1
|
Shan Q, Qiu J, Dong Z, Xu X, Zhang S, Ma J, Liu S. Lung Immune Cell Niches and the Discovery of New Cell Subtypes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405490. [PMID: 39401416 PMCID: PMC11615829 DOI: 10.1002/advs.202405490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/31/2024] [Indexed: 12/06/2024]
Abstract
Immune cells in the lungs are important for maintaining lung function. The importance of immune cells in defending against lung diseases and infections is increasingly recognized. However, a primary knowledge gaps in current studies of lung immune cells is the understanding of their subtypes and functional heterogeneity. Increasing evidence supports the existence of novel immune cell subtypes that engage in the complex crosstalk between lung-resident immune cells, recruited immune cells, and epithelial cells. Therefore, further studies on how immune cells respond to perturbations in the pulmonary microenvironment are warranted. This review explores the processes behind the formation of the immune cell niche during lung development, and the characteristics and cell interaction modes of several major lung-resident immune cells. It indicates that distinct lung microenvironments or inflammatory niches can mediate the formation of different cell subtypes. These findings summarize and clarify paths to identify new cell subtypes that originate from resident progenitor cells and recruited peripheral cells, which are remodeled by the pulmonary microenvironment. The development of new techniques combining transcriptome analysis and location information is essential for identifying new immune cell subtypes and their relative immune niches, as well as for uncovering the molecular mechanisms of immune cell-mediated lung homeostasis.
Collapse
Affiliation(s)
- Qing'e Shan
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- School of Public HealthShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
| | - Jiahuang Qiu
- Dongguan Key Laboratory of Environmental MedicineSchool of Public HealthGuangdong Medical UniversityDongguan523808P. R. China
| | - Zheng Dong
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- School of Public HealthShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
| | - Xiaotong Xu
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- School of Environmental SciencesUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Shuping Zhang
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- School of Public HealthShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
| | - Juan Ma
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- School of Environmental SciencesUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Sijin Liu
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- School of Public HealthShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- School of Environmental SciencesUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| |
Collapse
|
2
|
Moon HG, Eccles JD, Kim SJ, Kim KH, Kim YM, Rehman J, Lee H, Kanabar P, Christman JW, Ackerman SJ, Ascoli C, Kang H, Choi HS, Kim M, You S, Park GY. Complement C1q essential for aeroallergen sensitization via CSF1R + conventional dendritic cells type 2. J Allergy Clin Immunol 2023; 152:1141-1152.e2. [PMID: 37562753 PMCID: PMC10923196 DOI: 10.1016/j.jaci.2023.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/29/2023] [Accepted: 07/20/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Dendritic cells (DCs) are heterogeneous, comprising multiple subsets with unique functional specifications. Our previous work has demonstrated that the specific conventional type 2 DC subset, CSF1R+cDC2s, plays a critical role in sensing aeroallergens. OBJECTIVE It remains to be understood how CSF1R+cDC2s recognize inhaled allergens. We sought to elucidate the transcriptomic programs and receptor-ligand interactions essential for function of this subset in allergen sensitization. METHODS We applied single-cell RNA sequencing to mouse lung DCs. Conventional DC-selective knockout mouse models were employed, and mice were subjected to inhaled allergen sensitization with multiple readouts of asthma pathology. Under the clinical arm of this work, human lung transcriptomic data were integrated with mouse data, and bronchoalveolar lavage (BAL) specimens were collected from subjects undergoing allergen provocation, with samples assayed for C1q. RESULTS We found that C1q is selectively enriched in lung CSF1R+cDC2s, but not in other lung cDC2 or cDC1 subsets. Depletion of C1q in conventional DCs significantly attenuates allergen sensing and features of asthma. Additionally, we found that C1q binds directly to human dust mite allergen, and the C1q receptor CD91 (LRP1) is required for lung CSF1R+cDC2s to recognize the C1q-allergen complex and induce allergic lung inflammation. Lastly, C1q is enriched in human BAL samples following subsegmental allergen challenge, and human RNA sequencing data demonstrate close homology between lung IGSF21+DCs and mouse CSF1R+cDC2s. CONCLUSIONS C1q is secreted from the CSF1R+cDC2 subset among conventional DCs. Our data indicate that the C1q-LRP1 axis represents a candidate for translational therapeutics in the prevention and suppression of allergic lung inflammation.
Collapse
Affiliation(s)
- Hyung-Geun Moon
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago.
| | - Jacob D Eccles
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago
| | - Seung-Jae Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago
| | - Ki-Hyun Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago
| | - Young-Mee Kim
- Department of Pharmacology, University of Illinois College of Medicine, Chicago
| | - Jalees Rehman
- Department of Pharmacology, University of Illinois College of Medicine, Chicago
| | - Hyun Lee
- College of Pharmacy, University of Illinois at Chicago, Chicago
| | - Pinal Kanabar
- Research Informatics Core, University of Illinois at Chicago, Chicago
| | - John W Christman
- Section of Pulmonary, Critical Care, and Sleep Medicine, Columbus; Davis Heart and Lung Research Center, The Ohio State University, Columbus
| | - Steven J Ackerman
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago; Department of Medicine, University of Illinois at Chicago, Chicago
| | - Christian Ascoli
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Minhyung Kim
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles
| | - Sungyong You
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles
| | - Gye Young Park
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago; Jesse Brown Veterans Affairs Medical Center, Chicago.
| |
Collapse
|
3
|
Raith M, Swoboda I. Birch pollen-The unpleasant herald of spring. FRONTIERS IN ALLERGY 2023; 4:1181675. [PMID: 37255542 PMCID: PMC10225653 DOI: 10.3389/falgy.2023.1181675] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/24/2023] [Indexed: 06/01/2023] Open
Abstract
Type I respiratory allergies to birch pollen and pollen from related trees of the order Fagales are increasing in industrialized countries, especially in the temperate zone of the Northern hemisphere, but the reasons for this increase are still debated and seem to be multifaceted. While the most important allergenic molecules of birch pollen have been identified and characterized, the contribution of other pollen components, such as lipids, non-allergenic immunomodulatory proteins, or the pollen microbiome, to the development of allergic reactions are sparsely known. Furthermore, what also needs to be considered is that pollen is exposed to external influences which can alter its allergenicity. These external influences include environmental factors such as gaseous pollutants like ozone or nitrogen oxides or particulate air pollutants, but also meteorological events like changes in temperature, humidity, or precipitation. In this review, we look at the birch pollen from different angles and summarize current knowledge on internal and external influences that have an impact on the allergenicity of birch pollen and its interactions with the epithelial barrier. We focus on epithelial cells since these cells are the first line of defense in respiratory disease and are increasingly considered to be a regulatory tissue for the protection against the development of respiratory allergies.
Collapse
|
4
|
Satala D, Bras G, Kozik A, Rapala-Kozik M, Karkowska-Kuleta J. More than Just Protein Degradation: The Regulatory Roles and Moonlighting Functions of Extracellular Proteases Produced by Fungi Pathogenic for Humans. J Fungi (Basel) 2023; 9:jof9010121. [PMID: 36675942 PMCID: PMC9865821 DOI: 10.3390/jof9010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/17/2023] Open
Abstract
Extracellular proteases belong to the main virulence factors of pathogenic fungi. Their proteolytic activities plays a crucial role in the acquisition of nutrients from the external environment, destroying host barriers and defenses, and disrupting homeostasis in the human body, e.g., by affecting the functions of plasma proteolytic cascades, and playing sophisticated regulatory roles in various processes. Interestingly, some proteases belong to the group of moonlighting proteins, i.e., they have additional functions that contribute to successful host colonization and infection development, but they are not directly related to proteolysis. In this review, we describe examples of such multitasking of extracellular proteases that have been reported for medically important pathogenic fungi of the Candida, Aspergillus, Penicillium, Cryptococcus, Rhizopus, and Pneumocystis genera, as well as dermatophytes and selected endemic species. Additional functions of proteinases include supporting binding to host proteins, and adhesion to host cells. They also mediate self-aggregation and biofilm formation. In addition, fungal proteases affect the host immune cells and allergenicity, understood as the ability to stimulate a non-standard immune response. Finally, they play a role in the proper maintenance of cellular homeostasis. Knowledge about the multifunctionality of proteases, in addition to their canonical roles, greatly contributes to an understanding of the mechanisms of fungal pathogenicity.
Collapse
Affiliation(s)
- Dorota Satala
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Grazyna Bras
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
- Correspondence:
| |
Collapse
|
5
|
Robles TF, Bai S, Meng YY. Ozone Pollution, Perceived Support at Home, and Asthma Symptom Severity in the Adolescent Sample of the California Health Interview Survey. Int J Behav Med 2022; 30:398-408. [PMID: 35655059 PMCID: PMC10167194 DOI: 10.1007/s12529-022-10103-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Outdoor air pollution, including ozone (O3) pollution, and childhood family environments may interact and impact asthma exacerbations in children. Previous epidemiology studies have primarily focused on stress in the home, rather than support, and whether psychosocial factors modify the association between pollution and health outcomes, rather than whether pollution exposure modifies associations between psychosocial factors and health outcomes. METHODS Data from the cross-sectional 2003 representative, population-based California Health Interview Survey were linked with air quality monitoring data on O3 pollution from the California Air Resources Board. Adolescents (N = 209) ages 12-17 who reported an asthma diagnosis and lived within 5 mi of the nearest air monitoring station had linked O3 data for a 12-month period preceding the survey interview date. Adolescents reported perceived available support from an adult at home and frequency of asthma symptoms. RESULTS In unadjusted models, for adolescents living in high O3 pollution regions, greater perceived support was related to lower asthma symptom frequency. Follow-up analyses suggested that the most plausible interpretation of the interaction was that O3 exposure modified the association between perceived support and symptom frequency. O3 × perceived support interactions were not statistically significant after adjusting for covariates. CONCLUSIONS These data provide preliminary evidence that the association between the lack of support in the home environment and worse asthma symptoms may be stronger in areas with higher O3 exposure. Future work may benefit from incorporating personal pollution exposure assessments, comprehensive family environment assessments, and longitudinal follow-up of asthma exacerbations over time.
Collapse
Affiliation(s)
- Theodore F Robles
- Department of Psychology, University of California, 1285 Psychology Building, Box 951563, Los Angeles, CA, 90095-1563, USA.
| | - Sunhye Bai
- Department of Human Development and Family Studies, The Pennsylvania State University, 216 Health and Human Development Building, University Park, PA, 16802, USA
| | - Ying-Ying Meng
- Center for Health Policy Research, UCLA Fielding School of Public Health, 10960 Wilshire Blvd, Suite 1550, Box 957143, Los Angeles, CA, 90095-7143, USA
| |
Collapse
|
6
|
Involvement and therapeutic implications of airway epithelial barrier dysfunction in type 2 inflammation of asthma. Chin Med J (Engl) 2022; 135:519-531. [PMID: 35170505 PMCID: PMC8920422 DOI: 10.1097/cm9.0000000000001983] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Type 2 inflammation is a complex immune response and primary mechanism for several common allergic diseases including allergic rhinitis, allergic asthma, atopic dermatitis, and chronic rhinosinusitis with nasal polyps. It is the predominant type of immune response against helminths to prevent their tissue infiltration and induce their expulsion. Recent studies suggest that epithelial barrier dysfunction contributes to the development of type 2 inflammation in asthma, which may partly explain the increasing prevalence of asthma in China and around the globe. The epithelial barrier hypothesis has recently been proposed and has received great interest from the scientific community. The development of leaky epithelial barriers leads to microbial dysbiosis and the translocation of bacteria to inter- and sub-epithelial areas and the development of epithelial tissue inflammation. Accordingly, preventing the impairment and promoting the restoration of a deteriorated airway epithelial barrier represents a promising strategy for the treatment of asthma. This review introduces the interaction between type 2 inflammation and the airway epithelial barrier in asthma, the structure and molecular composition of the airway epithelial barrier, and the assessment of epithelial barrier integrity. The role of airway epithelial barrier disruption in the pathogenesis of asthma will be discussed. In addition, the possible mechanisms underlying the airway epithelial barrier dysfunction induced by allergens and environmental pollutants, and current treatments to restore the airway epithelial barrier are reviewed.
Collapse
|
7
|
De Leeuw E, Bosteels C, Lambrecht BN, Hammad H. Isolation of Conventional Murine Lung Dendritic Cell Subsets. Methods Mol Biol 2022; 2506:237-255. [PMID: 35771476 DOI: 10.1007/978-1-0716-2364-0_17] [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] [Indexed: 06/15/2023]
Abstract
In the lungs, immune cells make contact with different antigens every day. This requires an adequate immune response. Dendritic cells (DCs) form a dense network in the respiratory mucosa and continuously sample inhaled allergens. They play an important role in bridging innate and adaptive immunity. DCs are classically divided into plasmacytoid DCs (pDCs) and conventional DCs (cDCs). cDCs in the steady-state are further subdivided into cDC1s and cDC2s based on their ontogeny and distinct non-redundant functions. Recently, a hyperactivated state of cDC2s has been described that arises during inflammation, coined inflammatory cDC2s (inf-cDC2s) that phenotypically mimics monocyte-derived cells and has a hybrid cDC1/macrophage functional identity. This chapter describes different enrichment methods and a fluorescence-activated cell sorting protocol that in combination allow for discrimination and isolation of pure DC subsets from the murine lung. The chapter represents an up-to-date, universal framework that can be adapted to other tissues and species which is an added value in intra- and interspecies comparative research.
Collapse
Affiliation(s)
- Elisabeth De Leeuw
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Cédric Bosteels
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.
- Department of Internal Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
8
|
Merk VM, Brunner T. Immunosuppressive glucocorticoids at epithelial barriers in the regulation of anti-viral immune response. VITAMINS AND HORMONES 2021; 117:77-100. [PMID: 34420586 DOI: 10.1016/bs.vh.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The anti-inflammatory action of adrenal-derived glucocorticoids has been recognized since several decades. This knowledge has found broad application in the clinics and today synthetic glucocorticoids are widely used in the treatment of various inflammatory diseases. However, the use of synthetic glucocorticoids in the treatment of diseases associated with viral infections of epithelial surfaces, like the lung or the intestine, is still under debate and seems not as efficient as desired. Basic research on the anti-viral immune responses and on regulatory mechanisms in the prevention of immunopathological disorders, however, has led us back again to focus on endogenous glucocorticoid synthesis. It has become established that this synthesis is not restricted to the adrenal glands alone, but that numerous tissues also produce glucocorticoids in situ. Extra-adrenal derived glucocorticoids have the capacity to locally control and maintain immune homeostasis under steady-state and inflammatory conditions. Here, we discuss the current knowledge of extra-adrenal glucocorticoid synthesis in the lung and the intestine, and its role in the regulation of anti-viral immune responses.
Collapse
Affiliation(s)
- V M Merk
- Chair of Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - T Brunner
- Chair of Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
9
|
Nur Husna SM, Tan HTT, Md Shukri N, Mohd Ashari NS, Wong KK. Nasal Epithelial Barrier Integrity and Tight Junctions Disruption in Allergic Rhinitis: Overview and Pathogenic Insights. Front Immunol 2021; 12:663626. [PMID: 34093555 PMCID: PMC8176953 DOI: 10.3389/fimmu.2021.663626] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/22/2021] [Indexed: 12/20/2022] Open
Abstract
Allergic rhinitis (AR) is a common disorder affecting up to 40% of the population worldwide and it usually persists throughout life. Nasal epithelial barrier constitutes the first line of defense against invasion of harmful pathogens or aeroallergens. Cell junctions comprising of tight junctions (TJs), adherens junctions, desmosomes and hemidesmosomes form the nasal epithelial barrier. Impairment of TJ molecules plays causative roles in the pathogenesis of AR. In this review, we describe and discuss the components of TJs and their disruption leading to development of AR, as well as regulation of TJs expression by epigenetic changes, neuro-immune interaction, epithelial-derived cytokines (thymic stromal lymphopoietin, IL-25 and IL-33), T helper 2 (Th2) cytokines (IL-4, IL-5, IL-6 and IL-13) and innate lymphoid cells. These growing evidence support the development of novel therapeutic approaches to restore nasal epithelial TJs expression in AR patients.
Collapse
Affiliation(s)
- Siti Muhamad Nur Husna
- Department of Immunology, School of Medical Sciences Malaysia, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Hern-Tze Tina Tan
- Department of Immunology, School of Medical Sciences Malaysia, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Norasnieda Md Shukri
- Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Department of Otorhinolaryngology, Head and Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Noor Suryani Mohd Ashari
- Department of Immunology, School of Medical Sciences Malaysia, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences Malaysia, Universiti Sains Malaysia, Kubang Kerian, Malaysia.,Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
10
|
Pedan H, Janosova V, Hajtman A, Calkovsky V. Non-Reflex Defense Mechanisms of Upper Airway Mucosa: Possible Clinical Application. Physiol Res 2021; 69:S55-S67. [PMID: 32228012 DOI: 10.33549/physiolres.934404] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The sinonasal mucosa has an essential role in defense mechanisms of the upper respiratory tract. The innate immune system presents the primary defense against noxious microorganisms followed by induction of the adaptive immune mechanisms as a consequence of the presence of pathogens. This well-known activation of adaptive immune system in response to presence of the antigen on mucosal surfaces is now broadly applicated in vaccinology research. Prevention of infectious diseases belongs to substantial challenges in maintaining the population health. Non-invasive, easily applicable mucosal vaccination purposes various research opportunities that could be usable in daily practice. However, the existence of multiple limitations such as rapid clearance of vaccine from nasal mucosa by means of mucociliary transport represents a great challenge in development of safe and efficient vaccines. Here we give an updated view on nasal functions with focus on nasal mucosal immunity and its potential application in vaccination in nearly future.
Collapse
Affiliation(s)
- H Pedan
- Clinic of Otorhinolaryngology and Head and Neck Surgery, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, and Martin University Hospital, Martin, Slovak
| | | | | | | |
Collapse
|
11
|
Villena J, Kitazawa H. The Modulation of Mucosal Antiviral Immunity by Immunobiotics: Could They Offer Any Benefit in the SARS-CoV-2 Pandemic? Front Physiol 2020; 11:699. [PMID: 32670091 PMCID: PMC7326040 DOI: 10.3389/fphys.2020.00699] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023] Open
Abstract
Viral respiratory infections are of major importance because of their capacity to cause of a high degree of morbidity and mortality in high-risk populations, and to rapidly spread between countries. Perhaps the best example of this global threat is the infectious disease caused by the new SARS-CoV-2 virus, which has infected more than 4 million people worldwide, causing the death of 287,000 persons according to the WHO's situation report on May 13, 2020. The availability of therapeutic tools that would be used massively to prevent or mitigate the detrimental effects of emerging respiratory viruses on human health is therefore mandatory. In this regard, research from the last decade has reported the impact of the intestinal microbiota on the respiratory immunity. It was conclusively demonstrated how the variations in the intestinal microbiota affect the responses of respiratory epithelial cells and antigen presenting cells against respiratory virus attack. Moreover, the selection of specific microbial strains (immunobiotics) with the ability to modulate immunity in distal mucosal sites made possible the generation of nutritional interventions to strengthen respiratory antiviral defenses. In this article, the most important characteristics of the limited information available regarding the immune response against SARS-CoV-2 virus are revised briefly. In addition, this review summarizes the knowledge on the cellular and molecular mechanisms involved in the improvement of respiratory antiviral defenses by beneficial immunobiotic microorganisms such as Lactobacillus rhamnosus CRL1505. The ability of beneficial microorganisms to enhance type I interferons and antiviral factors in the respiratory tract, stimulate Th1 response and antibodies production, and regulate inflammation and coagulation activation during the course of viral infections reducing tissue damage and preserving lung functionally, clearly indicate the potential of immunobiotics to favorably influence the immune response against SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán, Argentina
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
12
|
Zheng R, Chen Y, Shi J, Wang K, Huang X, Sun Y, Yang Q. Combinatorial IL-17RB, ST2, and TSLPR Signaling in Dendritic Cells of Patients With Allergic Rhinitis. Front Cell Dev Biol 2020; 8:207. [PMID: 32309281 PMCID: PMC7145954 DOI: 10.3389/fcell.2020.00207] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Objectives Myeloid dendritic cells (DCs) in patients with allergic rhinitis (AR) express higher levels of IL-17RB, ST2, and TSLPR. However, their functional roles in DCs are much less clear. This study aimed to determine the combined effects of these three receptor signals on the T cell-polarizing function of DCs in AR patients. Methods Monocyte-derived DCs (mo-DCs) were generated and stimulated with Toll-like receptor (TLR) 1-9 ligands. Der.p1-induced mo-DCs were stimulated with different combinations of IL-25, IL-33, and TSLP to determine phenotypic characteristics and then co-cultured with CD4+ T cells to assess Th2 cytokine production. Expression levels of IL-17RB, ST2, and TSLPR on myeloid DCs (mDCs) from peripheral blood of AR and healthy subjects were detected to confirm the association of these receptors with disease severity. Results TLR ligands induced AR-derived mo-DCs to increase IL-17RB, ST2, and TSLPR expression by varying degrees; among these, Der.p1 was the strongest inducer. Der.p1-induced mo-DCs from AR showed increased OX40L expression. IL-25, IL-33, and TSLP (alone or in double combination) significantly increased OX40L expression on Der.p1-induced mo-DCs from AR, thereby increasing the production of IL-4, IL-5, and IL-13 in co-cultured CD4+ T cells; triple combination further enhanced these effects. The percentage of IL-17RB+ST2+TSLPR+ mDCs was increased in AR, higher in moderate to severe phase than in mild phase, and positively correlated with the percentages of IL-4+, IL-5+, and IL-13+ T cells. Conclusion A combination of IL-17RB, ST2, and TSLPR signals amplified the Th2-polarizing function of DCs and was associated with disease severity in AR patients.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yang Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianbo Shi
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, First People's Hospital of Foshan, Foshan, China
| | - Xuekun Huang
- Department of Otorhinolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yueqi Sun
- Department of Otolaryngology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qintai Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Scherzad A, Hagen R, Hackenberg S. Current Understanding of Nasal Epithelial Cell Mis-Differentiation. J Inflamm Res 2019; 12:309-317. [PMID: 31853193 PMCID: PMC6916682 DOI: 10.2147/jir.s180853] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022] Open
Abstract
The functional role of the respiratory epithelium is to generate a physical barrier. In addition, the epithelium supports the innate and acquired immune system through various cytokines and chemokines. However, epithelial cells are also involved in the pathogenesis of various respiratory diseases, some of which are mediated by increased permeability of the mucosal membrane or disturbed mucociliary transport. In addition, it has been shown that epithelial cells are involved in the development of inflammatory respiratory diseases. The following review article focuses on the aspects of epithelial mis-differentiation, in particular with respect to nasal mucosal barrier function, epithelial immunogenicity, nasal epithelial-mesenchymal transition and nasal microbiome.
Collapse
Affiliation(s)
- Agmal Scherzad
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, Würzburg97080, Germany
| | - Rudolf Hagen
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, Würzburg97080, Germany
| | - Stephan Hackenberg
- Department of Otorhinolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius Maximilian University of Wuerzburg, Würzburg97080, Germany
| |
Collapse
|
14
|
Bedke N, Swindle EJ, Molnar C, Holt PG, Strickland DH, Roberts GC, Morris R, Holgate ST, Davies DE, Blume C. A method for the generation of large numbers of dendritic cells from CD34+ hematopoietic stem cells from cord blood. J Immunol Methods 2019; 477:112703. [PMID: 31711888 PMCID: PMC6983936 DOI: 10.1016/j.jim.2019.112703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) play a central role in regulating innate and adaptive immune responses. It is well accepted that their regulatory functions change over the life course. In order to study DCs function during early life it is important to characterize the function of neonatal DCs. However, the availability of neonatal DCs is limited due to ethical reasons or relative small samples of cord blood making it difficult to perform large-scale experiments. Our aim was to establish a robust protocol for the generation of neonatal DCs from cord blood derived CD34+ hematopoietic stem cells. For the expansion of DC precursor cells we used a cytokine cocktail containing Flt-3 L, SCF, TPO, IL-3 and IL-6. The presence of IL-3 and IL-6 in the first 2 weeks of expansion culture was essential for the proliferation of DC precursor cells expressing CD14. After 4 weeks in culture, CD14+ precursor cells were selected and functional DCs were generated in the presence of GM-CSF and IL-4. Neonatal DCs were then stimulated with Poly(I:C) and LPS to mimic viral or bacterial infections, respectively. Poly(I:C) induced a higher expression of the maturation markers CD80, CD86 and CD40 compared to LPS. In line with literature data using cord blood DCs, our Poly(I:C) matured neonatal DCs cells showed a higher release of IL-12p70 compared to LPS matured neonatal DCs. Additionally, we demonstrated a higher release of IFN-γ, TNF-α, IL-1β and IL-6, but lower release of IL-10 in Poly(I:C) matured compared to LPS matured neonatal DCs derived from cord blood CD34+ hematopoietic stem cells. In summary, we established a robust protocol for the generation of large numbers of functional neonatal DCs. In line with previous studies, we showed that neonatal DCs generated form CD34+ cord blood progenitors have a higher inflammatory potential when exposed to viral than bacterial related stimuli. A robust protocol for the generation of high numbers of neonatal dendritic cells. IL-3 and IL-6 are crucial for the proliferation of cord blood CD34+ progenitors. Neonatal DCs have a higher inflammatory potential when exposed to viral stimuli. LPS induces higher release of IL-10 in neonatal DCs compared to Poly(I:C).
Collapse
Affiliation(s)
- Nicole Bedke
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Emily J Swindle
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Camelia Molnar
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Patrick G Holt
- Telethon Institute for Child Health Research, Centre for Child Health Research, University of Western Australia, Perth, Australia
| | - Deborah H Strickland
- Telethon Institute for Child Health Research, Centre for Child Health Research, University of Western Australia, Perth, Australia
| | - Graham C Roberts
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ruth Morris
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Stephen T Holgate
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Donna E Davies
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Cornelia Blume
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
| |
Collapse
|
15
|
Kao ST, Wang SD, Lin CC, Lin LJ. Jin Gui Shen Qi Wan, a traditional Chinese medicine, alleviated allergic airway hypersensitivity and inflammatory cell infiltration in a chronic asthma mouse model. JOURNAL OF ETHNOPHARMACOLOGY 2018; 227:181-190. [PMID: 30172058 DOI: 10.1016/j.jep.2018.08.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Asia, Jin Gui Shen Qi Wan (JGSQW) has been used for hundreds of years to treat asthma. AIM OF THE STUDY The traditional Chinese medicine JGSQW is composed of Rehmannia glutinosa, Dioscorea opposita, Cornus officinalis, Poria cocos, Paeonia suffruticosa, Alisma orientalis, Aconitum carmichaelii and Cinnamomum cassia. However, the immunological mechanism underlying the effect of JGSQW treatment on asthma remains unclear. This study examined whether JGSQW has the potential to reduce asthma symptoms in mice with chronic asthma induced by recurrent Dermatophagoides pteronyssinus (Der p) stimulation, as well as its immunoregulatory mechanisms. MATERIALS AND METHODS The airways of BALB/c mice were stimulated with Der p (i.t.) once per week (50 μL, 1 mg/mL) for 6 consecutive weeks, and the mice were fed JGSQW (1 g/kg) 30 min prior to the Der p stimulation. Three days after the last stimulation, the mice were sacrificed to evaluate the airway remodelling, infiltration of inflammatory cells, lung histological features, and total IgE in the blood. Additionally, after A549 cells were treated with JGSQW, loganin, or paeoniflorin for 30 min, 10 ng/mL IL-1β was added to stimulate the A549 cells to evaluate the effect of the medicine on the ICAM-1 gene expression after IL-1β stimulation. RESULTS JGSQW significantly reduced the Der p-induced infiltration of inflammatory cells into airways and decreased the total IgE and Der p-specific IgG1 in serum. Collagen assays and histopathological examinations showed that JGSQW reduced lung airway remodelling. Additionally, an electrophoretic mobility shift assay and immunohistochemical staining verified that JGSQW inhibited the NF-kB expression in airway epithelial cell nuclei. JGSQW, loganin, and paeoniflorin inhibited the ICAM-1 gene expression caused by the IL-1β stimulation of A549 cells, and loganin and paeoniflorin had the maximum inhibitory effect when mixed according to the combination of doses in JGSQW. CONCLUSION These results indicated that in the chronic asthma mouse model, JGSQW inhibits the infiltration of inflammatory cells into the airways and airway remodelling and exhibits specific immunoregulatory effects. JGSQW with certain doses of loganin and paeoniflorin inhibited ICAM-1 gene expression in epithelial cells.
Collapse
Affiliation(s)
- Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung 40402, Taiwan.
| | - Shulhn-Der Wang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| | - Chih-Che Lin
- Graduate Institute of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Li-Jen Lin
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| |
Collapse
|
16
|
Zheng R, Wang D, Wang K, Gao WX, Yang QT, Jiang LJ, Zhou M, Cao YJ, Shi J, Sun Y. Elevated expression of IL-17RB and ST2 on myeloid dendritic cells is associated with a Th2-skewed eosinophilic inflammation in nasal polyps. Clin Transl Allergy 2018; 8:50. [PMID: 30519393 PMCID: PMC6263180 DOI: 10.1186/s13601-018-0237-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022] Open
Abstract
Background Interleukin(IL)-25, IL-33, and thymic stromal lymphopoietin (TSLP) underlie the crosstalk between epithelial cells and dendritic cells (DCs) during the development of Th2 responses. This study aimed to measure the expressions of IL-17RB, ST2 and TSLPR, receptor of IL-25, IL-33, and TSLP respectively, on myeloid DCs in nasal polyps (NP) and evaluate their association with local Th2 inflammation and disease severity in patients with NP. Methods Samples were collected from 30 NP patients and 16 control subjects recruited prospectively. The mRNA expression of cytokines, including TSLP, IL-25 and IL-33, as well as interferon (IFN)-γ, IL-4, IL-5, IL-13 and IL-17A in NP and control tissues was examined by qualitative polymerase chain reaction (qPCR). The expression of IL-17RB, ST2 and TSLPR as well as other surface markers on myeloid DCs (mDCs) was examined by flow cytometry. Results Increased numbers of total and activated mDCs were found in NP patients. mDCs demonstrated significantly higher expression of IL-17RB, ST2 and TSLPR than those in control tissues. The activated mDCs exhibited up-regulations of OX40L and ICOSL, but down-regulation of PDL1 in NP. Moreover, the IL-17RB, ST2 and TSLPR levels on mDCs were positively correlated with IL-25, IL-33 and TSLP mRNA levels, respectively, in NP. Furthermore, IL-17RB and ST2 expressions on mDCs were correlated with the IL-5 mRNA level as well as eosinophil number in NP. Importantly, the IL-17RB expression on mDCs and the OX40L expression on activated mDCs in NP were positively correlated with CT score and total nasal symptom score. Conclusions Increased expressions of IL-17RB and ST2 on mDCs are associated with enhanced local Th2 inflammation in NP, suggesting that mDCs might play a role in IL-25- and IL-33-induced type 2 responses and eosinophilic inflammation in NP. Electronic supplementary material The online version of this article (10.1186/s13601-018-0237-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rui Zheng
- 1Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080 Guangdong China.,Guangzhou key Laboratory of Otorhinolarygology, Guangzhou, 510080 China
| | - Dan Wang
- 1Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080 Guangdong China.,Guangzhou key Laboratory of Otorhinolarygology, Guangzhou, 510080 China
| | - Kai Wang
- 3Department of Otorhinolaryngology-Head and Neck Surgery, First People's Hospital of Foshan, Foshan, 528000 China
| | - Wen-Xiang Gao
- 1Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080 Guangdong China.,Guangzhou key Laboratory of Otorhinolarygology, Guangzhou, 510080 China
| | - Qin-Tai Yang
- 4Department of Otorhinolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630 China
| | - Li-Jie Jiang
- 1Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080 Guangdong China.,Guangzhou key Laboratory of Otorhinolarygology, Guangzhou, 510080 China
| | - Min Zhou
- 1Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080 Guangdong China.,Guangzhou key Laboratory of Otorhinolarygology, Guangzhou, 510080 China
| | - Yu-Jie Cao
- 1Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080 Guangdong China.,Guangzhou key Laboratory of Otorhinolarygology, Guangzhou, 510080 China
| | - Jianbo Shi
- 1Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080 Guangdong China.,Guangzhou key Laboratory of Otorhinolarygology, Guangzhou, 510080 China
| | - Yueqi Sun
- 1Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080 Guangdong China.,Guangzhou key Laboratory of Otorhinolarygology, Guangzhou, 510080 China
| |
Collapse
|
17
|
Antigen-specific regulatory T-cell responses against aeroantigens and their role in allergy. Mucosal Immunol 2018; 11:1537-1550. [PMID: 29858582 DOI: 10.1038/s41385-018-0038-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 02/04/2023]
Abstract
The mucosal immune system of the respiratory tract is specialized to continuously monitor the external environment and to protect against invading pathogens, while maintaining tolerance to innocuous inhaled particles. Allergies result from a loss of tolerance against harmless antigens characterized by formation of allergen-specific Th2 cells and IgE. Tolerance is often described as a balance between harmful Th2 cells and various types of protective "regulatory" T cells. However, the identity of the protective T cells in healthy vs. allergic individuals or following successful allergen-specific therapy is controversially discussed. Recent technological progress enabling the identification of antigen-specific effector and regulatory T cells has significantly contributed to our understanding of tolerance. Here we discuss the experimental evidence for the various tolerance mechanisms described. We try to integrate the partially contradictory data into a new model proposing different mechanism of tolerance depending on the quality and quantity of the antigens as well as the way of antigen exposure. Understanding the basis of tolerance is essential for the rational design of novel and more efficient immunotherapies.
Collapse
|
18
|
Primary human nasal epithelial cells: a source of poly (I:C) LMW-induced IL-6 production. Sci Rep 2018; 8:11325. [PMID: 30054566 PMCID: PMC6063928 DOI: 10.1038/s41598-018-29765-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/13/2018] [Indexed: 01/06/2023] Open
Abstract
Infection plays a significant role in the relapse of chronic rhinosinusitis (CRS), however, the role of primary human nasal epithelial cells (HNECs) in this process is largely unknown. Here, we determined the effect of Toll-like receptor (TLR) agonists and inflammatory cytokines on mucosal barrier integrity and immune response of HNECs. TLR 1–9 agonists and inflammatory cytokines were applied to submerged and/or air-liquid interface (ALI) cultures of HNECs from CRS patients and controls for 24 hours. Interleukin-6 (IL-6) protein levels were determined by ELISA. Mucosal barrier integrity was measured via Transepithelial Electrical Resistance and passage of fluorescently-labelled dextrans. IL-1β and IFN- γ significantly increased IL-6 production in HNECs derived from CRS patients and controls, however, a dose-dependent effect was observed in CRS-derived HNECs only. Stimulation with Poly (I:C) LMW induced a 15 to 17 fold increase in IL-6 production by HNEC-ALI control cells (p < 0.05) and HNEC-ALI-CRS cells (p = 0.004) whilst a 2.5 fold increase was observed in CRS HNEC submerged cultures. Priming of cells with Poly (I:C) LMW reduced subsequent IL-6 secretion upon stimulation with TLR 2–4 agonists. Poly (I:C) LMW exerts a potent pro-inflammatory effect on HNECs and reduces a subsequent immune activation by TLR agonists.
Collapse
|
19
|
Wei T, Tang M. Biological effects of airborne fine particulate matter (PM 2.5) exposure on pulmonary immune system. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:195-201. [PMID: 29734103 DOI: 10.1016/j.etap.2018.04.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 05/21/2023]
Abstract
Airborne fine particulate matter (PM2.5) attracts more and more attention due to its environmental effects. The immune system appears to be a most sensitive target organ for the environmental pollutants. Inhaled PM2.5 can deposit in different compartments in the respiratory tract and interact with epithelial cells and resident immune cells. Exposed to PM2.5 can induce local or systematic inflammatory responses. This review focus on the effects of respiratory tract exposed to PM2.5. Firstly, we introduced the major emission sources, basic characteristics of PM2.5 and discussed its immunoadjuvant potential. Secondly, we elaborated the immune cells in the respiratory tract and the deposition of PM2.5 regarding the structural characteristics of the respiratory tract. Furthermore, we summarized the in vivo/vitro studies that revealed the immunotoxic effects of PM2.5 exposure to pulmonary cellular effectors and explored the contribution of PM2.5 exposure to the Th1/Th2 balance.
Collapse
Affiliation(s)
- Tingting Wei
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210009, PR China; Jiangsu key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210009, PR China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing 210009, PR China; Jiangsu key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210009, PR China.
| |
Collapse
|
20
|
Kim B, Kim TH. Fundamental role of dendritic cells in inducing Th2 responses. Korean J Intern Med 2018; 33:483-489. [PMID: 29502361 PMCID: PMC5943655 DOI: 10.3904/kjim.2016.227] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/05/2017] [Indexed: 12/24/2022] Open
Abstract
A mysterious puzzle in immunology is how the immune system decides what types of immune response to initiate against various stimuli. Although much is known about control of T helper 1 (Th1) and Th17 responses, the mechanisms that initiate Th2 responses remain obscure. Antigen-presenting cells, particularly dendritic cells (DCs), are mandatory for the induction of a Th cell response. Numerous studies have documented the organizing role of DCs in this process. The present review summarizes the fundamental roles of DCs in inducing Th2 responses.
Collapse
Affiliation(s)
- Byoungjae Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Korea University College of Medicine, Seoul, Korea
- Neuroscience Research Institute, Korea University College of Medicine, Seoul, Korea
| | - Tae Hoon Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Korea University College of Medicine, Seoul, Korea
- Allergy Immunology Center, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Pfeffer PE, Ho TR, Mann EH, Kelly FJ, Sehlstedt M, Pourazar J, Dove RE, Sandstrom T, Mudway IS, Hawrylowicz CM. Urban particulate matter stimulation of human dendritic cells enhances priming of naive CD8 T lymphocytes. Immunology 2018; 153:502-512. [PMID: 29044495 PMCID: PMC5838419 DOI: 10.1111/imm.12852] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 02/06/2023] Open
Abstract
Epidemiological studies have consistently shown associations between elevated concentrations of urban particulate matter (UPM) air pollution and exacerbations of asthma and chronic obstructive pulmonary disease, which are both associated with viral respiratory infections. The effects of UPM on dendritic cell (DC) -stimulated CD4 T lymphocytes have been investigated previously, but little work has focused on CD8 T-lymphocyte responses despite their importance in anti-viral immunity. To address this, we examined the effects of UPM on DC-stimulated naive CD8 T-cell responses. Expression of the maturation/activation markers CD83, CCR7, CD40 and MHC class I on human myeloid DCs (mDCs) was characterized by flow cytometry after stimulation with UPMin vitro in the presence/absence of granulocyte-macrophage colony-stimulating factor (GM-CSF). The capacity of these mDCs to stimulate naive CD8 T-lymphocyte responses in allogeneic co-culture was then assessed by measuring T-cell cytokine secretion using cytometric bead array, and proliferation and frequency of interferon-γ (IFN-γ)-producing T lymphocytes by flow cytometry. Treatment of mDCs with UPM increased expression of CD83 and CCR7, but not MHC class I. In allogeneic co-cultures, UPM treatment of mDCs enhanced CD8 T-cell proliferation and the frequency of IFN-γ+ cells. The secretion of tumour necrosis factor-α, interleukin-13, Granzyme A and Granzyme B were also increased. GM-CSF alone, and in concert with UPM, enhanced many of these T-cell functions. The PM-induced increase in Granzyme A was confirmed in a human experimental diesel exposure study. These data demonstrate that UPM treatment of mDCs enhances priming of naive CD8 T lymphocytes and increases production of pro-inflammatory cytokines. Such UPM-induced stimulation of CD8 cells may potentiate T-lymphocyte cytotoxic responses upon concurrent airway infection, increasing bystander damage to the airways.
Collapse
Affiliation(s)
- Paul E. Pfeffer
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
- Present address:
William Harvey Research InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Tzer R. Ho
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
| | - Elizabeth H. Mann
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
| | - Frank J. Kelly
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
- Environmental Research GroupMRC‐PHE Centre for Environment and HealthKing's College LondonLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental HazardsFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Maria Sehlstedt
- Division of MedicineDepartment of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
| | - Jamshid Pourazar
- Division of MedicineDepartment of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
| | - Rosamund E. Dove
- Environmental Research GroupMRC‐PHE Centre for Environment and HealthKing's College LondonLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental HazardsFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Thomas Sandstrom
- Division of MedicineDepartment of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
| | - Ian S. Mudway
- Environmental Research GroupMRC‐PHE Centre for Environment and HealthKing's College LondonLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental HazardsFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Catherine M. Hawrylowicz
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental HazardsFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| |
Collapse
|
22
|
Kao ST, Kuo YH, Wang SD, Hong HJ, Lin LJ. Analogous corticosteroids, 9A and EK100, derived from solid-state-cultured mycelium of Antrodia camphorata inhibit proinflammatory cytokine expression in macrophages. Cytokine 2018; 108:136-144. [PMID: 29605763 DOI: 10.1016/j.cyto.2018.03.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/23/2018] [Accepted: 03/23/2018] [Indexed: 12/13/2022]
Abstract
Antrodia camphorata mycelium is used in traditional Chinese medicine in Taiwan. The wild-type mycelium is rare and expensive, so a solid-state-cultured mycelium of A. camphorata (SCMAC) has been developed. Previous studies have found SCMAC to have anti-inflammatory effects. However, the immunomodulatory effects of SCMAC and of its active phytosterol compounds EK100 and 9A on asthma remain unknown. In this study, BALB/c mice were repeatedly exposed to Dermatogoides pteronyssinus (Der p) at 1-week intervals and were orally administered crude SCMAC extract before the Der p challenge. The mice were sacrificed 72 h after the last challenge to examine the airway remodeling, inflammation, and expression profiles of cytokines and various genes. Then, 30-µg/mL Der p-stimulated MH-S cells with 9A or EK100 were collected for real-time PCR analysis, and the effects of 9A and EK100 on macrophages were evaluated. The crude extract reduced Der p-induced airway hyperresponsiveness, total serum immunoglobulin E levels, and recruitment of inflammatory cells to the bronchoalveolar lavage fluid through cytokine downregulation and Th1/Th2/Th17 response modulation. Additionally, 9A and EK100 inhibited IL-1β and IL-6 expression in alveolar macrophages. These results indicate that the pharmacologically active compounds in a crude SCMAC extract exert synergistic effects on multiple targets to relieve asthma symptoms.
Collapse
Affiliation(s)
- Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung 40402, Taiwan.
| | - Yueh-Hsiung Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan; Department of Biotechnology, Asia University, Taichung 41354, Taiwan.
| | - Shulhn-Der Wang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Hong-Jye Hong
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Li-Jen Lin
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
23
|
Expression and regulation of immune-modulatory enzyme indoleamine 2,3-dioxygenase (IDO) by human airway epithelial cells and its effect on T cell activation. Oncotarget 2018; 7:57606-57617. [PMID: 27613847 PMCID: PMC5295376 DOI: 10.18632/oncotarget.11586] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/20/2016] [Indexed: 12/16/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) catalyzes the degradation of tryptophan, which plays a critical role in immune suppression through regulating the production of a series of metabolites that are generally referred to as kynurenines. It has become increasingly clear that epithelial cells (ECs) play an active role in maintaining lung homeostasis by modulating the function of immune cells via producing cytokines, chemokines, and anti-microbial mediators. In this study we assessed the regulation of IDO activity and expression in human primary ECs and EC lines under steady state conditions and in response to bacterial and allergenic stimuli. We also investigated the potential immune modulatory functions of IDO expression in human airway ECs. Our data clearly show that airway ECs produce IDO, which is down-regulated in response to allergens and TLR ligands while up-regulated in response to IFN-γ. Using gene silencing, we further demonstrate that IDO plays a key role in the EC-mediated suppression of antigen-specific and polyclonal proliferation of T cells. Interestingly, our data also show that ECs lose their inhibitory effect on T cell activation in response to different TLR agonists mimicking bacterial or viral infections. In conclusion, our work provides an understanding of how IDO is regulated in ECs as well as demonstrates that “resting” ECs can suppress T cell activation in an IDO dependent manner. These data provide new insight into how ECs, through the production of IDO, can influence downstream innate and adaptive responses as part of their function in maintaining immune homeostasis in the airways.
Collapse
|
24
|
Bosteels C, Lambrecht BN, Hammad H. Isolation of Conventional Murine Lung Dendritic Cell Subsets. ACTA ACUST UNITED AC 2018; 120:3.7B.1-3.7B.16. [PMID: 29512143 DOI: 10.1002/cpim.39] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The lungs are continuously exposed to environmental threats, requiring an adequate and stringent immune response of a heterogeneous set of effector cells. Dendritic cells (DCs) form a dense network in the respiratory mucosa and act as the central regulators of the different components of this response, both sensing the nature of the threats and precisely coordinating the effector mechanisms best suited for overcoming it. The DCs are classically subdivided in two main groups, plasmacytoid DCs (pDCs) and conventional DCs (cDCs), the latter being further subdivided into cDC1s and cDC2s based on ontogeny and their distinct non-redundant functions. This protocol provides different enrichment methods and represents an up-to-date, universal framework that uses a minimal set of highly specific lineage markers to discriminate and sort pure cDC subsets from the murine lung but also across tissues and species which is an added value in intra- and interspecies comparative research. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Cédric Bosteels
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
25
|
Respiratory sensitization: toxicological point of view on the available assays. Arch Toxicol 2017; 92:803-822. [DOI: 10.1007/s00204-017-2088-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/05/2017] [Indexed: 12/22/2022]
|
26
|
De Servi B, Ranzini F, Piqué N. Protective barrier properties of Rhinosectan ® spray (containing xyloglucan) on an organotypic 3D airway tissue model (MucilAir): results of an in vitro study. Allergy Asthma Clin Immunol 2017; 13:37. [PMID: 28811823 PMCID: PMC5553660 DOI: 10.1186/s13223-017-0209-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/02/2017] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND To evaluate barrier protective properties of Rhinosectan® spray, a medical device containing xyloglucan, on nasal epithelial cells (MucilAir). METHODS MucilAir-Nasal, a three-dimensional organotypic (with different cell types) airway tissue model, was treated with the medical device Rhinosectan® (30 µL) or with controls (Rhinocort-budesonide-or saline solution). The protective barrier effects of Rhinosectan® were evaluated by: TEER (trans-epithelial electrical resistance) (preservation of tight junctions), Lucifer Yellow assay (preservation of paracellular flux) and confocal immunofluorescence microscopy (localization of tight junction proteins). RESULTS Exposure of MucilAir with Rhinosectan® protected cell tight junctions (increases in TEER of 13.1% vs -6.3% with saline solution after 1 h of exposure), and preserved the paracellular flux, even after exposure with pro-inflammatory compounds (TNF-α and LPS from Pseudomonas aeruginosa 10). Results of confocal immunofluorescence microscopy demonstrated that, after treatment with the pro-inflammatory mixture, Rhinosectan® produced a slight relocation of zona occludens-1 in the cytosol compartment (while Rhinocort induced expression of zona-occludens-1), maintaining the localization of occludin (similarly to negative control). CONCLUSIONS Results of our study indicates that Rhinosectan® creates a protective physical barrier on nasal epithelial cells in vitro, allowing the avoidance of allergens and triggering factors, thus confirming the utility of this medical device in the management of nasal respiratory diseases, as rhinitis or rhinosinusitis.
Collapse
Affiliation(s)
| | | | - Núria Piqué
- Department of Microbiology and Parasitology, Diagonal Sud, Facultat de Farmàcia, Universitat de Barcelona (UB), Edifici A, Av Joan XXIII, 08028 Barcelona, Spain
| |
Collapse
|
27
|
Corthésy B, Bioley G. Therapeutic intranasal instillation of allergen-loaded microbubbles suppresses experimental allergic asthma in mice. Biomaterials 2017; 142:41-51. [PMID: 28727997 DOI: 10.1016/j.biomaterials.2017.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/20/2017] [Accepted: 07/09/2017] [Indexed: 12/18/2022]
Abstract
Despite proven efficiency, subcutaneous immunotherapy for aeroallergens is impaired by the duration of the protocol, the repeated injections and potential side-effects associated with the doses of allergen administered. Intranasal delivery of immunotherapeutic agents may overcome several of these drawbacks, provided that an efficient allergen delivery vehicle can be identified. This study evaluates whether intranasally delivered gas-filled microbubble (MB)-associated ovalbumin (OVA), used as a model allergen, can serve as a therapeutic treatment in a mouse model of established allergic asthma. Lung and systemic production of pro-tolerogenic markers, including Foxp3+ CD4 T cells, IL-10, and TGF-β, as well as the Th1-type cytokine IFN-γ, was observed after intranasal immunization with OVA-MB. Post-treatment, aerosol-sensitized mice exhibited the same pattern of markers. Moreover, decrease of eosinophils and neutrophils in BALs, lower frequencies of Th2 cytokine- and IL-17-producing CD4 T cells in lungs and reduced specific IgE in BALs and sera after allergen challenge were observed. Concomitantly, lung resistance and mucus production diminished in OVA-MB-treated animals. Thus, therapeutic intranasal administration of OVA-MBs in established experimental allergic asthma allows modulating pathology-associated immune and physiological parameters usually triggered after exposure to the allergen.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory, Division of Immunology and Allergy, University State Hospital (CHUV), Epalinges, Switzerland
| | - Gilles Bioley
- R&D Laboratory, Division of Immunology and Allergy, University State Hospital (CHUV), Epalinges, Switzerland.
| |
Collapse
|
28
|
Abstract
Apoptosis is an important component of normal tissue physiology, and the prompt removal of apoptotic cells is equally essential to avoid the undesirable consequences of their accumulation and disintegration. Professional phagocytes are highly specialized for engulfing apoptotic cells. The recent ability to track cells that have undergone apoptosis in situ has revealed a division of labor among the tissue resident phagocytes that sample them. Macrophages are uniquely programmed to process internalized apoptotic cell-derived fatty acids, cholesterol and nucleotides, as a reflection of their dominant role in clearing the bulk of apoptotic cells. Dendritic cells carry apoptotic cells to lymph nodes where they signal the emergence and expansion of highly suppressive regulatory CD4 T cells. A broad suppression of inflammation is executed through distinct phagocyte-specific mechanisms. A clever induction of negative regulatory nodes is notable in dendritic cells serving to simultaneously shut down multiple pathways of inflammation. Several of the genes and pathways modulated in phagocytes in response to apoptotic cells have been linked to chronic inflammatory and autoimmune diseases such as atherosclerosis, inflammatory bowel disease and systemic lupus erythematosus. Our collective understanding of old and new phagocyte functions after apoptotic cell phagocytosis demonstrates the enormity of ways to mediate immune suppression and enforce tissue homeostasis.
Collapse
Affiliation(s)
- J Magarian Blander
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
29
|
Wang J, Zhu W, Chen Y, Lin Z, Ma S. CD38 gene-modified dendritic cells inhibit murine asthma development by increasing IL-12 production and promoting Th1 cell differentiation. Mol Med Rep 2016; 14:4374-4382. [PMID: 27666020 DOI: 10.3892/mmr.2016.5756] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 08/31/2016] [Indexed: 11/06/2022] Open
Abstract
Predominant T helper (Th)2 and impaired Th1 cell polarization has a crucial role in the development of asthma. Cluster of differentiation (CD)38 is associated with the increased release of interleukin (IL)‑12 from dendritic cells (DCs) and DC‑induced Th1 cell polarization. However, whether CD38 expression affects DC function in asthma development remains unknown. In the current study, adenoviruses were constructed containing the murine CD38 gene. Overexpression of CD38 protein level in DCs induced from bone‑marrow derived DCs (BMDCs) by recombinant mouse granulocyte macrophage colony‑stimulating factor and IL‑4 was achieved through 24 h adenovirus infection. The results demonstrated that BMDCs with CD38 overexpression exhibited no phenotypic change; however, following stimulation with lipopolysaccharide (LPS), maturation and IL‑12 secretion were increased. In addition, CD38‑overexpressing BMDCs stimulated with LPS exhibited more effective Th1 cell differentiation. Mice that were administered CD38‑overexpressing BMDCs exhibited milder symptoms of asthma. Furthermore, decreased IL‑4, IL‑5 and IL‑13 levels were detected in bronchoalveolar lavage fluid (BALF), reduced immunoglobulin E levels were measured in the sera, and increased interferon‑γ was detected in BALF from the recipients of CD38‑overexpressing BMDCs. Increased phosphorylated‑p38 expression was also detected in LPS-stimulated CD38-overexpressing BMDCs, whereas pretreatment with a p38‑specific inhibitor was able to abolish the effects of LPS stimulation and CD38 overexpression on IL‑12 release and Th1 cell differentiation in BMDCs. These results suggested that CD38 may be involved in the DC function of alleviating asthma via restoration of the Th1/Th2 balance, thus providing a novel strategy for asthma therapy.
Collapse
Affiliation(s)
- Jiaoli Wang
- Department of Respiratory Medicine, Nanjing Medical University, Affiliated Hangzhou Hospital (Hangzhou First People's Hospital), Hangzhou, Zhejiang 310006, P.R. China
| | - Weiguo Zhu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Yinghu Chen
- Division of Infection Disease, Zhejiang Key Laboratory for Neonatal Diseases, Children Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhendong Lin
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Shenglin Ma
- Department of Oncology, Hangzhou First People's Hospital, Hangzhou Hospital Affiliated to Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
30
|
PathCellNet: Cell-type specific pathogen-response network explorer. J Immunol Methods 2016; 439:15-22. [PMID: 27659011 DOI: 10.1016/j.jim.2016.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/14/2016] [Accepted: 09/16/2016] [Indexed: 02/07/2023]
Abstract
Pathogen specific immune response is a complex interplay between several innate and adaptive immune cell-types. Innate immune cells play a critical role in pathogen recognition and initiating the antigen specific adaptive immune response. Despite specific functional roles of the innate immune cells, they share several anti-viral pathways. The question then becomes, what is the overlap in the transcriptional changes induced upon viral infections across different cell-types? Here we investigate the extent to which gene signatures are conserved across innate immune cell-types by performing a comparative analysis of transcriptomic data. Particularly, we integrate transcriptomic datasets measuring response of two innate immune cells (epithelial and dendritic cells) to influenza virus. The study reveals cell-type specific regulatory genes and a conserved network between the two cell-types. Additionally, novel functionally associated gene clusters are identified which are robustly defined across multiple independent studies. These gene clusters can be used in future investigation, and to facilitate their use we release PathCellNet (version 0), a cloud based tool to explore cell-type specific connectivity of user-defined genes. In the future, expansion of PathCellNet will allow exploration of cell-type specific responses across a variety of pathogens and cell-types.
Collapse
|
31
|
Kuo CH, Yang SN, Kuo HF, Lee MS, Huang MY, Huang SK, Lin YC, Hsieh CC, Hung CH. Cysteinyl leukotriene receptor antagonist epigenetically modulates cytokine expression and maturation of human myeloid dendritic cells. Pulm Pharmacol Ther 2016; 39:28-37. [PMID: 27312202 DOI: 10.1016/j.pupt.2016.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 05/29/2016] [Accepted: 06/12/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Cysteinyl leukotriene receptor antagonists are important controllers in treating asthma. Human myeloid DCs (mDCs) play critical roles in the pathogenesis of asthma. However, the effects of cysteinyl leukotriene receptor antagonist on human mDCs are unknown. METHODS To investigate the effects of cysteinyl leukotriene receptor antagonist on the function of human mDCs, circulating mDCs were isolated from six health subjects. Human mDCs were pretreated with montelukast and were stimulated with toll-like receptor (TLR) ligands lipopolysaccharide (LPS) or polyinosinic-polycytidylic acid (poly I:C). Tumor necrosis factor (TNF)-α and interleukin (IL)-10 were measured by ELISA. Intracellular signaling was investigated by pathway inhibitors, western blot and chromatin immunoprecipitation. Costimulatory molecules expression was investigated by flow cytometry. T cell polarization function of mDCs was investigated by measuring interferon (IFN)-γ, IL-13, IL-10 and IL-17A production by T cells using mDC/T cell coculture assay. RESULTS Montelukast suppressed TLR-mediated TNF-α expression via the NFκB-p65 and mitogen-activated protein kinase (MAPK)-JNK pathway, and enhanced TLR-mediated IL-10 expression via the MAPK-p38 pathway and epigenetic regulation by histone H3 acetylation. Montelukast suppressed LPS-induced CD80, CD86, CD40 and HLA-DR expression. Montelukast-treated mDCs suppressed IFN-γ and IL-13 production by T cells. CONCLUSION Cysteinyl leukotriene receptor antagonist alters the function of human mDCs by epigenetically modulating cytokine expression, suppressing costimulatory molecules expression and inhibiting the ability to initiate Th1/Th2 responses. The effects of cysteinyl leukotriene receptor antagonist on human mDCs can be an important mechanism in treating asthma.
Collapse
Affiliation(s)
- Chang-Hung Kuo
- Ta-Kuo Clinic, Kaohsiung, Taiwan; Department of Pediatrics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - San-Nan Yang
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Department of Pediatrics, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hsuan-Fu Kuo
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan; Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Min-Sheng Lee
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Pediatrics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Ming-Yii Huang
- Department of Radiation Oncology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shau-Ku Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Johns Hopkins University School of Medicine, Baltimore, MD, USA; National Health Research Institutes, Miaoli County, Taiwan
| | - Yi-Ching Lin
- Department of Laboratory Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chong-Chao Hsieh
- Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Hsing Hung
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan; Research Center of Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
32
|
Allen F, Tong AA, Huang AY. Unique Transcompartmental Bridge: Antigen-Presenting Cells Sampling across Endothelial and Mucosal Barriers. Front Immunol 2016; 7:231. [PMID: 27375624 PMCID: PMC4901051 DOI: 10.3389/fimmu.2016.00231] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/27/2016] [Indexed: 12/13/2022] Open
Abstract
Potentially harmful pathogens can gain access to tissues and organ systems through body sites that are in direct contact with the outside environment, such as the skin, the gut, and the airway mucosa. Antigen-presenting cells (APCs) represent a bridge between the innate and adaptive immunity, and their capacity for constant immune surveillance and rapid sampling of incoming pathogens and other potentially harmful antigens is central for mounting an effective and robust protective host response. The classical view is that APCs perform this task efficiently within the tissue to sense invading agents intra-compartmentally. However, recent data based on high resolution imaging support an additional transcompartmental surveillance behavior by APC by reaching across intact physical barriers. In this review, we summarize intravital microscopic evidences of APC to sample antigens transcompartmentally at the gut mucosa and other body sites.
Collapse
Affiliation(s)
- Frederick Allen
- Department of Pathology, Case Western Reserve University School of Medicine , Cleveland, OH , USA
| | - Alexander A Tong
- Department of Pathology, Case Western Reserve University School of Medicine , Cleveland, OH , USA
| | - Alex Y Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Angie Fowler AYA Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH, USA
| |
Collapse
|
33
|
Cao PP, Shi LL, Xu K, Yao Y, Liu Z. Dendritic cells in inflammatory sinonasal diseases. Clin Exp Allergy 2016; 46:894-906. [PMID: 27159777 DOI: 10.1111/cea.12755] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) are critical in linking the innate and adaptive immune responses, which have been implicated in the pathogenesis of many immune and inflammatory diseases as well as the development of tumours. The role of DCs in the pathophysiology of lung diseases has been widely studied. However, the phenotype, subset and function of DCs in upper airways under physiological or pathological conditions remain largely undefined. Allergic rhinitis (AR) and chronic rhinosinusitis (CRS) are two important upper airway diseases with a high worldwide prevalence. Aberrant innate and adaptive immune responses have been considered to play an important role in the pathogenesis of AR and CRS. To this end, understanding the function of DCs in shaping the immune responses in sinonasal mucosa is critical in exploring the pathogenic mechanisms underlying AR and CRS as well as in developing novel therapeutic strategies. This review summarizes the phenotype, subset, function and regulation of DCs in sinonasal mucosa, particularly in the setting of AR and CRS. Furthermore, this review discusses the perspectives for future research and potential clinical utility focusing on DC pathways in the context of AR and CRS.
Collapse
Affiliation(s)
- P-P Cao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - L-L Shi
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - K Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Yao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Z Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
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
|
35
|
Brand JD, Mathews JA, Kasahara DI, Wurmbrand AP, Shore SA. Regulation of IL-17A expression in mice following subacute ozone exposure. J Immunotoxicol 2016; 13:428-38. [PMID: 27043160 DOI: 10.3109/1547691x.2015.1120829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Exposure to subacute ozone (O3) causes pulmonary neutrophil recruitment. In mice, this recruitment requires IL-17A. Ozone also causes expression of IL-23 and IL-1, which can induce IL-17A. The purpose of this study was to examine the hypothesis that IL-23 and IL-1 contribute to IL-17A expression and subsequent neutrophil recruitment after subacute O3 exposure. Wild-type, IL-23(-/-), and Flt3l(-/-) mice were exposed to air or 0.3 ppm O3 for 72 h. Flt3l(-/-) mice lack conventional dendritic cells (cDC) that can express IL-23 and IL-1. Other wild-type mice were pre-treated with saline or the IL-1R1 antagonist anakinra prior to O3 exposure. After exposure, bronchoalveolar lavage (BAL) was performed and lung tissue harvested. The results indicated that pulmonary Il17a mRNA abundance and IL-17A(+) F4/80(+) cells were significantly reduced in O3-exposed IL-23(-/-) vs in wild-type mice. In contrast, anakinra had no effect on Il23a or Il17a pulmonary mRNA abundance or on BAL concentrations of the neutrophil survival factor G-CSF, but anakinra did reduce BAL neutrophil numbers, likely because anakinra also reduced BAL IL-6. Compared to air, O3 caused a significant increase in DC numbers in wild-type, but not in Flt3(-/-) mice. However, there was no significant difference in Il23a or Il17a mRNA abundance or in BAL neutrophil count in O3-exposed Flt3(-/-) vs in wild-type mice. From these results, it was concluded that IL-23 but not IL-1 contributes to the IL-17A expression induced by subacute O3 exposure. Induction of IL-23 by O3 does not appear to require cDC.
Collapse
Affiliation(s)
- Jeffrey D Brand
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Joel A Mathews
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - David I Kasahara
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Alison P Wurmbrand
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Stephanie A Shore
- a Molecular and Integrative Physiological Sciences Program, Department of Environmental Health , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| |
Collapse
|
36
|
Direct Delivery of Antigens to Dendritic Cells via Antibodies Specific for Endocytic Receptors as a Promising Strategy for Future Therapies. Vaccines (Basel) 2016; 4:vaccines4020008. [PMID: 27043640 PMCID: PMC4931625 DOI: 10.3390/vaccines4020008] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/15/2016] [Accepted: 03/18/2016] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are the most potent professional antigen presenting cells and are therefore indispensable for the control of immunity. The technique of antibody mediated antigen targeting to DC subsets has been the basis of intense research for more than a decade. Many murine studies have utilized this approach of antigen delivery to various kinds of endocytic receptors of DCs both in vitro and in vivo. Today, it is widely accepted that different DC subsets are important for the induction of select immune responses. Nevertheless, many questions still remain to be answered, such as the actual influence of the targeted receptor on the initiation of the immune response to the delivered antigen. Further efforts to better understand the induction of antigen-specific immune responses will support the transfer of this knowledge into novel treatment strategies for human diseases. In this review, we will discuss the state-of-the-art aspects of the basic principles of antibody mediated antigen targeting approaches. A table will also provide a broad overview of the latest studies using antigen targeting including addressed DC subset, targeted receptors, outcome, and applied coupling techniques.
Collapse
|
37
|
van de Laar L, Guilliams M, Tavernier S. Isolation of Conventional Dendritic Cells from Mouse Lungs. Methods Mol Biol 2016; 1423:139-152. [PMID: 27142014 DOI: 10.1007/978-1-4939-3606-9_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The lungs are in direct contact with the environment. Separated only by a thin layer of mucosa, the lung immune system is being exposed to dangers like pathogens, allergens, or pollutants. The lung dendritic cells form an elaborate network at the basolateral side of the epithelium and continuously sample antigens from the airway lumen. The conventional dendritic cells (cDCs) in the lung can be subdivided into two distinct subsets based on their ontogeny and are described to have distinct immunological functions. High-quality ex vivo isolation of these cells is required for experiments such as functional assays, transfer experiments, or transcriptomics and is crucial to further our knowledge concerning these subpopulations. In this chapter we describe a protocol for the isolation of both CD103(+) and CD11b(+) cDCs. In our protocol we compare different methods of cell isolation. We propose that the optimal isolation technique is based on the number of cells needed and the type of experiment that will be performed. If low cell numbers are required, simple flow cytometry-assisted cell sorting (FACS) is sufficient. In the case of high cell numbers that will be lysed or fixed upon sorting, positive selection of CD11c(+) cells followed by FACS can be utilized. Purification of cDCs through gradient selection and subsequent sorting is found to be optimal for experiments that require large amount of cells for functional assays.
Collapse
Affiliation(s)
- Lianne van de Laar
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent University, Technologiepark 927, 9052, Ghent, Belgium.
- Department of Internal Medicine, Ghent University, Ghent, Belgium.
| | - Martin Guilliams
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Simon Tavernier
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
- Department of Internal Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
38
|
Bachert C, Holtappels G. Pathophysiology of chronic rhinosinusitis, pharmaceutical therapy options. GMS CURRENT TOPICS IN OTORHINOLARYNGOLOGY, HEAD AND NECK SURGERY 2015; 14:Doc09. [PMID: 26770283 PMCID: PMC4702058 DOI: 10.3205/cto000124] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Research in immunology has brought great progress in knowledge of inflammatory processes in the last 2 decades, which also has an impact on the upper airways. Our understanding of the pathophysiology of chronic rhinosinusitis developed from a rather mechanistic point of view with a focus on narrow clefts and mucociliary clearance to the appreciation of a complex network of immunological pathways forming the basis of disease. We today differentiate various forms of inflammation, we start to understand complex immune-regulatory networks and the reasons for their failure, and have already developed innovative approaches for therapy for the most severely ill subjects. Due to this new knowledge in inflammation and remodeling processes within mucosal tissue, specifically on the key driving factors, new diagnostic tools and therapeutic approaches for chronic rhinosinusitis have developed; the differentiation of endotypes based on pathophysiological principles will be crucial for the use of innovative therapies, mostly humanized monoclonal antibodies. Several hundred of those antibodies are currently developed for various indications and will impact our specialty as well as pneumology to a great extent.
Collapse
Affiliation(s)
- Claus Bachert
- Department of Otolaryngology and Upper Airways Research Laboratory, University of Ghent, Belgium; Division of ENT Diseases, CLINTEC, Karolinska Institute, University of Stockholm, Sweden
| | - Gabriële Holtappels
- Department of Otolaryngology and Upper Airways Research Laboratory, University of Ghent, Belgium
| |
Collapse
|
39
|
Sundaram K, Mitra S, Gavrilin MA, Wewers MD. House Dust Mite Allergens and the Induction of Monocyte Interleukin 1β Production That Triggers an IκBζ-Dependent Granulocyte Macrophage Colony-Stimulating Factor Release from Human Lung Epithelial Cells. Am J Respir Cell Mol Biol 2015; 53:400-11. [PMID: 25629767 DOI: 10.1165/rcmb.2014-0370oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Asthma is a chronic lung disease characterized by inflammation centered upon bronchial epithelium. House dust mite is one of the most common respiratory allergens that trigger exacerbations of asthma. IκBζ (gene NFKBIZ) is a recently recognized member of the NF-κB family that can be induced in mononuclear phagocytes and lung epithelial cells and has been shown to play a prominent role in epithelial cell function. We therefore analyzed the role of IκBζ in regulating lung epithelial cell cytokine responses to house dust mite mix (HDM). We found that human bronchial epithelial cells express IκBζ and release IL-6 and granulocyte macrophage colony-stimulating factor (GMCSF) when cocultured with human monocytes and HDM. This response is blocked in the presence of IL-1 receptor antagonist (IL-1Ra), indicating that it is IL-1 mediated. Neither HDM-stimulated macrophages nor dendritic cells release IL-1β and subsequently induce cytokine release from the bronchial epithelial cells. Rhodobacter sphaeroides LPS (RS-LPS), a TLR4 antagonist, blocks the ability of HDM to induce IκBζ and release GMCSF from epithelial cells cocultured with monocytes. Additionally, human bronchial epithelial cells show no induction of IκBζ or cytokine responses to direct HDM stimulation. Finally, NFKBIZ small interfering RNA-mediated knockdown in the bronchial epithelial cells suppresses the release of IL-1-induced IL-6 and GMCSF. Our findings indicate a possible role for monocyte recruitment and lung epithelial cell IκBζ in mediating asthma associated inflammation. Thus, IκBζ, IL-1Ra, and RS-LPS deserve future study as potential modulators of house dust mite-induced asthma.
Collapse
Affiliation(s)
- Kruthika Sundaram
- Pulmonary, Allergy, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Department of Internal Medicine, Ohio State University Medical Center, Columbus, Ohio
| | - Srabani Mitra
- Pulmonary, Allergy, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Department of Internal Medicine, Ohio State University Medical Center, Columbus, Ohio
| | - Mikhail A Gavrilin
- Pulmonary, Allergy, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Department of Internal Medicine, Ohio State University Medical Center, Columbus, Ohio
| | - Mark D Wewers
- Pulmonary, Allergy, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Department of Internal Medicine, Ohio State University Medical Center, Columbus, Ohio
| |
Collapse
|
40
|
Jairaman A, Yamashita M, Schleimer RP, Prakriya M. Store-Operated Ca2+ Release-Activated Ca2+ Channels Regulate PAR2-Activated Ca2+ Signaling and Cytokine Production in Airway Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:2122-33. [PMID: 26238490 DOI: 10.4049/jimmunol.1500396] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/30/2015] [Indexed: 01/11/2023]
Abstract
The G-protein-coupled protease-activated receptor 2 (PAR2) plays an important role in the pathogenesis of various inflammatory and auto-immune disorders. In airway epithelial cells (AECs), stimulation of PAR2 by allergens and proteases triggers the release of a host of inflammatory mediators to regulate bronchomotor tone and immune cell recruitment. Activation of PAR2 turns on several cell signaling pathways of which the mobilization of cytosolic Ca(2+) is likely a critical but poorly understood event. In this study, we show that Ca(2+) release-activated Ca(2+) (CRAC) channels encoded by stromal interaction molecule 1 and Orai1 are a major route of Ca(2+) entry in primary human AECs and drive the Ca(2+) elevations seen in response to PAR2 activation. Activation of CRAC channels induces the production of several key inflammatory mediators from AECs including thymic stromal lymphopoietin, IL-6, and PGE2, in part through stimulation of gene expression via nuclear factor of activated T cells (NFAT). Furthermore, PAR2 stimulation induces the production of many key inflammatory mediators including PGE2, IL-6, IL-8, and GM-CSF in a CRAC channel-dependent manner. These findings indicate that CRAC channels are the primary mechanism for Ca(2+) influx in AECs and a vital checkpoint for the induction of PAR2-induced proinflammatory cytokines.
Collapse
Affiliation(s)
- Amit Jairaman
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - Megumi Yamashita
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - Robert P Schleimer
- Division of Allergy/Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| |
Collapse
|
41
|
Cell-contact dependent inhibition of monocytes by airway epithelial cells and reversion by infection with Respiratory Syncytial Virus. Immunobiology 2015; 220:1240-5. [PMID: 26153873 DOI: 10.1016/j.imbio.2015.06.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/17/2015] [Accepted: 06/22/2015] [Indexed: 01/01/2023]
Abstract
Airway epithelial cells (AEC) are the first line of defense against airborne infectious microbes and play an important role in regulating the local immune response. However, the interplay of epithelial cells and professional immune cells during both homeostasis and infection has only been partially studied. The present study was performed to determine how bronchial epithelial cells affect the activation of monocytes. Under healthy conditions, AECs were shown to inhibit reactivity of monocytes. We hypothesized that upon infection, monocytes might be released from inhibition by AECs. We report that direct contact of monocytes with unstimulated BEAS2B epithelial cells results in inhibition of TNF secretion by activated monocytes. In addition to the known soluble modulators, we show that cell contacts between epithelial cells and monocytes or macrophages also contribute to homeostatic inhibitory actions. We find AECs to express the inhibitory molecule PD-L1 and blockade of PD-L1 results in increased secretion of pro-inflammatory cytokines from monocytes. Contrary to the inhibitory activities during homeostasis, epithelial cells infected with Respiratory Syncitial Virus (RSV) induce a significant release of inhibition. However, release of inhibition was not due to modulation of PD-L1 expression in AECs. We conclude that airway epithelial cells control the reactivity of monocytes through direct and indirect interactions; however tonic inhibition can be reverted upon stimulation of AECs with RSV and thereof derived molecular patterns. The study confirms the important role of airway epithelial cells for local immune reactions.
Collapse
|
42
|
Akgün J, Schabussova I, Schwarzer M, Kozakova H, Kundi M, Wiedermann U. The Role of Alveolar Epithelial Type II-Like Cells in Uptake of Structurally Different Antigens and in Polarisation of Local Immune Responses. PLoS One 2015; 10:e0124777. [PMID: 25894334 PMCID: PMC4404363 DOI: 10.1371/journal.pone.0124777] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/05/2015] [Indexed: 01/17/2023] Open
Abstract
Background Our previous studies on intranasal tolerance induction demonstrated reduction of allergic responses with different allergen constructs. The underlying mechanisms varied depending on their conformation or size. Objective The aim of the present study was to compare the uptake of two structurally different allergen molecules within the respiratory tract following intranasal application. Methods The three-dimensional Bet v 1 (Bv1-Protein) and the T cell epitope peptide of Bet v 1 (Bv1-Peptide) were labelled with 5,6-Carboxyfluorescein (FAM) and their uptake was investigated in lung cells and cells of the nasal associated lymphoid tissue from naive and sensitised BALB/c mice. Phenotypic characterisation of FAM+ lung cells after antigen incubation in vitro and after intranasal application was performed by flow cytometry. Impact of Bv1-Protein and Bv1-Peptide on cytokine profiles and gene expression in vivo or in an alveolar epithelial type II (ATII) cell line were assessed in mono- and co-cultures with monocytes using ELISA and quantitative real-time PCR. Results Both antigens were taken up preferably by ATII-like cells (ATII-LCs) in naive mice, and by macrophages in sensitised mice. After intranasal application, Bv1-Peptide was taken up faster and more efficiently than Bv1-Protein. In vivo and in vitro experiments revealed that Bv1-Protein induced the transcription of thymic stromal lymphopoietin mRNA while Bv1-Peptide induced the transcription of IL-10 and MCP1 mRNA in ATII-LCs. Conclusion and Clinical Relevance Both tested antigens were taken up by ATII-LCs under steady state conditions and induced different polarisation of the immune responses. These data may have an important impact for the generation of novel and more effective prophylactic or therapeutic tools targeting the respiratory mucosa.
Collapse
Affiliation(s)
- Johnnie Akgün
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology, v.v.i., Academy of Sciences of the Czech Republic, Novy Hradek, Czech Republic
| | - Hana Kozakova
- Laboratory of Gnotobiology, Institute of Microbiology, v.v.i., Academy of Sciences of the Czech Republic, Novy Hradek, Czech Republic
| | - Michael Kundi
- Institute of Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Göteborg, Göteborg, Sweden
| |
Collapse
|
43
|
Alpaerts K, Buckinx R, Adriaensen D, Van Nassauw L, Timmermans JP. Identification and Putative Roles of Distinct Subtypes of Intestinal Dendritic Cells in Neuroimmune Communication: What can be Learned from Other Organ Systems? Anat Rec (Hoboken) 2015; 298:903-16. [DOI: 10.1002/ar.23106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 09/13/2014] [Accepted: 11/08/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Katrien Alpaerts
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Roeland Buckinx
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Dirk Adriaensen
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Luc Van Nassauw
- Laboratory of Human Anatomy and Embryology; Faculty of Medicine and Health Sciences; University of Antwerp; Antwerp Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| |
Collapse
|
44
|
Nasal Immunity, Rhinitis, and Rhinosinusitis. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00100-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
45
|
Palomares O, Crameri R, Rhyner C. The contribution of biotechnology toward progress in diagnosis, management, and treatment of allergic diseases. Allergy 2014; 69:1588-601. [PMID: 25307026 DOI: 10.1111/all.12533] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2014] [Indexed: 12/18/2022]
Abstract
'Biotechnology' has been intuitively used by humans since thousands of years for the production of foods, beverages, and drugs based on the experience without any scientific background. However, the golden era of this discipline emerged only during the second half of the last century. Incredible progresses have been achieved on all fields starting from the industrialization of the production of foods to the discovery of antibiotics, the decipherment of the genetic code, and rational approaches to understand and define the status we now call 'healthy'. The extremely complex interactions between genetic background, life style, and environmental factors influencing our continuously increasing life span have become more and more evident and steadily generate new questions which are only partly answered. Here, we try to summarize the contribution of biotechnology to our understanding, control, and cure of IgE-mediated allergic diseases. We are aware that a review of such a vast topic can never cover all aspects of the progress achieved in the different fields.
Collapse
Affiliation(s)
- O. Palomares
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | - R. Crameri
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Davos Switzerland
| | - C. Rhyner
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Davos Switzerland
| |
Collapse
|
46
|
Rossey I, Sedeyn K, De Baets S, Schepens B, Saelens X. CD8+ T cell immunity against human respiratory syncytial virus. Vaccine 2014; 32:6130-7. [PMID: 25223272 DOI: 10.1016/j.vaccine.2014.08.063] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/08/2014] [Accepted: 08/27/2014] [Indexed: 12/11/2022]
Abstract
Human respiratory syncytial virus (HRSV) was first discovered in the 1950s, but despite decades of research, a licensed vaccine against it is not available. Epidemiological studies indicate that antibodies directed against the fusion protein (F) partially correlate with protection. In addition, an F-specific monoclonal antibody is licensed as a prophylactic treatment in children who are at high risk of developing complications following HRSV infection. Therefore, most HRSV-oriented vaccination strategies focus on inducing a humoral immune response against F. In the quest for the development of a safe HRSV vaccine, the induction of a T cell immune response has received a lot less attention. T cell immunity directed against HRSV has not been associated unequivocally with protection against HRSV and CD4(+) T helper cell responses may even worsen disease due to HRSV. However, many studies support a protective role for CD8(+) T cells in clearance of HRSV from the lungs. In this review we highlight the clinical and experimental evidence in favor of a CD8(+) T lymphocyte-based vaccination strategy to protect against HRSV. First, we describe how T cell responses and T cell memory are induced in the lungs upon respiratory viral infection. HRSV has evolved mechanisms that hamper CD8(+) T cell priming and effector functions. We appraise the information on HRSV-specific CD8(+) T cell immunity gained from laboratory mouse studies, taking into account the advantages and limitations of this animal model and, where possible, the accordance with clinical evidence. Finally, we focus on recent efforts to develop T cell based vaccines against HRSV.
Collapse
Affiliation(s)
- Iebe Rossey
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium
| | - Koen Sedeyn
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium
| | - Sarah De Baets
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium
| | - Bert Schepens
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium
| | - Xavier Saelens
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium.
| |
Collapse
|
47
|
Qin Q, Wang Z, Pan P, Cao Z, Xia Q, Tan H, Hu C. Lung dendritic cells undergo maturation and polarization towards a T helper type 2-stimulating phenotype in a mouse model of asthma: Role of nerve growth factor. Exp Ther Med 2014; 8:1402-1408. [PMID: 25289030 PMCID: PMC4186495 DOI: 10.3892/etm.2014.1967] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 06/20/2014] [Indexed: 12/12/2022] Open
Abstract
Nerve growth factor (NGF) and dendritic cells (DCs) have been hypothesized to modulate T cell responses in a mouse model of asthma. However, whether NGF plays a role in regulating the maturation and polarization of lung DCs remains unclear. In the present study, the effect of NGF inhibition on the maturation and phenotype of lung DCs was investigated in a mouse model of asthma. BALB/c mice were sensitized and challenged with ovalbumin (OVA), and subsequently received anti-NGF treatment. At 24 h following the last challenge, airway responsiveness and inflammation were examined. The concentrations of NGF, interferon (IFN)-γ and interleukin (IL)-4 were analyzed. In addition, maturation and CD103 expression in the lung DCs were investigated. Anti-NGF treatment was found to significantly reduce airway hyperreactivity and inflammation in asthmatic mice. In addition, a subdued T helper 2 (Th2) response was observed, characterized by the downregulation of IL-4 and the upregulation of IFN-γ. Furthermore, the expression of the DC surface molecules, CD80, CD86 and major histocompatibility complex class II, as well as the proportion of lung CD103+ DCs, decreased in the OVA-sensitized and challenged mice. The proportion of lung CD103+ DCs also exhibited a positive correlation with the levels of plasma NGF in the mice. These results may provide an explanation for the role of NGF in amplifying the Th2 response in allergic diseases. Therefore, NGF may promote the maturation and polarization towards a Th2-stimulating phenotype of activated DCs, contributing to an amplification of the Th2 response in asthma.
Collapse
Affiliation(s)
- Qingwu Qin
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhan Wang
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Pinhua Pan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China ; Bronchial Asthma Research Center of Hunan Province, Changsha, Hunan 410008, P.R. China
| | - Zu Cao
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qing Xia
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hongyi Tan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China ; Bronchial Asthma Research Center of Hunan Province, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Severe asthma is a heterogeneous syndrome, encompassing several distinct clinical phenotypes. Different molecular and cellular pathways or endotypes determine the type of underlying airway inflammation in patients with severe asthma, which can be categorized as eosinophilic asthma (allergic and nonallergic) or noneosinophilic asthma (neutrophilic and paucigranulocytic). In this review, we discuss the potential role of macrolides in the treatment of severe asthma in adults. RECENT FINDINGS Maintenance treatment with low-dose macrolides such as erythromycin and azithromycin provides clinical benefit in several chronic neutrophilic airway diseases, including cystic fibrosis (CF), non-CF bronchiectasis and exacerbation-prone chronic obstructive pulmonary disease. Although several short-term studies of macrolides in mild-to-moderate asthma have failed to improve lung function, the AzIthromycin in Severe Asthma trial has demonstrated a significant reduction in the rate of exacerbations in patients with exacerbation-prone noneosinophilic severe asthma. As chronic macrolide use is associated with the risks of population antimicrobial resistance, this add-on treatment should be restricted to severe asthma patients at greatest unmet need despite optimal asthma management. SUMMARY Further clinical, translational and basic research is needed to better phenotype patients with severe asthma, to determine the risk-benefit ratio of macrolide maintenance treatment in neutrophilic severe asthma and to elucidate the principal mechanisms of action of macrolides.
Collapse
|
49
|
Weitnauer M, Schmidt L, Ng Kuet Leong N, Muenchau S, Lasitschka F, Eckstein V, Hübner S, Tuckermann J, Dalpke AH. Bronchial epithelial cells induce alternatively activated dendritic cells dependent on glucocorticoid receptor signaling. THE JOURNAL OF IMMUNOLOGY 2014; 193:1475-84. [PMID: 24965772 DOI: 10.4049/jimmunol.1400446] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Airway epithelial cells mount a tolerogenic microenvironment that reduces the proinflammatory potential of respiratory dendritic cells (DCs). We recently demonstrated that tracheal epithelial cells continuously secrete soluble mediators that affect the reactivity of local innate immune cells. Using transcriptional profiling, we now observed that conditioning of DCs by tracheal epithelial cells regulated 98 genes under homeostatic conditions. Among the most upregulated genes were Ms4a8a and Ym1, marker genes of alternatively activated myeloid cells. Ex vivo analysis of respiratory DCs from nonchallenged mice confirmed a phenotype of alternative activation. Bioinformatic analysis showed an overrepresentation of hormone-nuclear receptors within the regulated genes, among which was the glucocorticoid receptor. In line with a role for glucocorticoids, pharmacological blockade as well as genetic manipulation of the glucocorticoid receptor within DCs inhibited Ms4a8a and Ym1 expression as well as MHC class II and CD86 regulation upon epithelial cell conditioning. Within epithelial cell-conditioned medium, low amounts of glucocorticoids were present. Further analysis showed that airway epithelial cells did not produce glucocorticoids de novo, yet were able to reactivate inactive dehydrocorticosterone enzymatically. The results show that airway epithelial cells regulate local immune responses, and this modulation involves local production of glucocorticoids and induction of an alternative activation phenotype in DCs.
Collapse
Affiliation(s)
- Michael Weitnauer
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Lotte Schmidt
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, 69120 Heidelberg, Germany; Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Nathalie Ng Kuet Leong
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Stephanie Muenchau
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Volker Eckstein
- Department of Medicine V, University Hospital Heidelberg, 69120 Heidelberg, Germany; and
| | - Sabine Hübner
- Institute of General Zoology and Endocrinology, University of Ulm, 89081 Ulm, Germany
| | - Jan Tuckermann
- Institute of General Zoology and Endocrinology, University of Ulm, 89081 Ulm, Germany
| | - Alexander H Dalpke
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| |
Collapse
|
50
|
Guo HW, Yun CX, Hou GH, Du J, Huang X, Lu Y, Keller ET, Zhang J, Deng JG. Mangiferin attenuates TH1/TH2 cytokine imbalance in an ovalbumin-induced asthmatic mouse model. PLoS One 2014; 9:e100394. [PMID: 24955743 PMCID: PMC4067356 DOI: 10.1371/journal.pone.0100394] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 05/26/2014] [Indexed: 12/16/2022] Open
Abstract
Mangiferin is a major bioactive ingredient in Mangifera indica Linn. (Anacardiaceae) leaves. Aqueous extract of such leaves have been used as an indigenous remedy for respiratory diseases like asthma and coughing in traditional Chinese medicine. However, underlying molecular mechanisms of mangiferin on anti-asthma remain unclear. In our present study, we investigated the anti-asthmatic effect of mangiferin on Th1/Th2 cytokine profiles and explored its underlying immunoregulatory mechanism in mouse model of allergic asthma. Mangiferin significantly reduced the total inflammatory cell counts and eosinophil infiltration, decreased the production of ovalbumin-specific IgE in serum and PGD2 in BALF. The antibody array analysis showed that mangiferin down-regulated the levels of one group of cytokines/chemokines including Th2-related IL-4, IL-5, IL-13, and others IL-3, IL-9, IL-17, RANTES, TNF-α, but simultaneously up-regulated Th1-related IFN-γ, IL-2 and IL-10 and IL-12 expression in serum. Thus it attenuates the imbalance of Th1/Th2 cells ratio by diminishing the abnormal mRNA levels of Th1 cytokines (IFN-γ and IL-12) and Th2 cytokines (IL-4, IL-5 and IL-13). Finally, mangiferin substantially inhibited the activation and expression of STAT-6 and GATA-3 in excised lung tissues. Our results suggest that mangiferin can exert anti-asthmatic effect. The underlying mechanism may attribute to the modulation of Th1/Th2 cytokine imbalance via inhibiting the STAT6 signaling pathway.
Collapse
Affiliation(s)
- Hong-Wei Guo
- Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Longevity and Aging-related Disease, Chinese Ministry of Education, Nanning, Guangxi, China
- Guangxi Key Laboratory of Pharmacodynamic Studies of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Chen-Xia Yun
- School of Basic Medical Sciences, Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Guang-Han Hou
- Guangxi Key Laboratory of Pharmacodynamic Studies of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Jun Du
- Guangxi Key Laboratory of Pharmacodynamic Studies of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Xin Huang
- Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Longevity and Aging-related Disease, Chinese Ministry of Education, Nanning, Guangxi, China
| | - Yi Lu
- Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Longevity and Aging-related Disease, Chinese Ministry of Education, Nanning, Guangxi, China
| | - Evan T. Keller
- Department of Urology and Pathology, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jian Zhang
- Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Longevity and Aging-related Disease, Chinese Ministry of Education, Nanning, Guangxi, China
- * E-mail: (JZ); (JGD)
| | - Jia-Gang Deng
- Guangxi Key Laboratory of Pharmacodynamic Studies of Traditional Chinese Medicine, Nanning, Guangxi, China
- * E-mail: (JZ); (JGD)
| |
Collapse
|