea Pereira
- Department of Internal Medicine, School of Medicine, University of Arizona, Tucson, AZ, USA
| | - Xu Zhou
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Cheryl Bondy
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Barry M Pryor
- School of Plant Science, University of Arizona, Tucson, AZ, USA
| | - Jin Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, University of Arizona, Tucson, AZ, USA
| | - Yin Chen
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA.,Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
24
|
Li B, Zou Z, Meng F, Raz E, Huang Y, Tao A, Ai Y. Dust mite-derived Der f 3 activates a pro-inflammatory program in airway epithelial cells via PAR-1 and PAR-2. Mol Immunol 2019; 109:1-11. [PMID: 30836204 DOI: 10.1016/j.molimm.2019.02.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/23/2019] [Accepted: 02/20/2019] [Indexed: 11/17/2022]
Abstract
Protease activity of allergens has been suggested to be involved in the pathogenesis of allergic diseases. The major allergen Der f 3 from Dermatophagoides farinae harbors serine protease activity, but its immunopathogenesis remains unclear. This study aims to explore the effect of Der f 3 on the airway epithelial barrier and on the molecular pathways by which Der f 3 induces inflammation. RNA-seq was performed to identify differentially expressed genes in bronchial airway epithelial cells (AEC) between native Der f 3 and heat-inactivated (H) Der f 3, coupled with real-time PCR (RT-PCR) and ELISA for validation. Unlike other protease allergens such as that induce Th2-promoting alarmins (IL-25, IL-33, TSLP) in AECs, Der f 3 induced pro-inflammatory cytokines and chemokines including IL-6, IL-8 and GM-CSF, which are known to promote Th17 response. These pro-inflammatory mediators were induced by Der f 3 via the MAPK and NF-κB pathways as well as the store-operated calcium signaling. Gene silencing with small interfering RNA in A549 and BEAS-2B cells indicated that activation of AECs by Der f 3 was mainly dependent on protease-activated receptor 2 (PAR-2), while PAR-1 was also required for the full activation of AECs. Double knock-down of PAR-1 and PAR-2 largely impaired Der f 3-inducecd IL-8 production and subsequent signaling pathways. Our data suggest that Der f 3 induces pro-inflammatory mediators in human epithelial cell lines via the PARs-MAPK-NF-κB axis. Our results provide a molecular mechanism by which Der f 3 may trigger the Th17-skewed allergic response toward house dust mites.
Collapse
Affiliation(s)
- Bizhou Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, China; Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, China
| | - Zehong Zou
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Center for Inflammation, Immunity, & Immune-mediated Disease, Guangzhou Medical University, China
| | - Fanmei Meng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, China; Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, China
| | - Eyal Raz
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Center for Inflammation, Immunity, & Immune-mediated Disease, Guangzhou Medical University, China; Department of Medicine, University of California at San Diego, United States
| | - Yuye Huang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, China; Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, China
| | - Ailin Tao
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Center for Inflammation, Immunity, & Immune-mediated Disease, Guangzhou Medical University, China.
| | - Yuncan Ai
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, China; Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, China.
| |
Collapse
|
25
|
Heuberger DM, Schuepbach RA. Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb J 2019; 17:4. [PMID: 30976204 PMCID: PMC6440139 DOI: 10.1186/s12959-019-0194-8] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
Inflammatory diseases have become increasingly prevalent with industrialization. To address this, numerous anti-inflammatory agents and molecular targets have been considered in clinical trials. Among molecular targets, protease-activated receptors (PARs) are abundantly recognized for their roles in the development of chronic inflammatory diseases. In particular, several inflammatory effects are directly mediated by the sensing of proteolytic activity by PARs. PARs belong to the seven transmembrane domain G protein-coupled receptor family, but are unique in their lack of physiologically soluble ligands. In contrast with classical receptors, PARs are activated by N-terminal proteolytic cleavage. Upon removal of specific N-terminal peptides, the resulting N-termini serve as tethered activation ligands that interact with the extracellular loop 2 domain and initiate receptor signaling. In the classical pathway, activated receptors mediate signaling by recruiting G proteins. However, activation of PARs alternatively lead to the transactivation of and signaling through receptors such as co-localized PARs, ion channels, and toll-like receptors. In this review we consider PARs and their modulators as potential therapeutic agents, and summarize the current understanding of PAR functions from clinical and in vitro studies of PAR-related inflammation.
Collapse
Affiliation(s)
- Dorothea M Heuberger
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Surgical Research Division, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reto A Schuepbach
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Ueda Y, Nakagome K, Kobayashi T, Noguchi T, Soma T, Ohashi-Doi K, Tokuyama K, Nagata M. Dermatophagoides farinae Upregulates the Effector Functions of Eosinophils through αMβ2-Integrin and Protease-Activated Receptor-2. Int Arch Allergy Immunol 2019; 178:295-306. [PMID: 30630188 DOI: 10.1159/000495008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/31/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Even in subjects who are not sensitized to house dust mite (HDM), allergic symptoms can be aggravated by exposure to dust, suggesting that innate immune responses may be involved in these processes. Since eosinophils express pattern recognition receptors, HDM may directly upregulate eosinophil functions through these re ceptors. The objective of this study was to examine whether Dermatophagoides farinae (Df), a representative HDM, or Der f 1, a major allergen of Df, modifies the effector functions of eosinophils. METHODS Eosinophils isolated from the blood of healthy donors or allergic patients were stimulated with Df extract or Der f 1, and their adhesion to recombinant human intercellular adhesion molecule (ICAM)-1 was measured using eosinophil peroxidase assays. Generation of the eosinophil superoxide anion (O2-) was examined based on the superoxide dismutase-inhibitable reduction of cytochrome C. Eosinophil-derived neurotoxin (EDN) concentrations in cell media were measured by ELISA as a marker of degranulation. RESULTS Df extract or Der f 1 directly induced eosinophil adhesion to ICAM-1, O2- generation, and EDN release. Anti-αM- or anti-β2-integrin antibodies or protease-activated receptor (PAR)-2 antagonists suppressed the eosinophil adhesion, O2- generation, and EDN release induced by Df extract or Der f 1. Eosinophils from allergic patients showed higher adhesion to ICAM-1 than those from healthy donors. CONCLUSIONS These findings suggested that Df extract and Der f 1 directly activate eosinophil functions through αMβ2-integrin and PAR-2. Eosinophil activation by HDM may play roles in the aggravation of allergic symptoms, not only in HDM-sensitized patients, but also in nonsensitized patients.
Collapse
Affiliation(s)
- Yutaka Ueda
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan.,Department of Pediatrics, Saitama Medical University, Saitama, Japan.,Allergy Center, Saitama Medical University, Saitama, Japan
| | - Kazuyuki Nakagome
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan, .,Allergy Center, Saitama Medical University, Saitama, Japan,
| | - Takehito Kobayashi
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan.,Allergy Center, Saitama Medical University, Saitama, Japan
| | - Toru Noguchi
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan.,Allergy Center, Saitama Medical University, Saitama, Japan
| | - Tomoyuki Soma
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan.,Allergy Center, Saitama Medical University, Saitama, Japan
| | | | - Kenichi Tokuyama
- Department of Pediatrics, Saitama Medical University, Saitama, Japan.,Allergy Center, Saitama Medical University, Saitama, Japan
| | - Makoto Nagata
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan.,Allergy Center, Saitama Medical University, Saitama, Japan
| |
Collapse
|
27
|
Yee MC, Nichols HL, Polley D, Saifeddine M, Pal K, Lee K, Wilson EH, Daines MO, Hollenberg MD, Boitano S, DeFea KA. Protease-activated receptor-2 signaling through β-arrestin-2 mediates Alternaria alkaline serine protease-induced airway inflammation. Am J Physiol Lung Cell Mol Physiol 2018; 315:L1042-L1057. [PMID: 30335499 PMCID: PMC6337008 DOI: 10.1152/ajplung.00196.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/06/2018] [Accepted: 08/22/2018] [Indexed: 01/01/2023] Open
Abstract
Alternaria alternata is a fungal allergen associated with severe asthma and asthma exacerbations. Similarly to other asthma-associated allergens, Alternaria secretes a serine-like trypsin protease(s) that is thought to act through the G protein-coupled receptor protease-activated receptor-2 (PAR2) to induce asthma symptoms. However, specific mechanisms underlying Alternaria-induced PAR2 activation and signaling remain ill-defined. We sought to determine whether Alternaria-induced PAR2 signaling contributed to asthma symptoms via a PAR2/β-arrestin signaling axis, identify the protease activity responsible for PAR2 signaling, and determine whether protease activity was sufficient for Alternaria-induced asthma symptoms in animal models. We initially used in vitro models to demonstrate Alternaria-induced PAR2/β-arrestin-2 signaling. Alternaria filtrates were then used to sensitize and challenge wild-type, PAR2-/- and β-arrestin-2-/- mice in vivo. Intranasal administration of Alternaria filtrate resulted in a protease-dependent increase of airway inflammation and mucin production in wild-type but not PAR2-/- or β-arrestin-2-/- mice. Protease was isolated from Alternaria preparations, and select in vitro and in vivo experiments were repeated to evaluate sufficiency of the isolated Alternaria protease to induce asthma phenotype. Administration of a single isolated serine protease from Alternaria, Alternaria alkaline serine protease (AASP), was sufficient to fully activate PAR2 signaling and induce β-arrestin-2-/--dependent eosinophil and lymphocyte recruitment in vivo. In conclusion, Alternaria filtrates induce airway inflammation and mucus hyperplasia largely via AASP using the PAR2/β-arrestin signaling axis. Thus, β-arrestin-biased PAR2 antagonists represent novel therapeutic targets for treating aeroallergen-induced asthma.
Collapse
Affiliation(s)
- Michael C Yee
- Biomedical Sciences, Graduate Program, University of California Riverside , Riverside, California
| | - Heddie L Nichols
- Biomedical Sciences, Graduate Program, University of California Riverside , Riverside, California
| | - Danny Polley
- Cumming School of Medicine, Department of Physiology and Pharmacology and Department of Medicine, University of Calgary , Calgary, Alberta , Canada
| | - Mahmoud Saifeddine
- Cumming School of Medicine, Department of Physiology and Pharmacology and Department of Medicine, University of Calgary , Calgary, Alberta , Canada
| | - Kasturi Pal
- Biomedical Sciences, Graduate Program, University of California Riverside , Riverside, California
- Cell Molecular and Developmental Biology and Biochemistry Graduate Program, University of California Riverside , Riverside, California
| | - Kyu Lee
- Biomedical Sciences, Graduate Program, University of California Riverside , Riverside, California
- Molecular Biology Graduate Program, University of California Riverside , Riverside, California
| | - Emma H Wilson
- Biomedical Sciences, Graduate Program, University of California Riverside , Riverside, California
| | - Michael O Daines
- Department of Pediatrics, University of Arizona Health Sciences , Tucson, Arizona
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences , Tucson, Arizona
| | - Morley D Hollenberg
- Cumming School of Medicine, Department of Physiology and Pharmacology and Department of Medicine, University of Calgary , Calgary, Alberta , Canada
| | - Scott Boitano
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences , Tucson, Arizona
- Department of Physiology, University of Arizona Health Sciences , Tucson, Arizona
| | - Kathryn A DeFea
- Biomedical Sciences, Graduate Program, University of California Riverside , Riverside, California
- Cell Molecular and Developmental Biology and Biochemistry Graduate Program, University of California Riverside , Riverside, California
- Molecular Biology Graduate Program, University of California Riverside , Riverside, California
| |
Collapse
|
28
|
Zhang J, Chen J, Newton GK, Perrior TR, Robinson C. Allergen Delivery Inhibitors: A Rationale for Targeting Sentinel Innate Immune Signaling of Group 1 House Dust Mite Allergens through Structure-Based Protease Inhibitor Design. Mol Pharmacol 2018; 94:1007-1030. [PMID: 29976563 PMCID: PMC6064784 DOI: 10.1124/mol.118.112730] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/20/2018] [Indexed: 12/22/2022] Open
Abstract
Diverse evidence from epidemiologic surveys and investigations into the molecular basis of allergenicity have revealed that a small cadre of "initiator" allergens promote the development of allergic diseases, such as asthma, allergic rhinitis, and atopic dermatitis. Pre-eminent among these initiators are the group 1 allergens from house dust mites (HDM). In mites, group 1 allergens function as cysteine peptidase digestive enzymes to which humans are exposed by inhalation of HDM fecal pellets. Their protease nature confers the ability to activate high gain signaling mechanisms which promote innate immune responses, leading to the persistence of allergic sensitization. An important feature of this process is that the initiator drives responses both to itself and to unrelated allergens lacking these properties through a process of collateral priming. The clinical significance of group 1 HDM allergens in disease, their serodominance as allergens, and their IgE-independent bioactivities in innate immunity make these allergens interesting therapeutic targets in the design of new small-molecule interventions in allergic disease. The attraction of this new approach is that it offers a powerful, root-cause-level intervention from which beneficial effects can be anticipated by interference in a wide range of effector pathways associated with these complex diseases. This review addresses the general background to HDM allergens and the validation of group 1 as putative targets. We then discuss structure-based drug design of the first-in-class representatives of allergen delivery inhibitors aimed at neutralizing the proteolytic effects of HDM group 1 allergens, which are essential to the development and maintenance of allergic diseases.
Collapse
Affiliation(s)
- Jihui Zhang
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom (J.Z., J.C., C.R.); State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China (J.Z.); and Domainex Ltd., Chesterford Research Park, Saffron Walden, United Kingdom (G.K.N., T.R.P.)
| | - Jie Chen
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom (J.Z., J.C., C.R.); State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China (J.Z.); and Domainex Ltd., Chesterford Research Park, Saffron Walden, United Kingdom (G.K.N., T.R.P.)
| | - Gary K Newton
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom (J.Z., J.C., C.R.); State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China (J.Z.); and Domainex Ltd., Chesterford Research Park, Saffron Walden, United Kingdom (G.K.N., T.R.P.)
| | - Trevor R Perrior
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom (J.Z., J.C., C.R.); State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China (J.Z.); and Domainex Ltd., Chesterford Research Park, Saffron Walden, United Kingdom (G.K.N., T.R.P.)
| | - Clive Robinson
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom (J.Z., J.C., C.R.); State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, People's Republic of China (J.Z.); and Domainex Ltd., Chesterford Research Park, Saffron Walden, United Kingdom (G.K.N., T.R.P.)
| |
Collapse
|
29
|
Engebretsen K, Kezic S, Jakasa I, Hedengran A, Linneberg A, Skov L, Johansen J, Thyssen J. Effect of atopic skin stressors on natural moisturizing factors and cytokines in healthy adult epidermis. Br J Dermatol 2018; 179:679-688. [DOI: 10.1111/bjd.16487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2018] [Indexed: 12/29/2022]
Affiliation(s)
- K.A. Engebretsen
- National Allergy Research Centre; Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
- Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| | - S. Kezic
- Coronel Institute of Occupational Health; Academic Medical Center; Amsterdam Public Health Research Institute; University of Amsterdam; 1100 DE Amsterdam The Netherlands
| | - I. Jakasa
- Laboratory for Analytical Chemistry; Department of Chemistry and Biochemistry; Faculty of Food Technology and Biotechnology; University of Zagreb; Zagreb Croatia
| | - A. Hedengran
- Department of Clinical Biochemistry; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| | - A. Linneberg
- Research Centre for Prevention and Health; The Capital Region of Denmark; Copenhagen Denmark
- Department of Clinical Experimental Research; Rigshospitalet Glostrup Denmark
- Department of Clinical Medicine; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - L. Skov
- Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| | - J.D. Johansen
- National Allergy Research Centre; Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
- Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| | - J.P. Thyssen
- National Allergy Research Centre; Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
- Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| |
Collapse
|
30
|
Asosingh K, Weiss K, Queisser K, Wanner N, Yin M, Aronica M, Erzurum S. Endothelial cells in the innate response to allergens and initiation of atopic asthma. J Clin Invest 2018; 128:3116-3128. [PMID: 29911993 DOI: 10.1172/jci97720] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 05/01/2018] [Indexed: 01/03/2023] Open
Abstract
Protease-activated receptor 2 (PAR-2), an airway epithelial pattern recognition receptor (PRR), participates in the genesis of house dust mite-induced (HDM-induced) asthma. Here, we hypothesized that lung endothelial cells and proangiogenic hematopoietic progenitor cells (PACs) that express high levels of PAR-2 contribute to the initiation of atopic asthma. HDM extract (HDME) protease allergens were found deep in the airway mucosa and breaching the endothelial barrier. Lung endothelial cells and PACs released the Th2-promoting cytokines IL-1α and GM-CSF in response to HDME, and the endothelium had PAC-derived VEGF-C-dependent blood vessel sprouting. Blockade of the angiogenic response by inhibition of VEGF-C signaling lessened the development of inflammation and airway remodeling in the HDM model. Reconstitution of the bone marrow in WT mice with PAR-2-deficient bone marrow also reduced airway inflammation and remodeling. Adoptive transfer of PACs that had been exposed to HDME induced angiogenesis and Th2 inflammation with remodeling similar to that induced by allergen challenge. Our findings identify that lung endothelium and PACs in the airway sense allergen and elicit an angiogenic response that is central to the innate nonimmune origins of Th2 inflammation.
Collapse
Affiliation(s)
| | | | | | | | - Mei Yin
- Imaging Core, Lerner Research Institute, and
| | - Mark Aronica
- Department of Inflammation and Immunity.,Respiratory Institute, the Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil Erzurum
- Department of Inflammation and Immunity.,Respiratory Institute, the Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
31
|
Hosoki K, Brasier AR, Kurosky A, Boldogh I, Sur S. Reply: Protease Plays a Role in Ragweed Pollen-Induced Neutrophil Recruitment and Epithelial Barrier Disruption. Am J Respir Cell Mol Biol 2018; 56:272-273. [PMID: 28145773 DOI: 10.1165/rcmb.2016-0281le] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Koa Hosoki
- 1 University of Texas Medical Branch Galveston, Texas
| | | | | | | | - Sanjiv Sur
- 1 University of Texas Medical Branch Galveston, Texas
| |
Collapse
|
32
|
Ramu S, Menzel M, Bjermer L, Andersson C, Akbarshahi H, Uller L. Allergens produce serine proteases-dependent distinct release of metabolite DAMPs in human bronchial epithelial cells. Clin Exp Allergy 2017; 48:156-166. [PMID: 29210131 DOI: 10.1111/cea.13071] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 11/20/2017] [Accepted: 11/27/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND The respiratory epithelium is a major site for disease interaction with inhaled allergens. Additional to IgE-dependent effects, allergens contain proteases that may stimulate human bronchial epithelial cells (HBECs) through protease-activated receptors, causing the release of mediators important in driving Th2-mediated immune responses. OBJECTIVE We aimed to investigate whether different allergens induce metabolite DAMPs such as ATP and uric acid (UA) release in HBECs. METHODS HBECs (BEAS-2B cell line) were exposed to different allergen extracts; house dust mite (HDM), Alternaria alternata, Artemisia vulgaris and Betula pendula and UA, ATP, IL-8 and IL-33 release were measured. Allergen extracts were heat-inactivated or pre-incubated with serine (AEBSF) or cysteine (E64) protease inhibitors to study the involvement of protease activity in ATP, UA and IL-8 release. HDM-induced release of UA was studied in a mouse model of allergic inflammation. RESULTS All allergens caused dose-dependent rapid release of ATP and IL-8, but only HDM induced UA release from HBECs. HDM also caused release of UA in vivo in our mouse model of allergic inflammation. ATP release by all 4 allergen extracts was significantly reduced by heat-inactivation and by serine protease inhibitors. Similarly, the HDM-induced UA release was also abrogated by heat-inactivation of HDM extract and dependent on serine proteases. Furthermore, allergen-induced IL-8 mRNA expression was inhibited by serine protease inhibitors. CONCLUSIONS AND CLINICAL RELEVANCE ATP was released by all 4 allergens in HBECs supporting the role of ATP involvement in asthma pathology. However, HDM stands out by its capacity to cause UA release, which is of interest in view of the proposed role of UA in early initiation of allergic asthma. Although serine proteases may be involved in the activity of all the studied allergens, further work is warranted to explain the differences between HDM and the other 3 allergens regarding the effects on UA release.
Collapse
Affiliation(s)
- S Ramu
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - M Menzel
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - L Bjermer
- Skane University Hospital, Lund, Sweden
| | - C Andersson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - H Akbarshahi
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - L Uller
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
33
|
Yin SC, Liao EC, Ye CX, Chang CY, Tsai JJ. Effect of mite allergenic components on innate immune response: Synergy of protease (Group 1 & 3) and non-protease (Group 2 & 7) allergens. Immunobiology 2017; 223:443-448. [PMID: 29398016 DOI: 10.1016/j.imbio.2017.10.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 10/14/2017] [Indexed: 01/23/2023]
Abstract
The major mite allergenic components of protease allergens (group 1,3) and non-protease allergens (group 2,7) derived from Dermatophagoides peronyssinus (Dp) and D. farinae (Df) are reported to be capable of sensitizing 80-90% of mite-allergic patients. Although protease and non-protease allergens have been demonstrated to trigger innate and adaptive immune responses through epithelium activation, the simultaneous or sequential effects of both groups of allergens has not been reported. Since all allergens are present in the mite crude extracts, it is important to determine whether these allergens can synergistically trigger the immune responses to cause airway inflammation. A total of 60 house dust mite (HDM)-allergic asthmatic patients were recruited to analyze their serum-specific IgE response to both groups of allergens. Recombinant protease allergen (Der p1 and Der p3) and non-protease allergens (Der p2 and Der p7) were used to activate the human airway epithelium cell (Beas-2B). The cells were analyzed for mRNA expression of IL-6/IL-8 and the culture supernatants were analyzed for neutrophil chemotactic activity (NCA). The results showed 48/60 (80%) HDM-allergic patients were sensitized to all allergenic components of Der p1, Der p2, Der f1, and Der f2. Most of the allergic patients were sensitized to both groups of allergens simultaneously. The associations of Der p1 with Der p2 were 83.3% (50/60) and Der f1 with Der f2 were 80% (48/60). When Beas-2B cells were cultured with Der p2 in conjunction with Der p1 and Der p3, the results showed that there was increased expression of IL-6/IL-8 in comparison with culture with allergen alone. There was only a trivial effect on IL-6/IL-8 expression when Der p2 was co-cultured with Der p7. Similar findings were obtained in the NCA measurement. When Beas-2B was cultured with Der p2 in conjunction with Der p1 and Der p3, there was increased NCA in comparison with culture with allergen alone. There were also trivial effects when Der p2 was co-cultured with Der p7. The allergens (Der p2 and Der p3)-induced IL-6/IL-8 expression and NCA released from Beas-2B could be downregulated by dexamethasone and transcription factor inhibitor SP600125. The allergenic components derived from Dp and Df can sensitize allergic patients simultaneously and activate epithelium through protease allergens (group 1, 3) and non-protease allergen (group 2) synergistically.
Collapse
Affiliation(s)
- Sui-Chu Yin
- Center for Translational Medicine, Department of Medical Research, Taichung Veterans General Hospital, Taiwan
| | - En-Chih Liao
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chi-Xin Ye
- Center for Translational Medicine, Department of Medical Research, Taichung Veterans General Hospital, Taiwan; Section of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Asian University Hospital, Taichung, Taiwan
| | - Ching-Yun Chang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jaw-Ji Tsai
- Center for Translational Medicine, Department of Medical Research, Taichung Veterans General Hospital, Taiwan; Section of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Asian University Hospital, Taichung, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
34
|
Allergens with Protease Activity from House Dust Mites. Int J Mol Sci 2017; 18:ijms18071368. [PMID: 28653989 PMCID: PMC5535861 DOI: 10.3390/ijms18071368] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/13/2017] [Accepted: 06/21/2017] [Indexed: 11/29/2022] Open
Abstract
Globally, house dust mites (HDM) are one of the main sources of allergens causing Type I allergy, which has a high risk of progressing into a severe disabling disease manifestation such as allergic asthma. The strong protease activities of a number of these allergens are thought to be involved in several steps of the pathophysiology of this allergic disease. It has been a common notion that protease activity may be one of the properties that confers allergenicity to proteins. In this review we summarize and discuss the roles of the different HDM proteases in the development of Type I allergy.
Collapse
|
35
|
Jacquet A, Campisi V, Szpakowska M, Dumez ME, Galleni M, Chevigné A. Profiling the Extended Cleavage Specificity of the House Dust Mite Protease Allergens Der p 1, Der p 3 and Der p 6 for the Prediction of New Cell Surface Protein Substrates. Int J Mol Sci 2017; 18:ijms18071373. [PMID: 28654001 PMCID: PMC5535866 DOI: 10.3390/ijms18071373] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/16/2017] [Accepted: 06/21/2017] [Indexed: 12/22/2022] Open
Abstract
House dust mite (HDM) protease allergens, through cleavages of critical surface proteins, drastically influence the initiation of the Th2 type immune responses. However, few human protein substrates for HDM proteases have been identified so far, mainly by applying time-consuming target-specific individual studies. Therefore, the identification of substrate repertoires for HDM proteases would represent an unprecedented key step toward a better understanding of the mechanism of HDM allergic response. In this study, phage display screenings using totally or partially randomized nonameric peptide substrate libraries were performed to characterize the extended substrate specificities (P5–P4′) of the HDM proteases Der p 1, Der p 3 and Der p 6. The bioinformatics interface PoPS (Prediction of Protease Specificity) was then applied to define the proteolytic specificity profile of each protease and to predict new protein substrates within the human cell surface proteome, with a special focus on immune receptors. Specificity profiling showed that the nature of residues in P1 but also downstream the cleavage sites (P′ positions) are important for effective cleavages by all three HDM proteases. Strikingly, Der p 1 and Der p 3 display partially overlapping specificities. Analysis with PoPS interface predicted 50 new targets for the HDM proteases, including 21 cell surface receptors whose extracellular domains are potentially cleaved by Der p 1, Der p 3 and/or Der p 6. Twelve protein substrate candidates were confirmed by phage ELISA (enzyme linked immunosorbent assay). This extensive study of the natural protein substrate specificities of the HDM protease allergens unveils new cell surface target receptors for a better understanding on the role of these proteases in the HDM allergic response and paves the way for the design of specific protease inhibitors for future anti-allergic treatments.
Collapse
Affiliation(s)
- Alain Jacquet
- Faculty of Medicine, Division of Research Affairs, Chulalongkorn University, 10330 Bangkok, Thailand.
| | - Vincenzo Campisi
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg.
- Laboratoire des Macromolécules Biologiques, Centre for Protein Engineering (CIP), University of Liège, 4000 Liège, Belgium.
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Marie-Eve Dumez
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg.
- Laboratoire des Macromolécules Biologiques, Centre for Protein Engineering (CIP), University of Liège, 4000 Liège, Belgium.
| | - Moreno Galleni
- Laboratoire des Macromolécules Biologiques, Centre for Protein Engineering (CIP), University of Liège, 4000 Liège, Belgium.
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
36
|
Yamada Y, Matsumoto T. House Dust Mites Induce Production of Endothelin-1 and Matrix Metalloproteinase-9 in Keratinocytes via Proteinase-Activated Receptor-2 Activation. Int Arch Allergy Immunol 2017; 173:84-92. [PMID: 28586781 DOI: 10.1159/000473700] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/28/2017] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by skin barrier dysfunction and abnormal immune response. House dust mites (HDM) are a major source of allergens, some of which have cysteine and serine protease activities. Keratinocytes stimulated by HDM-derived proteases have been suggested to contribute to the pathogenesis of AD by producing various cytokines. However, whether keratinocytes contribute to the induction of pruritus in AD, especially by producing pruritus-related mediators upon stimulation with HDM-derived proteases, has not been fully elucidated. METHODS We examined whether the production of endothelin-1 (ET-1), matrix metalloproteinase (MMP)-2, and MMP-9 in keratinocytes can be induced by stimulation with Dermatophagoides farinae extracts, and if so, whether pretreatment with a protease inhibitor or proteinase-activated receptor-2 (PAR-2) antagonist affects the production of these mediators in keratinocytes. RESULTS Although MMP-2 levels were undetectable in the culture supernatants, the production of ET-1 and MMP-9 was increased upon stimulation with HDM extracts in a concentration- and time-dependent manner and suppressed by pretreatment of HDM extracts with serine protease inhibitor, but not with cysteine protease inhibitor. Mite-derived serine proteases also induced ET-1 and MMP-9 production in a concentration- and time-dependent manner. Moreover, pretreatment with a PAR-2 antagonist inhibited the production of ET-1 and MMP-9 in keratinocytes. CONCLUSION These results suggest that the activation of PAR-2 on keratinocytes by HDM-derived serine proteases induces the production of ET-1 and MMP-9, and may contribute to the induction of pruritus in AD.
Collapse
Affiliation(s)
- Yoshihito Yamada
- Drug Development Research Laboratories, Kyoto R&D Center, Maruho Co., Ltd., Kyoto, Japan
| | | |
Collapse
|
37
|
van Rijt LS, Utsch L, Lutter R, van Ree R. Oxidative Stress: Promoter of Allergic Sensitization to Protease Allergens? Int J Mol Sci 2017; 18:ijms18061112. [PMID: 28545251 PMCID: PMC5485936 DOI: 10.3390/ijms18061112] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 01/18/2023] Open
Abstract
Allergies arise from aberrant T helper type 2 responses to allergens. Several respiratory allergens possess proteolytic activity, which has been recognized to act as an adjuvant for the development of a Th2 response. Allergen source-derived proteases can activate the protease-activated receptor-2, have specific effects on immune cells by cleaving cell membrane-bound regulatory molecules, and can disrupt tight junctions. The protease activity can induce a non-allergen-specific inflammatory response in the airways, which will set the stage for an allergen-specific Th2 response. In this review, we will discuss the evidence for the induction of oxidative stress as an underlying mechanism in Th2 sensitization to proteolytic allergens. We will discuss recent data linking the proteolytic activity of an allergen to its potential to induce oxidative stress and how this can facilitate allergic sensitization. Based on experimental data, we propose that a less proficient anti-oxidant response to allergen-induced oxidative stress contributes to the susceptibility to allergic sensitization. Besides the effect of oxidative stress on the immune response, we will also discuss how oxidative stress can increase the immunogenicity of an allergen by chemical modification.
Collapse
Affiliation(s)
- Leonie S van Rijt
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Lara Utsch
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - René Lutter
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Ronald van Ree
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
- Department of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Yamada Y, Ueda Y, Nakamura A, Kanayama S, Tamura R, Hashimoto K, Matsumoto T, Ishii R. Immediate-type allergic and protease-mediated reactions are involved in scratching behaviour induced by topical application of Dermatophagoides farinae
extract in NC/Nga mice. Exp Dermatol 2017; 27:418-426. [DOI: 10.1111/exd.13322] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Yoshihito Yamada
- Drug Development Research Laboratories; Kyoto R&D Center; Maruho Co., Ltd.; Kyoto Japan
| | - Yuhki Ueda
- Drug Development Research Laboratories; Kyoto R&D Center; Maruho Co., Ltd.; Kyoto Japan
| | - Aki Nakamura
- Drug Development Research Laboratories; Kyoto R&D Center; Maruho Co., Ltd.; Kyoto Japan
| | - Shoji Kanayama
- Drug Development Research Laboratories; Kyoto R&D Center; Maruho Co., Ltd.; Kyoto Japan
| | - Rie Tamura
- Drug Development Research Laboratories; Kyoto R&D Center; Maruho Co., Ltd.; Kyoto Japan
| | - Kei Hashimoto
- Drug Development Research Laboratories; Kyoto R&D Center; Maruho Co., Ltd.; Kyoto Japan
| | - Tatsumi Matsumoto
- Drug Development Research Laboratories; Kyoto R&D Center; Maruho Co., Ltd.; Kyoto Japan
| | - Ritsuko Ishii
- Strategic Research Planning & Management Department; Kyoto R&D Center; Maruho Co., Ltd.; Kyoto Japan
| |
Collapse
|
39
|
Lee HS, Park DE, Song WJ, Park HW, Kang HR, Cho SH, Sohn SW. Effect of 1.8-Cineole in Dermatophagoides pteronyssinus-Stimulated Bronchial Epithelial Cells and Mouse Model of Asthma. Biol Pharm Bull 2017; 39:946-52. [PMID: 27251496 DOI: 10.1248/bpb.b15-00876] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
1.8-Cineole (eucalyptol) is a phytoncide, a volatile organic compound derived from plants. Phytoncides are known to have an anti-inflammatory effect. However, the effects of 1.8-cineole in house dust mite (HDM)-stimulated bronchial epithelial cells are poorly understood. The objective of this study was to assess the effect of 1.8-cineole in HDM-stimulated bronchial epithelial cells and in the HDM-induced murine asthma model. The purpose of the present study is to evaluate the anti-inflammatory effects and mechanism of 1.8-cineole action in HDM-induced airway inflammation. Human bronchial epithelial cells (HBECs) were cultured with Dermatophagoides pteronyssinus (Der p) and 1.8-cineole. Cytokine protein levels, phosphorylation of protein kinases, and intracellular Toll-like receptor 4 (TLR4) expressions were measured. In the murine model, BALB/C mice were sensitized with Der p and were exposed to Der p via intranasal route during the challenge period. 1.8-Cineole was given by inhalation 6 h before the each challenge. Treatment with 1.8-cineole inhibited the Der p-induced cytokine protein expression, phosphorylation of p38 mitogen-activated protein kinase (MAPK) and Akt and intracellular TLR4 expression in HBECs. In the Der p-induced mouse model, airway hyper-responsiveness (AHR) and the number of eosinophils in bronchoalveolar lavage fluid (BALF) was also significantly reduced by 1.8-cineole treatment. The treatment of 1.8-cineole inhibited the increased production of interleukin (IL)-4, IL-13 and IL-17A in BALF after Der p challenge. These results suggest that 1.8-cineole suppresses Der p-induced IL-8, IL-6 and granulocyte macrophage-colony stimulating factor (GM-CSF) production in HBECs. Finally, we confirmed that 1.8-cineole decreases AHR and eosinophilic airway inflammation in Der p-induced asthma mice.
Collapse
Affiliation(s)
- Hyun-Seung Lee
- Laboratory of Allergy and Clinical Immunology, Dongguk University Ilsan Hospital Medical Research Center
| | | | | | | | | | | | | |
Collapse
|
40
|
Ge S, Li T, Yao Q, Yan H, Huiyun Z, Zheng Y, Zhang B, He S. Expression of proteinase-activated receptor (PAR)-2 in monocytes from allergic patients and potential molecular mechanism. Cell Biol Toxicol 2016; 32:529-542. [PMID: 27423452 DOI: 10.1007/s10565-016-9353-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/05/2016] [Indexed: 02/05/2023]
Abstract
Serine proteases play an important role in inflammation via PARs. However, little is known of expression levels of PARs on monocytes of allergic patients, and influence of serine proteases and PARs on TNF-α secretion from monocytes. Using quantitative real-time PCR (qPCR) and flowcytometry techniques, we observed that the expression level of PAR-2 in monocytes of patients with allergic rhinitis and asthma was increased by 42.9 and 38.2 %. It was found that trypsin, thrombin, and tryptase induced up to 200, 320, and 310 % increase in TNF-α release from monocytes at 16 h, respectively. PAR-1 agonist peptide, SFLLR-NH2, and PAR-2 agonist peptide tc-LIGRLO-NH2 provoked up to 210 and 240 % increase in release of TNF-α. Since SCH 79797, a PAR-1 antagonist, and PD98059, an inhibitor of ERK inhibited thrombin- and SFLLR-NH2-induced TNF-α release, the action of thrombin is most likely through a PAR-1- and ERK-mediated signaling mechanism. Similarly, because FSLLRN-NH2, an inhibitor of PAR-2 diminished tryptase- and tc-LIGRLO-NH2-induced TNF-α release, the action of tryptase appears PAR-2 dependent. Moreover, in vivo study showed that both recombinant cockroach major allergens Per a 1 and Per a 7 provoked upregulation of PAR-2 and PAR-1 expression on CD14+ cells in OVA-sensitized mouse peritoneum. In conclusion, increased expression of PAR-2 in monocytes of AR and asthma implicates that PAR-2 likely play a role in allergy. PAR-2- and PAR-1-mediated TNF-α release from monocytes suggests that these unique protease receptors are involved in the pathogenesis of inflammation.
Collapse
Affiliation(s)
- Shuqing Ge
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Liaoning Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning, 121001, People's Republic of China
- Department of Dentistry, the First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, 121001, China
| | - Tao Li
- Department of Infectious Diseases, Shantou University Medical College, Shantou, 515031, China
| | - Qijian Yao
- Department of Infectious Diseases, Shantou University Medical College, Shantou, 515031, China
| | - Hongling Yan
- Clinical Research Centre, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zhang Huiyun
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Liaoning Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Yanshan Zheng
- Department of Infectious Diseases, Shantou University Medical College, Shantou, 515031, China
| | - Bin Zhang
- Department of Dentistry, the First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, 121001, China
| | - Shaoheng He
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Liaoning Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning, 121001, People's Republic of China.
| |
Collapse
|
41
|
Natarajan K, Gottipati KR, Berhane K, Samten B, Pendurthi U, Boggaram V. Proteases and oxidant stress control organic dust induction of inflammatory gene expression in lung epithelial cells. Respir Res 2016; 17:137. [PMID: 27770804 PMCID: PMC5075176 DOI: 10.1186/s12931-016-0455-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022] Open
Abstract
Background Persistant inflammatory responses to infectious agents and other components in organic dust underlie lung injury and development of respiratory diseases. Organic dust components responsible for eliciting inflammation and the mechanisms by which they cause lung inflammation are not fully understood. We studied the mechanisms by which protease activities in poultry dust extracts and intracellular oxidant stress induce inflammatory gene expression in A549 and Beas2B lung epithelial cells. Methods The effects of dust extracts on inflammatory gene expression were analyzed by quantitative polymerase chain reaction (qPCR), enzyme linked immunosorbent (ELISA) and western blot assays. Oxidant stress was probed by dihydroethidium (DHE) labeling, and immunostaining for 4-hydroxynonenal (4-HNE). Effects on interleukin-8 (IL-8) promoter regulation were determined by transient transfection assay. Results Dust extracts contained trypsin and elastase activities, and activated protease activated receptor (PAR)-1 and -2. Serine protease inhibitors and PAR-1 or PAR-2 knockdown suppressed inflammatory gene induction. Dust extract induction of IL-8 gene expression was associated with increased DHE-fluorescence and 4-HNE staining, and antioxidants suppressed inflammatory gene induction. Protease inhibitors and antioxidants suppressed protein kinase C and NF-κB activation and induction of IL-8 promoter activity in cells exposed to dust extract. Conclusions Our studies demonstrate that proteases and intracellular oxidants control organic dust induction of inflammatory gene expression in lung epithelial cells. Targeting proteases and oxidant stress may serve as novel approaches for the treatment of organic dust induced lung diseases. This is the first report on the involvement of oxidant stress in the induction of inflammatory gene expression by organic dust. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0455-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kartiga Natarajan
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708-3154, USA
| | - Koteswara R Gottipati
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708-3154, USA
| | - Kiflu Berhane
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708-3154, USA
| | - Buka Samten
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Usha Pendurthi
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708-3154, USA
| | - Vijay Boggaram
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708-3154, USA.
| |
Collapse
|
42
|
Ramachandran R, Altier C, Oikonomopoulou K, Hollenberg MD. Proteinases, Their Extracellular Targets, and Inflammatory Signaling. Pharmacol Rev 2016; 68:1110-1142. [PMID: 27677721 DOI: 10.1124/pr.115.010991] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Given that over 2% of the human genome codes for proteolytic enzymes and their inhibitors, it is not surprising that proteinases serve many physiologic-pathophysiological roles. In this context, we provide an overview of proteolytic mechanisms regulating inflammation, with a focus on cell signaling stimulated by the generation of inflammatory peptides; activation of the proteinase-activated receptor (PAR) family of G protein-coupled receptors (GPCR), with a mechanism in common with adhesion-triggered GPCRs (ADGRs); and by proteolytic ion channel regulation. These mechanisms are considered in the much wider context that proteolytic mechanisms serve, including the processing of growth factors and their receptors, the regulation of matrix-integrin signaling, and the generation and release of membrane-tethered receptor ligands. These signaling mechanisms are relevant for inflammatory, neurodegenerative, and cardiovascular diseases as well as for cancer. We propose that the inflammation-triggering proteinases and their proteolytically generated substrates represent attractive therapeutic targets and we discuss appropriate targeting strategies.
Collapse
Affiliation(s)
- Rithwik Ramachandran
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Christophe Altier
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Katerina Oikonomopoulou
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Morley D Hollenberg
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| |
Collapse
|
43
|
Bhagwat SS, Larsen AK, Seternes OM, Bang BE. Mixed exposure to bacterial lipopolysaccharide and seafood proteases augments inflammatory signalling in an airway epithelial cell model (A549). Toxicol Ind Health 2016; 32:1866-1874. [PMID: 26149191 DOI: 10.1177/0748233715590914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Seafood industry workers exhibit increased prevalence of respiratory symptoms due to exposure to bioaerosols containing a mixture of bioactive agents. In this study, a human pulmonary epithelial cell model (A549) was exposed to mixtures of bacterial lipopolysaccharide (LPS) and protease-activated receptor-2 (PAR-2) agonists H-Ser-Leu-Ile-Gly-Lys-Val-NH2 (SLIGKV-NH2), purified salmon ( Salmo salar) trypsin or purified king crab ( Paralithodes camtschaticus) trypsin. The inflammatory response was measured based on nuclear factor-kappa B (NF-κB) activation of transcription in a luciferase reporter gene assay and interleukin 8 (IL-8) secretion in an enzyme-linked immunosorbent assay. We observed that mixtures of SLIGKV-NH2 or trypsins with LPS augmented the activation of NF-κB and secretion of IL-8. The effect on IL-8 secretion was synergistic when both trypsins and LPS were used in the lower concentration range. The results demonstrate that exposure to mixtures of agents that are relevant to seafood industry workplaces may lead to increased inflammatory signalling compared with exposure to the individual agents alone. Furthermore, the results indicate that synergism may occur with the combined exposure to seafood trypsins and LPS and is most likely to occur when exposure to either agent is low.
Collapse
Affiliation(s)
- Sampada S Bhagwat
- 1 Department of Occupational and Environmental Medicine, University Hospital of North Norway, Tromsø, Norway.,2 Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Anett K Larsen
- 3 Department of Arctic and Marine Biology, Faculty of Biosciences, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ole-Morten Seternes
- 4 Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Berit E Bang
- 1 Department of Occupational and Environmental Medicine, University Hospital of North Norway, Tromsø, Norway.,2 Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
44
|
Matricardi PM, Kleine-Tebbe J, Hoffmann HJ, Valenta R, Hilger C, Hofmaier S, Aalberse RC, Agache I, Asero R, Ballmer-Weber B, Barber D, Beyer K, Biedermann T, Bilò MB, Blank S, Bohle B, Bosshard PP, Breiteneder H, Brough HA, Caraballo L, Caubet JC, Crameri R, Davies JM, Douladiris N, Ebisawa M, EIgenmann PA, Fernandez-Rivas M, Ferreira F, Gadermaier G, Glatz M, Hamilton RG, Hawranek T, Hellings P, Hoffmann-Sommergruber K, Jakob T, Jappe U, Jutel M, Kamath SD, Knol EF, Korosec P, Kuehn A, Lack G, Lopata AL, Mäkelä M, Morisset M, Niederberger V, Nowak-Węgrzyn AH, Papadopoulos NG, Pastorello EA, Pauli G, Platts-Mills T, Posa D, Poulsen LK, Raulf M, Sastre J, Scala E, Schmid JM, Schmid-Grendelmeier P, van Hage M, van Ree R, Vieths S, Weber R, Wickman M, Muraro A, Ollert M. EAACI Molecular Allergology User's Guide. Pediatr Allergy Immunol 2016; 27 Suppl 23:1-250. [PMID: 27288833 DOI: 10.1111/pai.12563] [Citation(s) in RCA: 535] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The availability of allergen molecules ('components') from several protein families has advanced our understanding of immunoglobulin E (IgE)-mediated responses and enabled 'component-resolved diagnosis' (CRD). The European Academy of Allergy and Clinical Immunology (EAACI) Molecular Allergology User's Guide (MAUG) provides comprehensive information on important allergens and describes the diagnostic options using CRD. Part A of the EAACI MAUG introduces allergen molecules, families, composition of extracts, databases, and diagnostic IgE, skin, and basophil tests. Singleplex and multiplex IgE assays with components improve both sensitivity for low-abundance allergens and analytical specificity; IgE to individual allergens can yield information on clinical risks and distinguish cross-reactivity from true primary sensitization. Part B discusses the clinical and molecular aspects of IgE-mediated allergies to foods (including nuts, seeds, legumes, fruits, vegetables, cereal grains, milk, egg, meat, fish, and shellfish), inhalants (pollen, mold spores, mites, and animal dander), and Hymenoptera venom. Diagnostic algorithms and short case histories provide useful information for the clinical workup of allergic individuals targeted for CRD. Part C covers protein families containing ubiquitous, highly cross-reactive panallergens from plant (lipid transfer proteins, polcalcins, PR-10, profilins) and animal sources (lipocalins, parvalbumins, serum albumins, tropomyosins) and explains their diagnostic and clinical utility. Part D lists 100 important allergen molecules. In conclusion, IgE-mediated reactions and allergic diseases, including allergic rhinoconjunctivitis, asthma, food reactions, and insect sting reactions, are discussed from a novel molecular perspective. The EAACI MAUG documents the rapid progression of molecular allergology from basic research to its integration into clinical practice, a quantum leap in the management of allergic patients.
Collapse
Affiliation(s)
- P M Matricardi
- Paediatric Pneumology and Immunology, Charitè Medical University, Berlin, Germany
| | - J Kleine-Tebbe
- Allergy & Asthma Center Westend, Outpatient Clinic Ackermann, Hanf, & Kleine-Tebbe, Berlin, Germany
| | - H J Hoffmann
- Department of Respiratory Diseases and Allergy, Institute of Clinical Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - R Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - C Hilger
- Department of Infection & Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - S Hofmaier
- Paediatric Pneumology and Immunology, Charitè Medical University, Berlin, Germany
| | - R C Aalberse
- Sanquin Research, Department of Immunopathology, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - I Agache
- Department of Allergy and Clinical Immunology, Faculty of Medicine, Transylvania University of Brasov, Brasov, Romania
| | - R Asero
- Ambulatorio di Allergologia, Clinica San Carlo, Paderno Dugnano, Italy
| | - B Ballmer-Weber
- Allergy Unit, Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - D Barber
- IMMA-School of Medicine, University CEU San Pablo, Madrid, Spain
| | - K Beyer
- Paediatric Pneumology and Immunology, Charitè Medical University, Berlin, Germany
| | - T Biedermann
- Department of Dermatology and Allergology, Technical University Munich, Munich, Germany
| | - M B Bilò
- Allergy Unit, Department of Internal Medicine, University Hospital Ospedali Riuniti di Ancona, Ancona, Italy
| | - S Blank
- Center of Allergy and Environment (ZAUM), Helmholtz Center Munich, Technical University of Munich, Munich, Germany
| | - B Bohle
- Division of Experimental Allergology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - P P Bosshard
- Allergy Unit, Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - H Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - H A Brough
- Paediatric Allergy, Department of Asthma, Allergy and Respiratory Science, King's College London, Guys' Hospital, London, UK
| | - L Caraballo
- Institute for Immunological Research, The University of Cartagena, Cartagena de Indias, Colombia
| | - J C Caubet
- Pediatric Allergy Unit, Department of Child and Adolescent, University Hospitals of Geneva, Geneva, Switzerland
| | - R Crameri
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Davos, Switzerland
| | - J M Davies
- School of Biomedical Sciences, Institute of Biomedical Innovation, Queensland University of Technology, Brisbane, Qld, Australia
| | - N Douladiris
- Allergy Unit, 2nd Paediatric Clinic, National & Kapodistrian University, Athens, Greece
| | - M Ebisawa
- Department of Allergy, Clinical Research Center for Allergology and Rheumatology, Sagamihara National Hospital, Kanagawa, Japan
| | - P A EIgenmann
- Pediatric Allergy Unit, Department of Child and Adolescent, University Hospitals of Geneva, Geneva, Switzerland
| | - M Fernandez-Rivas
- Allergy Department, Hospital Clinico San Carlos IdISSC, Madrid, Spain
| | - F Ferreira
- Division of Allergy and Immunology, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - G Gadermaier
- Division of Allergy and Immunology, Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - M Glatz
- Allergy Unit, Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
- Christine Kühne Center for Allergy Research and Education CK-CARE, Davos, Switzerland
| | - R G Hamilton
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - T Hawranek
- Department of Dermatology, Paracelsus Private Medical University, Salzburg, Austria
| | - P Hellings
- Department of Otorhinolaryngology, Academic Medical Center (AMC), Amsterdam, The Netherlands
- Department of Otorhinolaryngology, University Hospitals Leuven, Leuven, Belgium
| | - K Hoffmann-Sommergruber
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - T Jakob
- Department of Dermatology and Allergology, University Medical Center Giessen and Marburg, Justus Liebig University Giessen, Giessen, Germany
| | - U Jappe
- Division of Clinical and Molecular Allergology, Research Centre Borstel, Airway Research Centre North (ARCN), Member of the German Centre for Lung Research (DZL), Borstel, Germany
- Interdisciplinary Allergy Division, Department of Pneumology, University of Lübeck, Lübeck, Germany
| | - M Jutel
- Department of Clinical Immunology, 'ALL-MED' Medical Research Institute, Wrocław Medical University, Wrocław, Poland
| | - S D Kamath
- Molecular Allergy Research Laboratory, Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville City, Qld, Australia
| | - E F Knol
- Departments of Immunology and Dermatology/Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - P Korosec
- University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| | - A Kuehn
- Department of Infection & Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - G Lack
- King's College London, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
- Division of Asthma, Allergy and Lung Biology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - A L Lopata
- Department of Clinical Immunology, 'ALL-MED' Medical Research Institute, Wrocław Medical University, Wrocław, Poland
| | - M Mäkelä
- Skin and Allergy Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - M Morisset
- National Service of Immuno-Allergology, Centre Hospitalier Luxembourg (CHL), Luxembourg, UK
| | - V Niederberger
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - A H Nowak-Węgrzyn
- Pediatric Allergy and Immunology, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - N G Papadopoulos
- Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - E A Pastorello
- Unit of Allergology and Immunology, Niguarda Ca' Granda Hospital, Milan, Italy
| | - G Pauli
- Service de Pneumologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - T Platts-Mills
- Department of Microbiology & Immunology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - D Posa
- Paediatric Pneumology and Immunology, Charitè Medical University, Berlin, Germany
| | - L K Poulsen
- Allergy Clinic, Copenhagen University Hospital, Copenhagen, Denmark
| | - M Raulf
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Ruhr-University Bochum (IPA), Bochum, Germany
| | - J Sastre
- Allergy Division, Fundación Jimenez Díaz, Madrid, Spain
| | - E Scala
- Experimental Allergy Unit, IDI-IRCCS, Rome, Italy
| | - J M Schmid
- Department of Respiratory Diseases and Allergy, Institute of Clinical Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - P Schmid-Grendelmeier
- Allergy Unit, Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
- Christine Kühne Center for Allergy Research and Education CK-CARE, Davos, Switzerland
| | - M van Hage
- Department of Medicine Solna, Clinical Immunology and Allergy Unit, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - R van Ree
- Departments of Experimental Immunology and of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - S Vieths
- Department of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - R Weber
- School of Medicine, University of Colorado, Denver, CO, USA
- Department of Medicine, National Jewish Health Service, Denver, CO, USA
| | - M Wickman
- Sachs' Children's Hospital, Karolinska Institutet, Stockholm, Sweden
| | - A Muraro
- The Referral Centre for Food Allergy Diagnosis and Treatment Veneto Region, Department of Mother and Child Health, University of Padua, Padua, Italy
| | - M Ollert
- Department of Infection & Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
45
|
Papazian D, Hansen S, Würtzen PA. Airway responses towards allergens - from the airway epithelium to T cells. Clin Exp Allergy 2016; 45:1268-87. [PMID: 25394747 DOI: 10.1111/cea.12451] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The prevalence of allergic diseases such as allergic rhinitis is increasing, affecting up to 30% of the human population worldwide. Allergic sensitization arises from complex interactions between environmental exposures and genetic susceptibility, resulting in inflammatory T helper 2 (Th2) cell-derived immune responses towards environmental allergens. Emerging evidence now suggests that an epithelial dysfunction, coupled with inherent properties of environmental allergens, can be responsible for the inflammatory responses towards allergens. Several epithelial-derived cytokines, such as thymic stromal lymphopoietin (TSLP), IL-25 and IL-33, influence tissue-resident dendritic cells (DCs) as well as Th2 effector cells. Exposure to environmental allergens does not elicit Th2 inflammatory responses or any clinical symptoms in nonatopic individuals, and recent findings suggest that a nondamaged, healthy epithelium lowers the DCs' ability to induce inflammatory T-cell responses towards allergens. The purpose of this review was to summarize the current knowledge on which signals from the airway epithelium, from first contact with inhaled allergens all the way to the ensuing Th2-cell responses, influence the pathology of allergic diseases.
Collapse
Affiliation(s)
- D Papazian
- Department of Cancer & Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,ALK, Hørsholm, Denmark
| | - S Hansen
- Department of Cancer & Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
46
|
Williams PB, Barnes CS, Portnoy JM. Innate and Adaptive Immune Response to Fungal Products and Allergens. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2016; 4:386-95. [PMID: 26755096 DOI: 10.1016/j.jaip.2015.11.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/05/2015] [Accepted: 11/02/2015] [Indexed: 02/06/2023]
Abstract
Exposure to fungi and their products is practically ubiquitous, yet most of this is of little consequence to most healthy individuals. This is because there are a number of elaborate mechanisms to deal with these exposures. Most of these mechanisms are designed to recognize and neutralize such exposures. However, in understanding these mechanisms it has become clear that many of them overlap with our ability to respond to disruptions in tissue function caused by trauma or deterioration. These responses involve the innate and adaptive immune systems usually through the activation of nuclear factor kappa B and the production of cytokines that are considered inflammatory accompanied by other factors that can moderate these reactivities. Depending on different genetic backgrounds and the extent of activation of these mechanisms, various pathologies with resulting symptoms can ensue. Complicating this is the fact that these mechanisms can bias toward type 2 innate and adaptive immune responses. Thus, to understand what we refer to as allergens from fungal sources, we must first understand how they influence these innate mechanisms. In doing so it has become clear that many of the proteins that are described as fungal allergens are essentially homologues of our own proteins that signal or cause tissue disruptions.
Collapse
Affiliation(s)
- P Brock Williams
- Division of Allergy/Immunology, Children's Mercy Hospital, Kansas City, Mo
| | - Charles S Barnes
- Division of Allergy/Immunology, Children's Mercy Hospital, Kansas City, Mo
| | - Jay M Portnoy
- Division of Allergy/Immunology, Children's Mercy Hospital, Kansas City, Mo.
| | | |
Collapse
|
47
|
Breiteneder H. Grundlagen natürlicher Allergene. ALLERGOLOGIE 2016. [DOI: 10.1007/978-3-642-37203-2_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
48
|
Sircar G, Saha B, Mandal RS, Pandey N, Saha S, Gupta Bhattacharya S. Purification, Cloning and Immuno-Biochemical Characterization of a Fungal Aspartic Protease Allergen Rhi o 1 from the Airborne Mold Rhizopus oryzae. PLoS One 2015; 10:e0144547. [PMID: 26672984 PMCID: PMC4682942 DOI: 10.1371/journal.pone.0144547] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/19/2015] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Fungal allergy is considered as serious health problem worldwide and is increasing at an alarming rate in the industrialized areas. Rhizopus oyzae is a ubiquitously present airborne pathogenic mold and an important source of inhalant allergens for the atopic population of India. Here, we report the biochemical and immunological features of its 44 kDa sero-reactive aspartic protease allergen, which is given the official designation 'Rhi o 1'. METHOD The natural Rhi o 1 was purified by sequential column chromatography and its amino acid sequence was determined by mass spectrometry and N-terminal sequencing. Based on its amino acid sequence, the cDNA sequence was identified, cloned and expressed to produce recombinant Rhi o 1. The allergenic activity of rRhi o 1 was assessed by means of its IgE reactivity and histamine release ability. The biochemical property of Rhi o 1 was studied by enzyme assay. IgE-inhibition experiments were performed to identify its cross-reactivity with the German cockroach aspartic protease allergen Bla g 2. For precise characterization of the cross-reactive epitope, we used anti-Bla g 2 monoclonal antibodies for their antigenic specificity towards Rhi o 1. A homology based model of Rhi o 1 was built and mapping of the cross-reactive conformational epitope was done using certain in silico structural studies. RESULTS The purified natural nRhi o 1 was identified as an endopeptidase. The full length allergen cDNA was expressed and purified as recombinant rRhi o 1. Purified rRhi o 1 displayed complete allergenicity similar to the native nRhi o 1. It was recognized by the serum IgE of the selected mold allergy patients and efficiently induced histamine release from the sensitized PBMC cells. This allergen was identified as an active aspartic protease functional in low pH. The Rhi o 1 showed cross reactivity with the cockroach allergen Bla g 2, as it can inhibit IgE binding to rBla g 2 up to certain level. The rBla g 2 was also found to cross-stimulate histamine release from the effector cells sensitized with anti-Rhi o 1 serum IgE. This cross-reactivity was found to be mediated by a common mAb4C3 recognizable conformational epitope. Bioinformatic studies revealed high degree of structural resemblances between the 4C3 binding sites of both the allergens. CONCLUSION/SIGNIFICANCE The present study reports for the first time anew fungal aspartic protease allergen designated as Rhi o 1, which triggers IgE-mediated sensitization leading to various allergic diseases. Here we have characterized the recombinant Rhi o 1 and its immunological features including cross-reactive epitope information that will facilitate the component-resolved diagnosis of mold allergy.
Collapse
Affiliation(s)
- Gaurab Sircar
- Division of Plant Biology, Bose Institute (Main campus), 93/1 Acharya Prafulla Chandra Road, Kolkata– 700009, West Bengal, India
| | - Bodhisattwa Saha
- Division of Plant Biology, Bose Institute (Main campus), 93/1 Acharya Prafulla Chandra Road, Kolkata– 700009, West Bengal, India
| | - Rahul Shubhra Mandal
- Biomedical Informatics Center, National Institute of Cholera and Enteric Diseases, Kolkata-700010, West Bengal, India
| | - Naren Pandey
- Department of Allergy and Asthma, Belle Vue Clinic, 9, Dr. U. N. Brahmachari Street, Kolkata—700001, West Bengal, India
| | - Sudipto Saha
- Bioinformatics Centre, Bose Institute (Centenary Building), P 1/12, C. I. T. Road, Scheme–VIIM, Kolkata– 700054, West Bengal, India
| | - Swati Gupta Bhattacharya
- Division of Plant Biology, Bose Institute (Main campus), 93/1 Acharya Prafulla Chandra Road, Kolkata– 700009, West Bengal, India
| |
Collapse
|
49
|
Bouaziz A, Walgraffe D, Bouillot C, Herman J, Foguenne J, Gothot A, Louis R, Hentges F, Jacquet A, Mailleux AC, Chevigné A, Galleni M, Adam E, Dumez ME. Development of recombinant stable house dust mite allergen Der p 3 molecules for component-resolved diagnosis and specific immunotherapy. Clin Exp Allergy 2015; 45:823-34. [PMID: 25406386 DOI: 10.1111/cea.12452] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 07/30/2014] [Accepted: 09/10/2014] [Indexed: 01/22/2023]
Abstract
BACKGROUND The allergen Der p 3 is underrepresented in house dust mite (HDM) extracts probably due to autolysis. Recombinant stable molecule of the allergen is thus needed to improve the diagnosis of allergy and the safety and efficacy of immunotherapy. OBJECTIVE The current study reports the immunological characterization of two recombinant molecules of the HDM allergen Der p 3 as useful tools for diagnosis and immunotherapy. METHODS Recombinant mature (rDer p 3) and immature (proDer p 3) Der p 3 and their corresponding S196A mutants were produced in Pichia pastoris and purified. The stability, IgE-binding capacity and allergenicity of the different proteins were analysed and compared with those of the major mite allergen Der p 1 used as a reference. Additionally, the immunogenicity of the different allergens was evaluated in a murine model of Der p 3 sensitization. RESULTS Compared to the IgE reactivity to recombinant and natural Der p 3 (nDer p 3), the mean IgE binding of patient's sera to rDer p 3-S196A (50%) was higher. The poorly binding to nDer p 3 or rDer p 3 was due to autolysis of the allergen. Contrary to Der p 3, proDer p 3 displayed very weak IgE reactivity, as measured by sandwich ELISA and competitive inhibition, rat basophil leukaemia degranulation and human basophil activation assays. Moreover, proDer p 3 induced a TH 1-biased immune response that prevented allergic response in mice but retained Der p 3-specific T-cell reactivity. CONCLUSION rDer p 3-S196A should be used for the diagnosis of HDM allergy elicited by Der p 3, and proDer p 3 may represent a hypoallergen of Der p 3.
Collapse
Affiliation(s)
- A Bouaziz
- Macromolécules Biologiques, Centre d'ingénierie des Protéines, Université de Liège, Liège, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Son GY, Son A, Yang YM, Park W, Chang I, Lee JH, Shin DM. Airborne allergens induce protease activated receptor-2-mediated production of inflammatory cytokines in human gingival epithelium. Arch Oral Biol 2015; 61:138-43. [PMID: 26561723 DOI: 10.1016/j.archoralbio.2015.10.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 08/31/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE In reaching the airways inhaled allergens pass through and contact with the oral mucosa. Although they are often responsible for initiating asthmatic attacks, it is unknown whether airborne allergens can also trigger chronic inflammation of gingival epithelial cells leading to chronic periodontitis. In this study, we investigated the inflammatory responses of human gingival epithelial cells (HGECs) to airborne allergens, particularly German cockroach extract (GCE) with a focus on calcium signaling. DESIGN HGECs isolated from healthy donors were stimulated with GCE. Intracellular Ca(2+) concentration ([Ca(2+)]i) was measured with Fura-2-acetoxymethyl ester (Fura-2/AM) staining. Expression of inflammatory cytokines interleukin (IL)-8, IL-1β, IL-6, and NOD-like receptor family, pyridine domain-containing (NLRP) 3 was analyzed using reverse transcription-polymerase chain reaction (RT-PCR). RESULTS GCE promoted increase in the [Ca(2+)]i in a dose-dependent manner. Depletion of endoplasmic reticulum (ER) Ca(2+) by the ER Ca(2+) ATPase inhibitor thapsigargin (Tg) but not the depletion of extracellular Ca(2+) abolished the GCE-induced increase in [Ca(2+)]i. Treatment of phospholipase C (PLC) inhibitor (U73122) or 1,4,5-trisinositolphosphate (IP3) receptor inhibitor (2-APB) also prevented GCE-induced increase in [Ca(2+)]i. Protease activated receptor (PAR)-2 activation mainly mediated the GCE-induced increase in [Ca(2+)]i and enhanced the expression of IL-8, NLRP3, IL-1β, and IL-6 in HGECs. CONCLUSIONS GCE activates PAR-2, which can induce PLC/IP3-dependent Ca(2+) signaling pathway, ultimately triggering inflammation via the production of pro-inflammatory cytokines such as IL-1β, IL-6, IL-8, and NLRP 3 in HGECs.
Collapse
Affiliation(s)
- Ga-Yeon Son
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea; BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Aran Son
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Yu-Mi Yang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Wonse Park
- Department of Advanced General Dentistry, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Inik Chang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Jae-Ho Lee
- Department of Pediatric Dentistry, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Dong Min Shin
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Republic of Korea; BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Republic of Korea.
| |
Collapse
|